1
|
Duan X, Zhang L, Liu K, Guo K, You Y, Jia H, Zhou S, Han B. Macrophage-derived SPP1 exacerbate myocardial injury by interacting with fibroblasts in viral myocarditis. Biol Direct 2025; 20:30. [PMID: 40087693 PMCID: PMC11907792 DOI: 10.1186/s13062-025-00621-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 02/22/2025] [Indexed: 03/17/2025] Open
Abstract
BACKGROUND Viral myocarditis (VMC) is an inflammatory myocardial condition triggered by viral infections which involves pathogenic-related damage and immune-mediated damage. However, the precise immunopathogenic mechanisms underlying VMC remain elusive. METHODS We performed single-cell RNA sequencing on mouse hearts during the acute phase of CVB3-induced VMC. After manually annotating cell types, functional analyses of macrophage were performed by cell ratio changes, customized gene set module scoring and CellPhoneDB. Utilizing indirect co-culture experiments in vitro, the effects of macrophage-derived SPP1 on cardiac fibroblasts were investigated. Depletion of macrophages and inhibition of SPP1 expression in mice were carried out to study the effects of macrophage-derived SPP1 on cardiac function, inflammation levels, and myocardial injury in mice with VMC. RESULTS Our data revealed that macrophages are the major immune cells which infiltrate the heart during the acute phase of VMC, particularly a macrophage subpopulation which highly expresses Spp1 (Spp1+ macrophages) and exhibited characteristics of peripheral blood monocytes. Spp1+ macrophages communicate extensively with fibroblasts during VMC, and that SPP1 promotes fibroblast conversion to an inflammatory phenotype with high Ccl2/Ccl7 expression. This in turn increases monocyte chemotaxis to the heart. Besides, a partial depletion of macrophages in the early stages of VMC attenuated myocardial inflammation and myocardial injury in mice. Inhibition of SPP1 reduced cardiac macrophage infiltration, attenuated myocardial inflammation, and improved cardiac function in VMC mice. CONCLUSION Our findings suggested that Spp1+ macrophages could self-recruit, and macrophage-derived SPP1 exacerbated myocardial immune injury by promoting high Ccl2/Ccl7 expression in fibroblasts. Our study advances understandings of VMC pathogenesis, and provides novel insight into potential immunotherapies for VMC.
Collapse
Affiliation(s)
- Xiuyun Duan
- Department of Pediatric Cardiology, Cheeloo Colledge of Medicine, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
| | - Li Zhang
- Department of Pediatric Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- Shandong Provincial Clinical Research Center for Children's Health and Disease office, Shandong Provincial Hospital, Jinan, Shandong, China
- Medical Science and Technology Innovation Center, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Keyu Liu
- Department of Pediatric Cardiology, Cheeloo Colledge of Medicine, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
- Medical Science and Technology Innovation Center, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Kaiyin Guo
- Rui Jin Hospital Nanxiang Branch, Shanghai, 201802, China
| | - Yingnan You
- Department of Pediatric Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- Shandong Provincial Clinical Research Center for Children's Health and Disease office, Shandong Provincial Hospital, Jinan, Shandong, China
- Medical Science and Technology Innovation Center, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Hailin Jia
- Department of Pediatric Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- Shandong Provincial Clinical Research Center for Children's Health and Disease office, Shandong Provincial Hospital, Jinan, Shandong, China
- Medical Science and Technology Innovation Center, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Shan Zhou
- Department of Pediatric Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- Shandong Provincial Clinical Research Center for Children's Health and Disease office, Shandong Provincial Hospital, Jinan, Shandong, China
- Medical Science and Technology Innovation Center, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Bo Han
- Department of Pediatric Cardiology, Cheeloo Colledge of Medicine, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China.
- Department of Pediatric Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
- Shandong Provincial Clinical Research Center for Children's Health and Disease office, Shandong Provincial Hospital, Jinan, Shandong, China.
- Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China.
- Medical Science and Technology Innovation Center, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, China.
| |
Collapse
|
2
|
Li R, Li X, Zhang X, Yu J, Li Y, Ran S, Wang S, Luo Z, Zhao J, Hao Y, Zong J, Zheng K, Lai L, Zhang H, Huang P, Zhou C, Wu J, Ye W, Xia J. Macrophages in Cardiovascular Fibrosis: Novel Subpopulations, Molecular Mechanisms, and Therapeutic Targets. Can J Cardiol 2025; 41:309-322. [PMID: 39580052 DOI: 10.1016/j.cjca.2024.11.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 11/12/2024] [Accepted: 11/18/2024] [Indexed: 11/25/2024] Open
Abstract
Cardiovascular fibrosis is a common pathological process that contributes to the development and progression of various cardiovascular diseases. Despite being widely believed to be an irreversible and relentless process, preclinical models and clinical trials have shown that cardiovascular fibrosis is an extremely dynamic process. Additionally, as part of the innate immune system, macrophages are heterogeneous cells that are pivotal in tissue regeneration and fibrosis. They participate in fibroblast activation, extracellular matrix remodelling, and the regression of fibrosis. Although we have made some advances in understanding macrophages in cardiovascular fibrosis, a gap still remains between their identification and conversion into effective treatments. Moreover, the traditional M1-M2 paradigm faces many challenges because it does not sufficiently clarify macrophage diversity and their functions. Exploring novel macrophage-based therapies is urgent for cardiovascular fibrosis treatment. Single-cell techniques have shed light on identifying novel subpopulations that differ in function and molecular signature under steady-state and pathological conditions. In this review, we outline the developmental origins of macrophages, which underlie their functions; and recent technology development in the single-cell era. In addition, we describe the markers and mediators of the newly defined macrophage subpopulations and the molecular mechanisms involved to elucidate potential approaches for targeting macrophages in cardiovascular fibrosis.
Collapse
Affiliation(s)
- Ran Li
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaohan Li
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xi Zhang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jizhang Yu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuan Li
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuan Ran
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Song Wang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zilong Luo
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiulu Zhao
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yanglin Hao
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Junjie Zong
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kexiao Zheng
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Longyong Lai
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Han Zhang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Pinyan Huang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Cheng Zhou
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jie Wu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Weicong Ye
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Jiahong Xia
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China.
| |
Collapse
|
3
|
Li Q, Xie H, Guo Y, Li J, Qian J, Wu W. Formoterol Reduces the Pro-Inflammatory Phenotype by Enhancing the Activity of Glutaminase in Monocyte-Derived Macrophages in the CVB3-Induced Viral Myocarditis. Immun Inflamm Dis 2024; 12:e70073. [PMID: 39601476 PMCID: PMC11600452 DOI: 10.1002/iid3.70073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 08/13/2024] [Accepted: 11/06/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND Viral myocarditis (VMC) plays a significant role in heart failure, and there is currently a shortage of available targeted treatments. Macrophage phenotype and function are closely associated with the beta-2 adrenergic receptor (β2-AR). METHOD This research employed a BALB/c mouse model of VMC generated using Coxsackievirus B3 (CVB3), and the β2-AR agonist formoterol was administered as treatment. A bioinformatic analysis was conducted to identify the β2-AR in CCR2+MHCIIhigh monocyte-derived macrophages (MoMFs). Echocardiography and histopathological assessments were utilized to evaluate cardiac function and inflammation. The enzymatic activity of glutaminase (GLS) was quantified. Flow cytometry was employed to characterize the phenotype and function of the macrophages. RESULT Our study revealed that formoterol treatment effectively mitigated cardiac inflammation and fibrosis, improved cardiac function, and prolonged survival compared to the VMC group. Formoterol reduced the infiltration of CCR2+MHCIIhigh MoMFs in the heart, inhibited M1 phenotypic expression and activity, and reduced the percentage of Ly6Chigh monocytes in circulation. Additionally, formoterol stimulated M2 phenotypic expression and activity and increased the percentage of Ly6Clow monocytes in circulation. Additionally, the combination of NICB3344, a C-C motif chemokine receptor 2 inhibitor, with formoterol did not exhibit synergistic effects on reducing cardiac pathological scores or enhancing cardiac function. In vitro studies involving the use of lipopolysaccharide (LPS)-induced bone marrow-derived macrophages, revealed the ability of formoterol to suppress the M1 phenotype and functions induced by LPS while promoting the M2 phenotype and functions. Nevertheless, the observed effects were negated by the introduction of the GLS inhibitor BPTES. CONCLUSION Formoterol potentially serves as a significant metabolic regulator in the differentiation process of cardiac MoMFs, influencing this process by controlling GLS activity. Targeting β2-AR exhibits potential as an effective approach for managing VMC. It is essential to acknowledge that these findings were derived under specific experimental conditions, with the current conclusions predominantly based on animal models. Future research is necessary to further investigate the feasibility of formoterol in clinical practice.
Collapse
Affiliation(s)
- Quan‐liang Li
- Department of CardiologyThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Hua‐bao Xie
- Department of CardiologyThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Ying‐xin Guo
- Department of CardiologyThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Juan‐fen Li
- Department of CardiologyThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Jing Qian
- Department of CardiologyThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Wei‐Feng Wu
- Department of CardiologyThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co‐constructed by the Province and Ministry, Guangxi Medical UniversityNanningChina
| |
Collapse
|
4
|
Liu Q, Shang Y, Tao Z, Li X, Shen L, Zhang H, Liu Z, Rao Z, Yu X, Cao Y, Zeng L, Huang X. Coxsackievirus group B3 regulates ASS1-mediated metabolic reprogramming and promotes macrophage inflammatory polarization in viral myocarditis. J Virol 2024; 98:e0080524. [PMID: 39194244 PMCID: PMC11406948 DOI: 10.1128/jvi.00805-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 07/26/2024] [Indexed: 08/29/2024] Open
Abstract
Coxsackievirus group B3 (CVB3) belongs to the genus Enteroviruses of the family Picornaviridae and is the main pathogen underlying viral myocarditis (VMC). No specific therapeutic is available for this condition. Argininosuccinate synthase 1 (ASS1) is a key enzyme in the urea cycle that converts citrulline and aspartic acid to argininosuccinate. Here, we found that CVB3 and its capsid protein VP2 inhibit the autophagic degradation of ASS1 and that CVB3 consumes citrulline to upregulate ASS1, triggers urea cycle metabolic reprogramming, and then activates macrophages to develop pro-inflammatory polarization, thereby promoting the occurrence and development of VMC. Conversely, citrulline supplementation to prevent depletion can downregulate ASS1, rescue macrophage polarization, and alleviate the pathogenicity of VMC. These findings provide a new perspective on the occurrence and development of VMC, revealing ASS1 as a potential new target for treating this disease. IMPORTANCE Viral myocarditis (VMC) is a common and potentially life-threatening myocardial inflammatory disease, most commonly caused by CVB3 infection. So far, the pathogenesis of VMC caused by CVB3 is mainly focused on two aspects: one is the direct myocardial injury caused by a large number of viral replication in the early stage of infection, and the other is the local immune cell infiltration and inflammatory damage of the myocardium in the adaptive immune response stage. There are few studies on the early innate immunity of CVB3 infection in myocardial tissue, but the appearance of macrophages in the early stage of CVB3 infection suggests that they can play a regulatory role as early innate immune response cells in myocardial tissue. Here, we discovered a possible new mechanism of VMC caused by CVB3, revealed new drug targets for anti-CVB3, and discovered the therapeutic potential of citrulline for VMC.
Collapse
Affiliation(s)
- Qiong Liu
- The First Affiliated Hospital of Nanchang University and School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Yinpan Shang
- The First Affiliated Hospital of Nanchang University and School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Ziwei Tao
- The First Affiliated Hospital of Nanchang University and School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Xuan Li
- The First Affiliated Hospital of Nanchang University and School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Lu Shen
- The First Affiliated Hospital of Nanchang University and School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Hanchi Zhang
- The First Affiliated Hospital of Nanchang University and School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
- The Second Clinical Medical College, Nanchang University, Nanchang, China
| | - Zhili Liu
- The First Affiliated Hospital of Nanchang University and School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
- HuanKui Academy, Nanchang University, Nanchang, China
| | - Zhirong Rao
- The First Affiliated Hospital of Nanchang University and School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
- HuanKui Academy, Nanchang University, Nanchang, China
| | - Xiaomin Yu
- The First Affiliated Hospital of Nanchang University and School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Yanli Cao
- The First Affiliated Hospital of Nanchang University and School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Lingbing Zeng
- The First Affiliated Hospital of Nanchang University and School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
- The First Affiliated Hospital of Nanchang University, School of Public Health, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Xiaotian Huang
- The First Affiliated Hospital of Nanchang University and School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
| |
Collapse
|
5
|
Mohamud Y, Lin JC, Hwang SW, Bahreyni A, Wang ZC, Luo H. Coxsackievirus B3 Activates Macrophages Independently of CAR-Mediated Viral Entry. Viruses 2024; 16:1456. [PMID: 39339932 PMCID: PMC11437450 DOI: 10.3390/v16091456] [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: 08/23/2024] [Revised: 09/09/2024] [Accepted: 09/11/2024] [Indexed: 09/30/2024] Open
Abstract
Enteroviruses are a genus of small RNA viruses that are responsible for approximately one billion global infections annually. These infections range in severity from the common cold and flu-like symptoms to more severe diseases, such as viral myocarditis, pancreatitis, and neurological disorders, that continue to pose a global health challenge with limited therapeutic strategies currently available. In the current study, we sought to understand the interaction between coxsackievirus B3 (CVB3), which is a model enterovirus, and macrophage cells, as there is limited understanding of how this virus interacts with macrophage innate immune cells. Our study demonstrated that CVB3 can robustly activate macrophages without apparent viral replication in these cells. We also showed that myeloid cells lacked the viral entry receptor coxsackievirus and adenovirus receptor (CAR). However, the expression of exogenous CAR in RAW264.7 macrophages was unable to overcome the viral replication deficit. Interestingly, the CAR expression was associated with altered inflammatory responses during prolonged infection. Additionally, we identified the autophagy protein LC3 as a novel stimulus for macrophage activation. These findings provide new insights into the mechanisms of CVB3-induced macrophage activation and its implications for viral pathogenesis.
Collapse
Affiliation(s)
- Yasir Mohamud
- Centre for Heart Lung Innovation, University of British Columbia, St. Paul's Hospital, Vancouver, BC V6Z 1Y6, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6Z 1Y6, Canada
| | - Jingfei Carly Lin
- Centre for Heart Lung Innovation, University of British Columbia, St. Paul's Hospital, Vancouver, BC V6Z 1Y6, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6Z 1Y6, Canada
| | - Sinwoo Wendy Hwang
- Centre for Heart Lung Innovation, University of British Columbia, St. Paul's Hospital, Vancouver, BC V6Z 1Y6, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6Z 1Y6, Canada
| | - Amirhossein Bahreyni
- Centre for Heart Lung Innovation, University of British Columbia, St. Paul's Hospital, Vancouver, BC V6Z 1Y6, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6Z 1Y6, Canada
| | - Zhihan Claire Wang
- Centre for Heart Lung Innovation, University of British Columbia, St. Paul's Hospital, Vancouver, BC V6Z 1Y6, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6Z 1Y6, Canada
| | - Honglin Luo
- Centre for Heart Lung Innovation, University of British Columbia, St. Paul's Hospital, Vancouver, BC V6Z 1Y6, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6Z 1Y6, Canada
| |
Collapse
|
6
|
Favere K, Van Hecke M, Eens S, Bosman M, Stobbelaar K, Hotterbeekx A, Kumar-Singh S, L Delputte P, Fransen E, De Sutter J, Guns PJ, Roskams T, Heidbuchel H. The natural history of CVB3 myocarditis in C57BL/6J mice: an extended in-depth characterization. Cardiovasc Pathol 2024; 72:107652. [PMID: 38750778 DOI: 10.1016/j.carpath.2024.107652] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 05/03/2024] [Accepted: 05/06/2024] [Indexed: 06/24/2024] Open
Abstract
BACKGROUND AND AIMS Viral infections are the leading cause of myocarditis. Besides acute cardiac complications, late-stage sequelae such as myocardial fibrosis may develop, importantly impacting the prognosis. Coxsackievirus B3 (CVB)-induced myocarditis in mice is the most commonly used translational model to study viral myocarditis and has provided the majority of our current understanding of the disease pathophysiology. Nevertheless, the late stages of disease, encompassing fibrogenesis and arrhythmogenesis, have been underappreciated in viral myocarditis research to date. The present study investigated the natural history of CVB-induced myocarditis in C57BL/6J mice, expanding the focus beyond the acute phase of disease. In addition, we studied the impact of sex and inoculation dose on the disease course. METHODS AND RESULTS C57BL/6J mice (12 weeks old; n=154) received a single intraperitoneal injection with CVB to induce viral myocarditis, or vehicle (PBS) as control. Male mice (n=92) were injected with 5 × 105 (regular dose) (RD) or 5 × 106 (high dose) (HD) plaque-forming units of CVB, whereas female mice received the RD only. Animals were sacrificed 1, 2, 4, 8, and 11 weeks after CVB or PBS injection. Virally inoculated mice developed viral disease with a temporary decline in general condition and weight loss, which was less pronounced in female animals (P<.001). In male CVB mice, premature mortality occurred between days 8 and 23 after inoculation (RD: 21%, HD: 20%), whereas all female animals survived. Over the course of disease, cardiac inflammation progressively subsided, with faster resolution in female mice. There were no substantial group differences in the composition of the inflammatory cell infiltrates: predominance of cytotoxic T cells at day 7 and 14, and a switch from arginase1-reactive macrophages to iNOS-reactive macrophages from day 7 to 14 were the main findings. There was concomitant development and maturation of different patterns of myocardial fibrosis, with enhanced fibrogenesis in male mice. Virus was almost completely cleared from the heart by day 14. Serum biomarkers of cardiac damage and cardiac expression of remodeling genes were temporarily elevated during the acute phase of disease. Cardiac CTGF gene upregulation was less prolonged in female CVB animals. In vivo electrophysiology studies at weeks 8 and 11 demonstrated that under baseline conditions (i.e. in the absence of proarrhythmogenic drugs), ventricular arrhythmias could only be induced in CVB animals. The cumulative arrhythmia burden throughout the entire stimulation protocol was not significantly different between CVB and control groups. CONCLUSION CVB inoculation in C57BL/6J mice represents a model of acute self-limiting viral myocarditis, with progression to different patterns of myocardial fibrosis. Sex, but not inoculation dose, seems to modulate the course of disease.
Collapse
Affiliation(s)
- Kasper Favere
- Laboratory of Physiopharmacology, GENCOR, University of Antwerp, 2610 Antwerp, Belgium; Research Group Cardiovascular Diseases, GENCOR, University of Antwerp, 2610 Antwerp, Belgium; Department of Cardiology, Antwerp University Hospital, 2650 Antwerp, Belgium; Department of Internal Medicine, Ghent University, 9000 Ghent, Belgium.
| | - Manon Van Hecke
- Translational Cell & Tissue Research, Department of Imaging & Pathology, University of Leuven, 3000 Leuven, Belgium
| | - Sander Eens
- Laboratory of Physiopharmacology, GENCOR, University of Antwerp, 2610 Antwerp, Belgium; Research Group Cardiovascular Diseases, GENCOR, University of Antwerp, 2610 Antwerp, Belgium
| | - Matthias Bosman
- Laboratory of Physiopharmacology, GENCOR, University of Antwerp, 2610 Antwerp, Belgium
| | - Kim Stobbelaar
- Laboratory of Microbiology, Parasitology and Hygiene, University of Antwerp, 2610 Antwerp, Belgium
| | - An Hotterbeekx
- Molecular Pathology Group, FGGW-Laboratory of Cell Biology and Histology, University of Antwerp, 2610 Antwerp, Belgium
| | - Samir Kumar-Singh
- Molecular Pathology Group, FGGW-Laboratory of Cell Biology and Histology, University of Antwerp, 2610 Antwerp, Belgium
| | - Peter L Delputte
- Laboratory of Microbiology, Parasitology and Hygiene, University of Antwerp, 2610 Antwerp, Belgium
| | - Erik Fransen
- Centre for Medical Genetics, University of Antwerp, 2610 Antwerp, Belgium
| | - Johan De Sutter
- Department of Internal Medicine, Ghent University, 9000 Ghent, Belgium
| | - Pieter-Jan Guns
- Laboratory of Physiopharmacology, GENCOR, University of Antwerp, 2610 Antwerp, Belgium
| | - Tania Roskams
- Translational Cell & Tissue Research, Department of Imaging & Pathology, University of Leuven, 3000 Leuven, Belgium
| | - Hein Heidbuchel
- Research Group Cardiovascular Diseases, GENCOR, University of Antwerp, 2610 Antwerp, Belgium; Department of Cardiology, Antwerp University Hospital, 2650 Antwerp, Belgium
| |
Collapse
|
7
|
Wei J, Wang D, Cui C, Tan J, Peng M, Lu H. CXCL4/CXCR3 axis regulates cardiac fibrosis by activating TGF-β1/Smad2/3 signaling in mouse viral myocarditis. Immun Inflamm Dis 2024; 12:e1237. [PMID: 38577984 PMCID: PMC10996374 DOI: 10.1002/iid3.1237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 03/06/2024] [Accepted: 03/14/2024] [Indexed: 04/06/2024] Open
Abstract
BACKGROUND Severe myocarditis is often accompanied by cardiac fibrosis, but the underlying mechanism has not been fully elucidated. CXCL4 is a chemokine that has been reported to have pro-inflammatory and profibrotic functions. The exact role of CXCL4 in cardiac fibrosis remains unclear. METHODS Viral myocarditis (VMC) models were induced by intraperitoneal injection of Coxsackie B Type 3 (CVB3). In vivo, CVB3 (100 TCID50) and CVB3-AMG487 (CVB3: 100 TCID50; AMG487: 5 mg/kg) combination were administered in the VMC and VMC+AMG487 groups, respectively. Hematoxylin and eosin staining, severity score, Masson staining, and immunofluorescence staining were performed to measure myocardial morphology in VMC. Enzyme-linked immunosorbent assay (ELISA) and quantitative reverse transcription polymerase chain reaction (qRT-PCR) were performed to quantify inflammatory factors (IL-1β, IL-6, TNF-α, and CXCL4). Aspartate aminotransferase (AST), lactate dehydrogenase (LDH), and creatine kinase-myocardial band (CK-MB) levels were analyzed by commercial kits. CXCL4, CXCR3B, α-SMA, TGF-β1, Collagen I, and Collagen III were determined by Western blot and immunofluorescence staining. RESULTS In vivo, CVB3-AMG487 reduced cardiac injury, α-SMA, Collagen I and Collagen III levels, and collagen deposition in VMC+AMG487 group. Additionally, compared with VMC group, VMC+AMG group decreased the levels of inflammatory factors (IL-1β, IL-6, and TNF-α). In vitro, CXCL4/CXCR3B axis activation TGF-β1/Smad2/3 pathway promote mice cardiac fibroblasts differentiation. CONCLUSION CXCL4 acts as a profibrotic factor in TGF-β1/Smad2/3 pathway-induced cardiac fibroblast activation and ECM synthesis, and eventually progresses to cardiac fibrosis. Therefore, our findings revealed the role of CXCL4 in VMC and unveiled its underlying mechanism. CXCL4 appears to be a potential target for the treatment of VMC.
Collapse
Affiliation(s)
- Jing Wei
- Department of Laboratory Medicine, Nanjing First HospitalNanjing Medical UniversityNanjingChina
| | - Dan‐feng Wang
- Department of Laboratory MedicineJiangning Hospital Affiliated to Nanjing Medical UniversityNanjingChina
| | - Cong‐cong Cui
- Department of Laboratory MedicineJiangning Hospital Affiliated to Nanjing Medical UniversityNanjingChina
| | - Jia‐jia Tan
- Department of Laboratory MedicineJiangning Hospital Affiliated to Nanjing Medical UniversityNanjingChina
| | - Ming‐yu Peng
- Department of Laboratory MedicineJiangning Hospital Affiliated to Nanjing Medical UniversityNanjingChina
| | - Hong‐xiang Lu
- Department of Laboratory Medicine, Nanjing First HospitalNanjing Medical UniversityNanjingChina
- Department of Laboratory MedicineJiangning Hospital Affiliated to Nanjing Medical UniversityNanjingChina
| |
Collapse
|
8
|
Liu K, Han B. Role of immune cells in the pathogenesis of myocarditis. J Leukoc Biol 2024; 115:253-275. [PMID: 37949833 DOI: 10.1093/jleuko/qiad143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 10/15/2023] [Accepted: 10/24/2023] [Indexed: 11/12/2023] Open
Abstract
Myocarditis is an inflammatory heart disease that mostly affects young people. Myocarditis involves a complex immune network; however, its detailed pathogenesis is currently unclear. The diversity and plasticity of immune cells, either in the peripheral blood or in the heart, have been partially revealed in a number of previous studies involving patients and several kinds of animal models with myocarditis. It is the complexity of immune cells, rather than one cell type that is the culprit. Thus, recognizing the individual intricacies within immune cells in the context of myocarditis pathogenesis and finding the key intersection of the immune network may help in the diagnosis and treatment of this condition. With the vast amount of cell data gained on myocarditis and the recent application of single-cell sequencing, we summarize the multiple functions of currently recognized key immune cells in the pathogenesis of myocarditis to provide an immune background for subsequent investigations.
Collapse
Affiliation(s)
- Keyu Liu
- Department of Pediatric Cardiology, Shandong Provincial Hospital, Shandong University, Cheeloo Colledge of Medicine, No. 324 Jingwu Road, 250021, Jinan, China
| | - Bo Han
- Department of Pediatric Cardiology, Shandong Provincial Hospital, Shandong University, Cheeloo Colledge of Medicine, No. 324 Jingwu Road, 250021, Jinan, China
- Department of Pediatric Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324 Jingwu Road, 250021, Jinan, China
- Shandong Provincial Hospital, Shandong Provincial Clinical Research Center for Children' s Health and Disease office, No. 324 Jingwu Road, 250021, Jinan, China
| |
Collapse
|
9
|
Seropian IM, Cassaglia P, Miksztowicz V, González GE. Unraveling the role of galectin-3 in cardiac pathology and physiology. Front Physiol 2023; 14:1304735. [PMID: 38170009 PMCID: PMC10759241 DOI: 10.3389/fphys.2023.1304735] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 12/05/2023] [Indexed: 01/05/2024] Open
Abstract
Galectin-3 (Gal-3) is a carbohydrate-binding protein with multiple functions. Gal-3 regulates cell growth, proliferation, and apoptosis by orchestrating cell-cell and cell-matrix interactions. It is implicated in the development and progression of cardiovascular disease, and its expression is increased in patients with heart failure. In atherosclerosis, Gal-3 promotes monocyte recruitment to the arterial wall boosting inflammation and atheroma. In acute myocardial infarction (AMI), the expression of Gal-3 increases in infarcted and remote zones from the beginning of AMI, and plays a critical role in macrophage infiltration, differentiation to M1 phenotype, inflammation and interstitial fibrosis through collagen synthesis. Genetic deficiency of Gal-3 delays wound healing, impairs cardiac remodeling and function after AMI. On the contrary, Gal-3 deficiency shows opposite results with improved remodeling and function in other cardiomyopathies and in hypertension. Pharmacologic inhibition with non-selective inhibitors is also protective in cardiac disease. Finally, we recently showed that Gal-3 participates in normal aging. However, genetic absence of Gal-3 in aged mice exacerbates pathological hypertrophy and increases fibrosis, as opposed to reduced fibrosis shown in cardiac disease. Despite some gaps in understanding its precise mechanisms of action, Gal-3 represents a potential therapeutic target for the treatment of cardiovascular diseases and the management of cardiac aging. In this review, we summarize the current knowledge regarding the role of Gal-3 in the pathophysiology of heart failure, atherosclerosis, hypertension, myocarditis, and ischemic heart disease. Furthermore, we describe the physiological role of Gal-3 in cardiac aging.
Collapse
Affiliation(s)
- Ignacio M. Seropian
- Laboratorio de Patología Cardiovascular Experimental e Hipertensión Arterial, Instituto de Investigaciones Biomédicas (UCA-CONICET), Facultad de Ciencias Médicas Universidad Católica Argentina, Buenos Aires, Argentina
- Servicio de Hemodinamia, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| | - Pablo Cassaglia
- Departamento de Patología, Instituto de Salud Comunitaria, Universidad Nacional de Hurlingham, Buenos Aires, Argentina
| | - Verónica Miksztowicz
- Laboratorio de Patología Cardiovascular Experimental e Hipertensión Arterial, Instituto de Investigaciones Biomédicas (UCA-CONICET), Facultad de Ciencias Médicas Universidad Católica Argentina, Buenos Aires, Argentina
| | - Germán E. González
- Laboratorio de Patología Cardiovascular Experimental e Hipertensión Arterial, Instituto de Investigaciones Biomédicas (UCA-CONICET), Facultad de Ciencias Médicas Universidad Católica Argentina, Buenos Aires, Argentina
- Departamento de Patología, Instituto de Salud Comunitaria, Universidad Nacional de Hurlingham, Buenos Aires, Argentina
| |
Collapse
|
10
|
Zhang C, Li H, Han M. Effect of atrial fibrillation on plasma galectin-3 and soluble CD40 ligand levels in patients with ischemic cardiomyopathy. J Int Med Res 2023; 51:3000605231194457. [PMID: 37656969 PMCID: PMC10585127 DOI: 10.1177/03000605231194457] [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: 02/25/2023] [Accepted: 07/27/2023] [Indexed: 09/03/2023] Open
Abstract
OBJECTIVE To reveal the significance of plasma galectin-3 and soluble CD40 ligand (sCD40L) levels in patients with ischemic cardiomyopathy (ICM) combined with atrial fibrillation. METHODS In this case-control study, the case group comprised 60 patients with ICM combined with atrial fibrillation and the control group comprised patients with ICM without atrial fibrillation. Plasma galectin-3 and sCD40L levels, left atrial diameter (LAD), left ventricular ejection fraction (LVEF), and left ventricular diameter (LVD) were compared. RESULTS The plasma galectin-3 and sCD40L levels, LAD, and LVD were higher and the LVEF was lower in the case than control group. In the case group, the plasma galectin-3 and sCD40L levels were positively correlated with the LAD and LVD but negatively correlated with the LVEF. The area under the receiver operating characteristic curve of the plasma galectin-3 and sCD40L levels in the diagnosis of ICM combined with atrial fibrillation was 0.857 (95% confidence interval, 0.792-0.923) and 0.724 (95% confidence interval, 0.634-0.814), respectively. CONCLUSION The plasma galectin-3 and sCD40L levels are significantly elevated in patients with ICM combined with atrial fibrillation. Although both may have predictive value in the diagnosis of ICM combined with atrial fibrillation, galectin-3 may have the higher predictive value.
Collapse
Affiliation(s)
- Chi Zhang
- Department of Cardiovascular Medicine, The Eighth Hospital of Wuhan, Wuhan City, Hubei Province, China
| | - HongJun Li
- Department of Surgery, The Eighth Hospital of Wuhan, Wuhan City, Hubei Province, China
| | - Mei Han
- Department of Cardiovascular Medicine, The Eighth Hospital of Wuhan, Wuhan City, Hubei Province, China
| |
Collapse
|
11
|
Coulis G, Jaime D, Guerrero-Juarez C, Kastenschmidt JM, Farahat PK, Nguyen Q, Pervolarakis N, McLinden K, Thurlow L, Movahedi S, Hughes BS, Duarte J, Sorn A, Montoya E, Mozaffar I, Dragan M, Othy S, Joshi T, Hans CP, Kimonis V, MacLean AL, Nie Q, Wallace LM, Harper SQ, Mozaffar T, Hogarth MW, Bhattacharya S, Jaiswal JK, Golann DR, Su Q, Kessenbrock K, Stec M, Spencer MJ, Zamudio JR, Villalta SA. Single-cell and spatial transcriptomics identify a macrophage population associated with skeletal muscle fibrosis. SCIENCE ADVANCES 2023; 9:eadd9984. [PMID: 37418531 PMCID: PMC10328414 DOI: 10.1126/sciadv.add9984] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 06/05/2023] [Indexed: 07/09/2023]
Abstract
Macrophages are essential for skeletal muscle homeostasis, but how their dysregulation contributes to the development of fibrosis in muscle disease remains unclear. Here, we used single-cell transcriptomics to determine the molecular attributes of dystrophic and healthy muscle macrophages. We identified six clusters and unexpectedly found that none corresponded to traditional definitions of M1 or M2 macrophages. Rather, the predominant macrophage signature in dystrophic muscle was characterized by high expression of fibrotic factors, galectin-3 (gal-3) and osteopontin (Spp1). Spatial transcriptomics, computational inferences of intercellular communication, and in vitro assays indicated that macrophage-derived Spp1 regulates stromal progenitor differentiation. Gal-3+ macrophages were chronically activated in dystrophic muscle, and adoptive transfer assays showed that the gal-3+ phenotype was the dominant molecular program induced within the dystrophic milieu. Gal-3+ macrophages were also elevated in multiple human myopathies. These studies advance our understanding of macrophages in muscular dystrophy by defining their transcriptional programs and reveal Spp1 as a major regulator of macrophage and stromal progenitor interactions.
Collapse
Affiliation(s)
- Gerald Coulis
- Department of Physiology and Biophysics, University of California Irvine, Irvine, CA, USA
- Institute for Immunology, University of California Irvine, Irvine, CA, USA
| | - Diego Jaime
- Department of Physiology and Biophysics, University of California Irvine, Irvine, CA, USA
- Institute for Immunology, University of California Irvine, Irvine, CA, USA
| | | | - Jenna M. Kastenschmidt
- Department of Physiology and Biophysics, University of California Irvine, Irvine, CA, USA
- Institute for Immunology, University of California Irvine, Irvine, CA, USA
| | - Philip K. Farahat
- Department of Physiology and Biophysics, University of California Irvine, Irvine, CA, USA
- Institute for Immunology, University of California Irvine, Irvine, CA, USA
| | - Quy Nguyen
- Department of Biological Chemistry, University of California Irvine, Irvine, CA USA
| | | | - Katherine McLinden
- Department of Molecular Cell and Developmental Biology, University of California Los Angeles, Los Angeles, CA, USA
| | - Lauren Thurlow
- Department of Molecular Cell and Developmental Biology, University of California Los Angeles, Los Angeles, CA, USA
| | - Saba Movahedi
- Department of Physiology and Biophysics, University of California Irvine, Irvine, CA, USA
| | - Brandon S. Hughes
- Department of Physiology and Biophysics, University of California Irvine, Irvine, CA, USA
| | - Jorge Duarte
- Department of Physiology and Biophysics, University of California Irvine, Irvine, CA, USA
| | - Andrew Sorn
- Department of Physiology and Biophysics, University of California Irvine, Irvine, CA, USA
| | - Elizabeth Montoya
- Department of Physiology and Biophysics, University of California Irvine, Irvine, CA, USA
| | - Izza Mozaffar
- Department of Physiology and Biophysics, University of California Irvine, Irvine, CA, USA
| | - Morgan Dragan
- Department of Biological Chemistry, University of California Irvine, Irvine, CA USA
| | - Shivashankar Othy
- Department of Physiology and Biophysics, University of California Irvine, Irvine, CA, USA
- Institute for Immunology, University of California Irvine, Irvine, CA, USA
| | - Trupti Joshi
- Department of Health Management and Informatics, University of Missouri, Columbia, MO, USA
| | - Chetan P. Hans
- Department of Cardiovascular Medicine, University of Missouri, Columbia, MO USA
| | - Virginia Kimonis
- Department of Pediatrics, University of California Irvine, Irvine, CA, USA
| | - Adam L. MacLean
- Department of Quantitative and Computational Biology, University of Southern California, Los Angeles, CA, USA
| | - Qing Nie
- Department of Mathematics, Department of Developmental and Cell Biology, University of California Irvine, Irvine, CA, USA
| | - Lindsay M. Wallace
- Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, USA
| | - Scott Q. Harper
- Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, USA
- Department of Pediatrics, The Ohio State University, Columbus, OH, USA
| | - Tahseen Mozaffar
- Department of Neurology, University of California Irvine, Irvine, CA, USA
- Department of Pathology and Laboratory Medicine, University of California Irvine, Irvine, CA, USA
| | - Marshall W. Hogarth
- Children’s National Hospital, Research Center for Genetic Medicine, Washington, DC, USA
| | - Surajit Bhattacharya
- Children’s National Hospital, Research Center for Genetic Medicine, Washington, DC, USA
| | - Jyoti K. Jaiswal
- Children’s National Hospital, Research Center for Genetic Medicine, Washington, DC, USA
| | | | - Qi Su
- Regeneron Pharmaceuticals Inc., Tarrytown, NY, USA
| | - Kai Kessenbrock
- Department of Biological Chemistry, University of California Irvine, Irvine, CA USA
| | - Michael Stec
- Regeneron Pharmaceuticals Inc., Tarrytown, NY, USA
| | - Melissa J. Spencer
- Department of Neurology, University of California Los Angeles, Los Angeles, CA, USA
| | - Jesse R. Zamudio
- Department of Molecular Cell and Developmental Biology, University of California Los Angeles, Los Angeles, CA, USA
| | - S. Armando Villalta
- Department of Physiology and Biophysics, University of California Irvine, Irvine, CA, USA
- Institute for Immunology, University of California Irvine, Irvine, CA, USA
- Department of Neurology, University of California Irvine, Irvine, CA, USA
| |
Collapse
|
12
|
Banerjee D, Tian R, Cai S. The Role of Innate Immune Cells in Cardiac Injury and Repair: A Metabolic Perspective. Curr Cardiol Rep 2023; 25:631-640. [PMID: 37249739 PMCID: PMC10227821 DOI: 10.1007/s11886-023-01897-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/15/2023] [Indexed: 05/31/2023]
Abstract
PURPOSE OF REVIEW Recent technological advances have identified distinct subpopulations and roles of the cardiac innate immune cells, specifically macrophages and neutrophils. Studies on distinct metabolic pathways of macrophage and neutrophil in cardiac injury are expanding. Here, we elaborate on the roles of cardiac macrophages and neutrophils in concomitance with their metabolism in normal and diseased hearts. RECENT FINDINGS Single-cell techniques combined with fate mapping have identified the clusters of innate immune cell subpopulations present in the resting and diseased hearts. We are beginning to know about the presence of cardiac resident macrophages and their functions. Resident macrophages perform cardiac homeostatic roles, whereas infiltrating neutrophils and macrophages contribute to tissue damage during cardiac injury with eventual role in repair. Prior studies show that metabolic pathways regulate the phenotypes of the macrophages and neutrophils during cardiac injury. Profiling the metabolism of the innate immune cells, especially of resident macrophages during chronic and acute cardiac diseases, can further the understanding of cardiac immunometabolism.
Collapse
Affiliation(s)
- Durba Banerjee
- Department of Anesthesiology and Pain Medicine, University of Washington, 850 Republican St., Seattle, WA, 98109, USA
| | - Rong Tian
- Department of Anesthesiology and Pain Medicine, University of Washington, 850 Republican St., Seattle, WA, 98109, USA
| | - Shanshan Cai
- Department of Anesthesiology and Pain Medicine, University of Washington, 850 Republican St., Seattle, WA, 98109, USA.
| |
Collapse
|
13
|
Yip F, Lai B, Yang D. Role of Coxsackievirus B3-Induced Immune Responses in the Transition from Myocarditis to Dilated Cardiomyopathy and Heart Failure. Int J Mol Sci 2023; 24:ijms24097717. [PMID: 37175422 PMCID: PMC10178405 DOI: 10.3390/ijms24097717] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 04/16/2023] [Accepted: 04/20/2023] [Indexed: 05/15/2023] Open
Abstract
Dilated cardiomyopathy (DCM) is a cardiac disease marked by the stretching and thinning of the heart muscle and impaired left ventricular contractile function. While most patients do not develop significant cardiac diseases from myocarditis, disparate immune responses can affect pathological outcomes, including DCM progression. These altered immune responses, which may be caused by genetic variance, can prolong cytotoxicity, induce direct cleavage of host protein, or encourage atypical wound healing responses that result in tissue scarring and impaired mechanical and electrical heart function. However, it is unclear which alterations within host immune profiles are crucial to dictating the outcomes of myocarditis. Coxsackievirus B3 (CVB3) is a well-studied virus that has been identified as a causal agent of myocarditis in various models, along with other viruses such as adenovirus, parvovirus B19, and SARS-CoV-2. This paper takes CVB3 as a pathogenic example to review the recent advances in understanding virus-induced immune responses and differential gene expression that regulates iron, lipid, and glucose metabolic remodeling, the severity of cardiac tissue damage, and the development of DCM and heart failure.
Collapse
Affiliation(s)
- Fione Yip
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 2B5, Canada
- The Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, BC V6Z 1Y6, Canada
| | - Brian Lai
- The Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, BC V6Z 1Y6, Canada
| | - Decheng Yang
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 2B5, Canada
- The Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, BC V6Z 1Y6, Canada
| |
Collapse
|
14
|
Coulis G, Jaime D, Guerrero-Juarez C, Kastenschmidt JM, Farahat PK, Nguyen Q, Pervolarakis N, McLinden K, Thurlow L, Movahedi S, Duarte J, Sorn A, Montoya E, Mozaffar I, Dragan M, Othy S, Joshi T, Hans CP, Kimonis V, MacLean AL, Nie Q, Wallace LM, Harper SQ, Mozaffar T, Hogarth MW, Bhattacharya S, Jaiswal JK, Golann DR, Su Q, Kessenbrock K, Stec M, Spencer MJ, Zamudio JR, Villalta SA. Single-cell and spatial transcriptomics identify a macrophage population associated with skeletal muscle fibrosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.18.537253. [PMID: 37131694 PMCID: PMC10153153 DOI: 10.1101/2023.04.18.537253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
The monocytic/macrophage system is essential for skeletal muscle homeostasis, but its dysregulation contributes to the pathogenesis of muscle degenerative disorders. Despite our increasing knowledge of the role of macrophages in degenerative disease, it still remains unclear how macrophages contribute to muscle fibrosis. Here, we used single-cell transcriptomics to determine the molecular attributes of dystrophic and healthy muscle macrophages. We identified six novel clusters. Unexpectedly, none corresponded to traditional definitions of M1 or M2 macrophage activation. Rather, the predominant macrophage signature in dystrophic muscle was characterized by high expression of fibrotic factors, galectin-3 and spp1. Spatial transcriptomics and computational inferences of intercellular communication indicated that spp1 regulates stromal progenitor and macrophage interactions during muscular dystrophy. Galectin-3 + macrophages were chronically activated in dystrophic muscle and adoptive transfer assays showed that the galectin-3 + phenotype was the dominant molecular program induced within the dystrophic milieu. Histological examination of human muscle biopsies revealed that galectin-3 + macrophages were also elevated in multiple myopathies. These studies advance our understanding of macrophages in muscular dystrophy by defining the transcriptional programs induced in muscle macrophages, and reveal spp1 as a major regulator of macrophage and stromal progenitor interactions.
Collapse
Affiliation(s)
- Gerald Coulis
- Department of Physiology and Biophysics, University of California Irvine, USA
- Institute for Immunology, University of California Irvine, USA
| | - Diego Jaime
- Department of Physiology and Biophysics, University of California Irvine, USA
- Institute for Immunology, University of California Irvine, USA
| | - Christian Guerrero-Juarez
- Department of Mathematics, University of California Irvine, USA
- Department of Developmental and Cell Biology, University of California Irvine, USA
| | - Jenna M. Kastenschmidt
- Department of Physiology and Biophysics, University of California Irvine, USA
- Institute for Immunology, University of California Irvine, USA
| | - Philip K. Farahat
- Department of Physiology and Biophysics, University of California Irvine, USA
- Institute for Immunology, University of California Irvine, USA
| | - Quy Nguyen
- Department of Biological Chemistry, University of California Irvine, USA
| | | | - Katherine McLinden
- Department of Molecular Cell and Developmental Biology, University of California Los Angeles, USA
| | - Lauren Thurlow
- Department of Molecular Cell and Developmental Biology, University of California Los Angeles, USA
| | - Saba Movahedi
- Department of Physiology and Biophysics, University of California Irvine, USA
| | - Jorge Duarte
- Department of Physiology and Biophysics, University of California Irvine, USA
| | - Andrew Sorn
- Department of Physiology and Biophysics, University of California Irvine, USA
| | - Elizabeth Montoya
- Department of Physiology and Biophysics, University of California Irvine, USA
| | - Izza Mozaffar
- Department of Physiology and Biophysics, University of California Irvine, USA
| | - Morgan Dragan
- Department of Molecular Cell and Developmental Biology, University of California Los Angeles, USA
| | - Shivashankar Othy
- Department of Physiology and Biophysics, University of California Irvine, USA
- Institute for Immunology, University of California Irvine, USA
| | - Trupti Joshi
- Department of Health Management and Informatics, University of Missouri, Columbia, USA
| | - Chetan P. Hans
- Department of Cardiovascular Medicine, University of Missouri, Columbia, USA
| | | | - Adam L. MacLean
- Department of Quantitative and Computational Biology, University of Southern California, Los Angeles, USA
| | - Qing Nie
- Department of Mathematics, University of California Irvine, USA
- Department of Developmental and Cell Biology, University of California Irvine, USA
| | - Lindsay M. Wallace
- Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children’s Hospital
| | - Scott Q. Harper
- Department of Pediatrics, The Ohio State University, Columbus, OH, USA
- Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children’s Hospital
| | - Tahseen Mozaffar
- Department of Neurology, University of California Irvine, USA
- Department of Pathology and Laboratory Medicine, University of California Irvine, USA
| | - Marshall W. Hogarth
- Children’s National Hospital, Research Center for Genetic Medicine, Washington, DC, USA
| | - Surajit Bhattacharya
- Children’s National Hospital, Research Center for Genetic Medicine, Washington, DC, USA
| | - Jyoti K. Jaiswal
- Children’s National Hospital, Research Center for Genetic Medicine, Washington, DC, USA
| | | | - Qi Su
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, USA
| | - Kai Kessenbrock
- Department of Biological Chemistry, University of California Irvine, USA
| | - Michael Stec
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, USA
| | | | - Jesse R. Zamudio
- Department of Molecular Cell and Developmental Biology, University of California Los Angeles, USA
| | - S. Armando Villalta
- Department of Physiology and Biophysics, University of California Irvine, USA
- Institute for Immunology, University of California Irvine, USA
- Department of Neurology, University of California Irvine, USA
| |
Collapse
|
15
|
Mansour AA, Krautter F, Zhi Z, Iqbal AJ, Recio C. The interplay of galectins-1, -3, and -9 in the immune-inflammatory response underlying cardiovascular and metabolic disease. Cardiovasc Diabetol 2022; 21:253. [PMID: 36403025 PMCID: PMC9675972 DOI: 10.1186/s12933-022-01690-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 11/08/2022] [Indexed: 11/21/2022] Open
Abstract
Galectins are β-galactoside-binding proteins that bind and crosslink molecules via their sugar moieties, forming signaling and adhesion networks involved in cellular communication, differentiation, migration, and survival. Galectins are expressed ubiquitously across immune cells, and their function varies with their tissue-specific and subcellular location. Particularly galectin-1, -3, and -9 are highly expressed by inflammatory cells and are involved in the modulation of several innate and adaptive immune responses. Modulation in the expression of these proteins accompany major processes in cardiovascular diseases and metabolic disorders, such as atherosclerosis, thrombosis, obesity, and diabetes, making them attractive therapeutic targets. In this review we consider the broad cellular activities ascribed to galectin-1, -3, and -9, highlighting those linked to the progression of different inflammatory driven pathologies in the context of cardiovascular and metabolic disease, to better understand their mechanism of action and provide new insights into the design of novel therapeutic strategies.
Collapse
Affiliation(s)
- Adel Abo Mansour
- Institute of Cardiovascular Sciences (ICVS), College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Franziska Krautter
- Institute of Cardiovascular Sciences (ICVS), College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Zhaogong Zhi
- Institute of Cardiovascular Sciences (ICVS), College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Asif Jilani Iqbal
- Institute of Cardiovascular Sciences (ICVS), College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK.
| | - Carlota Recio
- Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Farmacología Molecular y Traslacional -BIOPharm, Universidad de Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Las Palmas, Spain.
| |
Collapse
|
16
|
Resident cardiac macrophages: Heterogeneity and function in health and disease. Immunity 2022; 55:1549-1563. [PMID: 36103852 DOI: 10.1016/j.immuni.2022.08.009] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 08/09/2022] [Accepted: 08/12/2022] [Indexed: 12/20/2022]
Abstract
Understanding tissue macrophage biology has become challenging in recent years due the ever-increasing complexity in macrophage-subset identification and functional characterization. This is particularly important within the myocardium, as we have come to understand that macrophages play multifaceted roles in cardiac health and disease, and heart disease remains the leading cause of death worldwide. Here, we review recent progress in the field, focusing on resident cardiac macrophage heterogeneity, origins, and functions at steady state and after injury. We stratify resident cardiac macrophage functions by the ability of macrophages to either directly influence cardiac physiology or indirectly influence cardiac physiology through orchestrating multi-cellular communication with cardiomyocytes and stromal and immune populations.
Collapse
|
17
|
Nie X, Fan J, Li H, Wang J, Xie R, Chen C, Wang DW. Identification of Cardiac CircRNAs in Mice With CVB3-Induced Myocarditis. Front Cell Dev Biol 2022; 10:760509. [PMID: 35198554 PMCID: PMC8859109 DOI: 10.3389/fcell.2022.760509] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 01/12/2022] [Indexed: 12/04/2022] Open
Abstract
Background: Viral myocarditis could initiate various immune response to the myocardium, resulting in myocyte damage and subsequent cardiac dysfunction. The expression profile and functions of circRNAs in this process are unknown. Methods: Fulminant myocarditis (FM) and non-FM models were induced by coxsackie B3 virus (CVB3) infection in A/J mice and C57BL/6 mice, respectively. CircRNAs expression profile was identified by RNA-seq. Quantitative RT-PCR, Spearman rank correlation, KEGG pathway, GO analysis, Western blot and flow cytometry were performed for functional analysis. Results: Severer inflammatory cell infiltration and cardiomyocyte necrosis were presented in CVB3-treated A/J mice than those in C57BL/6 mice. The dysregulated circRNAs in both of the mouse strains displayed strong correlation with the immune response, but dysregulated circRNAs in A/J mice were more prone to cardiac dysfunction. KEGG analysis indicated that the target genes of dysregulated circRNAs in A/J mice were mainly involved in viral infection, T cell and B cell receptor signaling pathways, while the target genes of dysregulated circRNAs in C57BL/6 mice were unrelated to immune pathways. Furthermore, knockdown of circArhgap32 that was downregulated in CVB3-treated A/J mice promoted cardiomyocyte apoptosis in vitro. Conclusion: Our data showed that cardiac circRNAs dysregulation is an important characteristic of viral myocarditis.
Collapse
Affiliation(s)
| | | | | | | | | | - Chen Chen
- *Correspondence: Chen Chen, ; Dao Wen Wang,
| | | |
Collapse
|
18
|
Koniari I, Artopoulou E, Velissaris D, Ainslie M, Mplani V, Karavasili G, Kounis N, Tsigkas G. Biomarkers in the clinical management of patients with atrial fibrillation and heart failure. J Geriatr Cardiol 2021; 18:908-951. [PMID: 34908928 PMCID: PMC8648548 DOI: 10.11909/j.issn.1671-5411.2021.11.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Atrial fibrillation (AF) and heart failure (HF) are two cardiovascular diseases with an increasing prevalence worldwide. These conditions share common pathophysiologiesand frequently co-exit. In fact, the occurrence of either condition can 'cause' the development of the other, creating a new patient group that demands different management strategies to that if they occur in isolation. Regardless of the temproral association of the two conditions, their presence is linked with adverse cardiovascular outcomes, increased rate of hospitalizations, and increased economic burden on healthcare systems. The use of low-cost, easily accessible and applicable biomarkers may hasten the correct diagnosis and the effective treatment of AF and HF. Both AF and HF effect multiple physiological pathways and thus a great number of biomarkers can be measured that potentially give the clinician important diagnostic and prognostic information. These will then guide patient centred therapeutic management. The current biomarkers that offer potential for guiding therapy, focus on the physiological pathways of miRNA, myocardial stretch and injury, oxidative stress, inflammation, fibrosis, coagulation and renal impairment. Each of these has different utility in current clinincal practice.
Collapse
Affiliation(s)
- Ioanna Koniari
- Manchester Heart Institute, Manchester University Foundation Trust, Manchester, United Kingdom
| | - Eleni Artopoulou
- Department of Internal Medicine, University Hospital of Patras, Patras, Greece
| | | | - Mark Ainslie
- Manchester Heart Institute, Manchester University Foundation Trust, Manchester, United Kingdom
- Division of Cardiovascular Sciences, University of Manchester
| | - Virginia Mplani
- Department of Cardiology, University Hospital of Patras, Patras, Greece
| | - Georgia Karavasili
- Manchester Heart Institute, Manchester University Foundation Trust, Manchester, United Kingdom
| | - Nicholas Kounis
- Department of Cardiology, University Hospital of Patras, Patras, Greece
| | - Grigorios Tsigkas
- Department of Cardiology, University Hospital of Patras, Patras, Greece
| |
Collapse
|
19
|
Histamine Deficiency Promotes Myofibroblasts Transformation from HDC-Expressing CD11b + Myeloid Cells in Injured Hearts Post Myocardial Infarction. J Cardiovasc Transl Res 2021; 15:621-634. [PMID: 34734351 DOI: 10.1007/s12265-021-10172-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 09/08/2021] [Indexed: 10/19/2022]
Abstract
Myocardial infarction (MI) is a significant contributor to the development of heart failure. Histidine decarboxylase (HDC), the unique enzyme that converts L-histidine to histamine, is highly expressed in CD11b+ immature myeloid cells. However, the relationship between HDC-expressing macrophages and cardiac myofibroblasts remains to be explained. Here, we demonstrate that the GFP (green fluorescent protein)-labeled HDC+CD11b+ myeloid precursors and their descendants could differentiate into fibroblast-like cells in myocardial interstitium. Furthermore, we prove that CD11b+Ly6C+ monocytes/macrophages, but not CD11b+Ly6G+ granulocytes, are identified as the main cellular source for bone marrow-derived myofibroblast transformation, which could be regulated via histamine H1 and H2 receptor-dependent signaling pathways. Using HDC knockout mice, we find that histamine deficiency promotes myofibroblast transformation from Ly6C+ macrophages and cardiac fibrosis partly through upregulating the expression of Krüppel-like factor 5 (KLF5). Taken together, our data uncover a central role of HDC in regulating bone marrow-derived macrophage-to-myofibroblast transformation but also identify a histamine receptor (HR)-KLF5 related signaling pathway that mediates myocardial fibrosis post-MI. CD11b+Ly6C+ monocytes/macrophages are the main cellular source for bone marrow-derived myofibroblast transformation. Histamine inhibits myofibroblasts transformation via H1R and H2R-dependent signaling pathways, and ameliorates cardiac fibrosis partly through upregulating KLF5 expression.
Collapse
|
20
|
Viruses in the Heart: Direct and Indirect Routes to Myocarditis and Heart Failure. Viruses 2021; 13:v13101924. [PMID: 34696354 PMCID: PMC8537553 DOI: 10.3390/v13101924] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 09/09/2021] [Accepted: 09/21/2021] [Indexed: 01/01/2023] Open
Abstract
Viruses are an underappreciated cause of heart failure. Indeed, several types of viral infections carry cardiovascular risks. Understanding shared and unique mechanisms by which each virus compromises heart function is critical to inform on therapeutic interventions. This review describes how the key viruses known to lead to cardiac dysfunction operate. Both direct host-damaging mechanisms and indirect actions on the immune systems are discussed. As viral myocarditis is a key pathologic driver of heart failure in infected individuals, this review also highlights the role of cytokine storms and inflammation in virus-induced cardiomyopathy.
Collapse
|
21
|
Khawaja A, Bromage DI. The innate immune response in myocarditis. Int J Biochem Cell Biol 2021; 134:105973. [PMID: 33831592 DOI: 10.1016/j.biocel.2021.105973] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 03/12/2021] [Accepted: 03/31/2021] [Indexed: 12/14/2022]
Abstract
Acute myocarditis is an inflammatory condition of the heart characterised by cellular injury and the influx of leucocytes, including neutrophils, monocytes, macrophages and lymphocytes. While this response is vital for tissue repair, excessive scar deposition and maladaptive ventricular remodelling can result in a legacy of heart failure. It is increasingly recognised as a clinical phenomenon due, in part, to increased availability of cardiac magnetic resonance imaging in patients presenting with chest pain in the absence of significant coronary artery disease. Emerging epidemiological evidence has associated myocarditis with poor outcomes in the context of left ventricular impairment, and even when the left ventricle is preserved outcomes are less benign than once thought. Despite this, our understanding of the contribution of the inflammatory response to the pathophysiology of acute myocarditis lags behind that of acute myocardial infarction, which is the vanguard cardiovascular condition for inflammation research. We recently reviewed monocyte and macrophage phenotype and function in acute myocardial infarction, concluding that their plasticity and heterogeneity might account for conflicting evidence from attempts to target specific leucocyte subpopulations. Here, we revise our understanding of myocardial inflammation, which is predominantly derived from myocardial infarction research, review experimental evidence for the immune response in acute myocarditis, focusing on innate immunity, and discuss potential future directions for immunotherapy research in acute myocarditis.
Collapse
Affiliation(s)
- Abdullah Khawaja
- School of Cardiovascular Medicine and Sciences, King's College London British Heart Foundation Centre of Excellence, James Black Centre, 125 Coldharbour Lane, London, SE5 9NU, UK
| | - Daniel I Bromage
- School of Cardiovascular Medicine and Sciences, King's College London British Heart Foundation Centre of Excellence, James Black Centre, 125 Coldharbour Lane, London, SE5 9NU, UK.
| |
Collapse
|
22
|
Fernandes F, Moreira CHV, Oliveira LC, Souza-Basqueira M, Ianni BM, Lorenzo CD, Ramires FJA, Nastari L, Cunha-Neto E, Ribeiro AL, Lopes RD, Keating SM, Sabino EC, Mady C. Galectin-3 Associated with Severe Forms and Long-term Mortality in Patients with Chagas Disease. Arq Bras Cardiol 2021; 116:248-256. [PMID: 33656072 PMCID: PMC7909980 DOI: 10.36660/abc.20190403] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 12/27/2019] [Indexed: 11/19/2022] Open
Abstract
Fundamento As características histopatológicas da doença de Chagas (DCC) são: presença de miocardite, destruição das fibras cardíacas e fibrose miocárdica. A Galectina-3 (Gal-3) é um biomarcador envolvido no mecanismo de fibrose e inflamação que pode ser útil para a estratificação de indivíduos com DCC por risco. Objetivos Nosso objetivo foi avaliar se níveis elevados de Gal-3 estão associados a formas graves de cardiomiopatia chagásica (CC) e são preditivos de mortalidade. Métodos Estudamos doadores de sangue (DS) positivos para anti-T. cruzi: não-CC-DS (187 DS sem CC com eletrocardiograma [ECG] e fração de ejeção do ventrículo esquerdo [FEVE] normais); CC-Não-Dis-DS (46 DS com CC e apresentando ECG anormal, mas FEVE normal); e 153 controles negativos correspondentes. Esta amostra foi composta por 97 pacientes com CC grave (CC-Dis). Usamos as correlações de Kruskall-Wallis e Spearman para testar a hipótese de associações, assumindo um p bicaudal <0,05 como significativo. Resultados O nível de Gal-3 foi de 12,3 ng/mL para não-CC-DS, 12,0 ng/mL para CC-Não-Dis-DS, 13,8 ng/mL para controles e 15,4 ng/mL para CC-Dis. FEVE <50 foi associada a níveis mais elevados de Gal-3 (p=0,0001). Em nosso modelo de regressão linear ajustado, encontramos associação entre os níveis de Gal-3 e os parâmetros do ecocardiograma em indivíduos positivos para T. cruzi. Nos pacientes CC-Dis, encontramos uma associação significativa de níveis mais elevados de Gal-3 (≥15,3 ng/mL) e morte ou transplante cardíaco em acompanhamento de cinco anos (Hazard ratio – HR 3,11; IC95% 1,21– 8,04; p=0,019). Conclusões Em pacientes com CC, níveis mais elevados de Gal-3 estiveram significativamente associados a formas graves da doença e maior taxa de mortalidade em longo prazo, o que significa que pode ser um meio efetivo para identificar pacientes de alto risco. (Arq Bras Cardiol. 2021; 116(2):248-256)
Collapse
Affiliation(s)
- Fábio Fernandes
- Universidade de São Paulo Faculdade de Medicina Hospital das Clínicas Instituto do Coração, São Paulo, SP- Brasil
| | | | | | | | - Barbara Maria Ianni
- Universidade de São Paulo Faculdade de Medicina Hospital das Clínicas Instituto do Coração, São Paulo, SP- Brasil
| | | | - Felix José Alvarez Ramires
- Universidade de São Paulo Faculdade de Medicina Hospital das Clínicas Instituto do Coração, São Paulo, SP- Brasil
| | - Luciano Nastari
- Universidade de São Paulo Faculdade de Medicina Hospital das Clínicas Instituto do Coração, São Paulo, SP- Brasil
| | - Edecio Cunha-Neto
- Universidade de São Paulo Faculdade de Medicina Hospital das Clínicas Instituto do Coração, São Paulo, SP- Brasil
| | - Antonio L Ribeiro
- Universidade Federal de Minas Gerais - Centro de Telessaúde - Hospital das Clínicas, Belo Horizonte, MG - Brasil
| | | | - Sheila M Keating
- Blood Systems Research Institute, San Francisco, Califórnia - EUA
| | | | - Charles Mady
- Universidade de São Paulo Faculdade de Medicina Hospital das Clínicas Instituto do Coração, São Paulo, SP- Brasil
| |
Collapse
|
23
|
Srejovic IM, Lukic ML. Galectin-3 in T cell-mediated immunopathology and autoimmunity. Immunol Lett 2021; 233:57-67. [PMID: 33753135 DOI: 10.1016/j.imlet.2021.03.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 03/17/2021] [Indexed: 01/05/2023]
Abstract
Galectin-3 (Gal-3) is the only member of galectin family able to form pentamers and heterodimers with chemokines. Its presence in various cells and tissues suggests variety of regulatory functions in physiological conditions, but increasing body of evidence indicates involvement of Gal-3 in pathological cascades of many diseases. Gal-3 exerts different, sometimes opposite, effects in various disorders or in different phases of the same disease. These differences in action of Gal-3 are related to the localization of Gal-3 in the cell, types of receptors through which it acts, or the types of cells that secrete it. As a regulator of immune response and T-cell activity, Gal-3 appears to have important role in development of autoimmunity mediated by T cells. Absence of Gal-3 in C57Bl6 mice favors Th2 mediated inflammatory myocarditis but attenuate fibrosis. Recent data also indicate Gal-3 involvement in development atherosclerosis. In pathogenesis of diabetes type 1 and autoimmune components of diabetes type 2 Gal-3 may have detrimental or protective role depending on its intracellular or extracellular localization. Gal-3 mediates autoimmune hepatic damage through activation of T-cells or natural killer T cells. Gal-3 is an important mediator in neurodevelopment, neuropathology and behavior due to its expression both in neurons and glial cells. All together, assessing the role of Gal-3 in immunopathology and autoimmunity it could be concluded that it is an important participant in pathogenesis, as well as promising monitoring marker and therapeutic target.
Collapse
Affiliation(s)
- Ivan M Srejovic
- University of Kragujevac, Faculty of Medical Sciences, Department of Physiology, Svetozara Markovica 69, 34000, Kragujevac, Serbia.
| | - Miodrag L Lukic
- University of Kragujevac, Faculty of Medical Sciences, Department of Physiology, Svetozara Markovica 69, 34000, Kragujevac, Serbia; University of Kragujevac, Faculty of Medical Sciences, Center for Molecular Medicine and Stem Cell Research, Svetozara Markovica 69, 34000, Kragujevac, Serbia.
| |
Collapse
|
24
|
Goetzke CC, Althof N, Neumaier HL, Heuser A, Kaya Z, Kespohl M, Klingel K, Beling A. Mitigated viral myocarditis in A/J mice by the immunoproteasome inhibitor ONX 0914 depends on inhibition of systemic inflammatory responses in CoxsackievirusB3 infection. Basic Res Cardiol 2021; 116:7. [PMID: 33523326 PMCID: PMC7851025 DOI: 10.1007/s00395-021-00848-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 01/12/2021] [Indexed: 12/13/2022]
Abstract
A preclinical model of troponin I-induced myocarditis (AM) revealed a prominent role of the immunoproteasome (ip), the main immune cell-resident proteasome isoform, in heart-directed autoimmunity. Viral infection of the heart is a known trigger of cardiac autoimmunity, with the ip enhancing systemic inflammatory responses after infection with a cardiotropic coxsackievirusB3 (CV). Here, we used ip-deficient A/J-LMP7-/- mice to investigate the role of ip-mediated effects on adaptive immunity in CV-triggered myocarditis and found no alteration of the inflammatory heart tissue damage or cardiac function in comparison to wild-type controls. Aiming to define the impact of the systemic inflammatory storm under the control of ip proteolysis during CV infection, we targeted the ip in A/J mice with the inhibitor ONX 0914 after the first cycle of infection, when systemic inflammation has set in, well before cardiac inflammation. During established acute myocarditis, the ONX 0914 treatment group had the same reduction in cardiac output as the controls, with inflammatory responses in heart tissue being unaffected by the compound. Based on these findings and with regard to the known anti-inflammatory role of ONX 0914 in CV infection, we conclude that the efficacy of ip inhibitors for CV-triggered myocarditis in A/J mice relies on their immunomodulatory effects on the systemic inflammatory reaction.
Collapse
Affiliation(s)
- Carl Christoph Goetzke
- Department of Pediatrics, Division of Pulmonology, Immunology and Critical Care Medicine, Charité-Universitätsmedizin, Berlin Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
- German Rheumatism Research Center (DRFZ), Leibniz Association, Berlin, Germany
- Berlin Institute of Health, Berlin, Germany
| | - Nadine Althof
- German Federal Institute for Risk Assessment, Berlin, Germany
| | - Hannah Louise Neumaier
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Institute of Biochemistry, Charitéplatz 1, 10117, Berlin, Germany
| | - Arndt Heuser
- Animal Phenotyping Platform, Max-Delbrueck-Center for Molecular Medicine, Berlin, Germany
| | - Ziya Kaya
- Universitätsklinikum Heidelberg, Medizinische Klinik für Innere Medizin III: Kardiologie, Angiologie und Pneumologie, Heidelberg, Germany
- Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK), Partner Side Heidelberg, Heidelberg, Germany
| | - Meike Kespohl
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Institute of Biochemistry, Charitéplatz 1, 10117, Berlin, Germany
- Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK), Partner Side Berlin, Berlin, Germany
| | - Karin Klingel
- Cardiopathology, Institute for Pathology and Neuropathology, University Hospital Tuebingen, Tuebingen, Germany
| | - Antje Beling
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Institute of Biochemistry, Charitéplatz 1, 10117, Berlin, Germany.
- Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK), Partner Side Berlin, Berlin, Germany.
| |
Collapse
|
25
|
Sansonetti M, Waleczek FJG, Jung M, Thum T, Perbellini F. Resident cardiac macrophages: crucial modulators of cardiac (patho)physiology. Basic Res Cardiol 2020; 115:77. [PMID: 33284387 PMCID: PMC7720787 DOI: 10.1007/s00395-020-00836-6] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 11/23/2020] [Indexed: 02/06/2023]
Abstract
Resident cardiac macrophages (rcMacs) are integral components of the myocardium where they have key roles for tissue homeostasis and in response to inflammation, tissue injury and remodelling. In this review, we summarize the current knowledge and limitations associated with the rcMacs studies. We describe their specific role and contribution in various processes such as electrical conduction, efferocytosis, inflammation, tissue development, remodelling and regeneration in both the healthy and the disease state. We also outline research challenges and technical complications associated with rcMac research. Recent technological developments and contemporary immunological techniques are now offering new opportunities to investigate the separate contribution of rcMac in respect to recruited monocytes and other cardiac cells. Finally, we discuss new therapeutic strategies, such as drugs or non-coding RNAs, which can influence rcMac phenotype and their response to inflammation. These novel approaches will allow for a deeper understanding of this cardiac endogenous cell type and might lead to the development of more specific and effective therapeutic strategies to boost the heart's intrinsic reparative capacity.
Collapse
Affiliation(s)
- M Sansonetti
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hanover, Germany
| | - F J G Waleczek
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hanover, Germany
| | - M Jung
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hanover, Germany
| | - T Thum
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hanover, Germany.
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany.
| | - F Perbellini
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hanover, Germany.
| |
Collapse
|
26
|
Zaman R, Hamidzada H, Epelman S. Exploring cardiac macrophage heterogeneity in the healthy and diseased myocardium. Curr Opin Immunol 2020; 68:54-63. [PMID: 33128959 DOI: 10.1016/j.coi.2020.09.005] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 09/15/2020] [Accepted: 09/17/2020] [Indexed: 12/24/2022]
Abstract
Cardiac macrophages maintain homeostasis and orchestrate response to disease. Utilization of genetic fate-mapping and single-cell RNA sequencing shifted the paradigm of macrophage heterogeneity from the canonical M1/M2 classification in favour of a nuanced approach that reconciles divergent origins, lifecycles, and transcriptional states. Here, we provide a conceptual framework to assess cardiac macrophage complexity that integrates transcriptional and functional heterogeneity that tracks with subset-specific markers (TIMD4 and CCR2). Our goal is to provide a starting point for researchers to dissect the functions of known resident cardiac macrophage subpopulations. We discuss recent advances and limitations in our understanding of cardiac macrophage diversity in ischemic injury, hypertension and myocarditis.
Collapse
Affiliation(s)
- Rysa Zaman
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada; Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, University of Toronto, Toronto, ON, Canada; Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Homaira Hamidzada
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada; Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, University of Toronto, Toronto, ON, Canada; Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Slava Epelman
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada; Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, University of Toronto, Toronto, ON, Canada; Department of Immunology, University of Toronto, Toronto, ON, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada; Peter Munk Cardiac Centre, Toronto General Hospital, Toronto, ON, Canada.
| |
Collapse
|
27
|
Xue YL, Zhang SX, Zheng CF, Li YF, Zhang LH, Su QY, Hao YF, Wang S, Li XW. Long non-coding RNA MEG3 inhibits M2 macrophage polarization by activating TRAF6 via microRNA-223 down-regulation in viral myocarditis. J Cell Mol Med 2020; 24:12341-12354. [PMID: 33047847 PMCID: PMC7686963 DOI: 10.1111/jcmm.15720] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Revised: 06/24/2020] [Accepted: 07/18/2020] [Indexed: 12/01/2022] Open
Abstract
Viral myocarditis (VMC) commonly triggers heart failure, for which no specific treatments are available. This study aims to explore the specific role of long non‐coding RNA (lncRNA) maternally expressed 3 (MEG3) in VMC. A VMC mouse model was induced by Coxsackievirus B3 (CVB3). Then, MEG3 and TNF receptor‐associated factor 6 (TRAF6) were silenced and microRNA‐223 (miR‐223) was over‐expressed in the VMC mice, followed by determination of ventricular ejection fraction (LVEF) and left ventricular fractional shortening (LVFS). Dual‐luciferase reporter assay was introduced to test the interaction among MEG3, TRAF6 and miR‐223. Macrophages were isolated from cardiac tissues and bone marrow, and polarization of M1 or M2 macrophages was induced. Then, the expressions of components of NLRP3 inflammatory body (NLRP3, ASC, Caspase‐1), M1 markers (CD86, iNOS and TNF‐α) and M2 markers (CD206, Arginase‐1 and Fizz‐1) were measured following MEG3 silencing. In the VMC mouse model, MEG3 and TRAF6 levels were obviously increased, while miR‐223 expression was significantly reduced. Down‐regulation of MEG3 resulted in the inhibition of TRAF6 by promoting miR‐223. TRAF6 was negatively correlated with miR‐223, but positively correlated with MEG3 expression. Down‐regulations of MEG3 or TRAF6 or up‐regulation of miR‐223 was observed to increase mouse weight, survival rate, LVEF and LVFS, while inhibiting myocarditis and inflammation via the NF‐κB pathway inactivation in VMC mice. Down‐regulation of MEG3 decreased M1 macrophage polarization and elevated M2 macrophage polarization by up‐regulating miR‐223. Collectively, down‐regulation of MEG3 leads to the inhibition of inflammation and induces M2 macrophage polarization via miR‐223/TRAF6/NF‐κB axis, thus alleviating VMC.
Collapse
Affiliation(s)
- Yu-Long Xue
- Department of Cardiovascular Medicine, Shanxi Dayi Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Sheng-Xiao Zhang
- Department of Rheumatology, the Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Chao-Feng Zheng
- Department of Genetics Laboratory, Linfen Maternity & Child Healthcare Hospital, Linfen, China
| | - Yu-Feng Li
- Department of Neurology and Stroke Center, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Li-Hui Zhang
- Department of Cardiovascular Medicine, Shanxi Dayi Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Qin-Yi Su
- Department of Rheumatology, the Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Yu-Fei Hao
- Department of Rheumatology, the Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Shu Wang
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Xue-Wen Li
- Department of Cardiovascular Medicine, Shanxi Dayi Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| |
Collapse
|
28
|
Hara A, Niwa M, Kanayama T, Noguchi K, Niwa A, Matsuo M, Kuroda T, Hatano Y, Okada H, Tomita H. Galectin-3: A Potential Prognostic and Diagnostic Marker for Heart Disease and Detection of Early Stage Pathology. Biomolecules 2020; 10:biom10091277. [PMID: 32899694 PMCID: PMC7565392 DOI: 10.3390/biom10091277] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 08/26/2020] [Accepted: 09/02/2020] [Indexed: 12/13/2022] Open
Abstract
The use of molecular biomarkers for the early detection of heart disease, before their onset of symptoms, is an attractive novel approach. Ideal molecular biomarkers, those that are both sensitive and specific to heart disease, are likely to provide a much earlier diagnosis, thereby providing better treatment outcomes. Galectin-3 is expressed by various immune cells, including mast cells, histiocytes and macrophages, and plays an important role in diverse physiological functions. Since galectin-3 is readily expressed on the cell surface, and is readily secreted by injured and inflammatory cells, it has been suggested that cardiac galectin-3 could be a marker for cardiac disorders such as cardiac inflammation and fibrosis, depending on the specific pathogenesis. Thus, galectin-3 may be a novel candidate biomarker for the diagnosis, analysis and prognosis of various cardiac diseases, including heart failure. The goals of heart disease treatment are to prevent acute onset and to predict their occurrence by using the ideal molecular biomarkers. In this review, we discuss and summarize recent developments of galectin-3 as a next-generation molecular biomarker of heart disease. Furthermore, we describe how galectin-3 may be useful as a diagnostic marker for detecting the early stages of various heart diseases, which may contribute to improved early therapeutic interventions.
Collapse
Affiliation(s)
- Akira Hara
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; (T.K.); (K.N.); (A.N.); (M.M.); (T.K.); (Y.H.); (H.T.)
- Correspondence: ; Tel.: +81-58-230-6225
| | - Masayuki Niwa
- Medical Education Development Center, Gifu University School of Medicine, Gifu 501-1194, Japan;
| | - Tomohiro Kanayama
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; (T.K.); (K.N.); (A.N.); (M.M.); (T.K.); (Y.H.); (H.T.)
| | - Kei Noguchi
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; (T.K.); (K.N.); (A.N.); (M.M.); (T.K.); (Y.H.); (H.T.)
| | - Ayumi Niwa
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; (T.K.); (K.N.); (A.N.); (M.M.); (T.K.); (Y.H.); (H.T.)
| | - Mikiko Matsuo
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; (T.K.); (K.N.); (A.N.); (M.M.); (T.K.); (Y.H.); (H.T.)
| | - Takahiro Kuroda
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; (T.K.); (K.N.); (A.N.); (M.M.); (T.K.); (Y.H.); (H.T.)
| | - Yuichiro Hatano
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; (T.K.); (K.N.); (A.N.); (M.M.); (T.K.); (Y.H.); (H.T.)
| | - Hideshi Okada
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan;
| | - Hiroyuki Tomita
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; (T.K.); (K.N.); (A.N.); (M.M.); (T.K.); (Y.H.); (H.T.)
| |
Collapse
|
29
|
Tschöpe C, Van Linthout S, Jäger S, Arndt R, Trippel T, Müller I, Elsanhoury A, Rutschow S, Anker SD, Schultheiss HP, Pauschinger M, Spillmann F, Pappritz K. Modulation of the acute defence reaction by eplerenone prevents cardiac disease progression in viral myocarditis. ESC Heart Fail 2020; 7:2838-2852. [PMID: 32662949 PMCID: PMC7405199 DOI: 10.1002/ehf2.12887] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 06/04/2020] [Accepted: 06/24/2020] [Indexed: 12/19/2022] Open
Abstract
Aims Left ventricular (LV) dysfunction in viral myocarditis is attributed to myocardial inflammation and fibrosis, inducing acute and long‐time cardiac damage. Interventions are not established. On the basis of the link between inflammation, fibrosis, aldosterone, and extracellular matrix regulation, we aimed to investigate the effect of an early intervention with the mineralocorticoid receptor antagonist (MRA) eplerenone on cardiac remodelling in a murine model of persistent coxsackievirus B3 (CVB3)‐induced myocarditis. Methods and results SWR/J mice were infected with 5 × 104 plaque‐forming units of CVB3 (Nancy strain) and daily treated either with eplerenone (200 mg/kg body weight) or with placebo starting from Day 1. At Day 8 or 28 post infection, mice were haemodynamically characterized and subsequently sacrificed for immunohistological and molecular biology analyses. Eplerenone did not influence CVB3 load. Already at Day 8, 1.8‐fold (P < 0.05), 1.4‐fold (P < 0.05), 3.2‐fold (P < 0.01), and 2.1‐fold (P < 0.001) reduction in LV intercellular adhesion molecule 1 expression, presence of monocytes/macrophages, oxidative stress, and apoptosis, respectively, was observed in eplerenone‐treated vs. untreated CVB3‐infected mice. In vitro, eplerenone led to 1.4‐fold (P < 0.01) and 1.2‐fold (P < 0.01) less CVB3‐induced cardiomyocyte oxidative stress and apoptosis. Furthermore, collagen production was 1.1‐fold (P < 0.05) decreased in cardiac fibroblasts cultured with medium of eplerenone‐treated vs. untreated CVB3‐infected HL‐1 cardiomyocytes. These ameliorations were in vivo translated into prevention of cardiac fibrosis, as shown by 1.4‐fold (P < 0.01) and 2.1‐fold (P < 0.001) lower collagen content in the LV of eplerenone‐treated vs. untreated CVB3‐infected mice at Days 8 and 28, respectively. This resulted in an early and long‐lasting improvement of LV dimension and function, as indicated by reduced LV end‐systolic volume and end‐diastolic volume, and an increase in LV contractility (dP/dtmax) and LV relaxation (dP/dtmin), respectively (P < 0.05). Conclusions Early intervention with the MRA eplerenone modulates the acute host and defence reaction and prevents cardiac disease progression in experimental CVB3‐induced myocarditis without aggravation of viral load. The findings advocate for an initiation of therapy of viral myocarditis as early as possible, even before the onset of inflammation‐induced myocardial dysfunction. This may also have implications for coronavirus disease‐19 therapy.
Collapse
Affiliation(s)
- Carsten Tschöpe
- Berlin Institute of Health Center for Regenerative Therapies and Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, Berlin, Germany.,German Center for Cardiovascular Research (DZHK), Partner site Berlin, Berlin, Germany.,Department of Cardiology, Charité-Universitätsmedizin Berlin, Campus Virchow Klinikum (CVK), Berlin, Germany
| | - Sophie Van Linthout
- Berlin Institute of Health Center for Regenerative Therapies and Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, Berlin, Germany.,German Center for Cardiovascular Research (DZHK), Partner site Berlin, Berlin, Germany
| | - Sebastian Jäger
- Department of Cardiology, Alexianer Hospital Hedwigshöhe, Berlin, Germany
| | - Robert Arndt
- Department of Emergency Medicine, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin (CBF), Berlin, Germany
| | - Tobias Trippel
- Department of Cardiology, Charité-Universitätsmedizin Berlin, Campus Virchow Klinikum (CVK), Berlin, Germany
| | - Irene Müller
- Berlin Institute of Health Center for Regenerative Therapies and Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, Berlin, Germany.,German Center for Cardiovascular Research (DZHK), Partner site Berlin, Berlin, Germany
| | - Ahmed Elsanhoury
- Berlin Institute of Health Center for Regenerative Therapies and Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, Berlin, Germany.,German Center for Cardiovascular Research (DZHK), Partner site Berlin, Berlin, Germany
| | - Susanne Rutschow
- Department of Cardiology, Angiology Johanniter-Kliniken, Stendal, Germany
| | - Stefan D Anker
- Berlin Institute of Health Center for Regenerative Therapies and Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, Berlin, Germany.,German Center for Cardiovascular Research (DZHK), Partner site Berlin, Berlin, Germany.,Department of Cardiology, Charité-Universitätsmedizin Berlin, Campus Virchow Klinikum (CVK), Berlin, Germany
| | | | - Matthias Pauschinger
- Department of Cardiology, Paracelsus University, Klinikum Nürnberg, Nürnberg, Germany
| | - Frank Spillmann
- Department of Cardiology, Charité-Universitätsmedizin Berlin, Campus Virchow Klinikum (CVK), Berlin, Germany
| | - Kathleen Pappritz
- Berlin Institute of Health Center for Regenerative Therapies and Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, Berlin, Germany.,German Center for Cardiovascular Research (DZHK), Partner site Berlin, Berlin, Germany
| |
Collapse
|
30
|
Abstract
Myocarditis (MCD) is a type of inflammatory disease in which inflammatory cells infiltrate the myocardium, leading to cardiac dysfunction, myocardial necrosis, and fibrosis. Although it has been reported that MCD is mediated by T cells, the immune system is complex and includes many types of immune cells that interact with one another. Through investigations of the inflammatory responses in MCD including myocardial necrosis, fibrosis, and arrhythmia, we have gained further insight into the pathogenesis of MCD. This article aims to discuss the diversity and the roles of immune cells involved in the pathogenesis of MCD. Moreover, immunotherapy for the treatment of MCD remains controversial, and further investigation is required to identify accurate immunotherapies for special cell types.
Collapse
|
31
|
Bouin A, Semler BL. Picornavirus Cellular Remodeling: Doubling Down in Response to Viral-Induced Inflammation. CURRENT CLINICAL MICROBIOLOGY REPORTS 2020; 7:31-37. [PMID: 32704466 PMCID: PMC7377643 DOI: 10.1007/s40588-020-00138-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
Purpose of Review To highlight recent findings on how picornavirus infections of the airways and cardiac tissues impact cellular inflammation and remodeling events. Recent Findings Recent published work has revealed that although many picornavirus infections appear to be initially asymptomatic, there are significant disease sequelae that result from chronic or persistent infections and the long-term, pathogenic effects on host tissues. Summary Because many acute picornavirus infections are asymptomatic, it is difficult to diagnose these pathologies at the early stages of disease. As a result, we must rely on preventative measures (i.e., vaccination) or discover novel treatments to reverse tissue damage and remodeling in affected individuals. Both of these strategies will require a comprehensive knowledge of virus-and cell-specific replication determinants and how these processes induce pathogenic effects in infected cells and tissues.
Collapse
Affiliation(s)
- Alexis Bouin
- Department of Microbiology & Molecular Genetics and Center for Virus Research, School of Medicine, University of California, Med Sci Bldg, Room B237, Irvine, CA 92697-4025, USA
| | - Bert L Semler
- Department of Microbiology & Molecular Genetics and Center for Virus Research, School of Medicine, University of California, Med Sci Bldg, Room B237, Irvine, CA 92697-4025, USA
| |
Collapse
|
32
|
Role of oxidative stress-related biomarkers in heart failure: galectin 3, α1-antitrypsin and LOX-1: new therapeutic perspective? Mol Cell Biochem 2019; 464:143-152. [DOI: 10.1007/s11010-019-03656-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 11/16/2019] [Indexed: 02/07/2023]
|
33
|
Ferrer MF, Thomas P, López Ortiz AO, Errasti AE, Charo N, Romanowski V, Gorgojo J, Rodriguez ME, Carrera Silva EA, Gómez RM. Junin Virus Triggers Macrophage Activation and Modulates Polarization According to Viral Strain Pathogenicity. Front Immunol 2019; 10:2499. [PMID: 31695702 PMCID: PMC6817498 DOI: 10.3389/fimmu.2019.02499] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 10/07/2019] [Indexed: 01/24/2023] Open
Abstract
The New World arenavirus Junin (JUNV) is the etiological agent of Argentine hemorrhagic fever (AHF). Previous studies of human macrophage infection by the Old-World arenaviruses Mopeia and Lassa showed that while the non-pathogenic Mopeia virus replicates and activates human macrophages, the pathogenic Lassa virus replicates but fails to activate human macrophages. Less is known in regard to the impact of New World arenavirus infection on the human macrophage immune response. Macrophage activation is critical for controlling infections but could also be usurped favoring immune evasion. Therefore, it is crucial to understand how the JUNV infection modulates macrophage plasticity to clarify its role in AHF pathogenesis. With this aim in mind, we compared infection with the attenuated Candid 1 (C#1) or the pathogenic P strains of the JUNV virus in human macrophage cultures. The results showed that both JUNV strains similarly replicated and induced morphological changes as early as 1 day post-infection. However, both strains differentially induced the expression of CD71, the receptor for cell entry, the activation and maturation molecules CD80, CD86, and HLA-DR and selectively modulated cytokine production. Higher levels of TNF-α, IL-10, and IL-12 were detected with C#1 strain, while the P strain induced only higher levels of IL-6. We also found that C#1 strain infection skewed macrophage polarization to M1, whereas the P strain shifted the response to an M2 phenotype. Interestingly, the MERTK receptor, that negatively regulates the immune response, was down-regulated by C#1 strain and up-regulated by P strain infection. Similarly, the target genes of MERTK activation, the cytokine suppressors SOCS1 and SOCS3, were also increased after P strain infection, in addition to IRF-1, that regulates type I IFN levels, which were higher with C#1 compared with P strain infection. Together, this differential activation/polarization pattern of macrophages elicited by P strain suggests a more evasive immune response and may have important implications in the pathogenesis of AHF and underpinning the development of new potential therapeutic strategies.
Collapse
Affiliation(s)
- María F Ferrer
- Laboratorio de Virus Animales, Instituto de Biotecnología y Biología Molecular, CONICET-Universidad Nacional de La Plata, La Plata, Argentina
| | - Pablo Thomas
- Laboratorio de Virus Animales, Instituto de Biotecnología y Biología Molecular, CONICET-Universidad Nacional de La Plata, La Plata, Argentina
| | - Aída O López Ortiz
- Laboratorio de Virus Animales, Instituto de Biotecnología y Biología Molecular, CONICET-Universidad Nacional de La Plata, La Plata, Argentina.,Laboratorio de Trombosis Experimental, Instituto de Medicina Experimental, CONICET-Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Andrea E Errasti
- Facultad de Medicina, Instituto de Farmacologia, University of Buenos Aries, Buenos Aires, Argentina
| | - Nancy Charo
- Laboratorio de Trombosis Experimental, Instituto de Medicina Experimental, CONICET-Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Victor Romanowski
- Laboratorio de Virus Animales, Instituto de Biotecnología y Biología Molecular, CONICET-Universidad Nacional de La Plata, La Plata, Argentina.,Global Viral Network, Baltimore, MD, United States
| | - Juan Gorgojo
- Centro de Investigación y Desarrollo en Fermentaciones Industriales, CONICET-Universidad Nacional de La Plata, La Plata, Argentina
| | - María E Rodriguez
- Centro de Investigación y Desarrollo en Fermentaciones Industriales, CONICET-Universidad Nacional de La Plata, La Plata, Argentina
| | - Eugenio A Carrera Silva
- Laboratorio de Trombosis Experimental, Instituto de Medicina Experimental, CONICET-Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Ricardo M Gómez
- Laboratorio de Virus Animales, Instituto de Biotecnología y Biología Molecular, CONICET-Universidad Nacional de La Plata, La Plata, Argentina.,Global Viral Network, Baltimore, MD, United States
| |
Collapse
|
34
|
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: 166] [Impact Index Per Article: 27.7] [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
|
35
|
Bao J, Sun T, Yue Y, Xiong S. Macrophage NLRP3 inflammasome activated by CVB3 capsid proteins contributes to the development of viral myocarditis. Mol Immunol 2019; 114:41-48. [PMID: 31336248 DOI: 10.1016/j.molimm.2019.07.012] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 06/28/2019] [Accepted: 07/11/2019] [Indexed: 12/18/2022]
Abstract
Viral myocarditis, mainly caused by enteroviruses specially coxsackievirus B3 (CVB3) infection, is a common clinical cardiovascular disease and characterized by cardiac massive inflammation. Our previous study showed that CVB3-induced myocardial NLRP3 contributed to the development of viral myocarditis. In this study, we found that beside of being up-regulated in myocardiocytes, NLPR3 was also obviously increased in the cardiac infiltrating macrophages. While whether this accumulated NLRP3 influences, macrophage inflammatory responses remains unknown. By adoptive transfer assays, we found that mice receiving NLRP3 up-regulated macrophages showed much more abundant cardiac IL-1β production and more severe myocardial inflammation, while those receiving NLRP3 down-regulated macrophages showed much less IL-1β production and milder myocarditis, indicating that NLRP3 up-regulated macrophages played a pathological role in CVB3-induced myocarditis. In addition, we further found that it was CVB3 capsid proteins VP1 (predominant) and VP2, but not viral RNAs, robustly triggered macrophage NLRP3 up-regulation and activation. Our study demonstrated macrophage NLRP3 inflammasome could be efficiently be activated by CVB3 capsid proteins, and contributed to the pathogenesis of viral myocarditis. It might provide some clues to the development of new therapeutic strategies based on macrophage NLRP3 modulation.
Collapse
Affiliation(s)
- Jingyin Bao
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, 215123, China
| | - Tianle Sun
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, 215123, China
| | - Yan Yue
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, 215123, China.
| | - Sidong Xiong
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, 215123, China.
| |
Collapse
|
36
|
Tatsumi K, Schmedes CM, Houston ER, Butler E, Mackman N, Antoniak S. Protease-activated receptor 4 protects mice from Coxsackievirus B3 and H1N1 influenza A virus infection. Cell Immunol 2019; 344:103949. [PMID: 31337508 DOI: 10.1016/j.cellimm.2019.103949] [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: 01/29/2019] [Revised: 06/20/2019] [Accepted: 07/02/2019] [Indexed: 02/07/2023]
Abstract
PAR4 is expressed by a variety of cells, including platelets, cardiac, lung and immune cells. We investigated the contribution of PAR4 to viral infections of the heart and lung. Toll-like receptor (TLR) 3-dependent immune responses were analyzed after co-stimulation of PAR4 in murine bone-marrow derived macrophages, embryonic fibroblasts and embryonic cardiomyocytes. In addition, we analyzed Coxsackievirus B3 (CVB3) or H1N1 influenza A virus (H1N1 IAV) infection of PAR4-/- (ΔPAR4) and wild-type (WT) mice. Lastly, we investigated the effect of platelet inhibition on H1N1 IAV infection. In vitro experiments revealed that PAR4 stimulation enhances the expression of TLR3-dependent CXCL10 expression and decreases TLR3-dependent NFκB-mediated proinflammatory gene expression. Furthermore, CVB3-infected ΔPAR4 mice exhibited a decreased anti-viral response and increased viral genomes in the heart leading to more pronounced CVB3 myocarditis compared to WT mice. Similarly, H1N1 IAV-infected ΔPAR4 mice had increased immune cell numbers and inflammatory mediators in the lung, and increased mortality compared with infected WT controls. The study showed that PAR4 protects mice from viral infections of the heart and lung.
Collapse
Affiliation(s)
- Kohei Tatsumi
- Department of Medicine, Thrombosis and Hemostasis Program, Division of Hematology and Oncology, UNC McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA
| | - Clare M Schmedes
- Department of Medicine, Thrombosis and Hemostasis Program, Division of Hematology and Oncology, UNC McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA
| | - E Reaves Houston
- Department of Medicine, Thrombosis and Hemostasis Program, Division of Hematology and Oncology, UNC McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA
| | - Emily Butler
- Department of Medicine, Thrombosis and Hemostasis Program, Division of Hematology and Oncology, UNC McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA
| | - Nigel Mackman
- Department of Medicine, Thrombosis and Hemostasis Program, Division of Hematology and Oncology, UNC McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA
| | - Silvio Antoniak
- Department of Pathology and Laboratory Medicine, UNC McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
37
|
Du X, Zhao W, Nguyen M, Lu Q, Kiriazis H. β-Adrenoceptor activation affects galectin-3 as a biomarker and therapeutic target in heart disease. Br J Pharmacol 2019; 176:2449-2464. [PMID: 30756388 PMCID: PMC6592856 DOI: 10.1111/bph.14620] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 12/11/2018] [Accepted: 01/29/2019] [Indexed: 12/12/2022] Open
Abstract
Myocardial fibrosis is a key histopathological component that drives the progression of heart disease leading to heart failure and constitutes a therapeutic target. Recent preclinical and clinical studies have implicated galectin-3 (Gal-3) as a pro-fibrotic molecule and a biomarker of heart disease and fibrosis. However, our knowledge is poor on the mechanism(s) that determine the blood level or regulate cardiac expression of Gal-3. Recent studies have demonstrated that enhanced β-adrenoceptor activity is a determinant of both circulating concentration and cardiac expression of Gal-3. Pharmacological or transgenic activation of β-adrenoceptors leads to increased blood levels of Gal-3 and up-regulated cardiac Gal-3 expression, effect that can be reversed with the use of β-adrenoceptor antagonists. Conversely, Gal-3 gene deletion confers protection against isoprenaline-induced cardiotoxicity and fibrogenesis. At the transcription level, β-adrenoceptor stimulation activates cardiac mammalian sterile-20-like kinase 1, a pivotal kinase of the Hippo signalling pathway, which is associated with Gal-3 up-regulation. Recent studies have suggested a role for the β-adrenoceptor-Hippo signalling pathway in the regulation of cardiac Gal-3 expression thereby contributing to the onset and progression of heart disease. This implies a therapeutic potential of the suppression of Gal-3 expression. In this review, we discuss the effects of β-adrenoceptor activity on Gal-3 as a biomarker and causative mediator in the setting of heart disease and point out pivotal knowledge gaps. LINKED ARTICLES: This article is part of a themed section on Adrenoceptors-New Roles for Old Players. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.14/issuetoc.
Collapse
Affiliation(s)
- Xiao‐Jun Du
- Experimental Cardiology LaboratoryBaker Heart and Diabetes InstituteMelbourneVICAustralia
- Department of Physiology and Pathophysiology, School of Medical SciencesXi'an Jiaotong University Health Science CenterXi'anChina
| | - Wei‐Bo Zhao
- Experimental Cardiology LaboratoryBaker Heart and Diabetes InstituteMelbourneVICAustralia
| | - My‐Nhan Nguyen
- Experimental Cardiology LaboratoryBaker Heart and Diabetes InstituteMelbourneVICAustralia
| | - Qun Lu
- Experimental Cardiology LaboratoryBaker Heart and Diabetes InstituteMelbourneVICAustralia
- Department of Cardiovascular Medicine, First HospitalXi'an Jiaotong University Health Science CenterXi'anChina
| | - Helen Kiriazis
- Experimental Cardiology LaboratoryBaker Heart and Diabetes InstituteMelbourneVICAustralia
| |
Collapse
|
38
|
Inflammation and fibrosis in murine models of heart failure. Basic Res Cardiol 2019; 114:19. [PMID: 30887214 DOI: 10.1007/s00395-019-0722-5] [Citation(s) in RCA: 263] [Impact Index Per Article: 43.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 02/12/2019] [Indexed: 02/07/2023]
Abstract
Heart failure is a consequence of various cardiovascular diseases and associated with poor prognosis. Despite progress in the treatment of heart failure in the past decades, prevalence and hospitalisation rates are still increasing. Heart failure is typically associated with cardiac remodelling. Here, inflammation and fibrosis are thought to play crucial roles. During cardiac inflammation, immune cells invade the cardiac tissue and modulate tissue-damaging responses. Cardiac fibrosis, however, is characterised by an increased amount and a disrupted composition of extracellular matrix proteins. As evidence exists that cardiac inflammation and fibrosis are potentially reversible in experimental and clinical set ups, they are interesting targets for innovative heart failure treatments. In this context, animal models are important as they mimic clinical conditions of heart failure patients. The advantages of mice in this respect are short generation times and genetic modifications. As numerous murine models of heart failure exist, the selection of a proper disease model for a distinct research question is demanding. To facilitate this selection, this review aims to provide an overview about the current understanding of the pathogenesis of cardiac inflammation and fibrosis in six frequently used murine models of heart failure. Hence, it compares the models of myocardial infarction with or without reperfusion, transverse aortic constriction, chronic subjection to angiotensin II or deoxycorticosterone acetate, and coxsackievirus B3-induced viral myocarditis in this context. It furthermore provides information about the clinical relevance and the limitations of each model, and, if applicable, about the recent advancements in their methodological proceedings.
Collapse
|
39
|
Stojanovic B, Milovanovic J, Arsenijevic A, Stojanovic B, Strazic Geljic I, Arsenijevic N, Jonjic S, Lukic ML, Milovanovic M. Galectin-3 Deficiency Facilitates TNF-α-Dependent Hepatocyte Death and Liver Inflammation in MCMV Infection. Front Microbiol 2019; 10:185. [PMID: 30800112 PMCID: PMC6376859 DOI: 10.3389/fmicb.2019.00185] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 01/23/2019] [Indexed: 12/12/2022] Open
Abstract
Galectin-3 (Gal-3) has a role in multiple inflammatory pathways. Various, opposite roles of Gal-3 in liver diseases have been described but there are no data about the role of Gal-3 in development of hepatitis induced with cytomegalovirus infection. In this study we aimed to clarify the role of Gal-3 in murine cytomegalovirus (MCMV)-induced hepatitis by using Gal-3-deficient (Gal-3 KO) mice. Here we provide the evidence that Gal-3 has the protective role in MCMV-induced hepatitis. Enhanced hepatitis manifested by more inflammatory and necrotic foci and serum level of ALT, enhanced apoptosis and necroptosis of hepatocytes and enhanced viral replication were detected in MCMV-infected Gal-3 deficient mice. NK cells does not contribute to more severe liver damage in MCMV-infected Gal-3 KO mice. Enhanced expression of TNF-α in the hepatocytes of Gal-3 KO mice after MCMV infection, abrogated hepatocyte death, and attenuated inflammation in the livers of Gal-3 KO mice after TNF-α blockade suggest that TNF-α plays the role in enhanced disease in Gal-3 deficient animals. Treatment with recombinant Gal-3 reduces inflammation and especially necrosis of hepatocytes in the livers of MCMV-infected Gal-3 KO mice. Our data highlight the protective role of Gal-3 in MCMV-induced hepatitis by attenuation of TNF-α-mediated death of hepatocytes.
Collapse
Affiliation(s)
- Bojana Stojanovic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia.,Faculty of Medical Sciences, Institute of Pathophysiology, University of Kragujevac, Kragujevac, Serbia
| | - Jelena Milovanovic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia.,Faculty of Medical Sciences, Institute of Histology, University of Kragujevac, Kragujevac, Serbia
| | - Aleksandar Arsenijevic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Bojan Stojanovic
- Department of Surgery, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Ivana Strazic Geljic
- Department for Histology and Embryology, Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Nebojsa Arsenijevic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Stipan Jonjic
- Department for Histology and Embryology, Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Miodrag L Lukic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Marija Milovanovic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| |
Collapse
|
40
|
Noguchi K, Tomita H, Kanayama T, Niwa A, Hatano Y, Hoshi M, Sugie S, Okada H, Niwa M, Hara A. Time-course analysis of cardiac and serum galectin-3 in viral myocarditis after an encephalomyocarditis virus inoculation. PLoS One 2019; 14:e0210971. [PMID: 30673749 PMCID: PMC6343901 DOI: 10.1371/journal.pone.0210971] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 01/06/2019] [Indexed: 11/19/2022] Open
Abstract
Galectin-3 is a β-galactoside-binding lectin which is important in cell proliferation and apoptotic regulation. Recently, serum galectin-3 has been shown to have prognostic value as a biomarker in heart failure. Encephalomyocarditis virus (EMCV) can cause severe myocarditis, congestive heart failure and dilated cardiomyopathy as well as encephalitis in various animals including mice. The pathophysiological role of galectin-3 in acute myocarditis following viral infection is not fully understood. The goal of this study is to determine the cardiac localization and the time-course of galectin-3 expression in heart failure after viral inoculation with EMCV. At 12, 24, 48, 96 hours, 7 and 10 days after intraperitoneal EMCV inoculation, animals were examined histologically and analyzed for the expression of galectin-3 and Iba1. Galectin-3 was up-regulated in degenerated fibrotic lesions of cardiac tissues 96 hours after viral inoculation and were followed by myocardial fibrosis. At the same time, Iba1 positive macrophages were observed within the inflammatory sites. A time-course correlation between the number of galectin-3 positive cells and the cardiac area of degenerated fibrotic lesions was detected—serum galectin-3 increased at 96 hours and correlated well with the number of cardiac galectin-3 positive cells. Our results indicate that galectin-3 expression may be a useful biomarker of cardiac fibrotic degeneration in acute myocarditis following viral infection. In addition, measuring serum galectin-3 levels might be an early diagnostic method for detecting cardiac degeneration in acute myocarditis.
Collapse
Affiliation(s)
- Kei Noguchi
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Hiroyuki Tomita
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Tomohiro Kanayama
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Ayumi Niwa
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Yuichiro Hatano
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Masato Hoshi
- Department of Biochemical and Analytical Sciences, Fujita Health University Graduate School of Health Sciences, Aichi, Japan
| | - Shigeyuki Sugie
- Department of Pathology, Asahi University Murakami Memorial Hospital, Gifu, Japan
| | - Hideshi Okada
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Masayuki Niwa
- Medical Science Division, United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, Gifu, Japan
| | - Akira Hara
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, Gifu, Japan
| |
Collapse
|
41
|
Beling A, Kespohl M. Proteasomal Protein Degradation: Adaptation of Cellular Proteolysis With Impact on Virus-and Cytokine-Mediated Damage of Heart Tissue During Myocarditis. Front Immunol 2018; 9:2620. [PMID: 30546359 PMCID: PMC6279938 DOI: 10.3389/fimmu.2018.02620] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 10/24/2018] [Indexed: 12/26/2022] Open
Abstract
Viral myocarditis is an inflammation of the heart muscle triggered by direct virus-induced cytolysis and immune response mechanisms with most severe consequences during early childhood. Acute and long-term manifestation of damaged heart tissue and disturbances of cardiac performance involve virus-triggered adverse activation of the immune response and both immunopathology, as well as, autoimmunity account for such immune-destructive processes. It is a matter of ongoing debate to what extent subclinical virus infection contributes to the debilitating sequela of the acute disease. In this review, we conceptualize the many functions of the proteasome in viral myocarditis and discuss the adaptation of this multi-catalytic protease complex together with its implications on the course of disease. Inhibition of proteasome function is already highly relevant as a strategy in treating various malignancies. However, cardiotoxicity and immune-related adverse effects have proven significant hurdles, representative of the target's wide-ranging functions. Thus, we further discuss the molecular details of proteasome-mediated activity of the immune response for virus-mediated inflammatory heart disease. We summarize how the spatiotemporal flexibility of the proteasome might be tackled for therapeutic purposes aiming to mitigate virus-mediated adverse activation of the immune response in the heart.
Collapse
Affiliation(s)
- Antje Beling
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Institute of Biochemistry, Berlin, Germany.,Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK), Partner Site Berlin, Berlin, Germany
| | - Meike Kespohl
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Institute of Biochemistry, Berlin, Germany.,Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK), Partner Site Berlin, Berlin, Germany
| |
Collapse
|
42
|
Laaf D, Bojarová P, Elling L, Křen V. Galectin-Carbohydrate Interactions in Biomedicine and Biotechnology. Trends Biotechnol 2018; 37:402-415. [PMID: 30413271 DOI: 10.1016/j.tibtech.2018.10.001] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 09/25/2018] [Accepted: 10/02/2018] [Indexed: 12/23/2022]
Abstract
Cellular communication events are mediated by interactions between cell-surface sugars and lectins, which are carbohydrate-binding proteins. Galectins are β-galactosyl-binding lectins that bridge molecules by their sugar moieties, forming a signaling and adhesion network. Severe changes in glycosylation and galectin expression accompany major processes in oncogenesis, cardiovascular disorders, and other pathologies, making galectins attractive therapeutic targets. Here we discuss advanced strategies of chemo-enzymatic carbohydrate synthesis for creating lead glycomimetics and (neo-)glycoconjugates for galectin-1 and -3 targeting in biomedicine and biotechnology. We will describe the challenges and bottlenecks on the route into biomedical and biotechnological practice and present the first clinical candidates. The coming era will see an exciting translation of selective well-defined high-affinity galectin ligands from bench to bedside.
Collapse
Affiliation(s)
- Dominic Laaf
- Laboratory for Biomaterials, Institute for Biotechnology and Helmholtz-Institute for Biomedical Engineering, RWTH Aachen University, Pauwelsstraße 20, D-52074 Aachen, Germany; Equally contributing authors
| | - Pavla Bojarová
- Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, CZ-14220 Prague 4, Czech Republic; Equally contributing authors
| | - Lothar Elling
- Laboratory for Biomaterials, Institute for Biotechnology and Helmholtz-Institute for Biomedical Engineering, RWTH Aachen University, Pauwelsstraße 20, D-52074 Aachen, Germany.
| | - Vladimír Křen
- Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, CZ-14220 Prague 4, Czech Republic.
| |
Collapse
|
43
|
Nguyen MN, Ziemann M, Kiriazis H, Su Y, Thomas Z, Lu Q, Donner DG, Zhao WB, Rafehi H, Sadoshima J, McMullen JR, El-Osta A, Du XJ. Galectin-3 deficiency ameliorates fibrosis and remodeling in dilated cardiomyopathy mice with enhanced Mst1 signaling. Am J Physiol Heart Circ Physiol 2018; 316:H45-H60. [PMID: 30387702 DOI: 10.1152/ajpheart.00609.2018] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Dilated cardiomyopathy (DCM) is a major cause of heart failure without effective therapy. Fibrogenesis plays a key role in the development of DCM, but little is known of the expression of the profibrotic factor galectin-3 (Gal-3) and its role in DCM pathophysiology. In a mouse DCM model with transgenic (TG) overexpression of mammalian sterile 20-like kinase 1 (Mst1), we studied Gal-3 expression and effects of the Gal-3 inhibitor modified citrus pectin (MCP) or Gal-3 gene knockout (KO). Gal-3 deletion in TG mice (TG/KO) was achieved by crossbreeding Mst1-TG mice with Gal-3 KO mice. The DCM phenotype was assessed by echocardiography and micromanometry. Cardiac expression of Gal-3 and fibrosis were determined. The cardiac transcriptome was profiled by RNA sequencing. Mst1-TG mice at 3-8 mo of age exhibited upregulated expression of Gal-3 by ~40-fold. TG mice had dilatation of cardiac chambers, suppressed left ventricular (LV) ejection fraction, poor LV contractility and relaxation, a threefold increase in LV collagen content, and upregulated fibrotic genes. Four-month treatment with MCP showed no beneficial effects. Gal-3 deletion in Mst1-TG mice attenuated chamber dilatation, organ congestion, and fibrogenesis. RNA sequencing identified profound disturbances by Mst1 overexpression in the cardiac transcriptome, which largely remained in TG/KO hearts. Gal-3 deletion in Mst1-TG mice, however, partially reversed the dysregulated transcriptional signaling involving extracellular matrix remodeling and collagen formation. We conclude that cardiac Mst1 activation leads to marked Gal-3 upregulation and transcriptome disturbances in the heart. Gal-3 deficiency attenuated cardiac remodeling and fibrotic signaling. NEW & NOTEWORTHY We found in a transgenic mouse dilated cardiomyopathy (DCM) model a pronounced upregulation of galectin-3 in cardiomyocytes. Galectin-3 gene deletion reduced cardiac fibrosis and fibrotic gene profiles and ameliorated cardiac remodeling and dysfunction. These benefits of galectin-3 deletion were in contrast to the lack of effect of treatment with the galectin-3 inhibitor modified citrus pectin. Our study suggests that suppression of galectin-3 mRNA expression could be used to treat DCM with high cardiac galectin-3 content.
Collapse
Affiliation(s)
- My-Nhan Nguyen
- Baker Heart and Diabetes Institute , Melbourne, Victoria , Australia.,Central Clinical School, Monash University , Melbourne, Victoria , Australia
| | - Mark Ziemann
- Department of Diabetes, Monash University , Melbourne, Victoria , Australia
| | - Helen Kiriazis
- Baker Heart and Diabetes Institute , Melbourne, Victoria , Australia
| | - Yidan Su
- Baker Heart and Diabetes Institute , Melbourne, Victoria , Australia
| | - Zara Thomas
- Baker Heart and Diabetes Institute , Melbourne, Victoria , Australia
| | - Qun Lu
- Baker Heart and Diabetes Institute , Melbourne, Victoria , Australia.,Health Science Center, Xian Jiaotong University , Xian , People's Republic of China
| | - Daniel G Donner
- Baker Heart and Diabetes Institute , Melbourne, Victoria , Australia
| | - Wei-Bo Zhao
- Baker Heart and Diabetes Institute , Melbourne, Victoria , Australia
| | - Haloom Rafehi
- Department of Diabetes, Monash University , Melbourne, Victoria , Australia
| | - Junichi Sadoshima
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School , Newark, New Jersey
| | - Julie R McMullen
- Baker Heart and Diabetes Institute , Melbourne, Victoria , Australia.,Central Clinical School, Monash University , Melbourne, Victoria , Australia
| | - Assam El-Osta
- Department of Diabetes, Monash University , Melbourne, Victoria , Australia.,Prince of Wales Hospital, The Chinese University of Hong Kong , Shatin, Hong Kong Special Administrative Region , People's Republic of China
| | - Xiao-Jun Du
- Baker Heart and Diabetes Institute , Melbourne, Victoria , Australia.,Central Clinical School, Monash University , Melbourne, Victoria , Australia.,Health Science Center, Xian Jiaotong University , Xian , People's Republic of China
| |
Collapse
|
44
|
Ferrer MF, Scharrig E, Charo N, Rípodas AL, Drut R, Carrera Silva EA, Nagel A, Nally JE, Montes de Oca DP, Schattner M, Gómez RM. Macrophages and Galectin 3 Control Bacterial Burden in Acute and Subacute Murine Leptospirosis That Determines Chronic Kidney Fibrosis. Front Cell Infect Microbiol 2018; 8:384. [PMID: 30425972 PMCID: PMC6218566 DOI: 10.3389/fcimb.2018.00384] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 10/11/2018] [Indexed: 12/15/2022] Open
Abstract
Previous studies have suggested that macrophages may contribute to acute Leptospira dissemination, as well as having a major role in kidney fibrosis. Our aim was to characterize the role of macrophages and galectin 3 (Gal-3) on the survival, clinical course, bacterial burden, interstitial nephritis, and chronic kidney fibrosis in Leptospira interrogans serovar Copenhageni (LIC)-induced experimental murine leptospirosis. C57BL/6J mice depleted of macrophages by liposome-encapsulated clodronate treatment and infected with LIC presented a higher bacterial burden, had reduced subacute nephritis and enhanced chronic kidney fibrosis relative to untreated, infected mice. Moreover, LIC infection in mice whose Gal-3 was disrupted (Lgals3−/–) had a higher bacterial burden and enhanced subacute nephritis and chronic kidney fibrosis when compared to C57BL/6J wild-type mice. Chronic fibrosis did not correlate with higher transcription levels of TGF-β1 or IL-13 in the kidneys. Kidney fibrosis was found in chronically infected rats as well as in wild infected rats. On the other hand, human fibroblast cultures exhibited enhanced differentiation to myofibroblasts after treatment with LIC. Our results demonstrate that macrophages and Gal-3 play a critical role in controlling the LIC burden but has a minor role in subsequent fibrosis. Instead, kidney fibrosis was better correlated with bacterial burden. Taken together, our results do not support a role for macrophages to disseminate leptospires during acute infection, nor in chronic kidney fibrosis.
Collapse
Affiliation(s)
- María F Ferrer
- Laboratory of Animal Viruses, Institute of Biotechnology and Molecular Biology, UNLP-CONICET, La Plata, Argentina
| | - Emilia Scharrig
- Laboratory of Animal Viruses, Institute of Biotechnology and Molecular Biology, UNLP-CONICET, La Plata, Argentina
| | - Nancy Charo
- Laboratory of Experimental Thrombosis, Institute of Experimental Medicine, National Academy of Medicine-CONICET, Buenos Aires, Argentina
| | | | - Ricardo Drut
- Division of Pathology, Children Hospital "Superiora Sor María Ludovica", La Plata, Argentina
| | - Eugenio A Carrera Silva
- Laboratory of Experimental Thrombosis, Institute of Experimental Medicine, National Academy of Medicine-CONICET, Buenos Aires, Argentina
| | - Ariel Nagel
- Biotechnology Institute, National Institute of Agricultural Technology (INTA-CONICET), Buenos Aires, Argentina
| | - Jarlath E Nally
- Infectious Bacterial Diseases Research Unit, National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Pullman, WA, United States
| | - Daniela P Montes de Oca
- Ecology, Genetics and Evolution Department, Exact and Natural Sciences Faculty, and Ecology, Genetics and Evolution Institute of Buenos Aires, UBA-CONICET, Buenos Aires, Argentina
| | - Mirta Schattner
- Laboratory of Experimental Thrombosis, Institute of Experimental Medicine, National Academy of Medicine-CONICET, Buenos Aires, Argentina
| | - Ricardo M Gómez
- Laboratory of Animal Viruses, Institute of Biotechnology and Molecular Biology, UNLP-CONICET, La Plata, Argentina
| |
Collapse
|
45
|
Rivadeneyra L, Charó N, Kviatcovsky D, de la Barrera S, Gómez RM, Schattner M. Role of neutrophils in CVB3 infection and viral myocarditis. J Mol Cell Cardiol 2018; 125:149-161. [PMID: 30393107 DOI: 10.1016/j.yjmcc.2018.08.029] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 08/05/2018] [Indexed: 12/11/2022]
Abstract
Coxsackievirus B3 (CVB3) is a globally prevalent enterovirus of the Picornaviridae family that is frequently associated with viral myocarditis (VM). Neutrophils, as first responders, may be key cells in determining viral disease outcomes; however, neutrophils have been poorly studied with respect to viral infection. Although neutrophils have been ascribed a relevant role in early cardiac inflammation, their precise role in CVB3 infection has not yet been evaluated. In this study, we aimed to determine if the interaction between human neutrophils and CVB3 could lead to viral replication and/or modulation of neutrophil survival and biological functions, and whether neutrophil depletion in a murine model has a beneficial or harmful effect on CVB3 infection. Our results show that CVB3 interacted with but did not replicate in human neutrophils. Neutrophils recognized CVB3 mainly through endosomal TLR-8, and infection triggered NFκB activation. Virus internalization resulted in increased cell survival, up-regulation of CD11b, enhanced adhesion to fibrinogen and fibronectin, and the secretion of IL-6, IL-1β, TNF-α, and IL-8. Supernatants from infected neutrophils exerted chemotactic activity partly mediated by IL-8. The infected neutrophils released myeloperoxidase and triggered neutrophil extracellular trap formation in the presence of TNF-α. In mice infected with CVB3, viral RNA was detected in neutrophils as well as in mononuclear cells. After neutrophil depletion, mice showed reduced VM reflected by a reduction in viral titers, cell exudates, and CCL-2 mRNA levels, as well as the abrogation of reactive cardiomyocyte hypertrophy. Our results indicate that neutrophils have relevant direct and indirect roles in the pathogenesis of CVB3-induced VM.
Collapse
Affiliation(s)
- Leonardo Rivadeneyra
- Laboratory of Experimental Thrombosis, Institute of Experimental Medicine-CONICET-ANM, Buenos Aires, Argentina.
| | - Nancy Charó
- Laboratory of Experimental Thrombosis, Institute of Experimental Medicine-CONICET-ANM, Buenos Aires, Argentina
| | - Denise Kviatcovsky
- Laboratory of Immunology of Respiratory Diseases, Institute of Experimental Medicine-CONICET-ANM, Buenos Aires, Argentina
| | - Silvia de la Barrera
- Laboratory of Immunology of Respiratory Diseases, Institute of Experimental Medicine-CONICET-ANM, Buenos Aires, Argentina.
| | - Ricardo Martín Gómez
- Biotechnology and Molecular Biology Institute, CONICET-UNLP, La Plata, Argentina.
| | - Mirta Schattner
- Laboratory of Experimental Thrombosis, Institute of Experimental Medicine-CONICET-ANM, Buenos Aires, Argentina.
| |
Collapse
|
46
|
Meyer IS, Goetzke CC, Kespohl M, Sauter M, Heuser A, Eckstein V, Vornlocher HP, Anderson DG, Haas J, Meder B, Katus HA, Klingel K, Beling A, Leuschner F. Silencing the CSF-1 Axis Using Nanoparticle Encapsulated siRNA Mitigates Viral and Autoimmune Myocarditis. Front Immunol 2018; 9:2303. [PMID: 30349538 PMCID: PMC6186826 DOI: 10.3389/fimmu.2018.02303] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 09/17/2018] [Indexed: 12/11/2022] Open
Abstract
Myocarditis is an inflammatory disease of the heart muscle most commonly caused by viral infection and often maintained by autoimmunity. Virus-induced tissue damage triggers chemokine production and, subsequently, immune cell infiltration with pro-inflammatory and pro-fibrotic cytokine production follows. In patients, the overall inflammatory burden determines the disease outcome. Following the aim to define specific molecules that drive both immunopathology and/or autoimmunity in inflammatory heart disease, here we report on increased expression of colony stimulating factor 1 (CSF-1) in patients with myocarditis. CSF-1 controls monocytes originating from hematopoietic stem cells and subsequent progenitor stages. Both, monocytes and macrophages are centrally involved in mediating tissue damage and fibrotic scarring in the heart. CSF-1 influences monocytes via engagement of CSF-1 receptor, and it is also produced by cells of the mononuclear phagocyte system themselves. Based on this, we sought to modulate the virus-triggered inflammatory response in an experimental model of Coxsackievirus B3-induced myocarditis by silencing the CSF-1 axis in myeloid cells using nanoparticle-encapsulated siRNA. siCSF-1 inverted virus-mediated immunopathology as reflected by lower troponin T levels, a reduction of accumulating myeloid cells in heart tissue and improved cardiac function. Importantly, pathogen control was maintained and the virus was efficiently cleared from heart tissue. Since viral heart disease triggers heart-directed autoimmunity, in a second approach we investigated the influence of CSF-1 upon manifestation of heart tissue inflammation during experimental autoimmune myocarditis (EAM). EAM was induced in Balb/c mice by immunization with a myocarditogenic myosin-heavy chain-derived peptide dissolved in complete Freund's adjuvant. siCSF-1 treatment initiated upon established disease inhibited monocyte infiltration into heart tissue and this suppressed cardiac injury as reflected by diminished cardiac fibrosis and improved cardiac function at later states. Mechanistically, we found that suppression of CSF-1 production arrested both differentiation and maturation of monocytes and their precursors in the bone marrow. In conclusion, during viral and autoimmune myocarditis silencing of the myeloid CSF-1 axis by nanoparticle-encapsulated siRNA is beneficial for preventing inflammatory tissue damage in the heart and preserving cardiac function without compromising innate immunity's critical defense mechanisms.
Collapse
Affiliation(s)
- Ingmar Sören Meyer
- Internal Medicine III, University Hospital Heidelberg, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg-Mannheim, Heidelberg, Germany
| | - Carl Christoph Goetzke
- Institute of Biochemistry, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Meike Kespohl
- Institute of Biochemistry, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Martina Sauter
- Cardiopathology, Institute for Pathology and Neuropathology, University Hospital Tuebingen, Tuebingen, Germany
| | - Arnd Heuser
- Max-Delbrueck-Center for Molecular Medicine Berlin, Berlin, Germany
| | - Volker Eckstein
- Internal Medicine V, University Hospital Heidelberg, Heidelberg, Germany
| | | | - Daniel G Anderson
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States.,Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA, United States.,Department of Chemical Engineering, Massachusetts Institute of Technology (MIT), Cambridge, MA, United States
| | - Jan Haas
- Internal Medicine III, University Hospital Heidelberg, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg-Mannheim, Heidelberg, Germany
| | - Benjamin Meder
- Internal Medicine III, University Hospital Heidelberg, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg-Mannheim, Heidelberg, Germany
| | - Hugo Albert Katus
- Internal Medicine III, University Hospital Heidelberg, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg-Mannheim, Heidelberg, Germany
| | - Karin Klingel
- Cardiopathology, Institute for Pathology and Neuropathology, University Hospital Tuebingen, Tuebingen, Germany
| | - Antje Beling
- Institute of Biochemistry, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Florian Leuschner
- Internal Medicine III, University Hospital Heidelberg, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg-Mannheim, Heidelberg, Germany
| |
Collapse
|
47
|
Kovacevic MM, Pejnovic N, Mitrovic S, Jovicic N, Petrovic I, Arsenijevic N, Lukic ML, Ljujic B. Galectin-3 deficiency enhances type 2 immune cell-mediated myocarditis in mice. Immunol Res 2018; 66:491-502. [PMID: 30099675 DOI: 10.1007/s12026-018-9013-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Experimental autoimmune myocarditis (EAM) is a mouse model of immune-mediated myocarditis and cardiomyopathy. The role of Galectin-3 (Gal-3), a β-galactoside-binding lectin, in autoimmune myocarditis has not been studied. Therefore, the aim of this study was to delineate the role of Gal-3 in myosin peptide-induced autoimmune myocarditis in mice. EAM was induced in relatively resistant C57BL/6J mice (wild type, WT) and in mice with a targeted deletion of Gal-3 gene (Gal-3KO) by immunization with myosin peptide MyHCα334-352. Gal-3KO mice developed more severe myocarditis and more pronounced heart hypertrophy than WT mice. Increased infiltration of CD45+ leucocytes, CD3+ T cells, F4/80+ macrophages, and eosinophils was observed in hearts of Gal-3KO mice compared to WT mice on day 21 after EAM induction. Moreover, hearts of Gal-3KO mice had more T helper type 2 (Th2) cells, alternatively activated M2 macrophages, higher amounts of IgG deposits, and higher serum levels of IL-4 and IL-33 than WT mice. Ablation of Gal-3 in Th1-dominant C57BL/6J mice that are relatively resistant to EAM resulted in more severe disease characterized by type 2 cardiac inflammation. The complex effects of Gal-3 on EAM progression might be important in the consideration of therapeutic options for the treatment of EAM.
Collapse
Affiliation(s)
- Marina Miletic Kovacevic
- Department of Histology and Embryology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Nada Pejnovic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
- Department of Pathophysiology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Slobodanka Mitrovic
- Department of Pathology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Nemanja Jovicic
- Department of Histology and Embryology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Ivica Petrovic
- Department of Pathophysiology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Nebojsa Arsenijevic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Miodrag L Lukic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia.
| | - Biljana Ljujic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
- Department of Genetics, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| |
Collapse
|
48
|
Zivlas C, Triposkiadis F, Psarras S, Giamouzis G, Skoularigis I, Chryssanthopoulos S, Kapelouzou A, Ramcharitar S, Barnes E, Papasteriadis E, Cokkinos D. Cystatin C and galectin-3 as therapeutic targets in heart failure. Ther Adv Cardiovasc Dis 2018; 12:233-235. [PMID: 29848191 DOI: 10.1177/1753944718778470] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Affiliation(s)
- Christos Zivlas
- Wiltshire Cardiac Centre, Great Western Hospitals NHS Foundation Trusts, Marlborough Road, Swindon, SN3 6BB, UK
| | | | - Stelios Psarras
- Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Gregory Giamouzis
- Department of Cardiology, Larissa University Hospital, Larissa, Greece
| | | | | | | | - Steve Ramcharitar
- Wiltshire Cardiac Centre, Great Western Hospitals NHS Foundation Trusts, Swindon, UK
| | - Edward Barnes
- Wiltshire Cardiac Centre, Great Western Hospitals NHS Foundation Trusts, Swindon, UK
| | | | - Dennis Cokkinos
- Biomedical Research Foundation, Academy of Athens, Athens, Greece
| |
Collapse
|
49
|
Nguyen MN, Su Y, Kiriazis H, Yang Y, Gao XM, McMullen JR, Dart AM, Du XJ. Upregulated galectin-3 is not a critical disease mediator of cardiomyopathy induced by β2-adrenoceptor overexpression. Am J Physiol Heart Circ Physiol 2018; 314:H1169-H1178. [DOI: 10.1152/ajpheart.00337.2017] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Preclinical studies have demonstrated that anti-galectin-3 (Gal-3) interventions are effective in attenuating cardiac remodeling, fibrosis, and dysfunction. We determined, in a transgenic (TG) mouse model of fibrotic cardiomyopathy, whether Gal-3 expression was elevated and whether Gal-3 played a critical role in disease development. We studied mice with fibrotic cardiomyopathy attributable to cardiac overexpression of human β2-adrenoceptors (β2-TG). Cardiac expression levels of Gal-3 and fibrotic or inflammatory genes were determined. The effect of Gal-3 inhibition in β2-TG mice was studied by treatment with Gal-3 inhibitors ( N-acetyllactosamine and modified citrus pectin) or by deletion of Gal-3 through crossing β2-TG and Gal-3 knockout mice. Changes in cardiomyopathy phenotypes were assessed by echocardiography and biochemical assays. In β2-TG mice at 3, 6, and 9 mo of age, upregulation of Gal-3 expression was observed at mRNA (~6- to 15-fold) and protein (~4- to 8-fold) levels. Treatment of β2-TG mice with N-acetyllactosamine (3 wk) or modified citrus pectin (3 mo) did not reverse cardiac fibrosis, inflammation, and cardiomyopathy. Similarly, Gal-3 gene deletion in β2-TG mice aged 3 and 9 mo did not rescue the cardiomyopathy phenotype. In conclusion, the β2-TG model of cardiomyopathy showed a robust upregulation of Gal-3 that correlated with disease severity, but Gal-3 inhibitors or Gal-3 gene deletion had no effect in halting myocardial fibrosis, remodeling, and dysfunction. Gal-3 may not be critical for cardiac fibrogenesis and remodeling in this cardiomyopathy model. NEW & NOTEWORTHY We showed a robust upregulation of cardiac galectin-3 (Gal-3) expression in a mouse model of cardiomyopathy attributable to cardiomyocyte-restricted transgenic activation of β2-adrenoceptors. However, pharmacological and genetic inhibition of Gal-3 did not confer benefit in this model, implying that Gal-3 may not be a critical disease mediator of cardiac remodeling in this model.
Collapse
Affiliation(s)
- My-Nhan Nguyen
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Yidan Su
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Helen Kiriazis
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Yan Yang
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Alfred Heart Centre, The Alfred Hospital, Melbourne, Victoria, Australia
| | - Xiao-Ming Gao
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Julie R. McMullen
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Anthony M. Dart
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Central Clinical School, Monash University, Melbourne, Victoria, Australia
- Alfred Heart Centre, The Alfred Hospital, Melbourne, Victoria, Australia
| | - Xiao-Jun Du
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Central Clinical School, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
50
|
Gou W, Zhang Z, Yang C, Li Y. MiR-223/Pknox1 axis protects mice from CVB3-induced viral myocarditis by modulating macrophage polarization. Exp Cell Res 2018. [PMID: 29524390 DOI: 10.1016/j.yexcr.2018.03.004] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Macrophage polarization plays a crucial role in regulating myocardial inflammation and injuries of coxsackievirus B3 (CVB3)-induced viral myocarditis (VM). It has been reported that miR-223 is a potent regulator of inflammatory responses that involved in macrophage polarization. However, the functional roles of miR-223 in CVB3-induced VM still remain unknown. Here, we found that miR-223 expression was significantly down-regulated in heart tissues and heart-infiltrating macrophages of CVB3-infected mice. Up-regulation of miR-223 in vivo protected the mice against CVB3-induced myocardial injuries characterized by the increased body weight and survival, enhanced left ventricular ejection fraction (LVEF) and left ventricular fractional shortening (LVFS), relieved inflammation, depressed creatine kinase-MB (CK-MB), lactate dehydrogenase (LDH) and aspartate transaminase (AST) levels, reduced production of interferon (IFN)-γ, interleukin (IL)- 6 as well as increased IL-10. We subsequently found that miR-233 up-regulation significantly suppressed the expression of M1 markers (iNOS, TNF-α and CD 86), and promoted the expression of M2 markers (Arginase-1, Fizz-1 and CD 206) in vivo and in vitro. Furthermore, we confirmed that miR-223 directly targeted Pknox1 to inhibit its expression, and the expression of Pknox1 was inversely correlated with miR-223 expression in heart tissues and heart-infiltrating macrophages of CVB3-infected mice. Gain-of-function analyses indicated that Pknox1 overexpression partially reversed the polarization phenotypes regulated by miR-223 overexpression. Taken together, the data suggest that miR-223 protects against CVB3-induced inflammation and myocardial damage, which may partly attribute to the regulation of macrophage polarization via targeting Pknox1.
Collapse
Affiliation(s)
- Weihui Gou
- PICU, First Hospital of Jilin University, 71 Xinmin Street, Chaoyang District, Changchun, Jilin 130021, China
| | - Zhen Zhang
- PICU, First Hospital of Jilin University, 71 Xinmin Street, Chaoyang District, Changchun, Jilin 130021, China
| | - Chunfeng Yang
- PICU, First Hospital of Jilin University, 71 Xinmin Street, Chaoyang District, Changchun, Jilin 130021, China
| | - Yumei Li
- PICU, First Hospital of Jilin University, 71 Xinmin Street, Chaoyang District, Changchun, Jilin 130021, China.
| |
Collapse
|