1
|
Liu M, Chen R, Zheng Z, Xu S, Hou C, Ding Y, Zhang M, Bao M, He B, Li S. Mechanisms of inflammatory microenvironment formation in cardiometabolic diseases: molecular and cellular perspectives. Front Cardiovasc Med 2025; 11:1529903. [PMID: 39877020 PMCID: PMC11772298 DOI: 10.3389/fcvm.2024.1529903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 12/26/2024] [Indexed: 01/31/2025] Open
Abstract
Cardiometabolic diseases (CMD) are leading causes of death and disability worldwide, with complex pathophysiological mechanisms in which inflammation plays a crucial role. This review aims to elucidate the molecular and cellular mechanisms within the inflammatory microenvironment of atherosclerosis, hypertension and diabetic cardiomyopathy. In atherosclerosis, oxidized low-density lipoprotein (ox-LDL) and pro-inflammatory cytokines such as Interleukin-6 (IL-6) and Tumor Necrosis Factor-alpha (TNF-α) activate immune cells contributing to foam cell formation and arterial wall thickening. Hypertension involves the activation of the renin-angiotensin system (RAS) alongside oxidative stress-induced endothelial dysfunction and local inflammation mediated by T cells. In diabetic cardiomyopathy, a high-glucose environment leads to the accumulation of advanced glycation end products (AGEs), activating the Receptor for Advanced Glycation Endproducts (RAGE) and triggering inflammatory responses that further damage cardiac and microvascular function. In summary, the inflammatory mechanisms in different types of metabolic cardiovascular diseases are complex and diverse; understanding these mechanisms deeply will aid in developing more effective individualized treatment strategies.
Collapse
Affiliation(s)
- Menghua Liu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Rumeng Chen
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Zhiwei Zheng
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Shuling Xu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Chunyan Hou
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Yining Ding
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Mengling Zhang
- School of Stomatology, Changsha Medical University, Changsha, China
| | - Meihua Bao
- Hunan key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, School of Pharmaceutical Science, Changsha Medical University, Changsha, China
| | - Binsheng He
- Hunan key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, School of Pharmaceutical Science, Changsha Medical University, Changsha, China
| | - Sen Li
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
2
|
Poto R, Marone G, Galli SJ, Varricchi G. Mast cells: a novel therapeutic avenue for cardiovascular diseases? Cardiovasc Res 2024; 120:681-698. [PMID: 38630620 PMCID: PMC11135650 DOI: 10.1093/cvr/cvae066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 11/28/2023] [Accepted: 01/08/2024] [Indexed: 04/19/2024] Open
Abstract
Mast cells are tissue-resident immune cells strategically located in different compartments of the normal human heart (the myocardium, pericardium, aortic valve, and close to nerves) as well as in atherosclerotic plaques. Cardiac mast cells produce a broad spectrum of vasoactive and proinflammatory mediators, which have potential roles in inflammation, angiogenesis, lymphangiogenesis, tissue remodelling, and fibrosis. Mast cells release preformed mediators (e.g. histamine, tryptase, and chymase) and de novo synthesized mediators (e.g. cysteinyl leukotriene C4 and prostaglandin D2), as well as cytokines and chemokines, which can activate different resident immune cells (e.g. macrophages) and structural cells (e.g. fibroblasts and endothelial cells) in the human heart and aorta. The transcriptional profiles of various mast cell populations highlight their potential heterogeneity and distinct gene and proteome expression. Mast cell plasticity and heterogeneity enable these cells the potential for performing different, even opposite, functions in response to changing tissue contexts. Human cardiac mast cells display significant differences compared with mast cells isolated from other organs. These characteristics make cardiac mast cells intriguing, given their dichotomous potential roles of inducing or protecting against cardiovascular diseases. Identification of cardiac mast cell subpopulations represents a prerequisite for understanding their potential multifaceted roles in health and disease. Several new drugs specifically targeting human mast cell activation are under development or in clinical trials. Mast cells and/or their subpopulations can potentially represent novel therapeutic targets for cardiovascular disorders.
Collapse
Affiliation(s)
- Remo Poto
- Department of Translational Medical Sciences, University of Naples Federico II, Via S. Pansini 5, Naples 80131, Italy
- World Allergy Organization (WAO), Center of Excellence (CoE), Via S. Pansini 5, Naples 80131, Italy
| | - Gianni Marone
- Department of Translational Medical Sciences, University of Naples Federico II, Via S. Pansini 5, Naples 80131, Italy
- World Allergy Organization (WAO), Center of Excellence (CoE), Via S. Pansini 5, Naples 80131, Italy
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Via S. Pansini 5, Naples 80131, Italy
- Institute of Experimental Endocrinology and Oncology ‘G. Salvatore’, National Research Council (CNR), Via S. Pansini 5, Naples 80131, Italy
| | - Stephen J Galli
- Department of Pathology and the Sean N. Parker Center for Allergy and Asthma Research, Stanford University School of Medicine, 291 Campus Dr, Stanford, CA, USA
- Department of Microbiology and Immunology, Stanford University School of Medicine, 291 Campus Dr, Stanford, CA, USA
| | - Gilda Varricchi
- Department of Translational Medical Sciences, University of Naples Federico II, Via S. Pansini 5, Naples 80131, Italy
- World Allergy Organization (WAO), Center of Excellence (CoE), Via S. Pansini 5, Naples 80131, Italy
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Via S. Pansini 5, Naples 80131, Italy
- Institute of Experimental Endocrinology and Oncology ‘G. Salvatore’, National Research Council (CNR), Via S. Pansini 5, Naples 80131, Italy
| |
Collapse
|
3
|
Obeagu EI. Diagnostic and prognostic significance of mast cell markers in HIV/AIDS: Current insights and future directions. Medicine (Baltimore) 2024; 103:e38117. [PMID: 38758896 PMCID: PMC11098248 DOI: 10.1097/md.0000000000038117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Accepted: 04/12/2024] [Indexed: 05/19/2024] Open
Abstract
Human immunodeficiency virus (HIV) infection continues to pose significant global health challenges, necessitating advancements in diagnostic and prognostic approaches to optimize disease management. While primarily recognized for their roles in allergic responses, mast cells have emerged as potential markers with diagnostic and prognostic significance in the context of HIV/AIDS. This paper aims to synthesize current insights and delineate future directions regarding the utility of mast cell markers in diagnosing HIV infection, predicting disease progression, and guiding therapeutic strategies. Mast cells, equipped with distinct markers such as tryptase, chymase, carboxypeptidase A3, and c-kit/CD117 receptors, exhibit tissue-specific expression patterns that offer potential as diagnostic indicators for HIV infection. Understanding the dynamics of these markers in different tissues and body fluids holds promise for accurate HIV diagnosis, disease staging, and monitoring treatment responses. Moreover, the prognostic significance of mast cell markers in HIV/AIDS lies in their potential to predict disease progression, immune dysregulation, and clinical outcomes. The integration of mast cell markers into clinical applications offers promising avenues for refining diagnostic assays, patient monitoring protocols, and therapeutic strategies in HIV/AIDS. Future research directions involve the development of novel diagnostic tools and targeted therapies based on mast cell-specific markers, potentially revolutionizing clinical practice and enhancing patient care in the management of HIV/AIDS. Continued investigations into mast cell markers' diagnostic and prognostic implications hold immense potential to advance our understanding and improve outcomes in HIV/AIDS management.
Collapse
|
4
|
Molderings GJ, Afrin LB. A survey of the currently known mast cell mediators with potential relevance for therapy of mast cell-induced symptoms. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023; 396:2881-2891. [PMID: 37243761 PMCID: PMC10567897 DOI: 10.1007/s00210-023-02545-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 05/18/2023] [Indexed: 05/29/2023]
Abstract
Mast cells (MCs) occupy a central role in immunological as well as non-immunological processes as reflected in the variety of the mediators by which MCs influence other cells. Published lists of MC mediators have all shown only subsets-usually quite small-of the full repertoire. The full repertoire of MC mediators released by exocytosis is comprehensively compiled here for the first time. The compilation of the data is essentially based on the largely cytokine-focused database COPE®, supplemented with data on the expression of substances in human MCs published in several articles, plus extensive research in the PubMed database. Three hundred and ninety substances could be identified as mediators of human MCs which can be secreted into the extracellular space by activation of the MC. This number might still be an underestimate of the actual number of MC mediators since, in principle, all substances produced by MCs can become mediators because of the possibility of their release by diffusion into the extracellular space, mast cell extracellular traps, and intercellular exchange via nanotubules. When human MCs release mediators in inappropriate manners, this may lead to symptoms in any or all organs/tissues. Thus, such MC activation disorders may clinically present with a myriad of potential combinations of symptoms ranging from trivial to disabling or even life-threatening. The present compilation can be consulted by physicians when trying to gain clarity about MC mediators which may be involved in patients with MC disease symptoms refractory to most therapies.
Collapse
Affiliation(s)
- Gerhard J Molderings
- Institute for Human Genetics, University Hospital of Bonn, Venusberg-Campus 1, D-53127, Bonn, Germany.
| | | |
Collapse
|
5
|
Pereira TCR, Fidale TM, Guimarães LC, Deconte SR, Herrera GC, Mundim AV, de Sales Cabral E, Lopes PR, de Souza FR, de Ulhôa Rocha Júnior LD, Silva ATF, Resende ES. Cardioprotective Effects of the 4-Week Aerobic Running Exercises Before Treatment with Doxorubicin in Rats. Cardiovasc Toxicol 2023; 23:265-277. [PMID: 37402033 DOI: 10.1007/s12012-023-09798-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 06/22/2023] [Indexed: 07/05/2023]
Abstract
Doxorubicin is associated with cardiotoxicity, and physical exercise seeks to minimize the toxic effects of doxorubicin through physiological cardiac remodeling, as well as the reduction of oxidative stress, evidenced by previous studies. This study aimed to analyze whether running training before treatment with doxorubicin influences tolerance to physical exertion and cardiotoxicity. Thirty-nine male Wistar rats, aged 90 days and weighing between 250 and 300 g, were divided into 4 groups: Control (C), Doxorubicin (D), Trained (T), and Trained + Doxorubicin (TD). Animals in groups T and DT were submitted to treadmill running for 3 weeks, 5 times a week at 18 m/min for 20-30 min before treatment with doxorubicin. Animals in groups D and DT received intraperitoneal injections of doxorubicin hydrochloride three times a week for two weeks, reaching a total cumulative dose of 7.50 mg/kg. Our results show an increase in total collagen fibers in the D group (p = 0.01), but no increase in the TD group, in addition to the attenuation of the number of cardiac mast cells in the animals in the TD group (p = 0.05). The animals in the TD group showed maintenance of tolerance to exertion compared to group D. Therefore, running training attenuated the cardiac damage caused by the treatment with doxorubicin, in addition to maintaining the tolerance to exertion in the rats.
Collapse
Affiliation(s)
- Talita Cristina Rodrigues Pereira
- Experimental Medicine Laboratory, Department of Medicine, Universidade Federal de Uberlândia-UFU, Uberlândia, MG, Brazil.
- , Uberlândia, Brazil.
| | - Thiago Montes Fidale
- Department of Medicine, Federal University of Catalão- UFCAT, Catalão-Goiás, Brazil
| | - Lucas Costa Guimarães
- Experimental Medicine Laboratory, Department of Medicine, Universidade Federal de Uberlândia-UFU, Uberlândia, MG, Brazil
| | - Simone Ramos Deconte
- Department of Physiology, Institute of Biomedical Sciences, Universidade Federal de Uberlândia-UFU, Uberlândia, MG, Brazil
| | | | | | - Eduardo de Sales Cabral
- Experimental Medicine Laboratory, Department of Medicine, Universidade Federal de Uberlândia-UFU, Uberlândia, MG, Brazil
| | - Paulo Ricardo Lopes
- Department of Physiology and Pathology, School of Dentistry-FOAr, Paulista State University "Julio de Mesquita Filho"-UNESP, Araraquara, SP, Brazil
| | - Fernanda Rodrigues de Souza
- Experimental Medicine Laboratory, Department of Medicine, Universidade Federal de Uberlândia-UFU, Uberlândia, MG, Brazil
| | | | - Alinne Tatiane Faria Silva
- Laboratory of Nanobiotechnology, Institute of Genetics and Biochemistry, Luiz Ricardo Goulart Filho, Universidade Federal de Uberlândia-UFU, Prof. Dr, Uberlândia, MG, Brazil
| | - Elmiro Santos Resende
- Graduate Program in Health Sciences-PPGCS, Faculty of Medicine, Universidade Federal de Uberlândia-UFU, Uberlândia, MG, Brazil
| |
Collapse
|
6
|
Harper RL, Fang F, San H, Negro A, St Hilaire C, Yang D, Chen G, Yu Z, Dmitrieva NI, Lanzer J, Davaine JM, Schwartzbeck R, Walts AD, Kovacic JC, Boehm M. Mast cell activation and degranulation in acute artery injury: A target for post-operative therapy. FASEB J 2023; 37:e23029. [PMID: 37310585 PMCID: PMC11095138 DOI: 10.1096/fj.202201745rr] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 05/10/2023] [Accepted: 06/01/2023] [Indexed: 06/14/2023]
Abstract
The increasing incidence of cardiovascular disease (CVD) has led to a significant ongoing need to address this surgically through coronary artery bypass grafting (CABG) and percutaneous coronary interventions (PCI). From this, there continues to be a substantial burden of mortality and morbidity due to complications arising from endothelial damage, resulting in restenosis. Whilst mast cells (MC) have been shown to have a causative role in atherosclerosis and other vascular diseases, including restenosis due to vein engraftment; here, we demonstrate their rapid response to arterial wire injury, recapitulating the endothelial damage seen in PCI procedures. Using wild-type mice, we demonstrate accumulation of MC in the femoral artery post-acute wire injury, with rapid activation and degranulation, resulting in neointimal hyperplasia, which was not observed in MC-deficient KitW-sh/W-sh mice. Furthermore, neutrophils, macrophages, and T cells were abundant in the wild-type mice area of injury but reduced in the KitW-sh/W-sh mice. Following bone-marrow-derived MC (BMMC) transplantation into KitW-sh/W-sh mice, not only was the neointimal hyperplasia induced, but the neutrophil, macrophage, and T-cell populations were also present in these transplanted mice. To demonstrate the utility of MC as a target for therapy, we administered the MC stabilizing drug, disodium cromoglycate (DSCG) immediately following arterial injury and were able to show a reduction in neointimal hyperplasia in wild-type mice. These studies suggest a critical role for MC in inducing the conditions and coordinating the detrimental inflammatory response seen post-endothelial injury in arteries undergoing revascularization procedures, and by targeting the rapid MC degranulation immediately post-surgery with DSCG, this restenosis may become a preventable clinical complication.
Collapse
Affiliation(s)
- Rebecca L Harper
- Translational Vascular Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Fang Fang
- Translational Vascular Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Hong San
- Translational Vascular Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Alejandra Negro
- Translational Vascular Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Cynthia St Hilaire
- Departments of Medicine and Bioengineering, Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Dan Yang
- Translational Vascular Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Guibin Chen
- Translational Vascular Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Zhen Yu
- Translational Vascular Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Natalia I Dmitrieva
- Translational Vascular Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Jan Lanzer
- Translational Vascular Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Jean-Michel Davaine
- Translational Vascular Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Robin Schwartzbeck
- Translational Vascular Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Avram D Walts
- Department of Medicine, National Jewish Health, Denver, Colorado, USA
| | - Jason C Kovacic
- The Zena and Michael A. Wiener Cardiovascular Institute, Mount Sinai School of Medicine, New York, New York, USA
- Victor Chang Cardiac Research Institute, Darlinghurst, Australia; St Vincent's Clinical School, University of NSW, Sydney, New South Wales, Australia
| | - Manfred Boehm
- Translational Vascular Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
7
|
Moriyama H, Endo J. Pathophysiological Involvement of Mast Cells and the Lipid Mediators in Pulmonary Vascular Remodeling. Int J Mol Sci 2023; 24:6619. [PMID: 37047587 PMCID: PMC10094825 DOI: 10.3390/ijms24076619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/24/2023] [Accepted: 03/30/2023] [Indexed: 04/05/2023] Open
Abstract
Mast cells are responsible for IgE-dependent allergic responses, but they also produce various bioactive mediators and contribute to the pathogenesis of various cardiovascular diseases, including pulmonary hypertension (PH). The importance of lipid mediators in the pathogenesis of PH has become evident in recent years, as exemplified by prostaglandin I2, the most central therapeutic target in pulmonary arterial hypertension. New bioactive lipids other than eicosanoids have also been identified that are associated with the pathogenesis of PH. However, it remains largely unknown how mast cell-derived lipid mediators are involved in pulmonary vascular remodeling. Recently, it has been demonstrated that mast cells produce epoxidized n-3 fatty acid (n-3 epoxides) in a degranulation-independent manner, and that n-3 epoxides produced by mast cells regulate the abnormal activation of pulmonary fibroblasts and suppress the progression of pulmonary vascular remodeling. This review summarizes the role of mast cells and bioactive lipids in the pathogenesis of PH. In addition, we introduce the pathophysiological role and therapeutic potential of n-3 epoxides, a mast cell-derived novel lipid mediator, in the pulmonary vascular remodeling in PH. Further knowledge of mast cells and lipid mediators is expected to lead to the development of innovative therapies targeting pulmonary vascular remodeling.
Collapse
Affiliation(s)
- Hidenori Moriyama
- Department of Cardiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku 160-8582, Tokyo, Japan
- Department of Cardiology, Tokyo Dental College Ichikawa General Hospital, 5-11-13 Sugano, Ichikawa 272-8513, Chiba, Japan
| | - Jin Endo
- Department of Cardiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku 160-8582, Tokyo, Japan
| |
Collapse
|
8
|
Autoantibodies to IgE can induce the release of proinflammatory and vasoactive mediators from human cardiac mast cells. Clin Exp Med 2022:10.1007/s10238-022-00861-w. [PMID: 35879625 PMCID: PMC10390627 DOI: 10.1007/s10238-022-00861-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 06/30/2022] [Indexed: 11/03/2022]
Abstract
Mast cells are multifunctional immune cells with complex roles in tissue homeostasis and disease. Cardiac mast cells (HCMCs) are strategically located within the human myocardium, in atherosclerotic plaques, in proximity to nerves, and in the aortic valve. HCMCs express the high-affinity receptor (FcεRI) for IgE and can be activated by anti-IgE and anti-FcεRI. Autoantibodies to IgE and/or FcεRI have been found in the serum of patients with a variety of immune disorders. We have compared the effects of different preparations of IgG anti-IgE obtained from patients with atopic dermatitis (AD) with rabbit IgG anti-IgE on the release of preformed (histamine and tryptase) and lipid mediators [prostaglandin D2 (PGD2) and cysteinyl leukotriene C4 (LTC4)] from HCMCs. Functional human IgG anti-IgE from one out of six AD donors and rabbit IgG anti-IgE induced the release of preformed (histamine, tryptase) and de novo synthesized mediators (PGD2 and LTC4) from HCMCs. Human IgG anti-IgE was more potent than rabbit IgG anti-IgE in inducing proinflammatory mediators from HCMCs. Human monoclonal IgE was a competitive antagonist of both human and rabbit IgG anti-IgE. Although functional anti-IgE autoantibodies rarely occur in patients with AD, when present, they can powerfully activate the release of proinflammatory and vasoactive mediators from HCMCs.
Collapse
|
9
|
Jin J, Jiang Y, Chakrabarti S, Su Z. Cardiac Mast Cells: A Two-Head Regulator in Cardiac Homeostasis and Pathogenesis Following Injury. Front Immunol 2022; 13:963444. [PMID: 35911776 PMCID: PMC9334794 DOI: 10.3389/fimmu.2022.963444] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 06/24/2022] [Indexed: 11/23/2022] Open
Abstract
Cardiac mast cells (CMCs) are multifarious immune cells with complex roles both in cardiac physiological and pathological conditions, especially in cardiac fibrosis. Little is known about the physiological importance of CMCs in cardiac homeostasis and inflammatory process. Therefore, the present review will summarize the recent progress of CMCs on origin, development and replenishment in the heart, including their effects on cardiac development, function and ageing under physiological conditions as well as the roles of CMCs in inflammatory progression and resolution. The present review will shed a light on scientists to understand cardioimmunology and to develop immune treatments targeting on CMCs following cardiac injury.
Collapse
Affiliation(s)
- Jing Jin
- International Genome Center, Jiangsu University, Zhenjiang, China
- Institute of Immunology, Jiangsu University, Zhenjiang, China
| | - Yuanyuan Jiang
- Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Subrata Chakrabarti
- Department of Pathology and Laboratory Medicine, Western University, London, ON, Canada
| | - Zhaoliang Su
- International Genome Center, Jiangsu University, Zhenjiang, China
- Institute of Immunology, Jiangsu University, Zhenjiang, China
- *Correspondence: Zhaoliang Su,
| |
Collapse
|
10
|
Nizawa T, Bhutto IA, Tiwari A, Grebe RR, Alt J, Rais R, Edwards MM, Lutty GA. Topical Ketotifen Fumarate Inhibits Choroidal Mast Cell Degranulation and Loss of Retinal Pigment Epithelial Cells in Rat Model for Geographic Atrophy. Transl Vis Sci Technol 2021; 10:37. [PMID: 34967831 PMCID: PMC8727493 DOI: 10.1167/tvst.10.14.37] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose This study evaluates whether topical ketotifen fumarate (KTF) can prevent geographic atrophy (GA)-like phenotypes in a rat model. Methods Pharmacokinetics (PKs) of KTF after topical administration twice daily for 5 days was analyzed in rat retina, retinal pigment epithelium (RPE)/choroid/sclera, and in plasma by an liquid chromatography tandem mass spectrometry (LC-MS/MS) method. Rats were then given hydrogel implants +/- 48/80 in the superior subconjunctival space and topically treated with 1% and 0.25% of KTF or phosphate buffer saline (PBS) twice daily. Rats were euthanized at 1, 2, 4, and 8 weeks postinjection. Choroidal mast cells (MCs) were stained with nonspecific esterase and the RPE monolayer was labeled with RPE65 and ZO-1 in whole mount choroids. Retinal and choroidal areas were determined in cryosections stained with picrosirius red. Dark-adapted electroretinogram (ERG) was also performed to evaluate retinal function. Results PK results showed the highest level of KTF (average 5.6 nM/mg) in the RPE/choroid/sclera in rats given topical 1% KTF. Topical 1% KTF significantly reduced choroidal MC degranulation at 1 week and 2 weeks (both P < 0.001) and RPE loss at 4 weeks (P < 0.001) as well as retinal and choroidal thinning (both P < 0.001) and reduction in ERG amplitude at 8 weeks (P < 0.05) compared to PBS. Similar results were obtained with 0.25% KTF. Conclusions Both 1% and 0.25% KTF eye drops effectively reduced MC degranulation, RPE loss, and retinal and choroidal thinning while preventing the decline of ERG amplitude in a GA-like rat model. These data suggest that topical KTF might be a new therapeutic drug for treating GA. Translational Relevance The results of this study demonstrate that topical KTF successfully reduced GA-like phenotypes in a rat model and may provide a novel therapy for GA.
Collapse
Affiliation(s)
- Tomohiro Nizawa
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Imran A Bhutto
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Anupama Tiwari
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Rhonda R Grebe
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jesse Alt
- Brain Science Institute, Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Rana Rais
- Brain Science Institute, Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Malia M Edwards
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Gerard A Lutty
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
11
|
Guimbal S, Cornuault L, Rouault P, Hollier PL, Chapouly C, Bats ML, Imbault J, Gadeau AP, Couffinhal T, Renault MA. Mast Cells Are the Trigger of Small Vessel Disease and Diastolic Dysfunction in Diabetic Obese Mice. Arterioscler Thromb Vasc Biol 2021; 41:e193-e207. [PMID: 33567863 DOI: 10.1161/atvbaha.121.315900] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
[Figure: see text].
Collapse
MESH Headings
- Animals
- Cell Degranulation
- Cells, Cultured
- Coronary Vessels/immunology
- Coronary Vessels/metabolism
- Coronary Vessels/pathology
- Diabetes Mellitus/genetics
- Diabetes Mellitus/immunology
- Diabetes Mellitus/metabolism
- Diastole
- Disease Models, Animal
- Female
- Heart Failure/etiology
- Heart Failure/immunology
- Heart Failure/metabolism
- Heart Failure/physiopathology
- Histamine Release
- Humans
- Mast Cells/immunology
- Mast Cells/metabolism
- Mast Cells/pathology
- Mice, Knockout
- Myocardium/immunology
- Myocardium/metabolism
- Myocardium/pathology
- Obesity/complications
- Obesity/immunology
- Obesity/metabolism
- Receptors, Leptin/deficiency
- Receptors, Leptin/genetics
- Ventricular Dysfunction, Left/etiology
- Ventricular Dysfunction, Left/immunology
- Ventricular Dysfunction, Left/metabolism
- Ventricular Dysfunction, Left/physiopathology
- Ventricular Function, Left
- Mice
Collapse
Affiliation(s)
- Sarah Guimbal
- University of Bordeaux, Inserm, Biology of Cardiovascular Diseases, U1034, CHU de Bordeaux, Pessac, France
| | - Lauriane Cornuault
- University of Bordeaux, Inserm, Biology of Cardiovascular Diseases, U1034, CHU de Bordeaux, Pessac, France
| | - Paul Rouault
- University of Bordeaux, Inserm, Biology of Cardiovascular Diseases, U1034, CHU de Bordeaux, Pessac, France
| | - Pierre-Louis Hollier
- University of Bordeaux, Inserm, Biology of Cardiovascular Diseases, U1034, CHU de Bordeaux, Pessac, France
| | - Candice Chapouly
- University of Bordeaux, Inserm, Biology of Cardiovascular Diseases, U1034, CHU de Bordeaux, Pessac, France
| | - Marie-Lise Bats
- University of Bordeaux, Inserm, Biology of Cardiovascular Diseases, U1034, CHU de Bordeaux, Pessac, France
| | - Julien Imbault
- University of Bordeaux, Inserm, Biology of Cardiovascular Diseases, U1034, CHU de Bordeaux, Pessac, France
| | - Alain-Pierre Gadeau
- University of Bordeaux, Inserm, Biology of Cardiovascular Diseases, U1034, CHU de Bordeaux, Pessac, France
| | - Thierry Couffinhal
- University of Bordeaux, Inserm, Biology of Cardiovascular Diseases, U1034, CHU de Bordeaux, Pessac, France
| | - Marie-Ange Renault
- University of Bordeaux, Inserm, Biology of Cardiovascular Diseases, U1034, CHU de Bordeaux, Pessac, France
| |
Collapse
|
12
|
Varricchi G, Marone G, Kovanen PT. Cardiac Mast Cells: Underappreciated Immune Cells in Cardiovascular Homeostasis and Disease. Trends Immunol 2020; 41:734-746. [DOI: 10.1016/j.it.2020.06.006] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 06/04/2020] [Accepted: 06/12/2020] [Indexed: 02/08/2023]
|
13
|
Kouhpeikar H, Delbari Z, Sathyapalan T, Simental-Mendía LE, Jamialahmadi T, Sahebkar A. The Effect of Statins through Mast Cells in the Pathophysiology of Atherosclerosis: a Review. Curr Atheroscler Rep 2020; 22:19. [PMID: 32458165 DOI: 10.1007/s11883-020-00837-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
PURPOSE OF REVIEW In this review, we discuss the evidence supporting the effects of statins on mast cells (MCs) in atherosclerosis and their molecular mechanism of action. RECENT FINDINGS Statins or HMG-CoA reductase inhibitors are known for their lipid-lowering properties and are widely used in the prevention and treatment of cardiovascular diseases. There is growing evidence that statins have an inhibitory effect on MCs, which contributes to the pleiotropic effect of statins in various diseases. MCs are one of the crucial effectors of the immune system which play an essential role in the pathogenesis of multiple disorders. Recent studies have shown that MCs are involved in the development of atherosclerotic plaques. MCs secrete various inflammatory cytokines (IL-6, IL4, TNF-α, and IFNγ) and inflammatory mediators (histamine, tryptase, proteoglycans) after activation by various stimulants. This, in turn, will exacerbate atherosclerosis. Statins suppress the activation of MCs via IgE inhibition which leads to inhibition of inflammatory mediators and cytokines which are involved in the development and progression of atherosclerosis. In keeping with this evidence presented here, MCs can be considered as one of the therapeutic targets for statins in the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Hamideh Kouhpeikar
- Department of hematology and blood bank, Tabas school of nursing, Birjand University of Medical Science, Birjand, Iran
| | - Zahra Delbari
- Inflammation and Inflammatory Diseases Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Thozhukat Sathyapalan
- Department of Academic Diabetes, Endocrinology and Metabolism, Hull York Medical School, University of Hull, Hull, HU3 2JZ, UK
| | | | - Tannaz Jamialahmadi
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Halal Research Center of IRI, FDA, Tehran, Iran. .,Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran. .,School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
14
|
Song Y, Lu M, Yuan H, Chen T, Han X. Mast cell-mediated neuroinflammation may have a role in attention deficit hyperactivity disorder (Review). Exp Ther Med 2020; 20:714-726. [PMID: 32742317 PMCID: PMC7388140 DOI: 10.3892/etm.2020.8789] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 04/29/2020] [Indexed: 12/12/2022] Open
Abstract
Attention deficit hyperactivity disorder (ADHD) is a common neurodevelopmental and behavioral disorder with a serious negative impact on the quality of life from childhood until adulthood, which may cause academic failure, family disharmony and even social unrest. The pathogenesis of ADHD has remained to be fully elucidated, leading to difficulties in the treatment of this disease. Genetic and environmental factors contribute to the risk of ADHD development. Certain studies indicated that ADHD has high comorbidity with allergic and autoimmune diseases, with various patients with ADHD having a high inflammatory status. Increasing evidence indicated that mast cells (MCs) are involved in the pathogenesis of brain inflammation and neuropsychiatric disorders. MCs may cause or aggravate neuroinflammation via the selective release of inflammatory factors, interaction with glial cells and neurons, activation of the hypothalamic-pituitary adrenal axis or disruption of the blood-brain barrier integrity. In the present review, the notion that MC activation may be involved in the occurrence and development of ADHD through a number of ways is discussed based on previously published studies. The association between MCs and ADHD appears to lack sufficient evidence at present and this hypothesis is considered to be worthy of further study, providing a novel perspective for the treatment of ADHD.
Collapse
Affiliation(s)
- Yuchen Song
- Institute of Pediatrics of Traditional Chinese Medicine, First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| | - Manqi Lu
- Institute of Pediatrics of Traditional Chinese Medicine, First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| | - Haixia Yuan
- Institute of Pediatrics of Traditional Chinese Medicine, First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| | - Tianyi Chen
- Institute of Pediatrics of Traditional Chinese Medicine, First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| | - Xinmin Han
- Institute of Pediatrics of Traditional Chinese Medicine, First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| |
Collapse
|
15
|
Mitry MA, Laurent D, Keith BL, Sira E, Eisenberg CA, Eisenberg LM, Joshi S, Gupte S, Edwards JG. Accelerated cardiomyocyte senescence contributes to late-onset doxorubicin-induced cardiotoxicity. Am J Physiol Cell Physiol 2020; 318:C380-C391. [PMID: 31913702 DOI: 10.1152/ajpcell.00073.2019] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Children surviving cancer and chemotherapy are at risk for adverse health events including heart failure that may be delayed by years. Although the early effects of doxorubicin-induced cardiotoxicity may be attributed to a direct effect on the cardiomyocytes, the mechanisms underlying the delayed or late effects (8-20 yr) are unknown. The goal of this project was to develop a model of late-onset doxorubicin-induced cardiotoxicity to better delineate the underlying pathophysiology responsible. The underlying hypothesis was that doxorubicin-induced "late-onset cardiotoxicity" was the result of mitochondrial dysfunction leading to cell failure and death. Wistar rats, 3-4 wk of age, were randomly assigned to vehicle or doxorubicin injection groups (1-45 mg/kg). Cardiovascular function was unaltered at the lower dosages (1-15 kg/mg), but beginning at 6 mo after injection significant cardiac degradation was observed in the 45 mg/kg group. Doxorubicin significantly increased myocardial mitochondrial DNA (mtDNA) damage. In contrast, in isolated c-kit left ventricular (LV) cells, doxorubicin treatment did not increase mtDNA damage. Biomarkers of senescence within the LV were significantly increased, suggesting accelerated aging of the LV. Doxorubicin also significantly increased LV histamine content suggestive of mast cell activation. With the use of flow cytometry, a significant expansion of the c-kit and stage-specific embryonic antigen 1 cell populations within the LV were concomitant with significant decreases in the circulating peripheral blood population of these cells. These results are consistent with the concept that doxorubicin induced significant damage to the cardiomyocyte population and that although the heart attempted to compensate it eventually succumbed to an inability for self-repair.
Collapse
Affiliation(s)
- Maria A Mitry
- Department of Physiology, New York Medical College, Valhalla, New York
| | - Dimitri Laurent
- Department of Physiology, New York Medical College, Valhalla, New York
| | - Britny L Keith
- Department of Physiology, New York Medical College, Valhalla, New York
| | - Elizabeth Sira
- Department of Physiology, New York Medical College, Valhalla, New York
| | - Carol A Eisenberg
- Department of Physiology, New York Medical College, Valhalla, New York
| | | | - Sachindra Joshi
- Department of Pharmacology, New York Medical College, Valhalla, New York
| | - Sachin Gupte
- Department of Pharmacology, New York Medical College, Valhalla, New York
| | - John G Edwards
- Department of Physiology, New York Medical College, Valhalla, New York
| |
Collapse
|
16
|
Abdel-Hamid AA, Firgany AEDL. Increased mast cell number is associated with a decrease in beta-cell mass and regeneration in type 2 diabetic rats. Acta Histochem 2019; 121:508-515. [PMID: 31014904 DOI: 10.1016/j.acthis.2019.04.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 04/16/2019] [Accepted: 04/17/2019] [Indexed: 11/27/2022]
Abstract
The role of mast cells (MCs) in type 2 diabetes (T2D) is not thoroughly studied as much as in T1D. Therefore in the current study we investigated correlation between these cells and various parameters of islets of Langerhans (IOL) in rats which were equally divided (n = 40) into; control and diabetic groups. We detected a significantly increased (p < 0.05) MC count (MCC) in the diabetic IOL compared to the control, together with a noticeable intra-islet seeding of these cells which displayed a tryptase positive immunostaining. A significant positive correlation (p < 0.05) between MCC and the % of glucagon cells per islet was detected in DM, unlike mass of the islets, mass of β-cells, and % of β-cells per islet which were negatively correlated with MCC. Similarly, there was a negative correlation between MCC with β-cell proliferation and neogenesis frequency in DM. This highlights the potential association between the increased MC number and the diminished islet`s mass as well as regeneration which may fasten the progression of T2D.
Collapse
|
17
|
Mohajeri M, Kovanen PT, Bianconi V, Pirro M, Cicero AFG, Sahebkar A. Mast cell tryptase - Marker and maker of cardiovascular diseases. Pharmacol Ther 2019; 199:91-110. [PMID: 30877022 DOI: 10.1016/j.pharmthera.2019.03.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 03/01/2019] [Indexed: 12/14/2022]
Abstract
Mast cells are tissue-resident cells, which have been proposed to participate in various inflammatory diseases, among them the cardiovascular diseases (CVDs). For mast cells to be able to contribute to an inflammatory process, they need to be activated to exocytose their cytoplasmic secretory granules. The granules contain a vast array of highly bioactive effector molecules, the neutral protease tryptase being the most abundant protein among them. The released tryptase may act locally in the inflamed cardiac or vascular tissue, so contributing directly to the pathogenesis of CVDs. Moreover, a fraction of the released tryptase reaches the systemic circulation, thereby serving as a biomarker of mast cell activation. Actually, increased levels of circulating tryptase have been found to associate with CVDs. Here we review the biological relevance of the circulating tryptase as a biomarker of mast cell activity in CVDs, with special emphasis on the relationship between activation of mast cells in their tissue microenvironments and the pathophysiological pathways of CVDs. Based on the available in vitro and in vivo studies, we highlight the potential molecular mechanisms by which tryptase may contribute to the pathogenesis of CVDs. Finally, the synthetic and natural inhibitors of tryptase are reviewed for their potential utility as therapeutic agents in CVDs.
Collapse
Affiliation(s)
- Mohammad Mohajeri
- Department of Medical Biotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Vanessa Bianconi
- Unit of Internal Medicine, Department of Medicine, University of Perugia, Perugia, Italy
| | - Matteo Pirro
- Unit of Internal Medicine, Department of Medicine, University of Perugia, Perugia, Italy
| | - Arrigo F G Cicero
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
18
|
Kritikou E, van der Heijden T, Swart M, van Duijn J, Slütter B, Wezel A, Smeets HJ, Maffia P, Kuiper J, Bot I. Hypercholesterolemia Induces a Mast Cell-CD4 + T Cell Interaction in Atherosclerosis. THE JOURNAL OF IMMUNOLOGY 2019; 202:1531-1539. [PMID: 30683705 DOI: 10.4049/jimmunol.1800648] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 12/20/2018] [Indexed: 11/19/2022]
Abstract
Mast cells (MCs) are potent innate immune cells that aggravate atherosclerosis through the release of proinflammatory mediators inside atherosclerotic plaques. Similarly, CD4+ T cells are constituents of the adaptive immune response and accumulate within the plaques following lipid-specific activation by APCs. Recently it has been proposed that these two cell types can interact in a direct manner. However, no indication of such an interaction has been investigated in the context of atherosclerosis. In our study, we aimed to examine whether MCs can act as APCs in atherosclerosis, thereby modulating CD4+ T cell responses. We observed that MCs increased their MHC class II expression under hyperlipidemic conditions both in vivo and in vitro. Furthermore, we showed that MCs can present Ags in vivo via MHC class II molecules. Serum from high-fat diet-fed mice also enhanced the expression of the costimulatory molecule CD86 on cultured MCs, whereas OVA peptide-loaded MCs increased OT-II CD4+ T cell proliferation in vitro. The aortic CD4+ and TH1 cell content of atherosclerotic mice that lack MCs was reduced as compared with their wild-type counterparts. Importantly, we identified MCs that express HLA-DR in advanced human atheromata, indicating that these cells are capable of Ag presentation within human atherosclerotic plaques. Therefore, in this artice, we show that MCs may directly modulate adaptive immunity by acting as APCs in atherosclerosis.
Collapse
Affiliation(s)
- Eva Kritikou
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, 2300 RA Leiden, the Netherlands;
| | - Thomas van der Heijden
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, 2300 RA Leiden, the Netherlands
| | - Maarten Swart
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, 2300 RA Leiden, the Netherlands
| | - Janine van Duijn
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, 2300 RA Leiden, the Netherlands
| | - Bram Slütter
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, 2300 RA Leiden, the Netherlands
| | - Anouk Wezel
- Department of Surgery, Haaglanden Medical Center Westeinde, 2501 CK The Hague, the Netherlands
| | - Harm J Smeets
- Department of Surgery, Haaglanden Medical Center Westeinde, 2501 CK The Hague, the Netherlands
| | - Pasquale Maffia
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8TA, United Kingdom.,Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8TA, United Kingdom; and.,Department of Pharmacy, University of Naples Federico II, 80138 Naples, Italy
| | - Johan Kuiper
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, 2300 RA Leiden, the Netherlands
| | - Ilze Bot
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, 2300 RA Leiden, the Netherlands
| |
Collapse
|
19
|
Šteiner I, Stejskal V, Žáček P. Mast cells in calcific aortic stenosis. Pathol Res Pract 2018; 214:163-168. [DOI: 10.1016/j.prp.2017.07.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 06/26/2017] [Accepted: 07/19/2017] [Indexed: 12/24/2022]
|
20
|
Kempuraj D, Selvakumar GP, Thangavel R, Ahmed ME, Zaheer S, Raikwar SP, Iyer SS, Bhagavan SM, Beladakere-Ramaswamy S, Zaheer A. Mast Cell Activation in Brain Injury, Stress, and Post-traumatic Stress Disorder and Alzheimer's Disease Pathogenesis. Front Neurosci 2017; 11:703. [PMID: 29302258 PMCID: PMC5733004 DOI: 10.3389/fnins.2017.00703] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 11/30/2017] [Indexed: 12/30/2022] Open
Abstract
Mast cells are localized throughout the body and mediate allergic, immune, and inflammatory reactions. They are heterogeneous, tissue-resident, long-lived, and granulated cells. Mast cells increase their numbers in specific site in the body by proliferation, increased recruitment, increased survival, and increased rate of maturation from its progenitors. Mast cells are implicated in brain injuries, neuropsychiatric disorders, stress, neuroinflammation, and neurodegeneration. Brain mast cells are the first responders before microglia in the brain injuries since mast cells can release prestored mediators. Mast cells also can detect amyloid plaque formation during Alzheimer's disease (AD) pathogenesis. Stress conditions activate mast cells to release prestored and newly synthesized inflammatory mediators and induce increased blood-brain barrier permeability, recruitment of immune and inflammatory cells into the brain and neuroinflammation. Stress induces the release of corticotropin-releasing hormone (CRH) from paraventricular nucleus of hypothalamus and mast cells. CRH activates glial cells and mast cells through CRH receptors and releases neuroinflammatory mediators. Stress also increases proinflammatory mediator release in the peripheral systems that can induce and augment neuroinflammation. Post-traumatic stress disorder (PTSD) is a traumatic-chronic stress related mental dysfunction. Currently there is no specific therapy to treat PTSD since its disease mechanisms are not yet clearly understood. Moreover, recent reports indicate that PTSD could induce and augment neuroinflammation and neurodegeneration in the pathogenesis of neurodegenerative diseases. Mast cells play a crucial role in the peripheral inflammation as well as in neuroinflammation due to brain injuries, stress, depression, and PTSD. Therefore, mast cells activation in brain injury, stress, and PTSD may accelerate the pathogenesis of neuroinflammatory and neurodegenerative diseases including AD. This review focusses on how mast cells in brain injuries, stress, and PTSD may promote the pathogenesis of AD. We suggest that inhibition of mast cells activation and brain cells associated inflammatory pathways in the brain injuries, stress, and PTSD can be explored as a new therapeutic target to delay or prevent the pathogenesis and severity of AD.
Collapse
Affiliation(s)
- Duraisamy Kempuraj
- Department of Neurology and Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, United States
- Harry S. Truman Memorial Veteran's Hospital, United States Department of Veterans Affairs, Columbia, MO, United States
| | - Govindhasamy P. Selvakumar
- Department of Neurology and Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, United States
- Harry S. Truman Memorial Veteran's Hospital, United States Department of Veterans Affairs, Columbia, MO, United States
| | - Ramasamy Thangavel
- Department of Neurology and Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, United States
- Harry S. Truman Memorial Veteran's Hospital, United States Department of Veterans Affairs, Columbia, MO, United States
| | - Mohammad E. Ahmed
- Department of Neurology and Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, United States
- Harry S. Truman Memorial Veteran's Hospital, United States Department of Veterans Affairs, Columbia, MO, United States
| | - Smita Zaheer
- Department of Neurology and Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, United States
| | - Sudhanshu P. Raikwar
- Department of Neurology and Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, United States
- Harry S. Truman Memorial Veteran's Hospital, United States Department of Veterans Affairs, Columbia, MO, United States
| | - Shankar S. Iyer
- Department of Neurology and Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, United States
- Harry S. Truman Memorial Veteran's Hospital, United States Department of Veterans Affairs, Columbia, MO, United States
| | - Sachin M. Bhagavan
- Department of Neurology and Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, United States
| | - Swathi Beladakere-Ramaswamy
- Department of Neurology and Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, United States
| | - Asgar Zaheer
- Department of Neurology and Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, United States
- Harry S. Truman Memorial Veteran's Hospital, United States Department of Veterans Affairs, Columbia, MO, United States
| |
Collapse
|
21
|
Kovanen PT, Bot I. Mast cells in atherosclerotic cardiovascular disease – Activators and actions. Eur J Pharmacol 2017; 816:37-46. [DOI: 10.1016/j.ejphar.2017.10.013] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 10/06/2017] [Accepted: 10/09/2017] [Indexed: 12/19/2022]
|
22
|
McLeod DS, Bhutto I, Edwards MM, Gedam M, Baldeosingh R, Lutty GA. Mast Cell-Derived Tryptase in Geographic Atrophy. Invest Ophthalmol Vis Sci 2017; 58:5887-5896. [PMID: 29164232 PMCID: PMC5699534 DOI: 10.1167/iovs.17-22989] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 10/14/2017] [Indexed: 11/25/2022] Open
Abstract
Purpose Our previous study demonstrated significantly more degranulating mast cells (MCs) in choroids from subjects with age-related macular degeneration compared to aged controls. This study examined the immunolocalization of tryptase, the most abundant MC secretory granule-derived serine protease, in aged control eyes and eyes with geographic atrophy (GA). Methods Postmortem human eyes with and without GA were obtained from the National Disease Research Interchange. Tissue was fixed, cryopreserved, sectioned, and immunostained with a monoclonal antibody against tryptase. Sections were imaged on a Zeiss 710 Confocal Microscope. Results In the posterior pole of all aged control eyes, tryptase was confined to choroidal MCs, which were located primarily in Sattler's layer. In eyes with GA, many MCs were located in the inner choroid near choriocapillaris and Bruch's membrane (BM). Tryptase was found not only in MCs but also diffusely around them in stroma, suggesting they had degranulated. In contrast with aged control eyes, eyes with GA also had strong tryptase staining in BM. Tryptase was observed within BM in regions of RPE atrophy, at the border of atrophy, and extending well into the nonatrophic region. Conclusions Our results demonstrate that tryptase, released during choroidal MC degranulation, binds to BM in GA in advance of RPE atrophy. Tryptase activates MMPs that can degrade extracellular matrix (ECM) and basement membrane components found in BM. ECM modifications are likely to have a profound effect on the function and health of RPE and choroidal thinning in GA.
Collapse
Affiliation(s)
- D. Scott McLeod
- Wilmer Ophthalmological Institute, Johns Hopkins Hospital, Baltimore, Maryland, United States
| | - Imran Bhutto
- Wilmer Ophthalmological Institute, Johns Hopkins Hospital, Baltimore, Maryland, United States
| | - Malia M. Edwards
- Wilmer Ophthalmological Institute, Johns Hopkins Hospital, Baltimore, Maryland, United States
| | - Manasee Gedam
- Wilmer Ophthalmological Institute, Johns Hopkins Hospital, Baltimore, Maryland, United States
| | - Rajkumar Baldeosingh
- Wilmer Ophthalmological Institute, Johns Hopkins Hospital, Baltimore, Maryland, United States
| | - Gerard A. Lutty
- Wilmer Ophthalmological Institute, Johns Hopkins Hospital, Baltimore, Maryland, United States
| |
Collapse
|
23
|
Ngkelo A, Richart A, Kirk JA, Bonnin P, Vilar J, Lemitre M, Marck P, Branchereau M, Le Gall S, Renault N, Guerin C, Ranek MJ, Kervadec A, Danelli L, Gautier G, Blank U, Launay P, Camerer E, Bruneval P, Menasche P, Heymes C, Luche E, Casteilla L, Cousin B, Rodewald HR, Kass DA, Silvestre JS. Mast cells regulate myofilament calcium sensitization and heart function after myocardial infarction. J Exp Med 2017; 213:1353-74. [PMID: 27353089 PMCID: PMC4925026 DOI: 10.1084/jem.20160081] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2016] [Accepted: 05/12/2016] [Indexed: 11/24/2022] Open
Abstract
Ngkelo et al. use a mast cell–deficient mouse model to reveal a protective role of mast cells in myocardial infarction, through regulation of the cardiac contractile machinery. Acute myocardial infarction (MI) is a severe ischemic disease responsible for heart failure and sudden death. Inflammatory cells orchestrate postischemic cardiac remodeling after MI. Studies using mice with defective mast/stem cell growth factor receptor c-Kit have suggested key roles for mast cells (MCs) in postischemic cardiac remodeling. Because c-Kit mutations affect multiple cell types of both immune and nonimmune origin, we addressed the impact of MCs on cardiac function after MI, using the c-Kit–independent MC-deficient (Cpa3Cre/+) mice. In response to MI, MC progenitors originated primarily from white adipose tissue, infiltrated the heart, and differentiated into mature MCs. MC deficiency led to reduced postischemic cardiac function and depressed cardiomyocyte contractility caused by myofilament Ca2+ desensitization. This effect correlated with increased protein kinase A (PKA) activity and hyperphosphorylation of its targets, troponin I and myosin-binding protein C. MC-specific tryptase was identified to regulate PKA activity in cardiomyocytes via protease-activated receptor 2 proteolysis. This work reveals a novel function for cardiac MCs modulating cardiomyocyte contractility via alteration of PKA-regulated force–Ca2+ interactions in response to MI. Identification of this MC-cardiomyocyte cross-talk provides new insights on the cellular and molecular mechanisms regulating the cardiac contractile machinery and a novel platform for therapeutically addressable regulators.
Collapse
Affiliation(s)
- Anta Ngkelo
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS-970, Centre de Recherche Cardiovasculaire, Université Paris Descartes, Sorbonne Paris Cité, F-75015 Paris, France
| | - Adèle Richart
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS-970, Centre de Recherche Cardiovasculaire, Université Paris Descartes, Sorbonne Paris Cité, F-75015 Paris, France
| | - Jonathan A Kirk
- Division of Cardiology, Johns Hopkins Medical Institutions, Baltimore, MD 212015
| | - Philippe Bonnin
- INSERM, U965, Hôpital Lariboisière-Fernand-Widal, Assistance Publique Hôpitaux de Paris, F-75010 Paris, France
| | - Jose Vilar
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS-970, Centre de Recherche Cardiovasculaire, Université Paris Descartes, Sorbonne Paris Cité, F-75015 Paris, France
| | - Mathilde Lemitre
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS-970, Centre de Recherche Cardiovasculaire, Université Paris Descartes, Sorbonne Paris Cité, F-75015 Paris, France
| | - Pauline Marck
- INSERM, UMR-1048, Institut des Maladies Métaboliques et Cardiovasculaires, F-31004 Toulouse, France
| | - Maxime Branchereau
- INSERM, UMR-1048, Institut des Maladies Métaboliques et Cardiovasculaires, F-31004 Toulouse, France
| | - Sylvain Le Gall
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS-970, Centre de Recherche Cardiovasculaire, Université Paris Descartes, Sorbonne Paris Cité, F-75015 Paris, France
| | - Nisa Renault
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS-970, Centre de Recherche Cardiovasculaire, Université Paris Descartes, Sorbonne Paris Cité, F-75015 Paris, France
| | - Coralie Guerin
- National Cytometry Platform, Department of Infection and Immunity, Luxembourg Institute of Health, L-4354 Esch-sur-Alzette, Luxembourg
| | - Mark J Ranek
- Division of Cardiology, Johns Hopkins Medical Institutions, Baltimore, MD 212015
| | - Anaïs Kervadec
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS-970, Centre de Recherche Cardiovasculaire, Université Paris Descartes, Sorbonne Paris Cité, F-75015 Paris, France
| | - Luca Danelli
- Laboratoire d'Excellence INFLAMEX, Université Paris Diderot, Sorbonne Paris Cité, F-75018 Paris, France INSERM, U1149, F-75018 Paris, France Centre National de la Recherche Scientifique (CNRS) ERL 8252, F-75018 Paris, France
| | - Gregory Gautier
- Laboratoire d'Excellence INFLAMEX, Université Paris Diderot, Sorbonne Paris Cité, F-75018 Paris, France INSERM, U1149, F-75018 Paris, France
| | - Ulrich Blank
- Laboratoire d'Excellence INFLAMEX, Université Paris Diderot, Sorbonne Paris Cité, F-75018 Paris, France INSERM, U1149, F-75018 Paris, France Centre National de la Recherche Scientifique (CNRS) ERL 8252, F-75018 Paris, France
| | - Pierre Launay
- Laboratoire d'Excellence INFLAMEX, Université Paris Diderot, Sorbonne Paris Cité, F-75018 Paris, France INSERM, U1149, F-75018 Paris, France
| | - Eric Camerer
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS-970, Centre de Recherche Cardiovasculaire, Université Paris Descartes, Sorbonne Paris Cité, F-75015 Paris, France
| | - Patrick Bruneval
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS-970, Centre de Recherche Cardiovasculaire, Université Paris Descartes, Sorbonne Paris Cité, F-75015 Paris, France Hôpital European George Pompidou, Assistance Publique Hôpitaux de Paris, F-75015 Paris, France
| | - Philippe Menasche
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS-970, Centre de Recherche Cardiovasculaire, Université Paris Descartes, Sorbonne Paris Cité, F-75015 Paris, France Hôpital European George Pompidou, Assistance Publique Hôpitaux de Paris, F-75015 Paris, France
| | - Christophe Heymes
- INSERM, UMR-1048, Institut des Maladies Métaboliques et Cardiovasculaires, F-31004 Toulouse, France
| | - Elodie Luche
- STROMALab, Etablissement Français du Sang, INSERM U1031, CNRS ERL 5311, Université de Toulouse, F-31004 Toulouse, France
| | - Louis Casteilla
- STROMALab, Etablissement Français du Sang, INSERM U1031, CNRS ERL 5311, Université de Toulouse, F-31004 Toulouse, France
| | - Béatrice Cousin
- STROMALab, Etablissement Français du Sang, INSERM U1031, CNRS ERL 5311, Université de Toulouse, F-31004 Toulouse, France
| | - Hans-Reimer Rodewald
- Division of Cellular Immunology, German Cancer Research Center, D-69120 Heidelberg, Germany
| | - David A Kass
- Division of Cardiology, Johns Hopkins Medical Institutions, Baltimore, MD 212015
| | - Jean-Sébastien Silvestre
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS-970, Centre de Recherche Cardiovasculaire, Université Paris Descartes, Sorbonne Paris Cité, F-75015 Paris, France
| |
Collapse
|
24
|
Role of the immune system in cardiac tissue damage and repair following myocardial infarction. Inflamm Res 2017; 66:739-751. [PMID: 28600668 DOI: 10.1007/s00011-017-1060-4] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 05/17/2017] [Accepted: 06/01/2017] [Indexed: 12/24/2022] Open
Abstract
INTRODUCTION The immune system plays a crucial role in the initiation, development, and resolution of inflammation following myocardial infarction (MI). The lack of oxygen and nutrients causes the death of cardiomyocytes and leads to the exposure of danger-associated molecular patterns that are recognized by the immune system to initiate inflammation. RESULTS At the initial stage of post-MI inflammation, the immune system further damages cardiac tissue to clear cell debris. The excessive production of reactive oxygen species (ROS) by immune cells and the inability of the anti-oxidant system to neutralize ROS cause oxidative stress that further aggravates inflammation. On the other hand, the cells of both innate and adaptive immune system and their secreted factors are critically instrumental in the very dynamic and complex processes of regulating inflammation and mediating cardiac repair. CONCLUSIONS It is important to decipher the balance between detrimental and beneficial effects of the immune system in MI. This enables us to identify better therapeutic targets for reducing the infarct size, sustaining the cardiac function, and minimizing the likelihood of heart failure. This review discusses the role of both innate and adaptive immune systems in cardiac tissue damage and repair in experimental models of MI.
Collapse
|
25
|
Sage AP, Mallat Z. Readapting the adaptive immune response - therapeutic strategies for atherosclerosis. Br J Pharmacol 2017; 174:3926-3939. [PMID: 28052311 DOI: 10.1111/bph.13700] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 12/13/2016] [Accepted: 12/20/2016] [Indexed: 02/06/2023] Open
Abstract
Cardiovascular diseases remain a major global health issue, with the development of atherosclerosis as a major underlying cause. Our treatment of cardiovascular disease has improved greatly over the past three decades, but much remains to be done reduce disease burden. Current priorities include reducing atherosclerosis advancement to clinically significant stages and preventing plaque rupture or erosion. Inflammation and involvement of the adaptive immune system influences all these aspects and therefore is one focus for future therapeutic development. The atherosclerotic vascular wall is now recognized to be invaded from both sides (arterial lumen and adventitia), for better or worse, by the adaptive immune system. Atherosclerosis is also affected at several stages by adaptive immune responses, overall providing many opportunities to target these responses and to reduce disease progression. Protective influences that may be defective in diseased individuals include humoral responses to modified LDL and regulatory T cell responses. There are many strategies in development to boost these pathways in humans, including vaccine-based therapies. The effects of various existing adaptive immune targeting therapies, such as blocking critical co-stimulatory pathways or B cell depletion, on cardiovascular disease are beginning to emerge with important consequences for both autoimmune disease patients and the potential for wider use of such therapies. Entering the translation phase for adaptive immune targeting therapies is an exciting and promising prospect. LINKED ARTICLES This article is part of a themed section on Targeting Inflammation to Reduce Cardiovascular Disease Risk. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.22/issuetoc and http://onlinelibrary.wiley.com/doi/10.1111/bcp.v82.4/issuetoc.
Collapse
Affiliation(s)
- Andrew P Sage
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Ziad Mallat
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge, UK.,INSERM U970, Paris Cardiovascular Research Center, Paris, France, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| |
Collapse
|
26
|
Wu J, Grassia G, Cambrook H, Ialenti A, MacRitchie N, Carberry J, Wadsworth RM, Lawrence C, Kennedy S, Maffia P. Perivascular mast cells regulate vein graft neointimal formation and remodeling. PeerJ 2015; 3:e1192. [PMID: 26312183 PMCID: PMC4548472 DOI: 10.7717/peerj.1192] [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] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Accepted: 07/26/2015] [Indexed: 01/26/2023] Open
Abstract
Objective. Emerging evidence suggests an important role for mast cells in vein graft failure. This study addressed the hypothesis that perivascular mast cells regulate in situ vascular inflammatory and proliferative responses and subsequent vein graft neointimal lesion formation, using an optimized local mast cell reconstitution method. Methods and Results. Neointimal hyperplasia was induced by insertion of a vein graft into the right carotid artery in wild type and mast cell deficient KitW−sh/W−sh mice. In some experiments, mast cells were reconstituted systemically (tail vein injection of bone marrow-derived mast cells) or locally (directly into the right neck area) prior to vein grafting. Vein graft neointimal lesion formation was significantly (P < 0.05) reduced in KitW−sh/W−sh mice. Mast cell deficiency reduced the number of proliferating cells, and inhibited L-selectin, CCL2, M-CSF and MIP-3α expression in the vein grafts. Local but not systemic mast cell reconstitution restored a perivascular mast cell population that subsequently promoted neointimal formation in mast cell deficient mice. Conclusion. Our data demonstrate that perivascular mast cells play a key role in promoting neointima formation by inducing local acute inflammatory and proliferative responses. These results suggest that ex vivo intraoperative targeting of mast cells may have therapeutic potential for the prevention of pathological vein graft remodeling.
Collapse
Affiliation(s)
- Junxi Wu
- Strathclyde Institute of Pharmacy & Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| | - Gianluca Grassia
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, United Kingdom.,Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Helen Cambrook
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, United Kingdom
| | - Armando Ialenti
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Neil MacRitchie
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, United Kingdom
| | - Jaclyn Carberry
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, United Kingdom
| | - Roger M Wadsworth
- Strathclyde Institute of Pharmacy & Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| | - Catherine Lawrence
- Strathclyde Institute of Pharmacy & Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| | - Simon Kennedy
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, United Kingdom
| | - Pasquale Maffia
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, United Kingdom.,Department of Pharmacy, University of Naples Federico II, Naples, Italy.,Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, United Kingdom
| |
Collapse
|