1
|
Wang X, Liu Q, Cheng P, Yang T, Zhao T, Liu M, Dai E, Sha W, Yuan J, Rong J, Qu H, Zhou H. LuQi formula ameliorates pressure overload-induced heart failure by regulating macrophages and regulatory T cells. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156527. [PMID: 40118747 DOI: 10.1016/j.phymed.2025.156527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 02/11/2025] [Accepted: 02/15/2025] [Indexed: 03/23/2025]
Abstract
BACKGROUND Inflammatory macrophages in failing myocardium secrete CCL17, which targets CCR4 in immunosuppressive Tregs and inhibits the intracellular second messenger ARRB2-mediated cardiac chemotaxis. Traditional Chinese medicine (TCM) LuQi formula (LQF) is safe and effective in treating heart failure (HF). This study aims to elucidate the cardioprotective mechanism of LQF through its modulation of cardiac macrophages and Tregs. METHODS In vivo, the HF mouse model was established via transverse aortic constriction (TAC), with the superagonistic anti-CD28 monoclonal antibody (CD28-SA)-induced Tregs expansion as a positive control. Proteomics analysis elucidated the core link of LQF in anti-HF. In vitro, bone marrow-derived macrophages (BMDMs) were isolated, and Naive CD4+T cells were sorted and stimulated to differentiate into Tregs. The pharmacological mechanism of LQF was confirmed through histological and molecular biology experiments. RESULTS Proteomics reveals that LQF modulates the immune microenvironment of failing myocardium. We revealed that LQF inhibited cardiac inflammatory macrophage infiltration and NF-κB (p50, p65)/CCL17 axis expression, and promoted cardiac Tregs recruitment against HF, with the comparable efficacy of CD-SA28-induced Tregs expansion. Mechanistically, LQF inhibited the NF-κB activator 1-induced NF-κB (p50, p65)/CCL17 axis overexpression, and JSH-23 (NF-κB Inhibitor) abolished NF-κB (p50, p65)/CCL17 axis expression in inflammatory macrophages. Furthermore, the inhibition of CCL17 expression in inflammatory macrophages by LQF was found to be mediated by NF-κB (p50, p65). LQF concentration-dependently promoted Tregs CD73/Foxp3 axis expression, enhancing Tregs immunosuppressive function. LQF activated CCR4-ARRB2 complex and CCR4/ARRB2 axis expression in Tregs. Although AZD2098 (CCR4 Inhibitor) blocked CCR4 expression and CCR4-ARRB2 complex, LQF promoted ARRB2-mediated Tregs cardiac chemotaxis independent of the CCR4. We revealed that NF-κB p50SEP337-CCL17, NF-κB p65SEP536-CCL17, and CCR4-ARRB2 highly bound subunit interface targets. Molecular docking analysis demonstrated that the LQF's active ingredients exhibit good binding affinity with the NF-κB (p50, p65) /CCL17 axis in macrophages and Foxp3 in Tregs. CONCLUSION LQF has the potential to enhance the cardiac immune microenvironment and effectively prevent and treat HF by modulating both innate and adaptive immune responses. It achieves this by inhibiting the infiltration of inflammatory macrophages, suppressing the NF-κB (p50, p65)/CCL17 axis, and promoting Tregs recruitment. The active ingredients of LQF provide valuable candidate compounds for developing new anti-HF drugs. Furthermore, CD-28SA-induced Tregs expansion showed cardioprotective effects in TAC-induced non-ischemic HF models.
Collapse
Affiliation(s)
- Xinting Wang
- Institute of Cardiovascular Disease of Integrated Traditional Chinese and Western medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Qian Liu
- Institute of Cardiovascular Disease of Integrated Traditional Chinese and Western medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Peipei Cheng
- Institute of Cardiovascular Disease of Integrated Traditional Chinese and Western medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Tianshu Yang
- Department of Cardiovascular Disease, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, ShangHai 200040, China
| | - Tingyao Zhao
- Guanghua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200050, China
| | - Meng Liu
- Guanghua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200050, China
| | - Enrui Dai
- Institute of Cardiovascular Disease of Integrated Traditional Chinese and Western medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Wanjing Sha
- Institute of Cardiovascular Disease of Integrated Traditional Chinese and Western medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jinfeng Yuan
- Institute of Cardiovascular Disease of Integrated Traditional Chinese and Western medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jingfeng Rong
- Institute of Cardiovascular Disease of Integrated Traditional Chinese and Western medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Huiyan Qu
- Institute of Cardiovascular Disease of Integrated Traditional Chinese and Western medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Hua Zhou
- Institute of Cardiovascular Disease of Integrated Traditional Chinese and Western medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
2
|
Qi W, Fu L, Wu Q, Yang P. Novel biomarkers related to mitochondrial permeability transition driven-necrosis in hypertrophic cardiomyopathy. J Cardiol 2025:S0914-5087(25)00106-6. [PMID: 40306461 DOI: 10.1016/j.jjcc.2025.04.008] [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: 11/25/2024] [Revised: 04/17/2025] [Accepted: 04/22/2025] [Indexed: 05/02/2025]
Abstract
BACKGROUND The role of mitochondrial permeability transition driven necrosis-related genes (MPTDN-RGs) in hypertrophic cardiomyopathy (HCM) is unclear. This investigation combined transcriptomics and Mendelian randomization (MR) analysis to explore the association of MPTDN-RGs with HCM. METHODS GSE36961 (training set), GSE141910 (validation set), and GSE174691 (single-cell dataset) were retrieved from Gene Expression Omnibus (GEO) database. This study is based on scRNA-seq and transcriptome sequencing (mRNA Sequencing, mRNA-seq) data combined with MR, and use MPTDN-RGs to identify genes of HCM. RESULTS Based on 51 interaction genes overlapped by 250 module genes and 154 differentially expressed genes, the top 10 genes within protein-protein interaction (PPI) core network were regarded as candidate genes. ITGB2 and STAT3 were screened out as genes by multiple analysis methods. MR results revealed that ITGB2 was a risk factor, while STAT3 was a protective factor for HCM. Gene set enrichment analysis (GSEA) indicated that ITGB2 and STAT3 were involved in complement and coagulation cascade. Moreover, ITGB2 had the strongest positive and significant correlations with myeloid-derived suppressor cells and chemokine receptor. Single cell analysis showed that STAT3 was highly expressed in endothelial cells, while ITGB2 was significantly greater in dendritic cells. During the process of differentiation, the expression of ITGB2 and STAT3 were decreased, and dendritic cells gradually differentiated and matured to play a role in immune function. CONCLUSION To our knowledge, this is the first study to identify the novel genes related to MPTDN in HCM by combining transcriptomics and MR analysis. Two key genes play a critical role in HCM. ITGB2 and STAT3 deserve further investigation as potential therapeutic targets for HCM.
Collapse
Affiliation(s)
- Wenqiang Qi
- Department of Cardiovascular Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China; Department of Cardiovascular Medicine, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
| | - Linghua Fu
- Department of Cardiovascular Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Qinghua Wu
- Department of Cardiovascular Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China.
| | - Pingping Yang
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China; Jiangxi Provincial Sub-center of National Clinical Medical Research Center for Metabolic Diseases, Nanchang, Jiangxi, China.
| |
Collapse
|
3
|
Mikolajczyk TP, Śliwa T, Guzik TJ. Immunometabolic switch in hypertension: how dendritic cell mineralocorticoid receptors drive Th17 polarization and blood pressure control. Eur Heart J 2025; 46:1352-1354. [PMID: 39842875 PMCID: PMC11973550 DOI: 10.1093/eurheartj/ehae863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2025] Open
Affiliation(s)
- Tomasz P Mikolajczyk
- Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
- Center for Medical Genomics OMICRON, Jagiellonian University Medical College, Krakow, Poland
| | - Tomasz Śliwa
- Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Tomasz J Guzik
- Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
- Center for Medical Genomics OMICRON, Jagiellonian University Medical College, Krakow, Poland
- BHF Centre for Research Excellence, Centre for Cardiovascular Sciences, The University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| |
Collapse
|
4
|
Xiang L, Yin G, Gong Z, Lv X, Feng C, Liu L, Abdu FA, Shi T, Zhang W, Alifu J, Xu X, Dai Y, Che W, Weng X. IL-22 Attenuates Pressure Overload-Induced Heart Failure and Inflammation. J Cardiovasc Transl Res 2025:10.1007/s12265-025-10613-2. [PMID: 40195212 DOI: 10.1007/s12265-025-10613-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Accepted: 03/25/2025] [Indexed: 04/09/2025]
Abstract
Heart failure (HF) due to left ventricular (LV) dysfunction remains a major global health challenge, with inflammation driving its progression under chronic pressure overload, such as hypertension. This study explored the role of interleukin-22 (IL-22), a cytokine associated with tissue protection, in HF induced by transverse aortic constriction (TAC). IL-22 knockout (KO) mice exhibited exacerbated HF, marked by worsened LV hypertrophy, heightened inflammation, and impaired cardiac function compared to wild-type controls. Conversely, treatment with recombinant IL-22Fc improved LV function, reduced inflammatory cell infiltration, and alleviated cardiac remodeling and inflammation. These findings demonstrate that IL-22 plays a critical role in regulating inflammation and cardiac remodeling in pressure overload-induced HF. Targeting IL-22 may represent a promising therapeutic strategy to alleviate HF progression and associated pulmonary complications.
Collapse
Affiliation(s)
- Lanqing Xiang
- Department of Cardiology, Clinical Medical College of Shanghai Tenth People'S Hospital, Nanjing Medical University, Shanghai, China
- Department of Cardiology, Shanghai Tenth People'S Hospital, Tongji University School of Medicine, Shanghai, China
| | - Guoqing Yin
- Department of Cardiology, Shanghai Tenth People'S Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zifan Gong
- The Second Clinical Medical School of Nanjing Medical University, Nanjing, China
| | - Xian Lv
- Department of Cardiology, Shanghai Tenth People'S Hospital, Tongji University School of Medicine, Shanghai, China
| | - Cailin Feng
- Department of Cardiology, Shanghai Tenth People'S Hospital, Tongji University School of Medicine, Shanghai, China
| | - Lu Liu
- Department of Cardiology, Shanghai Tenth People'S Hospital, Tongji University School of Medicine, Shanghai, China
| | - Fuad A Abdu
- Department of Cardiology, Shanghai Tenth People'S Hospital, Tongji University School of Medicine, Shanghai, China
| | - Tingting Shi
- Department of Cardiology, Shanghai Tenth People'S Hospital, Tongji University School of Medicine, Shanghai, China
| | - Wen Zhang
- Department of Cardiology, Shanghai Tenth People'S Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jiasuer Alifu
- Department of Cardiology, Shanghai Tenth People'S Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiaojiang Xu
- Department of Pathology & Laboratory Medicine, School of Medicine, Tulane University, New Orleans, LA, 70112, USA
| | - Yuxiang Dai
- Department of Cardiology, State Key Laboratory of Cardiovascular Diseases, Shanghai Institute of Cardiovascular Diseases, NHC Key Laboratory of Ischemic Heart Diseases China, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Wenliang Che
- Department of Cardiology, Clinical Medical College of Shanghai Tenth People'S Hospital, Nanjing Medical University, Shanghai, China.
- Department of Cardiology, Shanghai Tenth People'S Hospital, Tongji University School of Medicine, Shanghai, China.
- Department of Cardiology, Shanghai Tenth People's Hospital Chongming Branch, Shanghai, China.
| | - Xinyu Weng
- Department of Cardiology, State Key Laboratory of Cardiovascular Diseases, Shanghai Institute of Cardiovascular Diseases, NHC Key Laboratory of Ischemic Heart Diseases China, Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
5
|
Wang YL, Zhu H, Pan YT, Shang D, Du LJ, Bai L, Zhu SW, Lin WZ, Zhang XY, Lu HX, Bi C, Liu Y, Liu Y, Xiao H, Qian YC, Zhou B, Li RG, Duan SZ. Dendritic cell mineralocorticoid receptor controls blood pressure by regulating T helper 17 differentiation: role of the Plcβ1/4-Stat5-NF-κB pathway. Eur Heart J 2025; 46:1335-1351. [PMID: 39498862 DOI: 10.1093/eurheartj/ehae670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 01/31/2024] [Accepted: 09/19/2024] [Indexed: 11/07/2024] Open
Abstract
BACKGROUND AND AIMS Dendritic cells (DCs) are closely related to blood pressure (BP) regulation. Mineralocorticoid receptor (MR) is an important drug target for antihypertensive treatment. However, the role of DC MR in the pathogenesis of hypertension has not been fully elucidated. This study aimed to determine the role of DC MR in BP regulation and to explore the mechanism. METHODS Renal biopsy and peripheral blood samples were collected from hypertensive patients (HTN) for immunostaining and flow cytometry. Dendritic cell MR knockout (DCMRKO) mice, DC MR overexpressing (DCMROV) mice, DCMROV/IL-17A knockout (DCMROV/IL-17AKO) mice and finerenone-treated C57BL/6 mice were infused with angiotensin II (Ang II) to establish hypertensive models. Western blotting, chromatin immunoprecipitation, co-immunoprecipitation, and in vivo DC depletion or adoptive transfer were used to delineate the functional importance of DC MR in hypertension development. RESULTS Mineralocorticoid receptor antagonists (spironolactone and finerenone) suppressed DC aggregation and activation, as well as hypertension in HTN and mice. Compared with littermate control (LC) mice, dendritic cell MR knockout mice had strikingly decreased BPs and attenuated target organ damage after Ang II infusion. Flow cytometry showed that DC MR deficiency mitigated Ang II-induced DC activation and T helper 17 (Th17) cell differentiation. RNA sequencing revealed that MR-deficient DCs had elevated expression of Plcβ1 and Plcβ4, knockdown of which reversed the inhibitory effect of MR deficiency on DC activation and Th17 differentiation. Adoptive transfer of MR-deficient DCs protected Ang II-induced hypertension, whereas knockdown of Plcβ1/4 eliminated the protective effects. At the molecular level, MR negatively regulated Plcβ1/4, which recruited SHP-1 to inactivate of Stat5 activity, resulting in enhanced NF-κB activation and Th17 polarization. Furthermore, DCMROV mice manifested more elevated BPs and target organ damage than control mice after Ang II infusion, and these differences were abolished in DCMROV/IL-17AKO mice. Finally, MR antagonists decreased the aggregation of Th17 in HTN and mice. CONCLUSIONS Dendritic cell MR plays important roles in the pathogenesis of hypertension by regulating Th17 through Plcβ1/4-Stat5-NF-κB signalling, and blockade of DC MR is beneficial for treating hypertension.
Collapse
Affiliation(s)
- Yong-Li Wang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, 241 West Huaihai Road, Shanghai 200030, China
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, 166 North Qiutao Rd, Hangzhou 310000, China
| | - Hong Zhu
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200123, China
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai 200125, China
| | - Yi-Tong Pan
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, 241 West Huaihai Road, Shanghai 200030, China
| | - Da Shang
- Division of Nephrology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Lin-Juan Du
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai 200125, China
| | - Lan Bai
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai 200125, China
| | - Shi-Wei Zhu
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, 241 West Huaihai Road, Shanghai 200030, China
| | - Wen-Zhen Lin
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai 200125, China
| | - Xing-Yu Zhang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, 241 West Huaihai Road, Shanghai 200030, China
| | - Hai-Xia Lu
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai 200125, China
| | - Chao Bi
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai 200125, China
| | - Yuan Liu
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai 200125, China
| | - Yan Liu
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai 200125, China
| | - Hui Xiao
- Key Laboratory of Immune Response and Immunotherapy, Shanghai Institute of Immunity and Infection, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200032, China
| | - You-Cun Qian
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Bin Zhou
- CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Ruo-Gu Li
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, 241 West Huaihai Road, Shanghai 200030, China
| | - Sheng-Zhong Duan
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, 166 North Qiutao Rd, Hangzhou 310000, China
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai 200125, China
- State Key Laboratory of Transvascular Implantation Devices, The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou 310000, China
| |
Collapse
|
6
|
Jiang Y, Liu Y, Huang H, Zhao T, Zhao Z, Gao Y. Effect of RAS mutations and related immune characteristics on the prognosis of patients with MSI-H/dMMR colorectal cancer. Cancer Immunol Immunother 2025; 74:78. [PMID: 39891700 PMCID: PMC11787098 DOI: 10.1007/s00262-024-03926-9] [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: 10/15/2024] [Accepted: 12/16/2024] [Indexed: 02/03/2025]
Abstract
PURPOSE Microsatellite high instability/deficient mismatch repair (MSI-H/dMMR) colorectal cancer (CRC) has an active tumor microenvironment, rendering it more sensitive to immune checkpoint inhibitors. Given that studies involving patients with MSI-H colorectal cancer with RAS mutations are scarce, we explored the effect of RAS mutations on the TME in patients with MSI-H/dMMR cancer and identified potential prognostic factors. METHODS Seventy-five patients diagnosed with MSI-H/dMMR colorectal cancer were retrospectively enrolled and divided into RAS-mutant and -wild-type groups. The expression levels of CD11c+ dendritic cells, CD4+ T cells, CD8+ T cells, and regulatory T cell (Treg) markers were detected, and prognostic factors were analyzed. RESULTS RAS-mutant MSI-H colorectal patients were more likely to have: (1) higher platelet values; (2) shorter disease-free survival (DFS); (3) lower infiltrated numbers of CD11c+ dendritic cells, CD4+ T lymphocytes, and CD8+ T lymphocytes, and higher infiltrated numbers of Foxp3+ Treg cells. In MSI-H/dMMR CRC patients: (1) the high CD11c + , CD4 +, and CD8 + cells infiltration group had longer DFS than the low-infiltration group, and Foxp3 + cells infiltration was not significantly correlated with DFS; (2) the RAS mutation status, number of CD11c+ cells infiltrated, and carbohydrate antigen 19-9 (CA19-9) level were the potential prognostic factors. CONCLUSION RAS mutations in patients with MSI-H/dMMR CRC may reduce the infiltration of CD11c+ dendritic cells, CD4+ T cells, and CD8+ T cells, and increase the infiltration of Foxp3+ Treg cells to affect the tumor microenvironment of patients. RAS gene status, CD11c + cells infiltration, and CA19-9 level were potential prognostic factors for MSI-H/dMMR CRC.
Collapse
Affiliation(s)
- Yupeng Jiang
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Yuyao Liu
- Department of Oncology, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, 410004, China
| | - Hong Huang
- Guilin Medical University, Guilin, 541000, China
| | - Tiantian Zhao
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Zengyi Zhao
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Yawen Gao
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China.
| |
Collapse
|
7
|
Song M, Ivkov R, Korangath P. Dendritic cell activation by iron oxide nanoparticles depends on the extracellular environment. NANOSCALE ADVANCES 2024; 7:209-218. [PMID: 39569333 PMCID: PMC11575603 DOI: 10.1039/d4na00561a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 11/10/2024] [Indexed: 11/22/2024]
Abstract
Nanoparticles can exert immune modulating effects in a host depending on composition, mode of administration, and type of disease. Although the specific mechanisms of nanoparticle-induced immune responses remain unclear, their uptake by macrophages and other phagocytic innate immune cells is considered to be a key event. Our objective here was to ascertain if nanoparticle-mediated activation of dendritic cells (DCs) occurs in vitro or in vivo when exposed to hydroxyethyl starch-coated iron oxide nanoparticles. For the present studies, our choice of nanoparticles, animal model, and experimental design is motivated by our previously published observations that systemic exposure can induce antitumor adaptive immune responses in mouse models of metastatic breast cancer. Here, we began by assessing the potential toxicity of systemic exposure to commercially available starch-coated Bionized Nanoferrite® nanoparticles (BP) by measuring body weight, complete blood count, and enzyme parameters in healthy FVB/NJ mice after repeated BP dosing. We observed no evidence of toxicity at doses up to 25 mg Fe per mouse, five-fold higher than those used in subsequent in vivo experiments. We then measured the expression of surface maturation markers (CD86, MHC II) in DCs incubated with BP in vitro. Although DCs cultured with BP revealed high levels of nanoparticle uptake, neither JAWSII dendritic cells nor bone marrow derived dendritic cells (BMDCs) showed significant changes in marker expression to indicate stimulation of maturation and effector function. To assess whether BP interactions in vivo produced different effects, we analyzed CD80, CD86, and MHC II expression of DCs recovered from the livers, spleens, bone marrows, and lymph nodes of mice injected once with BP (5 mg Fe). Interestingly, only DCs in spleens and bone marrow cells responded to BP exposure. DCs recovered from other organs showed no evidence of increased activation. These findings highlight complex interactions between living systems and nanoparticles, and their potential to mediate context-specific and selective activation of innate immune cells. Our study also emphasizes that results obtained from in vitro experiments must be interpreted with caution, as they may not faithfully represent responses in living systems.
Collapse
Affiliation(s)
- Mason Song
- Department of Biomedical Engineering, Whiting School of Engineering, Johns Hopkins University Baltimore 21218 USA
| | - Robert Ivkov
- Department of Radiation Oncology and Molecular Radiation Sciences, School of Medicine, Johns Hopkins University 1550 Orleans Street, Cancer Research Building - II, Rm 416 Baltimore MD 21231 USA
- Department of Oncology, Sydney Kimmel Comprehensive Cancer Center, School of Medicine, Johns Hopkins University Baltimore MD 21231 USA
- Department of Mechanical Engineering, Whiting School of Engineering, Johns Hopkins University Baltimore 21218 USA
- Department of Materials Science and Engineering, Whiting School of Engineering, Johns Hopkins University Baltimore 21218 USA
| | - Preethi Korangath
- Department of Radiation Oncology and Molecular Radiation Sciences, School of Medicine, Johns Hopkins University 1550 Orleans Street, Cancer Research Building - II, Rm 416 Baltimore MD 21231 USA
- Department of Oncology, Sydney Kimmel Comprehensive Cancer Center, School of Medicine, Johns Hopkins University Baltimore MD 21231 USA
| |
Collapse
|
8
|
Bhattarai U, Xu R, He X, Pan L, Niu Z, Wang D, Zeng H, Chen JX, Clemmer JS, Chen Y. High selenium diet attenuates pressure overload-induced cardiopulmonary oxidative stress, inflammation, and heart failure. Redox Biol 2024; 76:103325. [PMID: 39197316 PMCID: PMC11399737 DOI: 10.1016/j.redox.2024.103325] [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: 06/27/2024] [Revised: 08/14/2024] [Accepted: 08/19/2024] [Indexed: 09/01/2024] Open
Abstract
Selenium (Se) deficiency is associated with the development of Keshan disease, a cardiomyopathy associated with massive cardiac immune cell infiltration that can lead to heart failure (HF). The purpose of this study was to determine whether high Se diet can attenuate systolic overload-induced cardiopulmonary inflammation and HF. Briefly, transverse aortic constriction (TAC)-induced cardiopulmonary oxidative stress, inflammation, left ventricular (LV) dysfunction, and pulmonary remodeling were determined in male mice fed with either high Se diet or normal Se diet. High Se diet had no detectable effect on LV structure and function in mice under control conditions, but high Se diet significantly protected mice from TAC-induced LV hypertrophy, dysfunction, increase of lung weight, and right ventricular hypertrophy. As compared with mice treated with normal Se diet, high Se diet also reduced TAC-induced LV cardiomyocyte hypertrophy, fibrosis, leukocyte infiltration, pulmonary inflammation, pulmonary fibrosis, and pulmonary micro-vessel muscularization. In addition, high Se diet significantly ameliorated TAC-induced accumulation and activation of pulmonary F4/80+ macrophages, and activation of dendritic cells. Interestingly, high Se diet also significantly attenuated TAC-induced activation of pulmonary CD4+ and CD8+ T cells. Moreover, we found that TAC caused a significant increase in cardiac and pulmonary ROS production, increases of 4-hydroxynonenal (4-HNE) and 3-nitrotyrosine (3-NT), as well as a compensatory increases of LV glutathione peroxidase 1 (GPX1) and 4 (GPX4) in mice fed with normal Se diet. Above changes were diminished in mice fed with high Se diet. Collectively, these data demonstrated that high Se diet significantly attenuated systolic pressure overload-induced cardiac oxidative stress, inflammation, HF development, and consequent pulmonary inflammation and remodeling.
Collapse
Affiliation(s)
- Umesh Bhattarai
- Department of Physiology and Biophysics, School of Medicine, University of Mississippi Medical Center, Jackson, MS, United States
| | - Rui Xu
- Department of Physiology and Biophysics, School of Medicine, University of Mississippi Medical Center, Jackson, MS, United States
| | - Xiaochen He
- Department of Physiology and Biophysics, School of Medicine, University of Mississippi Medical Center, Jackson, MS, United States
| | - Lihong Pan
- Department of Physiology and Biophysics, School of Medicine, University of Mississippi Medical Center, Jackson, MS, United States
| | - Ziru Niu
- Department of Physiology and Biophysics, School of Medicine, University of Mississippi Medical Center, Jackson, MS, United States
| | - Dongzhi Wang
- Department of Physiology and Biophysics, School of Medicine, University of Mississippi Medical Center, Jackson, MS, United States
| | - Heng Zeng
- Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center, Jackson, MS, United States
| | - Jian-Xiong Chen
- Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center, Jackson, MS, United States
| | - John S Clemmer
- Department of Physiology and Biophysics, School of Medicine, University of Mississippi Medical Center, Jackson, MS, United States
| | - Yingjie Chen
- Department of Physiology and Biophysics, School of Medicine, University of Mississippi Medical Center, Jackson, MS, United States.
| |
Collapse
|
9
|
Wang D, Weng X, Yue W, Shang L, Wei Y, Clemmer JS, Xu Y, Chen Y. CD8 T cells promote heart failure progression in mice with preexisting left ventricular dysfunction. Front Immunol 2024; 15:1472133. [PMID: 39324134 PMCID: PMC11422781 DOI: 10.3389/fimmu.2024.1472133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Accepted: 08/22/2024] [Indexed: 09/27/2024] Open
Abstract
Introduction Even under the standard medical care, patients with left ventricular (LV) failure or heart failure (HF) often progress to pulmonary hypertension and right ventricular (RV) hypertrophy. We previously showed that inflammation and regulatory T cells (Tregs) modulate HF progression in mice with preexisting LV failure. The main objective of this study is to determine the role of CD8+ T cells in modulating LV failure and the consequent pulmonary inflammation and RV hypertrophy in mice with preexisting LV failure. Methods Mice with LV failure produced by transverse aortic constriction (TAC) were randomized to depletion of cytotoxic CD8+ T cells, Tregs, or both using specific blocking antibodies. Cardiac function, lung inflammation, fibrosis, vascular remodeling, and right ventricular remodeling were determined. Results LV failure caused pulmonary inflammation, fibrosis, vascular remodeling, and RV hypertrophy. Depletion of CD8+ T cells significantly attenuated above changes in mice with preexisting LV failure. LV failure was associated with increased CD4+ and CD8+ T cell activation, and increased ratios of activated T cells to Tregs. Treg depletion exacerbated lung inflammation and HF progression, as well as lung CD4+ and CD8+ T cell infiltration and activation in HF mice. However, CD8+ T cells depletion rescue these mice from exacerbated lung inflammation and RV hypertrophy after Treg depletion. Discussion Our findings demonstrate an important role of CD8+ T cells in promoting pulmonary inflammation and RV hypertrophy in mice with preexisting LV failure. Depletion of CD8+ T cells also rescued HF mice from the exacerbated HF progression by Treg depletion.
Collapse
Affiliation(s)
- Dongzhi Wang
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, United States
- Department of Cardiology, Tenth People’s Hospital, Tongji University, Shanghai, China
| | - Xinyu Weng
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Lillehei Heart Institute, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Wenhui Yue
- Department of Cardiology, Tenth People’s Hospital, Tongji University, Shanghai, China
| | - Linlin Shang
- Department of Cardiology, Tenth People’s Hospital, Tongji University, Shanghai, China
- Department of Clinical Pharmacy, School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Yidong Wei
- Department of Cardiology, Tenth People’s Hospital, Tongji University, Shanghai, China
| | - John S. Clemmer
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, United States
| | - Yawei Xu
- Department of Cardiology, Tenth People’s Hospital, Tongji University, Shanghai, China
| | - Yingjie Chen
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, United States
- Lillehei Heart Institute, University of Minnesota Medical School, Minneapolis, MN, United States
| |
Collapse
|
10
|
Huang X, Hu L, Long Z, Wang X, Wu J, Cai J. Hypertensive Heart Disease: Mechanisms, Diagnosis and Treatment. Rev Cardiovasc Med 2024; 25:93. [PMID: 39076964 PMCID: PMC11263885 DOI: 10.31083/j.rcm2503093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 11/19/2023] [Accepted: 11/22/2023] [Indexed: 07/31/2024] Open
Abstract
Hypertensive heart disease (HHD) presents a substantial global health burden, spanning a spectrum from subtle cardiac functional alterations to overt heart failure. In this comprehensive review, we delved into the intricate pathophysiological mechanisms governing the onset and progression of HHD. We emphasized the significant role of neurohormonal activation, inflammation, and metabolic remodeling in HHD pathogenesis, offering insights into promising therapeutic avenues. Additionally, this review provided an overview of contemporary imaging diagnostic tools for precise HHD severity assessment. We discussed in detail the current potential treatments for HHD, including pharmacologic, lifestyle, and intervention devices. This review aimed to underscore the global importance of HHD and foster a deeper understanding of its pathophysiology, ultimately contributing to improved public health outcomes.
Collapse
Affiliation(s)
- Xuewei Huang
- Department of Cardiology, The Third Xiangya Hospital, Central South University, 410013 Changsha, Hunan, China
| | - Lizhi Hu
- Xiangya School of Medicine, Central South University, 410013 Changsha, Hunan, China
| | - Zhuojun Long
- Xiangya School of Medicine, Central South University, 410013 Changsha, Hunan, China
| | - Xinyao Wang
- Xiangya School of Medicine, Central South University, 410013 Changsha, Hunan, China
| | - Junru Wu
- Department of Cardiology, The Third Xiangya Hospital, Central South University, 410013 Changsha, Hunan, China
| | - Jingjing Cai
- Department of Cardiology, The Third Xiangya Hospital, Central South University, 410013 Changsha, Hunan, China
| |
Collapse
|
11
|
Toma AI, Shah D, Roth D, Oliver Piña J, Hymel L, Turner T, Kamalakar A, Liu K, Bartsch P, Jacobs L, D'Souza R, Liotta D, Botchwey E, Willett NJ, Goudy SL. Harnessing Bilayer Biomaterial Delivery of FTY720 as an Immunotherapy to Accelerate Oral Wound Healing. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.22.573096. [PMID: 38187740 PMCID: PMC10769397 DOI: 10.1101/2023.12.22.573096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Orofacial clefts are the most common craniofacial congenital anomaly. Following cleft palate repair, up to 60% of surgeries have wound healing complications leading to oronasal fistula (ONF), a persistent connection between the roof of the mouth and the nasal cavity. The current gold standard methods for ONF repair use human allograft tissues; however, these procedures have risks of graft infection and/or rejection, requiring surgical revisions. Immunoregenerative therapies present a novel alternative approach to harness the body's immune response and enhance the wound healing environment. We utilized a repurposed FDA-approved immunomodulatory drug, FTY720, to reduce the egress of lymphocytes and induce immune cell fate switching toward pro-regenerative phenotypes. Here, we engineered a bilayer biomaterial system using Tegaderm™, a liquid-impermeable wound dressing, to secure and control the delivery of FTY720- nanofiber scaffolds (FTY720-NF). We optimized release kinetics of the bilayer FTY720-NF to sustain drug release for up to 7d with safe, efficacious transdermal absorption and tissue biodistribution. Through comprehensive immunophenotyping, our results illustrate a pseudotime pro-regenerative state transition in recruited hybrid immune cells to the wound site. Additional histological assessments established a significant difference in full thickness ONF closure in mice on Day 7 following treatment with bilayer FTY720-NF, compared to controls. These findings demonstrate the utility of immunomodulatory strategies for oral wound healing, better positing the field to develop more efficacious treatment options for pediatric patients. One Sentence Summary Local delivery of bilayer FTY720-nanofiber scaffolds in an ONF mouse model promotes complete wound closure through modulation of pro-regenerative immune and stromal cells.
Collapse
|
12
|
He X, Xu R, Pan L, Bhattarai U, Liu X, Zeng H, Chen JX, Hall ME, Chen Y. Inhibition of NK1.1 signaling attenuates pressure overload-induced heart failure, and consequent pulmonary inflammation and remodeling. Front Immunol 2023; 14:1215855. [PMID: 37554327 PMCID: PMC10405176 DOI: 10.3389/fimmu.2023.1215855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 07/05/2023] [Indexed: 08/10/2023] Open
Abstract
Background Inflammation contributes to heart failure (HF) development, the progression from left ventricular failure to pulmonary remodeling, and the consequent right ventricular hypertrophy and failure. NK1.1 plays a critical role in Natural killer (NK) and NK T (NKT) cells, but the role of NK1.1 in HF development and progression is unknown. Methods We studied the effects of NK1.1 inhibition on transverse aortic constriction (TAC)-induced cardiopulmonary inflammation, HF development, and HF progression in immunocompetent male mice of C57BL/6J background. Results We found that NK1.1+ cell-derived interferon gamma+ (IFN-γ+) was significantly increased in pulmonary tissues after HF. In addition, anti-NK1.1 antibodies simultaneously abolished both NK1.1+ cells, including the NK1.1+NK and NK1.1+NKT cells in peripheral blood, spleen, and lung tissues, but had no effect on cardiopulmonary structure and function under control conditions. However, systemic inhibition of NK1.1 signaling by anti-NK1.1 antibodies significantly rescued mice from TAC-induced left ventricular inflammation, fibrosis, and failure. Inhibition of NK1.1 signaling also significantly attenuated TAC-induced pulmonary leukocyte infiltration, fibrosis, vessel remodeling, and consequent right ventricular hypertrophy. Moreover, inhibition of NK1.1 signaling significantly reduced TAC-induced pulmonary macrophage and dendritic cell infiltration and activation. Conclusions Our data suggest that inhibition of NK1.1 signaling is effective in attenuating systolic overload-induced cardiac fibrosis, dysfunction, and consequent pulmonary remodeling in immunocompetent mice through modulating the cardiopulmonary inflammatory response.
Collapse
Affiliation(s)
- Xiaochen He
- Department of Physiology and Biophysics, University of Mississippi Medical Center, School of Medicine, Jackson, MS, United States
| | - Rui Xu
- Department of Physiology and Biophysics, University of Mississippi Medical Center, School of Medicine, Jackson, MS, United States
| | - Lihong Pan
- Department of Physiology and Biophysics, University of Mississippi Medical Center, School of Medicine, Jackson, MS, United States
| | - Umesh Bhattarai
- Department of Physiology and Biophysics, University of Mississippi Medical Center, School of Medicine, Jackson, MS, United States
| | - Xiaoguang Liu
- Department of Physiology and Biophysics, University of Mississippi Medical Center, School of Medicine, Jackson, MS, United States
- College of Sports and Health, Guangzhou Sport University, Guangzhou, China
| | - Heng Zeng
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, School of Medicine, Jackson, MS, United States
| | - Jian-Xiong Chen
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, School of Medicine, Jackson, MS, United States
| | - Michael E. Hall
- Department of Physiology and Biophysics, University of Mississippi Medical Center, School of Medicine, Jackson, MS, United States
- Department of Medicine, University of Mississippi Medical Center, School of Medicine, Jackson, MS, United States
| | - Yingjie Chen
- Department of Physiology and Biophysics, University of Mississippi Medical Center, School of Medicine, Jackson, MS, United States
| |
Collapse
|
13
|
Warde KM, Smith LJ, Liu L, Stubben CJ, Lohman BK, Willett PW, Ammer JL, Castaneda-Hernandez G, Imodoye SO, Zhang C, Jones KD, Converso-Baran K, Ekiz HA, Barry M, Clay MR, Kiseljak-Vassiliades K, Giordano TJ, Hammer GD, Basham KJ. Senescence-induced immune remodeling facilitates metastatic adrenal cancer in a sex-dimorphic manner. NATURE AGING 2023; 3:846-865. [PMID: 37231196 PMCID: PMC11534150 DOI: 10.1038/s43587-023-00420-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 04/12/2023] [Indexed: 05/27/2023]
Abstract
Aging markedly increases cancer risk, yet our mechanistic understanding of how aging influences cancer initiation is limited. Here we demonstrate that the loss of ZNRF3, an inhibitor of Wnt signaling that is frequently mutated in adrenocortical carcinoma, leads to the induction of cellular senescence that remodels the tissue microenvironment and ultimately permits metastatic adrenal cancer in old animals. The effects are sexually dimorphic, with males exhibiting earlier senescence activation and a greater innate immune response, driven in part by androgens, resulting in high myeloid cell accumulation and lower incidence of malignancy. Conversely, females present a dampened immune response and increased susceptibility to metastatic cancer. Senescence-recruited myeloid cells become depleted as tumors progress, which is recapitulated in patients in whom a low myeloid signature is associated with worse outcomes. Our study uncovers a role for myeloid cells in restraining adrenal cancer with substantial prognostic value and provides a model for interrogating pleiotropic effects of cellular senescence in cancer.
Collapse
Affiliation(s)
- Kate M Warde
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Lorenzo J Smith
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Lihua Liu
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Chris J Stubben
- Bioinformatics Shared Resource, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Brian K Lohman
- Bioinformatics Shared Resource, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Parker W Willett
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Julia L Ammer
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Michigan, Ann Arbor, MI, USA
| | | | - Sikiru O Imodoye
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Chenge Zhang
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Kara D Jones
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Kimber Converso-Baran
- Frankel Cardiovascular Center Physiology and Phenotyping Core, University of Michigan, Ann Arbor, MI, USA
| | - H Atakan Ekiz
- Department of Molecular Biology and Genetics, Izmir Institute of Technology, Urla Izmir, Turkey
| | - Marc Barry
- Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - Michael R Clay
- Department of Pathology, University of Colorado School of Medicine at Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Katja Kiseljak-Vassiliades
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado School of Medicine at Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Thomas J Giordano
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Michigan, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Endocrine Oncology Program, Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Gary D Hammer
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Michigan, Ann Arbor, MI, USA
- Endocrine Oncology Program, Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Kaitlin J Basham
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
14
|
Cohen CD, Rousseau ST, Bermea KC, Bhalodia A, Lovell JP, Dina Zita M, Čiháková D, Adamo L. Myocardial Immune Cells: The Basis of Cardiac Immunology. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:1198-1207. [PMID: 37068299 PMCID: PMC10111214 DOI: 10.4049/jimmunol.2200924] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 01/14/2023] [Indexed: 04/19/2023]
Abstract
The mammalian heart is characterized by the presence of striated myocytes, which allow continuous rhythmic contraction from early embryonic development until the last moments of life. However, the myocardium contains a significant contingent of leukocytes from every major class. This leukocyte pool includes both resident and nonresident immune cells. Over recent decades, it has become increasingly apparent that the heart is intimately sensitive to immune signaling and that myocardial leukocytes exhibit an array of critical functions, both in homeostasis and in the context of cardiac adaptation to injury. Here, we systematically review current knowledge of all major leukocyte classes in the heart, discussing their functions in health and disease. We also highlight the connection between the myocardium, immune cells, lymphoid organs, and both local and systemic immune responses.
Collapse
Affiliation(s)
- Charles D. Cohen
- Cardiac Immunology Laboratory, Department of Medicine, Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Sylvie T. Rousseau
- Cardiac Immunology Laboratory, Department of Medicine, Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Kevin C. Bermea
- Cardiac Immunology Laboratory, Department of Medicine, Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Aashik Bhalodia
- Cardiac Immunology Laboratory, Department of Medicine, Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Jana P. Lovell
- Cardiac Immunology Laboratory, Department of Medicine, Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Marcelle Dina Zita
- Cardiac Immunology Laboratory, Department of Medicine, Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Daniela Čiháková
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, United States of America
| | - Luigi Adamo
- Cardiac Immunology Laboratory, Department of Medicine, Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| |
Collapse
|
15
|
Benson LN, Guo Y, Deck K, Mora C, Liu Y, Mu S. The link between immunity and hypertension in the kidney and heart. Front Cardiovasc Med 2023; 10:1129384. [PMID: 36970367 PMCID: PMC10034415 DOI: 10.3389/fcvm.2023.1129384] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 02/20/2023] [Indexed: 03/11/2023] Open
Abstract
Hypertension is the primary cause of cardiovascular disease, which is a leading killer worldwide. Despite the prevalence of this non-communicable disease, still between 90% and 95% of cases are of unknown or multivariate cause ("essential hypertension"). Current therapeutic options focus primarily on lowering blood pressure through decreasing peripheral resistance or reducing fluid volume, but fewer than half of hypertensive patients can reach blood pressure control. Hence, identifying unknown mechanisms causing essential hypertension and designing new treatment accordingly are critically needed for improving public health. In recent years, the immune system has been increasingly implicated in contributing to a plethora of cardiovascular diseases. Many studies have demonstrated the critical role of the immune system in the pathogenesis of hypertension, particularly through pro-inflammatory mechanisms within the kidney and heart, which, eventually, drive a myriad of renal and cardiovascular diseases. However, the precise mechanisms and potential therapeutic targets remain largely unknown. Therefore, identifying which immune players are contributing to local inflammation and characterizing pro-inflammatory molecules and mechanisms involved will provide promising new therapeutic targets that could lower blood pressure and prevent progression from hypertension into renal or cardiac dysfunction.
Collapse
Affiliation(s)
- Lance N. Benson
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, United States
| | | | | | | | | | - Shengyu Mu
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, United States
| |
Collapse
|
16
|
Bhattarai U, He X, Xu R, Liu X, Pan L, Sun Y, Chen JX, Chen Y. IL-12α deficiency attenuates pressure overload-induced cardiac inflammation, hypertrophy, dysfunction, and heart failure progression. Front Immunol 2023; 14:1105664. [PMID: 36860846 PMCID: PMC9969090 DOI: 10.3389/fimmu.2023.1105664] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 01/30/2023] [Indexed: 02/15/2023] Open
Abstract
IL-12α plays an important role in modulating inflammatory response, fibroblast proliferation and angiogenesis through modulating macrophage polarization or T cell function, but its effect on cardiorespiratory fitness is not clear. Here, we studied the effect of IL-12α on cardiac inflammation, hypertrophy, dysfunction, and lung remodeling in IL-12α gene knockout (KO) mice in response to chronic systolic pressure overload produced by transverse aortic constriction (TAC). Our results showed that IL-12α KO significantly ameliorated TAC-induced left ventricular (LV) failure, as evidenced by a smaller decrease of LV ejection fraction. IL-12α KO also exhibited significantly attenuated TAC-induced increase of LV weight, left atrial weight, lung weight, right ventricular weight, and the ratios of them in comparison to body weight or tibial length. In addition, IL-12α KO showed significantly attenuated TAC-induced LV leukocyte infiltration, fibrosis, cardiomyocyte hypertrophy, and lung inflammation and remodeling (such as lung fibrosis and vessel muscularization). Moreover, IL-12α KO displayed significantly attenuated TAC-induced activation of CD4+ T cells and CD8+ T cells in the lung. Furthermore, IL-12α KO showed significantly suppressed accumulation and activation of pulmonary macrophages and dendritic cells. Taken together, these findings indicate that inhibition of IL-12α is effective in attenuating systolic overload-induced cardiac inflammation, heart failure development, promoting transition from LV failure to lung remodeling and right ventricular hypertrophy.
Collapse
Affiliation(s)
- Umesh Bhattarai
- Department of Physiology and Biophysics, School of Medicine, University of Mississippi Medical Center, Jackson, MS, United States
| | - Xiaochen He
- Department of Physiology and Biophysics, School of Medicine, University of Mississippi Medical Center, Jackson, MS, United States
| | - Rui Xu
- Department of Physiology and Biophysics, School of Medicine, University of Mississippi Medical Center, Jackson, MS, United States
| | - Xiaoguang Liu
- Department of Physiology and Biophysics, School of Medicine, University of Mississippi Medical Center, Jackson, MS, United States
- College of Sports and Health, Guangzhou Sport University, Guangzhou, China
| | - Lihong Pan
- Department of Physiology and Biophysics, School of Medicine, University of Mississippi Medical Center, Jackson, MS, United States
| | - Yuxiang Sun
- Department of Nutrition, Texas A&M University, College Station, TX, United States
| | - Jian-Xiong Chen
- Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center, Jackson, MS, United States
| | - Yingjie Chen
- Department of Physiology and Biophysics, School of Medicine, University of Mississippi Medical Center, Jackson, MS, United States
| |
Collapse
|
17
|
Ngwenyama N, Kaur K, Bugg D, Theall B, Aronovitz M, Berland R, Panagiotidou S, Genco C, Perrin MA, Davis J, Alcaide P. Antigen presentation by cardiac fibroblasts promotes cardiac dysfunction. NATURE CARDIOVASCULAR RESEARCH 2022; 1:761-774. [PMID: 36092510 PMCID: PMC9451034 DOI: 10.1038/s44161-022-00116-7] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 07/08/2022] [Indexed: 11/09/2022]
Abstract
Heart failure (HF) is a leading cause of morbidity and mortality. Studies in animal models and patients with HF revealed a prominent role for CD4+ T cell immune responses in the pathogenesis of HF and highlighted an active crosstalk between cardiac fibroblasts and IFNγ producing CD4+ T cells that results in profibrotic myofibroblast transformation. Whether cardiac fibroblasts concomitantly modulate pathogenic cardiac CD4+ T cell immune responses is unknown. Here we show report that murine cardiac fibroblasts express major histocompatibility complex type II (MHCII) in two different experimental models of cardiac inflammation. We demonstrate that cardiac fibroblasts take up and process antigens for presentation to CD4+ T cells via MHCII induced by IFNγ. Conditional deletion of MhcII in cardiac fibroblasts ameliorates cardiac remodelling and dysfunction induced by cardiac pressure overload. Collectively, we demonstrate that cardiac fibroblasts function as antigen presenting cells (APCs) and contribute to cardiac fibrosis and dysfunction through IFNγ induced MHCII.
Collapse
Affiliation(s)
| | - Kuljeet Kaur
- Department of Immunology, Tufts University, Boston, MA, USA
| | - Darrian Bugg
- Departments of Lab Medicine-Pathology & Bioengineering, University of Washington, Seattle, WA, USA
| | - Brandon Theall
- Department of Immunology, Tufts University, Boston, MA, USA
| | - Mark Aronovitz
- Department of Immunology, Tufts University, Boston, MA, USA
| | - Robert Berland
- Department of Immunology, Tufts University, Boston, MA, USA
| | - Smaro Panagiotidou
- Developmental, Molecular and Chemical Biology, Tufts University, Boston, MA, USA
| | - Caroline Genco
- Department of Immunology, Tufts University, Boston, MA, USA
| | - Mercio A. Perrin
- Developmental, Molecular and Chemical Biology, Tufts University, Boston, MA, USA
| | - Jennifer Davis
- Departments of Lab Medicine-Pathology & Bioengineering, University of Washington, Seattle, WA, USA
| | - Pilar Alcaide
- Department of Immunology, Tufts University, Boston, MA, USA
| |
Collapse
|
18
|
da Purificação NRC, Garcia VB, Frez FCV, Sehaber CC, Lima KRDA, de Oliveira Lima MF, de Carvalho Vasconcelos R, de Araujo AA, de Araújo Júnior RF, Lacchini S, de Oliveira F, Perles JVCM, Zanoni JN, de Sousa Lopes MLD, Clebis NK. Combined use of systemic quercetin, glutamine and alpha-tocopherol attenuates myocardial fibrosis in diabetic rats. Biomed Pharmacother 2022; 151:113131. [PMID: 35643067 DOI: 10.1016/j.biopha.2022.113131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 05/11/2022] [Accepted: 05/13/2022] [Indexed: 11/02/2022] Open
Abstract
This study aimed to analyze the effects of the quercetin (100 mg/kg), 1% glutamine and 1% α-tocopherol antioxidants in the myocardium of rats with streptozotocin-induced diabetes mellitus. Twenty male rats were subdivided into four groups (n = 5): N (normoglycemic); D (diabetic); NT (normoglycemic treated with antioxidants); and DT (diabetic treated with antioxidants) treated for 60 days. Clinical parameters, oxidative stress markers, inflammatory cytokines, myocardial collagen fibers and immunoexpression of superoxide dismutase 1 (SOD-1), glutathione peroxidase-1 (GPx-1), interleukin-1β (IL-1-β), transforming growth factor-beta (TGF-β), and fibroblast growth factor-2 (FGF-2) were evaluated. Results showed reduced body weight, hyperphagia, polydipsia and hyperglycemic state in groups D and DT. The levels of glutathione (GSH) were higher in NT and DT compared to N (p < 0.01) and D (p < 0.001) groups, respectively. Greater GSH levels were found in DT when compared to N animals (p < 0.001). In DT, there was an increase in IL-10 in relation to N, D and NT (p < 0.05), while GPx-1 expression was similar to N and lower compared to D (p < 0.001). TGF-β expression in DT was greater than N (p < 0.001) group, whereas FGF-2 in DT was higher than in the other groups (p < 0.001). A significant reduction in collagen fibers (type I) was found in DT compared to D (p < 0.05). The associated administration of quercetin, glutamine and α-tocopherol increased the levels of circulating interleukin-10 (IL-10) and GSH, and reduced the number of type I collagen fibers. Combined use of systemic quercetin, glutamine and alpha-tocopherol attenuates myocardial fibrosis in diabetic rats.
Collapse
Affiliation(s)
| | | | | | | | - Kaio Ramon De Aguiar Lima
- Postgraduate Program in Functional & Structural Biology, Departament of Morphology, UFRN, Natal, RN, Brazil
| | | | | | - Aurigena Antunes de Araujo
- Postgraduate Program in Pharmaceutical Sciences, Postgraduate Program in Dental Sciences, Department of Pharmacology and Biophysical, UFRN, Natal, RN, Brazil.
| | - Raimundo Fernandes de Araújo Júnior
- Postgraduate Program in Health Sciences, Postgraduate Program in Functional & Structural Biology, Departament of Morphology, UFRN, Natal, RN, Brazil
| | - Silvia Lacchini
- Postgraduate Program in Morphology Science, Departamento of Anatomy, São Paulo University, São Paulo, SP, Brazil
| | - Flávia de Oliveira
- Departament of Biocience, Federal University of São Paulo (UNIFESP), Santos, SP, Brazil
| | | | | | | | - Naianne Kelly Clebis
- Postgraduate Program in Functional & Structural Biology, Departament of Morphology, UFRN, Natal, RN, Brazil
| |
Collapse
|
19
|
Lamy R, Wolf M, Bispo C, Clay SM, Zheng S, Wolfreys F, Pan P, Chan MF. Characterization of Recruited Mononuclear Phagocytes following Corneal Chemical Injury. Int J Mol Sci 2022; 23:ijms23052574. [PMID: 35269717 PMCID: PMC8910730 DOI: 10.3390/ijms23052574] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 02/18/2022] [Accepted: 02/23/2022] [Indexed: 02/07/2023] Open
Abstract
Mononuclear phagocytes (MP) have central importance in innate immunity, inflammation, and fibrosis. Recruited MPs, such as macrophages, are plastic cells and can switch from an inflammatory to a restorative phenotype during the healing process. However, the role of the MPs in corneal wound healing is not completely understood. The purpose of this study is to characterize the kinetics of recruited MPs and evaluate the role of macrophage metalloelastase (MMP12) in the healing process, using an in vivo corneal chemical injury model. Unwounded and wounded corneas of wild-type (WT) and Mmp12-/- mice were collected at 1, 3, and 6 days after chemical injury and processed for flow cytometry analysis. Corneal MP phenotype significantly changed over time with recruited Ly6Chigh (proinflammatory) cells being most abundant at 1 day post-injury. Ly6Cint cells were highly expressed at 3 days post-injury and Ly6Cneg (patrolling) cells became the predominant cell type at 6 days post-injury. CD11c+ dendritic cells were abundant in corneas from Mmp12-/- mice at 6 days post-injury. These findings show the temporal phenotypic plasticity of recruited MPs and provide valuable insight into the role of the MPs in the corneal repair response, which may help guide the future development of MP-targeted therapies.
Collapse
Affiliation(s)
- Ricardo Lamy
- Department of Ophthalmology, University of California, San Francisco, CA 94158, USA; (R.L.); (M.W.); (S.M.C.); (S.Z.); (F.W.); (P.P.)
| | - Marie Wolf
- Department of Ophthalmology, University of California, San Francisco, CA 94158, USA; (R.L.); (M.W.); (S.M.C.); (S.Z.); (F.W.); (P.P.)
| | - Claudia Bispo
- UCSF Parnassus Flow Cytometry Core Facility, University of California, San Francisco, CA 94143, USA;
| | - Selene M. Clay
- Department of Ophthalmology, University of California, San Francisco, CA 94158, USA; (R.L.); (M.W.); (S.M.C.); (S.Z.); (F.W.); (P.P.)
| | - Siyu Zheng
- Department of Ophthalmology, University of California, San Francisco, CA 94158, USA; (R.L.); (M.W.); (S.M.C.); (S.Z.); (F.W.); (P.P.)
| | - Finn Wolfreys
- Department of Ophthalmology, University of California, San Francisco, CA 94158, USA; (R.L.); (M.W.); (S.M.C.); (S.Z.); (F.W.); (P.P.)
| | - Peipei Pan
- Department of Ophthalmology, University of California, San Francisco, CA 94158, USA; (R.L.); (M.W.); (S.M.C.); (S.Z.); (F.W.); (P.P.)
| | - Matilda F. Chan
- Department of Ophthalmology, University of California, San Francisco, CA 94158, USA; (R.L.); (M.W.); (S.M.C.); (S.Z.); (F.W.); (P.P.)
- Francis I. Proctor Foundation, University of California, San Francisco, CA 94158, USA
- Correspondence:
| |
Collapse
|
20
|
Theall B, Alcaide P. The heart under pressure: immune cells in fibrotic remodeling. CURRENT OPINION IN PHYSIOLOGY 2022; 25:100484. [PMID: 35224321 PMCID: PMC8881013 DOI: 10.1016/j.cophys.2022.100484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The complex syndrome of heart failure (HF) is characterized by increased left ventricular pressures. Cardiomyocytes increase in size, cardiac fibroblasts transform and make extracellular matrix, and leukocytes infiltrate the cardiac tissue and alter cardiomyocyte and cardiac fibroblast function. Here we review recent advances in our understanding of the cellular composition of the heart during homeostasis and in response to cardiac pressure overload, with an emphasis on immune cell communication with cardiac fibroblasts and its consequences in cardiac remodeling.
Collapse
Affiliation(s)
- Brandon Theall
- Department of Immunology, Tufts University School of Medicine, Boston, MA,Immunology Program, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA
| | - Pilar Alcaide
- Department of Immunology, Tufts University School of Medicine, Boston, MA,Immunology Program, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA
| |
Collapse
|
21
|
Codden CJ, Chin MT. Common and Distinctive Intercellular Communication Patterns in Human Obstructive and Nonobstructive Hypertrophic Cardiomyopathy. Int J Mol Sci 2022; 23:946. [PMID: 35055131 PMCID: PMC8780670 DOI: 10.3390/ijms23020946] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 01/12/2022] [Accepted: 01/13/2022] [Indexed: 01/27/2023] Open
Abstract
Hypertrophic Cardiomyopathy (HCM) is a common inherited disorder characterized by unexplained left ventricular hypertrophy with or without left ventricular outflow tract (LVOT) obstruction. Single-nuclei RNA-sequencing (snRNA-seq) of both obstructive and nonobstructive HCM patient samples has revealed alterations in communication between various cell types, but no direct and integrated comparison between the two HCM phenotypes has been reported. We performed a bioinformatic analysis of HCM snRNA-seq datasets from obstructive and nonobstructive patient samples to identify differentially expressed genes and distinctive patterns of intercellular communication. Differential gene expression analysis revealed 37 differentially expressed genes, predominantly in cardiomyocytes but also in other cell types, relevant to aging, muscle contraction, cell motility, and the extracellular matrix. Intercellular communication was generally reduced in HCM, affecting the extracellular matrix, growth factor binding, integrin binding, PDGF binding, and SMAD binding, but with increases in adenylate cyclase binding, calcium channel inhibitor activity, and serine-threonine kinase activity in nonobstructive HCM. Increases in neuron to leukocyte and dendritic cell communication, in fibroblast to leukocyte and dendritic cell communication, and in endothelial cell communication to other cell types, largely through changes in the expression of integrin-β1 and its cognate ligands, were also noted. These findings indicate both common and distinct physiological mechanisms affecting the pathogenesis of obstructive and nonobstructive HCM and provide opportunities for the personalized management of different HCM phenotypes.
Collapse
Affiliation(s)
- Christina J. Codden
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA 02111, USA;
| | - Michael T. Chin
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA 02111, USA;
- Tufts Hypertrophic Cardiomyopathy Center and Research Institute, Boston, MA 02111, USA
| |
Collapse
|
22
|
Gajić D, Despotović S, Koprivica I, Miljković Đ, Saksida T. Ethyl Pyruvate Ameliorates Experimental Autoimmune Myocarditis. Biomolecules 2021; 11:biom11121768. [PMID: 34944410 PMCID: PMC8698267 DOI: 10.3390/biom11121768] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 11/17/2021] [Accepted: 11/23/2021] [Indexed: 02/07/2023] Open
Abstract
Ethyl pyruvate (EP) has profound anti-inflammatory and immunomodulatory properties. Here, its effects were determined on experimental autoimmune myocarditis (EAM) induced in mice by heart-specific myosin-alpha heavy chain peptide immunization. EP was applied intraperitoneally, daily, starting with the immunization. Severity of EAM was determined by histological assessment of immune cell infiltrates into the heart. Cells were phenotypically characterized by flow cytometry. Concentration of cytokines in cell culture supernatants and sera was determined by ELISA. EP reduced the infiltration of immune cells into the heart and lessened heart inflammation. Smaller number of total immune cells, as well as of CD11b+ and CD11c+ cells were isolated from the hearts of EP-treated mice. A reduced number of antigen-presenting cells, detected by anti-CD11c, MHC class II and CD86 antibodies, as well as of T helper (Th)1 and Th17 cells, detected by anti-CD4, IFN-γ and IL-17 antibodies, was determined in mediastinal lymph nodes draining the heart, in parallel. In the spleen, only the number of CD11c+ cells were reduced, but not of the other examined populations, thus implying limited systemic effect of EP. Reduced production of IFN-γ and IL-17 by myosin-alpha heavy chain peptide-restimulated cells of the lymph nodes draining the site of immunization was observed in EP-treated mice. Our results clearly imply that EP restrains autoimmunity in EAM. Therapeutic application of EP in the treatment of myocarditis in humans should be addressed in the forthcoming studies.
Collapse
Affiliation(s)
- Dragica Gajić
- Department of Immunology, Institute for Biological Research “Siniša Stanković”—National Institute of Republic of Serbia, University of Belgrade, Bulevar Despota Stefana 142, 11000 Belgrade, Serbia; (D.G.); (I.K.); (T.S.)
| | - Sanja Despotović
- Faculty of Medicine, Institute of Histology and Embryology, University of Belgrade, Dr Subotića 9, 11000 Belgrade, Serbia;
| | - Ivan Koprivica
- Department of Immunology, Institute for Biological Research “Siniša Stanković”—National Institute of Republic of Serbia, University of Belgrade, Bulevar Despota Stefana 142, 11000 Belgrade, Serbia; (D.G.); (I.K.); (T.S.)
| | - Đorđe Miljković
- Department of Immunology, Institute for Biological Research “Siniša Stanković”—National Institute of Republic of Serbia, University of Belgrade, Bulevar Despota Stefana 142, 11000 Belgrade, Serbia; (D.G.); (I.K.); (T.S.)
- Correspondence: ; Tel.: +381-11-2078390
| | - Tamara Saksida
- Department of Immunology, Institute for Biological Research “Siniša Stanković”—National Institute of Republic of Serbia, University of Belgrade, Bulevar Despota Stefana 142, 11000 Belgrade, Serbia; (D.G.); (I.K.); (T.S.)
| |
Collapse
|
23
|
Characterization of Transverse Aortic Constriction in Mice Based on the Specific Recruitment of Leukocytes to the Hypertrophic Myocardium and the Aorta Ascendens. Mediators Inflamm 2021; 2021:1376859. [PMID: 34776787 PMCID: PMC8580661 DOI: 10.1155/2021/1376859] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 09/07/2021] [Accepted: 10/13/2021] [Indexed: 11/17/2022] Open
Abstract
Transverse aortic constriction (TAC) is a model that mimics pressure overload-induced left ventricular (LV) hypertrophy in mice. Alterations in immune cell functionality can promote cardiac and vascular remodeling. In the present study, we characterized the time course in innate immune cell dynamics in response to TAC in the different tissues of mice. It was determined whether TAC induces a characteristic leukocyte-driven immune response in the myocardium, aorta ascendens and descendens, spleen, blood, and draining lymph nodes supported by cytokine-driven chemotaxis in mice at 3, 6, and 21 days following surgery. We used complex flow cytometry staining combinations to characterize the various innate immune cell subsets and a multiplex array to determine cytokine concentrations in the serum. The results of the current study indicated that leukocytes accumulate in the myocardium and aorta ascendens in response to TAC. The leukocyte dynamics in the myocardium were dominated by the Ly6Clow macrophages with an early accumulation, whereas the response in the aorta ascendens was characterized by a long-lasting proinflammatory phenotype driven by Ly6Chigh macrophages, neutrophils, and activated DCs. In contrast to the high-pressure environment of the aorta ascendens, the tissue of the aorta descendens did not react to TAC with any leukocyte increase. The levels of proinflammatory cytokines in the blood were elevated in response to TAC, indicating a systemic reaction. Moreover, our findings strongly suggest that cardiac macrophages could origin from splenic pools and reach the site of the inflammation via the blood. Based on the current findings, it can be concluded that the high-pressure conditions in the aorta ascendens cause a characteristic immune response, dominated by the accumulation of leukocytes and the activation of DCs that varies in comparison to the immune cell dynamics in the myocardium and the aorta descendens.
Collapse
|
24
|
Elmadbouh I, Singla DK. BMP-7 Attenuates Inflammation-Induced Pyroptosis and Improves Cardiac Repair in Diabetic Cardiomyopathy. Cells 2021; 10:2640. [PMID: 34685620 PMCID: PMC8533936 DOI: 10.3390/cells10102640] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 09/22/2021] [Accepted: 09/28/2021] [Indexed: 01/15/2023] Open
Abstract
In the present study, we investigated a novel signaling target in diabetic cardiomyopathy where inflammation induces caspase-1-dependent cell death, pyroptosis, involving Nek7-GBP5 activators to activate the NLRP3 inflammasome, destabilizes cardiac structure and neovascularization. Furthermore, we explored the therapeutic ability of bone morphogenetic protein-7 (BMP-7) to attenuate these adverse effects. C57BL/6J mice (n = 16 mice/group) were divided into: control (200 mg/kg, 0.9% saline intraperitoneal injection, i.p.); Streptozotocin (STZ) and STZ-BMP-7 groups (STZ, 200 mg/kg, i.p. injection). After 6 weeks, heart function was examined with echocardiography, and mice were sacrificed. Immunostaining, Western blotting, H&E, and Masson's trichrome staining was performed on heart tissues. STZ-induced diabetic cardiomyopathy significantly increased inflammasome formation (TLR4, NLRP3, Nek7, and GBP5), pyroptosis markers (caspase-1, IL-1β, and IL-18), inflammatory cytokines (IL-6 and TNF-α), MMP9, and infiltration of monocytes (CD14), macrophage (iNOS), and dendritic cells (CD11b and CD11c) (p < 0.05). Moreover, a significant endothelial progenitor cells (EPCs) dysfunction (c-Kit/FLk-1, CD31), adverse cardiac remodeling, and reduction in left ventricular (LV) heart function were observed in STZ versus control (p < 0.05). Treatment with BMP-7 significantly reduced inflammasome formation, pyroptosis, and inflammatory cytokines and infiltrated inflammatory cells. In addition, BMP-7 treatment enhanced EPC markers and neovascularization and subsequently improved cardiac remodeling in a diabetic heart. Moreover, a significant improvement in LV heart function was achieved after BMP-7 administration relative to diabetic mice (p < 0.05). In conclusion, BMP-7 attenuated inflammation-induced pyroptosis, adverse cardiac remodeling, and improved heart function via the TLR4-NLRP3 inflammasome complex activated by novel signaling Nek7/GBP5. Our BMP-7 pre-clinical studies of mice could have significant potential as a future therapy for diabetic patients.
Collapse
Affiliation(s)
| | - Dinender K. Singla
- Division of Metabolic and Cardiovascular Sciences, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA;
| |
Collapse
|
25
|
Liu X, Shi GP, Guo J. Innate Immune Cells in Pressure Overload-Induced Cardiac Hypertrophy and Remodeling. Front Cell Dev Biol 2021; 9:659666. [PMID: 34368120 PMCID: PMC8343105 DOI: 10.3389/fcell.2021.659666] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 06/28/2021] [Indexed: 12/23/2022] Open
Abstract
Pressure overload and heart failure are among the leading causes of cardiovascular morbidity and mortality. Accumulating evidence suggests that inflammatory cell activation and release of inflammatory mediators are of vital importance during the pathogenesis of these cardiac diseases. Yet, the roles of innate immune cells and subsequent inflammatory events in these processes remain poorly understood. Here, we outline the possible underlying mechanisms of innate immune cell participation, including mast cells, macrophages, monocytes, neutrophils, dendritic cells, eosinophils, and natural killer T cells in these pathological processes. Although these cells accumulate in the atrium or ventricles at different time points after pressure overload, their cardioprotective or cardiodestructive activities differ from each other. Among them, mast cells, neutrophils, and dendritic cells exert detrimental function in experimental models, whereas eosinophils and natural killer T cells display cardioprotective activities. Depending on their subsets, macrophages and monocytes may exacerbate cardiodysfunction or negatively regulate cardiac hypertrophy and remodeling. Pressure overload stimulates the secretion of cytokines, chemokines, and growth factors from innate immune cells and even resident cardiomyocytes that together assist innate immune cell infiltration into injured heart. These infiltrates are involved in pro-hypertrophic events and cardiac fibroblast activation. Immune regulation of cardiac innate immune cells becomes a promising therapeutic approach in experimental cardiac disease treatment, highlighting the significance of their clinical evaluation in humans.
Collapse
Affiliation(s)
- Xin Liu
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Guo-Ping Shi
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
| | - Junli Guo
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
- Hainan Provincial Key Laboratory for Tropical Cardiovascular Diseases Research & Key Laboratory of Emergency and Trauma of Ministry of Education, Institute of Cardiovascular Research of the First Affiliated Hospital, Hainan Medical University, Haikou, China
| |
Collapse
|
26
|
Bocchino M, Zanotta S, Capitelli L, Galati D. Dendritic Cells Are the Intriguing Players in the Puzzle of Idiopathic Pulmonary Fibrosis Pathogenesis. Front Immunol 2021; 12:664109. [PMID: 33995394 PMCID: PMC8121252 DOI: 10.3389/fimmu.2021.664109] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 04/15/2021] [Indexed: 12/12/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is the most devastating progressive interstitial lung disease that remains refractory to treatment. Pathogenesis of IPF relies on the aberrant cross-talk between injured alveolar cells and myofibroblasts, which ultimately leads to an aberrant fibrous reaction. The contribution of the immune system to IPF remains not fully explored. Recent evidence suggests that both innate and adaptive immune responses may participate in the fibrotic process. Dendritic cells (DCs) are the most potent professional antigen-presenting cells that bridge innate and adaptive immunity. Also, they exert a crucial role in the immune surveillance of the lung, where they are strategically placed in the airway epithelium and interstitium. Immature DCs accumulate in the IPF lung close to areas of epithelial hyperplasia and fibrosis. Conversely, mature DCs are concentrated in well-organized lymphoid follicles along with T and B cells and bronchoalveolar lavage of IPF patients. We have recently shown that all sub-types of peripheral blood DCs (including conventional and plasmacytoid DCs) are severely depleted in therapy naïve IPF patients. Also, the low frequency of conventional CD1c+ DCs is predictive of a worse prognosis. The purpose of this mini-review is to focus on the main evidence on DC involvement in IPF pathogenesis. Unanswered questions and opportunities for future research ranging from a better understanding of their contribution to diagnosis and prognosis to personalized DC-based therapies will be explored.
Collapse
Affiliation(s)
- Marialuisa Bocchino
- Respiratory Medicine Division, Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
| | - Serena Zanotta
- Hematology-Oncology and Stem Cell Transplantation Unit, Department of Hematology and Developmental Therapeutics, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Naples, Italy
| | - Ludovica Capitelli
- Respiratory Medicine Division, Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
| | - Domenico Galati
- Hematology-Oncology and Stem Cell Transplantation Unit, Department of Hematology and Developmental Therapeutics, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Naples, Italy
| |
Collapse
|
27
|
Ngwenyama N, Kirabo A, Aronovitz M, Velázquez F, Carrillo-Salinas F, Salvador AM, Nevers T, Amarnath V, Tai A, Blanton RM, Harrison DG, Alcaide P. Isolevuglandin-Modified Cardiac Proteins Drive CD4+ T-Cell Activation in the Heart and Promote Cardiac Dysfunction. Circulation 2021; 143:1242-1255. [PMID: 33463362 PMCID: PMC7987774 DOI: 10.1161/circulationaha.120.051889] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 12/11/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND Despite the well-established association between T-cell-mediated inflammation and nonischemic heart failure, the specific mechanisms triggering T-cell activation during the progression of heart failure and the antigens involved are poorly understood. We hypothesized that myocardial oxidative stress induces the formation of isolevuglandin (IsoLG)-modified proteins that function as cardiac neoantigens to elicit CD4+ T-cell receptor (TCR) activation and promote heart failure. METHODS We used transverse aortic constriction in mice to trigger myocardial oxidative stress and T-cell infiltration. We profiled the TCR repertoire by mRNA sequencing of intramyocardial activated CD4+ T cells in Nur77GFP reporter mice, which transiently express GFP on TCR engagement. We assessed the role of antigen presentation and TCR specificity in the development of cardiac dysfunction using antigen presentation-deficient MhcII-/- mice and TCR transgenic OTII mice that lack specificity for endogenous antigens. We detected IsoLG protein adducts in failing human hearts. We also evaluated the role of reactive oxygen species and IsoLGs in eliciting T-cell immune responses in vivo by treating mice with the antioxidant TEMPOL and the IsoLG scavenger 2-hydroxybenzylamine during transverse aortic constriction, and ex vivo in mechanistic studies of CD4+ T-cell proliferation in response to IsoLG-modified cardiac proteins. RESULTS We discovered that TCR antigen recognition increases in the left ventricle as cardiac dysfunction progresses and identified a limited repertoire of activated CD4+ T-cell clonotypes in the left ventricle. Antigen presentation of endogenous antigens was required to develop cardiac dysfunction because MhcII-/- mice reconstituted with CD4+ T cells and OTII mice immunized with their cognate antigen were protected from transverse aortic constriction-induced cardiac dysfunction despite the presence of left ventricle-infiltrated CD4+ T cells. Scavenging IsoLGs with 2-hydroxybenzylamine reduced TCR activation and prevented cardiac dysfunction. Mechanistically, cardiac pressure overload resulted in reactive oxygen species-dependent dendritic cell accumulation of IsoLG protein adducts, which induced robust CD4+ T-cell proliferation. CONCLUSIONS Our study demonstrates an important role of reactive oxygen species-induced formation of IsoLG-modified cardiac neoantigens that lead to TCR-dependent CD4+ T-cell activation within the heart.
Collapse
Affiliation(s)
| | - Annet Kirabo
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Mark Aronovitz
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA
| | | | | | | | - Tania Nevers
- Department of Immunology, Tufts University, Boston, MA
| | - Venkataraman Amarnath
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, TN
| | - Albert Tai
- Department of Immunology, Tufts University, Boston, MA
| | - Robert M. Blanton
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA
| | - David G. Harrison
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Pilar Alcaide
- Department of Immunology, Tufts University, Boston, MA
| |
Collapse
|
28
|
Shang L, Yue W, Wang D, Weng X, Hall ME, Xu Y, Hou M, Chen Y. Systolic overload-induced pulmonary inflammation, fibrosis, oxidative stress and heart failure progression through interleukin-1β. J Mol Cell Cardiol 2020; 146:84-94. [PMID: 32712269 DOI: 10.1016/j.yjmcc.2020.07.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 06/28/2020] [Accepted: 07/16/2020] [Indexed: 10/23/2022]
Abstract
Chronic heart failure is associated with increased interleukin-1β (IL-1β), leukocyte infiltration, and fibrosis in the heart and lungs. Here we further studied the role of IL-1β in the transition from left heart failure to pulmonary hypertension and right ventricular hypertrophy in mice with existing left heart failure produced by transverse aortic constriction. We demonstrated that transverse aortic constriction-induced heart failure was associated with increased lung inflammation and cleaved IL-1β, and inhibition of IL-1β signaling using blocking antibodies of clone B122 effectively attenuated further decrease of left ventricular systolic function in mice with existing heart failure. We found that inhibition of IL-1β attenuated lung inflammation, inflammasome activation, fibrosis, oxidative stress, and right ventricular hypertrophy. IL-1β blocking antibodies of clone B122 also significantly attenuated lung T cell activation. Together, these data indicate that IL-1β signaling exerts a causal role for heart failure progression, or the transition from left heart failure to lung remodeling and right heart hypertrophy.
Collapse
Affiliation(s)
- Linlin Shang
- Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| | - Wenhui Yue
- Department of Cardiology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Dongzhi Wang
- Department of Cardiology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xinyu Weng
- Department of Cardiology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China; Lillehei Heart Institute and Cardiovascular Division, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Michael E Hall
- Department of Physiology & Biophysics, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Yawei Xu
- Department of Cardiology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Mingxiao Hou
- Shenyang Pharmaceutical University, Shenyang, Liaoning, China.
| | - Yingjie Chen
- Lillehei Heart Institute and Cardiovascular Division, University of Minnesota Medical School, Minneapolis, MN 55455, USA; Department of Physiology & Biophysics, University of Mississippi Medical Center, Jackson, MS 39216, USA
| |
Collapse
|
29
|
Dendritic cells are crucial for cardiovascular remodeling and modulate neutrophil gelatinase-associated lipocalin expression upon mineralocorticoid receptor activation. J Hypertens 2020; 37:1482-1492. [PMID: 31033725 DOI: 10.1097/hjh.0000000000002067] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
BACKGROUND Adaptive immunity is crucial in cardiovascular and renal inflammation/fibrosis upon hyperactivation of mineralocorticoid receptor. We have previously demonstrated that dendritic cells can respond to mineralocorticoid receptor activation, and the neutrophil gelatinase-associated lipocalin (NGAL) in dendritic cells is highly increased during aldosterone (Aldo)/mineralocorticoid receptor-dependent cardiovascular damage. However, the interrelationship among dendritic cells, target organs inflammation/fibrosis induced by mineralocorticoid receptor, and NGAL-dependence remains unknown. OBJECTIVE We studied the role of dendritic cells in mineralocorticoid receptor-dependent tissue remodeling and whether NGAL can modulate the inflammatory response of dendritic cells after mineralocorticoid receptor activation. METHODS Cardiovascular and renal remodeling induced by Aldo and high-salt diet [nephrectomy-Aldo-salt (NAS) model] were analyzed in CD11c.DOG mice, a model which allows dendritic cells ablation by using diphtheria toxin. In addition, in-vitro studies in NGAL-knock out dendritic cells were performed to determine the immunomodulatory role of NGAL upon Aldo treatment. RESULTS The ablation of dendritic cells prevented the development of cardiac hypertrophy, perivascular fibrosis, and the overexpression of NGAL, brain natriuretic peptide, and two profibrotic factors induced by NAS: collagen 1A1 and connective tissue growth factor. We determined that dendritic cells were not required to prevent renal hypertrophy/fibrosis induced by NAS. Between different immune cells analyzed, we observed that NGAL abundance was higher in antigen-presenting cells, while in-vitro studies showed that mineralocorticoid receptor stimulation in dendritic cells favored NGAL and IL-23 expression (p19 and p40 subunits), which are involved in the development of fibrosis and the Th17-driven response, respectively. CONCLUSION NGAL produced by dendritic cells may play a pivotal role in the activation of adaptive immunity that leads to cardiovascular fibrosis during mineralocorticoids excess.
Collapse
|
30
|
Okyere AD, Tilley DG. Leukocyte-Dependent Regulation of Cardiac Fibrosis. Front Physiol 2020; 11:301. [PMID: 32322219 PMCID: PMC7156539 DOI: 10.3389/fphys.2020.00301] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 03/17/2020] [Indexed: 12/24/2022] Open
Abstract
Cardiac fibrosis begins as an intrinsic response to injury or ageing that functions to preserve the tissue from further damage. Fibrosis results from activated cardiac myofibroblasts, which secrete extracellular matrix (ECM) proteins in an effort to replace damaged tissue; however, excessive ECM deposition leads to pathological fibrotic remodeling. At this extent, fibrosis gravely disturbs myocardial compliance, and ultimately leads to adverse outcomes like heart failure with heightened mortality. As such, understanding the complexity behind fibrotic remodeling has been a focal point of cardiac research in recent years. Resident cardiac fibroblasts and activated myofibroblasts have been proven integral to the fibrotic response; however, several findings point to additional cell types that may contribute to the development of pathological fibrosis. For one, leukocytes expand in number after injury and exhibit high plasticity, thus their distinct role(s) in cardiac fibrosis is an ongoing and controversial field of study. This review summarizes current findings, focusing on both direct and indirect leukocyte-mediated mechanisms of fibrosis, which may provide novel targeted strategies against fibrotic remodeling.
Collapse
Affiliation(s)
- Ama Dedo Okyere
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Douglas G Tilley
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| |
Collapse
|
31
|
Haybar H, Maleki Behzad M, Shahrabi S, Ansari N, Saki N. Expression of Blood Cells Associated CD Markers and Cardiovascular Diseases: Clinical Applications in Prognosis. Lab Med 2020; 51:122-142. [PMID: 31340048 DOI: 10.1093/labmed/lmz049] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Cardiovascular diseases (CVDs) are a major cause of mortality worldwide. The results of various studies have shown that abnormality in the frequency and function of blood cells can be involved in CVD complications. In this review, we have focused on abnormalities in the expression of the CD (cluster of differentiation) markers of blood cells to assess the association of these abnormalities with CVD prognosis. METHODS We identified the relevant literature through a PubMed search (1990-2018) of English-language articles using the terms "Cardiovascular diseases", "CD markers", "leukocytes", "platelets", and "endothelial cells". RESULTS There is a variety of mechanisms for the effect of CD-marker expressions on CVDs prognosis, ranging from proinflammatory processes to dysfunctional effects in blood cells. CONCLUSION Considering the possible effects of CD-marker expression on CVDs prognosis, particularly prognosis of acute myocardial infarction and atherosclerosis, long-term studies in large cohorts are required to identify the prognostic value of CD markers and to target them with appropriate therapeutic agents.
Collapse
Affiliation(s)
- Habib Haybar
- Atherosclerosis Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Masumeh Maleki Behzad
- Thalassemia and Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Saeid Shahrabi
- Department of Biochemistry and Hematology, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Narges Ansari
- Isfahan Bone Metabolic Disorders Research Center, Department of Internal Medicine, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Najmaldin Saki
- Thalassemia and Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
32
|
Wu J, Liu M, Mang G, Yu S, Chen Q, Li T, Wang Y, Meng Y, Tang X, Zheng Y, Sun Y, Zhang M, Yu B. Protosappanin A protects against experimental autoimmune myocarditis, and induces metabolically reprogrammed tolerogenic DCs. Pharmacol Res 2019; 146:104269. [PMID: 31078745 DOI: 10.1016/j.phrs.2019.104269] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 04/08/2019] [Accepted: 05/08/2019] [Indexed: 02/07/2023]
Abstract
Autoimmune myocarditis is an immune-mediated myocardial injury that evolves into dilated cardiomyopathy (DCM). Protosappanin A (PrA), an immunosuppressive compound, induces immune tolerance in cardiac transplantation. However, whether PrA confers protective immunosuppression on experimental autoimmune myocarditis (EAM) is unknown. In this study, PrA treatment remarkably suppressed cardiac inflammatory cell infiltration and ameliorated cardiac remodeling in EAM mice. Additionally, PrA treatment reduced splenic T cells response, and induced expansion of immunosuppressive regulatory T cells (Tregs). Meanwhile, PrA induced the splenic dendritic cells (DCs) into a tolerogenic state with reduced co-stimulatory molecules, increased the production of tolerogenic cytokines in vivo. PrA also reprogrammed the metabolism of splenic DCs to a more glycolytic phenotype. To further investigate the effect of PrA on the functional and metabolic phenotype of DCs, the compound was added into the in vitro culture of MyHC-α-loaded DCs. These cells switched to a tolerogenic state and a metabolic profile similar to that found in cells during in ex vivo experiments. Treatment with glycolytic inhibitor 2-DG significantly reversed PrA-mediated DC tolerogenic properties, suggesting that glycolysis is indispensable for PrA-conditioned DCs to maintain their tolerogenic properties. Notably, PrA-conditioned DC vaccinations dampened EAM progress, and promoted Tregs expansion. Similarly, tolerogenic and metabolic patterns were also observed in PrA-modified human DC. In conclusion, PrA endows DC with a tolerogenic profile via glycolytic reprogramming, thereby inducing expansion of immunosuppressive Tregs, and preventing EAM progress. Our results suggested that PrA may confer immunosuppressive and protective effects on EAM by metabolically reprogramming DCs, which could contribute to the development of a new potential immunotherapy for the treatment of EAM and immune-related disorders.
Collapse
Affiliation(s)
- Jian Wu
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China; The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang Province, China
| | - Mingyang Liu
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China; The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang Province, China
| | - Ge Mang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China; The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang Province, China
| | - Shan Yu
- Department of Pathology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Qi Chen
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China; The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang Province, China
| | - Tingting Li
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China; The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang Province, China
| | - Yongchen Wang
- Department of General Practice, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Ying Meng
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China; The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang Province, China
| | - XinYue Tang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China; The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang Province, China
| | - Yang Zheng
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China; The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang Province, China
| | - Yong Sun
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China; The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang Province, China
| | - Maomao Zhang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China; The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang Province, China.
| | - Bo Yu
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China; The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang Province, China
| |
Collapse
|
33
|
Carrillo-Salinas FJ, Ngwenyama N, Anastasiou M, Kaur K, Alcaide P. Heart Inflammation: Immune Cell Roles and Roads to the Heart. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 189:1482-1494. [PMID: 31108102 DOI: 10.1016/j.ajpath.2019.04.009] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 03/29/2019] [Accepted: 04/18/2019] [Indexed: 02/08/2023]
Abstract
Heart failure (HF) has been traditionally viewed as a disease of the cardiac muscle associated with systemic inflammation. Burgeoning evidence implicates immune effector mechanisms that include immune cell activation and trafficking to the heart. Immune cell infiltration in the myocardium can have adverse effects in the heart and contribute to the pathogenesis of HF. Both innate and adaptive immunity operate sequentially, and the specificity of these responses depends on the initial trigger sensed by the heart. Although the role of the immune system in the initial inflammatory response to infection and injury is well studied, what sets the trajectory to HF from different etiologies and the role of immunity once HF has been established is less understood. Herein, we review experimental and clinical knowledge of cardiac inflammation induced by different triggers that often result in HF from different etiologies. We focus on the mechanisms of immune cell activation systemically and on the pathways immune cells use to traffic to the heart.
Collapse
Affiliation(s)
| | - Njabulo Ngwenyama
- Department of Immunology, Tufts University School of Medicine, Boston, Massachusetts; Sackler School for Graduate Studies Immunology Program, Tufts University School of Medicine, Boston, Massachusetts
| | - Marina Anastasiou
- Department of Immunology, Tufts University School of Medicine, Boston, Massachusetts
| | - Kuljeet Kaur
- Department of Immunology, Tufts University School of Medicine, Boston, Massachusetts
| | - Pilar Alcaide
- Department of Immunology, Tufts University School of Medicine, Boston, Massachusetts; Sackler School for Graduate Studies Immunology Program, Tufts University School of Medicine, Boston, Massachusetts.
| |
Collapse
|
34
|
Blanton RM, Carrillo-Salinas FJ, Alcaide P. T-cell recruitment to the heart: friendly guests or unwelcome visitors? Am J Physiol Heart Circ Physiol 2019; 317:H124-H140. [PMID: 31074651 DOI: 10.1152/ajpheart.00028.2019] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Myocardial inflammation can lead to lethal acute or chronic heart failure (HF). T lymphocytes (T cells), have been reported in the inflamed heart in different etiologies of HF, and more recent studies support that different T-cell subsets play distinct roles in the heart depending on the inflammation-triggering event. T cells follow sequential steps to extravasate into tissues, but their specific recruitment to the heart is determined by several factors. These include differences in T-cell responsiveness to specific chemokines in the heart environment, as well as differences in the expression of adhesion molecules in response to distinct stimuli, which regulate T-cell recruitment to the heart and have consequences in cardiac remodeling and function. This review focuses on recent advances in our understanding of the role T cells play in the heart, including its critical role for host defense to virus and myocardial healing postischemia, and its pathogenic role in chronic ischemic and nonischemic HF. We discuss a variety of mechanisms that contribute to the inflammatory damage to the heart, as well as regulatory mechanisms that limit the magnitude of T-cell-mediated inflammation. We also highlight areas in which further research is needed to understand the role T cells play in the heart and distinguish the findings reported in experimental animal models and how they may translate to clinical observations in the human heart.
Collapse
Affiliation(s)
- Robert M Blanton
- Molecular Cardiology Research Institute, Tufts Medical Center , Boston, Massachusetts
| | | | - Pilar Alcaide
- Department of Immunology, Tufts University School of Medicine, Boston, Massachusetts
| |
Collapse
|
35
|
Yue W, Tong L, Liu X, Weng X, Chen X, Wang D, Dudley SC, Weir EK, Ding W, Lu Z, Xu Y, Chen Y. Short term Pm2.5 exposure caused a robust lung inflammation, vascular remodeling, and exacerbated transition from left ventricular failure to right ventricular hypertrophy. Redox Biol 2019; 22:101161. [PMID: 30861460 PMCID: PMC6411784 DOI: 10.1016/j.redox.2019.101161] [Citation(s) in RCA: 142] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Revised: 03/04/2019] [Accepted: 03/05/2019] [Indexed: 11/17/2022] Open
Abstract
Heart failure (HF) is the single largest cause for increased hospitalization after fine particulate matter (PM2.5) exposure. Patients with left HF often progress to right ventricular (RV) failure even with optimal medical care. An increase of PM2.5 of 10 μg per cubic meter was associated with a 76% increase in the risk of death from cardiovascular disease in 4 years' period. However, the role and mechanism of PM2.5 in HF progression are not known. Here we investigated the role of PM2.5 exposure in mice with existing HF mice produced by transverse aortic constriction (TAC). TAC-induced HF caused lung inflammation, vascular remodeling and RV hypertrophy. We found increased PM2.5 profoundly exacerbated lung oxidative stress in mice with existing left HF. To our surprise, PM2.5 exposure had no effect on LV hypertrophy and function, but profoundly exacerbated lung inflammation, vascular remodeling, and RV hypertrophy in mice with existing left HF. These striking findings demonstrate that PM2.5 and/or air pollution is a critical factor for overall HF progression by regulating lung oxidative stress, inflammation and remodeling as well as RV hypertrophy. Improving air quality may save HF patients from a dismal fate.
Collapse
Affiliation(s)
- Wenhui Yue
- Department of Cardiology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Lei Tong
- College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiaohong Liu
- Shanxi Provincial People's Hospital, Taiyuan, China
| | - Xinyu Weng
- Department of Cardiology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xiaoyu Chen
- College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Dongzhi Wang
- Department of Cardiology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Samuel C Dudley
- Cardiovascular Division and Lillehei Heart Institute, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - E Kenneth Weir
- Cardiovascular Division and Lillehei Heart Institute, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Wenjun Ding
- College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhongbing Lu
- College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Yawei Xu
- Department of Cardiology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China.
| | - Yingjie Chen
- Cardiovascular Division and Lillehei Heart Institute, University of Minnesota Medical School, Minneapolis, MN 55455, USA.
| |
Collapse
|
36
|
Liu X, Yang L, Kwak D, Hou L, Shang R, Meyer C, Panoskaltsis-Mortari A, Xu X, Weir EK, Chen Y. Profound Increase of Lung Airway Resistance in Heart Failure: a Potential Important Contributor for Dyspnea. J Cardiovasc Transl Res 2019; 12:271-279. [PMID: 30680546 DOI: 10.1007/s12265-019-9864-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 01/03/2019] [Indexed: 12/18/2022]
Abstract
Dyspnea is a major symptom of heart failure (HF). Here, we have studied the lung remodeling and airway resistance in HF mice. We demonstrated that aortic banding-induced HF caused a dramatic decrease of lung compliance and an increase of lung airway resistance. The decrease of lung compliance was correlated with the increased lung weight in a linear fashion (γ2 = 0.824). An HF-induced increase of lung airway resistance and a decrease of lung compliance were almost identical in anesthetized mice and in the isolated lungs from these mice. HF caused profound lung fibrosis in mice with increased lung weight. Moreover, HF patients of NYHA class III-IV showed increased lung density as revealed by high-resolution CT scanning. These data indicate that lung compliance and lung airway resistance may be useful in determining lung remodeling after HF, and lung structure changes may contribute to dyspnea in HF.
Collapse
Affiliation(s)
- Xiaohong Liu
- Shanxi Provincial People's Hospital, Taiyuan, China
| | - Liuqing Yang
- Cardiovascular Division and Lillehei Heart Institute, University of Minnesota Medical School, Minneapolis, MN, 55455, USA.,Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Dongmin Kwak
- Cardiovascular Division and Lillehei Heart Institute, University of Minnesota Medical School, Minneapolis, MN, 55455, USA.,Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Lei Hou
- Cardiovascular Division and Lillehei Heart Institute, University of Minnesota Medical School, Minneapolis, MN, 55455, USA.,Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Ruru Shang
- Shanxi Provincial People's Hospital, Taiyuan, China
| | - Carolyn Meyer
- Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Angela Panoskaltsis-Mortari
- Cardiovascular Division and Lillehei Heart Institute, University of Minnesota Medical School, Minneapolis, MN, 55455, USA.,Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Xin Xu
- Department of Exercise Rehabilitation, Shanghai University of Sport, Shanghai, 200438, China
| | | | - Yingjie Chen
- Cardiovascular Division and Lillehei Heart Institute, University of Minnesota Medical School, Minneapolis, MN, 55455, USA. .,Department of Medicine, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
37
|
Abstract
Previous studies have suggested the involvement of CD4 + T lymphocytes in cardiac remodelling. T-bet can direct Th1 lineage commitment. This study aimed to investigate the functional significance of T-bet in cardiac remodelling induced by pressure overload using T-bet global knockout rats. Increased T-bet levels were observed in rodent and human hypertrophied hearts. T-bet deficiency resulted in a less severe hypertrophic phenotype in rats. CD4 + T-lymphocyte reconstitution in T-bet-/- rats resulted in aggravated cardiac remodelling. T-cell homing molecule expression and cytokine secretion were altered in T-bet-deficient rat hearts. Administration of exogenous interferon-γ (IFN-γ) offset T-bet deficiency-mediated cardioprotection. Cardiomyocytes cultured in T-bet-/- CD4 + T-cell-conditioned media showed a reduced hypertrophic response after hypertrophic stimuli, which was abolished by an IFN-γ-neutralizing antibody. Taken together, our findings show that T-bet deficiency attenuates pressure overload-induced cardiac remodelling in rats. Specifically, targeting T-bet in T cells may be of great importance for the treatment of pathological cardiac remodelling and heart failure.
Collapse
|
38
|
EZH2 Inhibition Ameliorates Transverse Aortic Constriction-Induced Pulmonary Arterial Hypertension in Mice. Can Respir J 2018; 2018:9174926. [PMID: 29854032 PMCID: PMC5960552 DOI: 10.1155/2018/9174926] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 01/08/2018] [Indexed: 12/30/2022] Open
Abstract
Background EPZ005687 is a selective inhibiter of methyltransferase EZH2. In this article, we investigated the protective role and mechanism of EPZ005687 in transverse aortic constriction-induced pulmonary arterial hypertension in mice. Methods We assigned 15 (6–8 weeks old) male balb/c mice to 3 groups randomly: Sham control + DMSO group, TAC + DMSO group, and TAC + EPZ005687 group (10 mg kg−1, once a week for 4 weeks). On day 28 following TAC operation, the right ventricular systolic blood pressure (RVSBP) was measured, and lung tissues were collected for laboratory examinations (DHE, Western blot, real-time PCR, and ChIP). Results Murine PAH model was successfully created by TAC operation as evidenced by increased RVSBP and hypertrophic right ventricle. Compared with the sham control, TAC-induced PAH markedly upregulated the expression of EZH2 and ROS deposition in lungs in PAH mice. The inhibiter of methyltransferase EZH2, EPZ005687 significantly inhibits the development of TAC-induced PAH in an EZH2-SOD1-ROS dependent manner. Conclusion Our data identified that EZH2 serves a fundamental role in TAC-induced PAH, and administration of EPZ005687 might represent a novel therapeutic target for the treatment of TAC-induced PAH.
Collapse
|
39
|
Koentges C, Pepin ME, Müsse C, Pfeil K, Alvarez SVV, Hoppe N, Hoffmann MM, Odening KE, Sossalla S, Zirlik A, Hein L, Bode C, Wende AR, Bugger H. Gene expression analysis to identify mechanisms underlying heart failure susceptibility in mice and humans. Basic Res Cardiol 2017; 113:8. [PMID: 29288409 DOI: 10.1007/s00395-017-0666-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 12/19/2017] [Indexed: 12/27/2022]
Abstract
Genetic factors are known to modulate cardiac susceptibility to ventricular hypertrophy and failure. To determine how strain influences the transcriptional response to pressure overload-induced heart failure (HF) and which of these changes accurately reflect the human disease, we analyzed the myocardial transcriptional profile of mouse strains with high (C57BL/6J) and low (129S1/SvImJ) susceptibility for HF development, which we compared to that of human failing hearts. Following transverse aortic constriction (TAC), C57BL/6J mice developed overt HF while 129S1/SvImJ did not. Despite a milder aortic constriction, impairment of ejection fraction and ventricular remodeling (dilation, fibrosis) was more pronounced in C57BL/6J mice. Similarly, changes in myocardial gene expression were more robust in C57BL/6J (461 genes) compared to 129S1/SvImJ mice (71 genes). When comparing these patterns to human dilated cardiomyopathy (1344 genes), C57BL/6J mice tightly grouped to human hearts. Overlay and bioinformatic analysis of the transcriptional profiles of C57BL/6J mice and human failing hearts identified six co-regulated genes (POSTN, CTGF, FN1, LOX, NOX4, TGFB2) with established link to HF development. Pathway enrichment analysis identified angiotensin and IGF-1 signaling as most enriched putative upstream regulator and pathway, respectively, shared between TAC-induced HF in C57BL/6J mice and in human failing hearts. TAC-induced heart failure in C57BL/6J mice more closely reflects the gene expression pattern of human dilated cardiomyopathy compared to 129S1/SvImJ mice. Unbiased as well as targeted gene expression and pathway analyses identified periostin, angiotensin signaling, and IGF-1 signaling as potential causes of increased HF susceptibility in C57BL/6J mice and as potentially useful drug targets for HF treatment.
Collapse
Affiliation(s)
- Christoph Koentges
- Cardiology and Angiology I, Heart Center, Freiburg University, Hugstetter Str. 55, 79106, Freiburg, Germany
| | - Mark E Pepin
- Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham, 901 19th Street South, BMR2 Rm 506, Birmingham, AL, 35294, USA
| | - Carolyn Müsse
- Cardiology and Angiology I, Heart Center, Freiburg University, Hugstetter Str. 55, 79106, Freiburg, Germany
| | - Katharina Pfeil
- Cardiology and Angiology I, Heart Center, Freiburg University, Hugstetter Str. 55, 79106, Freiburg, Germany
| | - Sonia V Viteri Alvarez
- Cardiology and Angiology I, Heart Center, Freiburg University, Hugstetter Str. 55, 79106, Freiburg, Germany
| | - Natalie Hoppe
- Cardiology and Angiology I, Heart Center, Freiburg University, Hugstetter Str. 55, 79106, Freiburg, Germany
| | - Michael M Hoffmann
- Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Institute for Clinical Chemistry and Laboratory Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Katja E Odening
- Cardiology and Angiology I, Heart Center, Freiburg University, Hugstetter Str. 55, 79106, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Samuel Sossalla
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Andreas Zirlik
- Cardiology and Angiology I, Heart Center, Freiburg University, Hugstetter Str. 55, 79106, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Lutz Hein
- Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Institute of Experimental and Clinical Pharmacology, BIOSS Center for Biological Signaling Studies, University of Freiburg, Freiburg, Germany
| | - Christoph Bode
- Cardiology and Angiology I, Heart Center, Freiburg University, Hugstetter Str. 55, 79106, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Adam R Wende
- Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham, 901 19th Street South, BMR2 Rm 506, Birmingham, AL, 35294, USA.
| | - Heiko Bugger
- Cardiology and Angiology I, Heart Center, Freiburg University, Hugstetter Str. 55, 79106, Freiburg, Germany. .,Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
40
|
Gröschel C, Sasse A, Röhrborn C, Monecke S, Didié M, Elsner L, Kruse V, Bunt G, Lichtman AH, Toischer K, Zimmermann WH, Hasenfuß G, Dressel R. T helper cells with specificity for an antigen in cardiomyocytes promote pressure overload-induced progression from hypertrophy to heart failure. Sci Rep 2017; 7:15998. [PMID: 29167489 PMCID: PMC5700082 DOI: 10.1038/s41598-017-16147-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 11/08/2017] [Indexed: 12/13/2022] Open
Abstract
We investigated whether CD4+-T cells with specificity for an antigen in cardiomyocytes promote the progression from hypertrophy to heart failure in mice with increased pressure load due to transverse aortic constriction (TAC). OT-II mice expressing a transgenic T cell receptor (TCR) with specificity for ovalbumin (OVA) on CD4+-T cells and cMy-mOVA mice expressing OVA on cardiomyocytes were crossed. The resulting cMy-mOVA-OT-II mice did not display signs of spontaneous autoimmunity despite the fact that their OVA-specific CD4+-T cells were not anergic. After TAC, progression to heart failure was significantly accelerated in cMy-mOVA-OT-II compared to cMy-mOVA mice. No OVA-specific antibodies were induced in response to TAC in cMy-mOVA-OT-II mice, yet more CD3+ T cells infiltrated their myocardium when compared with TAC-operated cMy-mOVA mice. Systemically, the proportion of activated CD4+-T cells with a Th1 and Th17 cytokine profile was increased in cMy-mOVA-OT-II mice after TAC. Thus, T helper cells with specificity for an antigen in cardiomyocytes can directly promote the progression of heart failure in response to pressure overload independently of autoantibodies.
Collapse
Affiliation(s)
- Carina Gröschel
- Institute of Cellular and Molecular Immunology, University Medical Center Göttingen, Göttingen, Germany.,DZHK (German Center for Cardiovascular Research), partner site Göttingen, Göttingen, Germany
| | - André Sasse
- Institute of Cellular and Molecular Immunology, University Medical Center Göttingen, Göttingen, Germany
| | - Charlotte Röhrborn
- Institute of Cellular and Molecular Immunology, University Medical Center Göttingen, Göttingen, Germany
| | - Sebastian Monecke
- Institute of Cellular and Molecular Immunology, University Medical Center Göttingen, Göttingen, Germany.,DZHK (German Center for Cardiovascular Research), partner site Göttingen, Göttingen, Germany
| | - Michael Didié
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Göttingen, Germany.,Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Göttingen, Germany.,Department of Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany
| | - Leslie Elsner
- Institute of Cellular and Molecular Immunology, University Medical Center Göttingen, Göttingen, Germany
| | - Vanessa Kruse
- Institute of Cellular and Molecular Immunology, University Medical Center Göttingen, Göttingen, Germany
| | - Gertrude Bunt
- Clinical Optical Microscopy, Department of Neuropathology, University Medical Center Göttingen, Göttingen, Germany
| | - Andrew H Lichtman
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Karl Toischer
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Göttingen, Germany.,Department of Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany
| | - Wolfram-Hubertus Zimmermann
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Göttingen, Germany.,Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Göttingen, Germany
| | - Gerd Hasenfuß
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Göttingen, Germany.,Department of Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany
| | - Ralf Dressel
- Institute of Cellular and Molecular Immunology, University Medical Center Göttingen, Göttingen, Germany. .,DZHK (German Center for Cardiovascular Research), partner site Göttingen, Göttingen, Germany.
| |
Collapse
|
41
|
Brown SM, Smith CE, Meuth AI, Khan M, Aroor AR, Cleeton HM, Meininger GA, Sowers JR, DeMarco VG, Chandrasekar B, Nistala R, Bender SB. Dipeptidyl Peptidase-4 Inhibition With Saxagliptin Ameliorates Angiotensin II-Induced Cardiac Diastolic Dysfunction in Male Mice. Endocrinology 2017; 158:3592-3604. [PMID: 28977602 PMCID: PMC5659692 DOI: 10.1210/en.2017-00416] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 07/25/2017] [Indexed: 12/28/2022]
Abstract
Activation of the renin-angiotensin-aldosterone system is common in hypertension and obesity and contributes to cardiac diastolic dysfunction, a condition for which no treatment currently exists. In light of recent reports that antihyperglycemia incretin enhancing dipeptidyl peptidase (DPP)-4 inhibitors exert cardioprotective effects, we examined the hypothesis that DPP-4 inhibition with saxagliptin (Saxa) attenuates angiotensin II (Ang II)-induced cardiac diastolic dysfunction. Male C57BL/6J mice were infused with either Ang II (500 ng/kg/min) or vehicle for 3 weeks receiving either Saxa (10 mg/kg/d) or placebo during the final 2 weeks. Echocardiography revealed Ang II-induced diastolic dysfunction, evidenced by impaired septal wall motion and prolonged isovolumic relaxation, coincident with aortic stiffening. Ang II induced cardiac hypertrophy, coronary periarterial fibrosis, TRAF3-interacting protein 2 (TRAF3IP2)-dependent proinflammatory signaling [p-p65, p-c-Jun, interleukin (IL)-17, IL-18] associated with increased cardiac macrophage, but not T cell, gene expression. Flow cytometry revealed Ang II-induced increases of cardiac CD45+F4/80+CD11b+ and CD45+F4/80+CD11c+ macrophages and CD45+CD4+ lymphocytes. Treatment with Saxa reduced plasma DPP-4 activity and abrogated Ang II-induced cardiac diastolic dysfunction independent of aortic stiffening or blood pressure. Furthermore, Saxa attenuated Ang II-induced periarterial fibrosis and cardiac inflammation, but not hypertrophy or cardiac macrophage infiltration. Analysis of Saxa-induced changes in cardiac leukocytes revealed Saxa-dependent reduction of the Ang II-mediated increase of cardiac CD11c messenger RNA and increased cardiac CD8 gene expression and memory CD45+CD8+CD44+ lymphocytes. In summary, these results demonstrate that DPP-4 inhibition with Saxa prevents Ang II-induced cardiac diastolic dysfunction, fibrosis, and inflammation associated with unique shifts in CD11c-expressing leukocytes and CD8+ lymphocytes.
Collapse
Affiliation(s)
- Scott M. Brown
- Research, Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri 65201
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri 65211
| | - Cassandra E. Smith
- Research, Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri 65201
- Division of Endocrinology, Diabetes, and Metabolism, University of Missouri School of Medicine, Columbia, Missouri 65212
| | - Alex I. Meuth
- Research, Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri 65201
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri 65211
| | - Maloree Khan
- Research, Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri 65201
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri 65211
| | - Annayya R. Aroor
- Research, Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri 65201
- Division of Endocrinology, Diabetes, and Metabolism, University of Missouri School of Medicine, Columbia, Missouri 65212
| | - Hannah M. Cleeton
- Research, Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri 65201
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri 65211
| | - Gerald A. Meininger
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri 65211
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri 65212
| | - James R. Sowers
- Research, Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri 65201
- Division of Endocrinology, Diabetes, and Metabolism, University of Missouri School of Medicine, Columbia, Missouri 65212
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri 65211
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri 65212
| | - Vincent G. DeMarco
- Research, Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri 65201
- Division of Endocrinology, Diabetes, and Metabolism, University of Missouri School of Medicine, Columbia, Missouri 65212
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri 65212
| | - Bysani Chandrasekar
- Research, Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri 65201
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri 65211
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri 65212
- Division of Cardiovascular Medicine, Department of Medicine, University of Missouri School of Medicine, Columbia, Missouri 65212
| | - Ravi Nistala
- Research, Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri 65201
- Division of Nephrology, University of Missouri School of Medicine, Columbia, Missouri 65212
| | - Shawn B. Bender
- Research, Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri 65201
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri 65211
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri 65211
| |
Collapse
|
42
|
Trial J, Heredia CP, Taffet GE, Entman ML, Cieslik KA. Dissecting the role of myeloid and mesenchymal fibroblasts in age-dependent cardiac fibrosis. Basic Res Cardiol 2017; 112:34. [PMID: 28478479 DOI: 10.1007/s00395-017-0623-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 04/27/2017] [Indexed: 12/24/2022]
Abstract
Aging is associated with increased cardiac interstitial fibrosis and diastolic dysfunction. Our previous study has shown that mesenchymal fibroblasts in the C57BL/6J (B6J) aging mouse heart acquire an inflammatory phenotype and produce higher levels of chemokines. Monocyte chemoattractant protein-1 (MCP-1) secreted by these aged fibroblasts promotes leukocyte uptake into the heart. Some of the monocytes that migrate into the heart polarize into M2a macrophages/myeloid fibroblasts. The number of activated mesenchymal fibroblasts also increases with age, and consequently, both sources of fibroblasts contribute to fibrosis. Here, we further investigate mechanisms by which inflammation influences activation of myeloid and mesenchymal fibroblasts and their collagen synthesis. We examined cardiac fibrosis and heart function in three aged mouse strains; we compared C57BL/6J (B6J) with two other strains that have reduced inflammation via different mechanisms. Aged C57BL/6N (B6N) hearts are protected from oxidative stress and fibroblasts derived from them do not develop an inflammatory phenotype. Likewise, these mice have preserved diastolic function. Aged MCP-1 null mice on the B6J background (MCP-1KO) are protected from elevated leukocyte infiltration; they develop moderate but reduced fibrosis and diastolic dysfunction. Based on these studies, we further delineated the role of resident versus monocyte-derived M2a macrophages in myeloid-dependent fibrosis and found that the number of monocyte-derived M2a (but not resident) macrophages correlates with age-related fibrosis and diastolic dysfunction. In conclusion, we have found that ROS and inflammatory mediators are necessary for activation of fibroblasts of both developmental origins, and prevention of either led to better functional outcomes.
Collapse
Affiliation(s)
- JoAnn Trial
- Division of Cardiovascular Sciences and the DeBakey Heart Center, Department of Medicine, Baylor College of Medicine, One Baylor Plaza, M.S. BCM620, Houston, TX, 77030, USA
| | - Celia Pena Heredia
- Division of Cardiovascular Sciences and the DeBakey Heart Center, Department of Medicine, Baylor College of Medicine, One Baylor Plaza, M.S. BCM620, Houston, TX, 77030, USA
| | - George E Taffet
- Division of Cardiovascular Sciences and the DeBakey Heart Center, Department of Medicine, Baylor College of Medicine, One Baylor Plaza, M.S. BCM620, Houston, TX, 77030, USA
| | - Mark L Entman
- Division of Cardiovascular Sciences and the DeBakey Heart Center, Department of Medicine, Baylor College of Medicine, One Baylor Plaza, M.S. BCM620, Houston, TX, 77030, USA.,Houston Methodist, Houston, TX, USA
| | - Katarzyna A Cieslik
- Division of Cardiovascular Sciences and the DeBakey Heart Center, Department of Medicine, Baylor College of Medicine, One Baylor Plaza, M.S. BCM620, Houston, TX, 77030, USA.
| |
Collapse
|