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
|
Li G, He W, Wang DW. Immune cell dynamics in heart failure: implicated mechanisms and therapeutic targets. ESC Heart Fail 2025; 12:1739-1758. [PMID: 39905753 PMCID: PMC12055366 DOI: 10.1002/ehf2.15238] [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: 10/24/2024] [Revised: 01/05/2025] [Accepted: 01/21/2025] [Indexed: 02/06/2025] Open
Abstract
The relationship between heart failure (HF) and immune activation has garnered significant interest. Studies highlight the critical role of inflammation in HF, affecting cardiac structure and function. Despite promising anti-inflammatory therapies, clinical trials have faced challenges, indicating an incomplete understanding of immune mechanisms in HF. Immune cells, which are key cytokine sources, are pivotal in HF progression. In this review, the authors provide a comprehensive overview of the complex role of different types of immune cells and their cell subtypes in HF. In addition, the authors summarize the available targets and animal experimental evidence for targeting immune cells for the treatment of HF. Future research directions will focus on the roles of immune cells and their interrelationships at different stages of HF, aiming to develop more targeted therapeutic strategies that can achieve more precise interventions in the pathological process of HF.
Collapse
Affiliation(s)
- Gen Li
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhan430000China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological DisordersWuhan430000China
| | - Wu He
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhan430000China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological DisordersWuhan430000China
| | - Dao Wen Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhan430000China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological DisordersWuhan430000China
| |
Collapse
|
3
|
Wang S, Pei G, Shen J, Fang Z, Chen T, Wang L, Cheng H, Li H, Pei H, Feng Q, Fan Q, He C, Fu C, Guo Y, Wei Q. Pulsed electromagnetic fields treatment ameliorates cardiac function after myocardial infarction in mice and pigs. J Adv Res 2025:S2090-1232(25)00263-2. [PMID: 40250556 DOI: 10.1016/j.jare.2025.04.019] [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: 05/23/2024] [Revised: 09/21/2024] [Accepted: 04/15/2025] [Indexed: 04/20/2025] Open
Abstract
INTRODUCTION Ischemic heart disease (IHD) is a prominent contributor to mortality worldwide, with myocardial infarction (MI) representing its most severe manifestation. Pulsed electromagnetic fields (PEMF) treatment shows promise for treating IHD. Nevertheless, the therapeutic impact and underlying mechanism of PEMF in MI are not fully understood. OBJECTIVES To investigate the efficacy, safety, and mechanisms of PEMF for MI. METHODS We established MI models in both mice and pigs and performed serial echocardiography and cardiac magnetic resonance follow-up to demonstrate the benefit of PEMF treatment after MI. The pathological environment after myocardial infarction was simulated in vitro to observe changes in various cells exposed to PEMF. Gene knockout (TLR4-/-) mice and inhibitors were used to compare the differences in the efficacy of PEMF treatment relative to that of gene knockout/inhibitor treatments. Agonists were used to further explore the mechanism of PEMF treatment. RESULTS In post-MI mice, PEMF treatment enhanced cardiac function and reduced scar formation. PEMF reduced the macrophage inflammatory response, improved cardiomyocyte survival in an inflammatory environment, and decreased collagen secretion by fibroblasts in vitro. Importantly, in the clinically relevant porcine model, PEMF treatment inhibited the inflammatory response and alleviated adverse left ventricular remodeling. Moreover, PEMF could exert therapeutic effects similar to those of gene knockout or inhibitor treatments. In the presence of TLR4 knockout or pyrrolidine dithiocarbamate (an NF-κB inhibitor) administration, PEMF could still improve cardiac function in post-MI mice. Mechanistically, the anti-inflammatory effect of PEMF was reversed when RS09 (a TLR4 agonist) was administered, and the antifibrotic effect of PEMF was attenuated after treatment with SRI-011381 (a TGF-β signaling pathway agonist). CONCLUSIONS PEMF treatment exhibits considerable promise as a noninvasive physical therapy modality, warranting further investigation into its potential implications for managing patients with IHD.
Collapse
Affiliation(s)
- Shiqi Wang
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu 610041, China; Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Gaiqin Pei
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu 610041, China; Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jiayu Shen
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhi Fang
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Tingyu Chen
- Department of Radiology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Lu Wang
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu 610041, China; Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hongxin Cheng
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu 610041, China; Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hanbin Li
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu 610041, China; Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hongliang Pei
- School of Mechanical Engineering, Sichuan University, Chengdu 610041, China
| | - Qipu Feng
- Animal Experiment Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qingwen Fan
- School of Mechanical Engineering, Sichuan University, Chengdu 610041, China
| | - Chengqi He
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu 610041, China; Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Chenying Fu
- State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China; Aging and Geriatric Mechanism Laboratory, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Yingqiang Guo
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Quan Wei
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu 610041, China; Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
4
|
Wang L, Lin S, Wei Q, Li T, Mo Q, Bai R, Feng J, Zhan A, Yang X, Rong X, Guo J. Preventive Administration of Quercetin Promotes Survival and Reduces Adverse Ventricular Remodeling after myocardial infarction through facilitating M2-like macrophage polarization. Int Immunopharmacol 2025; 151:114296. [PMID: 39983422 DOI: 10.1016/j.intimp.2025.114296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 02/06/2025] [Accepted: 02/11/2025] [Indexed: 02/23/2025]
Abstract
Myocardial infarction (MI) remains a leading cause of global health burden, and adverse cardiac remodeling after MI seriously affects patient recovery. Macrophages play an important role in the cardiac remodeling post-MI. Quercitrin (Que), a bioflavonoid commonly found in fruits, vegetables, and various Chinese medicines, possesses a therapeutic effect in MI, but whether it has a role in the prevention of MI is unclear. This study investigated the potential preventive value and mechanism of Que against MI. In this study, we treated adult male C57BL/6 mice with Que for 2 weeks and then constructed the MI model. We found that pre-treatment with Que improved cardiac fractional shortening and ejection fraction, and elevated the survival of mice after MI. In addition, pre-administration of Que attenuated cardiac hypertrophy and diminished the infarct size of the heart post-MI. Picrosirius red staining of heart sections and detection of fibrosis markers' levels by real-time polymerase chain reaction and western blot analyses revealed that Que repressed cardiac fibrosis after MI. Que pre-administration inhibited the levels of inflammatory factors and the infiltration of inflammatory cells, and increased the proportion of M2-like macrophages in the infarcted area of the heart. Furthermore, we found that Que pre-treatment polarized macrophage from M1-like to M2-like, which promoted the proliferation, migration, and activation of cardiac fibroblasts in vitro. Collectively, these data demonstrated that pre-administration Que promoted survival and reduced adverse ventricular remodeling after MI partially through modifying macrophage polarization. This provides an experimental basis for the future application of Que in cardiovascular diseases including MI.
Collapse
Affiliation(s)
- Lexun Wang
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China; Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine; Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, Guangdong Province, China
| | - Shaolin Lin
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China; Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine; Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, Guangdong Province, China
| | - Quxing Wei
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China; Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine; Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, Guangdong Province, China
| | - Tongjun Li
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China; Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine; Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, Guangdong Province, China
| | - Quqian Mo
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China; Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine; Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, Guangdong Province, China
| | - Ruining Bai
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China; Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine; Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, Guangdong Province, China
| | - Jiaojiao Feng
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China; Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine; Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, Guangdong Province, China
| | - Angyu Zhan
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China; Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine; Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, Guangdong Province, China
| | - Xiao Yang
- Department of Clinical Laboratory, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong Province, China
| | - Xianglu Rong
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China; Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine; Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, Guangdong Province, China
| | - Jiao Guo
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China; Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine; Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, Guangdong Province, China.
| |
Collapse
|
5
|
Koenig AL, Kadyrov FF, Amrute JM, Yang S, Weinheimer CJ, Nigro JM, Kovacs A, Smith G, Lavine KJ. Genetic Mapping of Monocyte Fate Decisions Following Myocardial Infarction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.12.24.573263. [PMID: 39974922 PMCID: PMC11838486 DOI: 10.1101/2023.12.24.573263] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Inflammation contributes to the pathogenesis of myocardial infarction and heart failure and represents a viable therapeutic target. Monocytes and their progeny are highly abundant and display incredible functional diversity, serving as key determinants of myocardial inflammation and tissue repair. Much remains to be learned regarding mechanisms and signaling events that instruct monocyte fate decisions. We devised a genetic lineage tracing strategy using Ccr2 crERT2 Rosa26 LSL-tdTomato mice in combination with single cell RNA-sequencing to map the differentiation trajectories of monocytes that infiltrate the heart after reperfused myocardial infarction. Monocytes are recruited to the heart early after injury and give rise to transcriptionally distinct and spatially restricted macrophage and dendritic cell-like subsets that are specified prior to extravasation and chronically persist within the myocardium. Pseudotime analysis predicted two differentiation trajectories of monocyte-derived macrophages that are partitioned into the border and infarct zones, respectively. Among these trajectories, we show that macrophages expressing a type I IFN responsive signature are an intermediate population that gives rise to MHC-II hi macrophages, are localized within the border zone, and promote myocardial protection. Collectively, these data uncover new complexities of monocyte differentiation in the infarcted heart and suggest that modulating monocyte fate decisions may have clinical implications.
Collapse
|
6
|
Sharma I, Bhardwaj S, Karwasra R, Kaushik D, Sharma S. The Emergence of Nanotechnology in the Prognosis and Treatment of Myocardial Infarctions. RECENT PATENTS ON NANOTECHNOLOGY 2025; 19:35-55. [PMID: 37904554 DOI: 10.2174/1872210517666230721123453] [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: 02/16/2023] [Revised: 06/08/2023] [Accepted: 06/16/2023] [Indexed: 11/01/2023]
Abstract
Myocardial infarction (MI), commonly known as a heart attack, is a critical cardiovascular condition associated with high morbidity and mortality rates worldwide. Despite significant advancements in traditional treatment modalities, there remains a need for innovative approaches to improve the prognosis and treatment outcomes of MI. The emergence of nanotechnology has provided a promising avenue for revolutionizing the management of this life-threatening condition. This manuscript aims to explore the role of nanotechnology in the prognosis and treatment of myocardial infarctions. Nanotechnology offers unique advantages in the field of cardiovascular medicine, including targeted drug delivery, precise imaging and diagnosis, regenerative medicine approaches, biosensors and monitoring, and the integration of therapy and diagnostics (theragnostic). One of the key advantages of nanotechnology is the ability to deliver therapeutic agents directly to the affected site. Nanoparticles can be engineered to carry drugs specifically to damaged heart tissue, enhancing their efficacy while minimizing off-target effects. Additionally, nanoparticles can serve as contrast agents, facilitating high-resolution imaging and accurate diagnosis of infarcted heart tissue. Furthermore, nanotechnology-based regenerative approaches show promise in promoting tissue healing and regeneration after MI. Nanomaterials can provide scaffolding structures or release growth factors to stimulate the growth of new blood vessels and support tissue repair. This regenerative potential holds significant implications for restoring cardiac function and minimizing long-term complications. Nanotechnology also enables real-time monitoring of critical parameters within the heart, such as oxygen levels, pH, and electrical activity, through the utilization of nanoscale devices and sensors. This capability allows for the early detection of complications and facilitates timely interventions. Moreover, the integration of therapy and diagnostics through nanotechnology- based platforms, known as theragnostic, holds tremendous potential. Nanoparticles can simultaneously deliver therapeutic agents while providing imaging capabilities, enabling personalized treatment strategies tailored to individual patients. This manuscript will review the recent advancements, clinical trials, and patents in nanotechnology for the prognosis and treatment of myocardial infarctions. By leveraging nanotechnology's unique properties and applications, researchers and clinicians can develop innovative therapeutic approaches that enhance patient outcomes, improve prognosis, and ultimately revolutionize the management of myocardial infarctions.
Collapse
Affiliation(s)
- Isha Sharma
- Department of Pharmaceutical Sciences, Gurugram University, Gurugram, 122018, India
| | - Shivani Bhardwaj
- ICAR- Central Institute for Research on Buffaloes Hisar, Haryana, 125001, India
| | - Ritu Karwasra
- Central Council for Research in Unani Medicine, Ministry of Ayush, Govt. of India, New Delhi, 110058, India
| | - Dhirender Kaushik
- Department of Pharmaceutical Sciences, Gurugram University, Gurugram, 122018, India
| | - Shivkant Sharma
- Department of Pharmaceutical Sciences, Gurugram University, Gurugram, 122018, India
| |
Collapse
|
7
|
Silvain J, Materne C, Zeitouni M, Procopi N, Guedeney P, Brugier D, Galier S, Lhomme M, Ponnaiah M, Guillas I, Kc P, Dahik VD, Frisdal E, Vicaut E, Lesnik P, Rahoual G, Le Goff W, Montalescot G, Kerneis M, Guerin M. Defective biological activities of high-density lipoprotein identify patients at highest risk of recurrent cardiovascular event. Eur J Prev Cardiol 2024:zwae356. [PMID: 39506545 DOI: 10.1093/eurjpc/zwae356] [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: 08/23/2024] [Revised: 10/07/2024] [Accepted: 11/05/2024] [Indexed: 11/08/2024]
Abstract
AIMS Low cholesterol efflux capacity and elevated levels of Interleukin-1ß (IL-1ß) are both associated with residual cardiovascular risk in patients with acute myocardial infarction (MI) and may be used as new biomarkers to identify patients at higher cardiovascular risk. METHODS We evaluated potential synergetic effect of cholesterol efflux capacity and IL-1ß on recurrent major adverse cardiovascular events (MACE) at one-year in 2012 patients with acute ST- segment elevation MI who underwent primary percutaneous coronary intervention. In addition, we evaluated the contribution to residual risk of HDL biological functions from 20 patients of the two extreme subgroups, focusing on cholesterol efflux capacity and anti-inflammatory properties. RESULTS Patients with MACE during the first year after the MI had significantly lower serum cholesterol efflux capacity as compared to those without recurrent events and higher level of IL-1ß, both associations were confirmed after multivariate analysis. We found an inverse relationship between CEC and circulating levels of the inflammatory markers IL-1ß, defining a very high risk (Low CEC/High IL-1ß) and a low risk (High CEC/Low IL-1ß) group of patients. Patients combining Low CEC/High IL-1ß exhibited the highest risk of recurrent MACE at one year showing an additive prognostic value of these biomarkers, regardless of all the other clinical or biological factors. In this very high-risk subgroup, patients exhibited reduced HDL-efflux capacity and defective ABCA1 and SR-BI with enhanced pro-inflammatory activity as a potential explanation for our clinical findings. CONCLUSION Impaired cholesterol efflux capacity and elevated IL-1β synergistically increase the residual cardiovascular risk in MI patients, which could be explained by reduced HDL-efflux capacity and enhanced HDL pro-inflammatory activity.
Collapse
Affiliation(s)
- Johanne Silvain
- ACTION Study Group, Institut de Cardiologie Hôpital Pitié-Salpêtrière (AP-HP), F-75013 Paris, France
- Sorbonne University, INSERM Unité de recherche sur les maladies cardiovasculaires, le métabolisme et la nutrition, UMR_S1166-ICAN F-75013 Paris, France
| | - Clément Materne
- Sorbonne University, INSERM Unité de recherche sur les maladies cardiovasculaires, le métabolisme et la nutrition, UMR_S1166-ICAN F-75013 Paris, France
| | - Michel Zeitouni
- ACTION Study Group, Institut de Cardiologie Hôpital Pitié-Salpêtrière (AP-HP), F-75013 Paris, France
- Sorbonne University, INSERM Unité de recherche sur les maladies cardiovasculaires, le métabolisme et la nutrition, UMR_S1166-ICAN F-75013 Paris, France
| | - Niki Procopi
- ACTION Study Group, Institut de Cardiologie Hôpital Pitié-Salpêtrière (AP-HP), F-75013 Paris, France
- Sorbonne University, INSERM Unité de recherche sur les maladies cardiovasculaires, le métabolisme et la nutrition, UMR_S1166-ICAN F-75013 Paris, France
| | - Paul Guedeney
- ACTION Study Group, Institut de Cardiologie Hôpital Pitié-Salpêtrière (AP-HP), F-75013 Paris, France
- Sorbonne University, INSERM Unité de recherche sur les maladies cardiovasculaires, le métabolisme et la nutrition, UMR_S1166-ICAN F-75013 Paris, France
| | - Delphine Brugier
- ACTION Study Group, Institut de Cardiologie Hôpital Pitié-Salpêtrière (AP-HP), F-75013 Paris, France
- Sorbonne University, INSERM Unité de recherche sur les maladies cardiovasculaires, le métabolisme et la nutrition, UMR_S1166-ICAN F-75013 Paris, France
| | - Sophie Galier
- Sorbonne University, INSERM Unité de recherche sur les maladies cardiovasculaires, le métabolisme et la nutrition, UMR_S1166-ICAN F-75013 Paris, France
| | - Marie Lhomme
- Foundation for Innovation in Cardiometabolism and Nutrition (IHU ICAN, ICAN OMICS and ICAN I/O), F-75013 Paris, France
| | - Maharajah Ponnaiah
- Foundation for Innovation in Cardiometabolism and Nutrition (IHU ICAN, ICAN OMICS and ICAN I/O), F-75013 Paris, France
| | - Isabelle Guillas
- Sorbonne University, INSERM Unité de recherche sur les maladies cardiovasculaires, le métabolisme et la nutrition, UMR_S1166-ICAN F-75013 Paris, France
| | - Pukar Kc
- Sorbonne University, INSERM Unité de recherche sur les maladies cardiovasculaires, le métabolisme et la nutrition, UMR_S1166-ICAN F-75013 Paris, France
| | - Veronica D Dahik
- Sorbonne University, INSERM Unité de recherche sur les maladies cardiovasculaires, le métabolisme et la nutrition, UMR_S1166-ICAN F-75013 Paris, France
| | - Eric Frisdal
- Sorbonne University, INSERM Unité de recherche sur les maladies cardiovasculaires, le métabolisme et la nutrition, UMR_S1166-ICAN F-75013 Paris, France
| | - Eric Vicaut
- ACTION Study Group, Institut de Cardiologie Hôpital Pitié-Salpêtrière (AP-HP), F-75013 Paris, France
- Unité de Recherche Clinique, Hôpital Fernand Widal (AP-HP), Paris, France. SAMM - Statistique, Analyse et Modélisation Multidisciplinaire EA 4543, Université Paris 1 Panthéon Sorbonne, France
| | - Philippe Lesnik
- Sorbonne University, INSERM Unité de recherche sur les maladies cardiovasculaires, le métabolisme et la nutrition, UMR_S1166-ICAN F-75013 Paris, France
| | - Ghilas Rahoual
- ACTION Study Group, Institut de Cardiologie Hôpital Pitié-Salpêtrière (AP-HP), F-75013 Paris, France
| | - Wilfried Le Goff
- Sorbonne University, INSERM Unité de recherche sur les maladies cardiovasculaires, le métabolisme et la nutrition, UMR_S1166-ICAN F-75013 Paris, France
| | - Gilles Montalescot
- ACTION Study Group, Institut de Cardiologie Hôpital Pitié-Salpêtrière (AP-HP), F-75013 Paris, France
- Sorbonne University, INSERM Unité de recherche sur les maladies cardiovasculaires, le métabolisme et la nutrition, UMR_S1166-ICAN F-75013 Paris, France
| | - Mathieu Kerneis
- ACTION Study Group, Institut de Cardiologie Hôpital Pitié-Salpêtrière (AP-HP), F-75013 Paris, France
- Sorbonne University, INSERM Unité de recherche sur les maladies cardiovasculaires, le métabolisme et la nutrition, UMR_S1166-ICAN F-75013 Paris, France
| | - Maryse Guerin
- Sorbonne University, INSERM Unité de recherche sur les maladies cardiovasculaires, le métabolisme et la nutrition, UMR_S1166-ICAN F-75013 Paris, France
| |
Collapse
|
8
|
Choudhury P, Kandula N, Kosuru R, Adena SKR. Nanomedicine: A great boon for cardiac regenerative medicine. Eur J Pharmacol 2024; 982:176969. [PMID: 39218342 DOI: 10.1016/j.ejphar.2024.176969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 08/28/2024] [Accepted: 08/29/2024] [Indexed: 09/04/2024]
Abstract
Cardiovascular disease (CVD) represents a significant global health challenge, remaining the leading cause of illness and mortality worldwide. The adult heart's limited regenerative capacity poses a major obstacle in repairing extensive damage caused by conditions like myocardial infarction. In response to these challenges, nanomedicine has emerged as a promising field aimed at improving treatment outcomes through innovative drug delivery strategies. Nanocarriers, such as nanoparticles (NPs), offer a revolutionary approach by facilitating targeted delivery of therapeutic agents directly to the heart. This precise delivery system holds immense potential for treating various cardiac conditions by addressing underlying mechanisms such as inflammation, oxidative stress, cell death, extracellular matrix remodeling, prosurvival signaling, and angiogenic pathways associated with ischemia-reperfusion injury. In this review, we provide a concise summary of the fundamental mechanisms involved in cardiac remodeling and regeneration. We explore how nanoparticle-based drug delivery systems can effectively target the afore-mentioned mechanisms. Furthermore, we discuss clinical trials that have utilized nanoparticle-based drug delivery systems specifically designed for cardiac applications. These trials demonstrate the potential of nanomedicine in clinical settings, paving the way for future advancements in cardiac therapeutics through precise and efficient drug delivery. Overall, nanomedicine holds promise in revolutionizing the treatment landscape of cardiovascular diseases by offering targeted and effective therapeutic strategies that address the complex pathophysiology of cardiac injuries.
Collapse
Affiliation(s)
- Priyanka Choudhury
- Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Nirupama Kandula
- Department of Microbiology, GSL Medical College, Rajahmahendravaram, Andhra Pradesh, 533296, India
| | - Ramoji Kosuru
- Versiti Blood Research Institute, Milwaukee, WI, 53226, USA
| | - Sandeep Kumar Reddy Adena
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh, 221005, India.
| |
Collapse
|
9
|
Nayak TK, Parasania D, Tilley DG. Adrenergic orchestration of immune cell dynamics in response to cardiac stress. J Mol Cell Cardiol 2024; 196:115-124. [PMID: 39303854 PMCID: PMC12059806 DOI: 10.1016/j.yjmcc.2024.09.010] [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: 06/28/2024] [Revised: 08/30/2024] [Accepted: 09/10/2024] [Indexed: 09/22/2024]
Abstract
Immune cells contribute approximately 5-10 % of the heart's total cell population, including several myeloid cell and lymphocyte cell subsets, which, despite their relatively small percentages, play important roles in cardiac homeostasis and remodeling responses to various forms of injury and long-term stress. Pathological cardiac stress activates the sympathetic nervous system (SNS), resulting in the release of the catecholamines epinephrine and norepinephrine either systemically or from sympathetic nerve terminals within various lymphoid organs. Acting at α- or β-adrenergic receptors (αAR, βAR), catecholamines regulate immune cell hematopoiesis, egress and migration in response to stress. Classically, αAR stimulation tends to promote inflammatory responses while βAR stimulation has typically been shown to be immunosuppressive, though the effects can be nuanced depending on the immune cells subtype, the site of regulation and pathophysiological context. Herein, we will discuss several facets of SNS-mediated regulation of immune cells and their response to cardiac stress, including: catecholamine response to cardiovascular stress and action at their receptors, adrenergic regulation of hematopoiesis, immune cell retention and release from the bone marrow, adrenergic regulation of splenic immune cells and their retention, as well as adrenergic regulation of immune cell recruitment to the injured heart, including neutrophils, monocytes and macrophages. A particular focus will be given to βAR-mediated effects on myeloid cells in response to acute or chronic cardiac stress.
Collapse
Affiliation(s)
- Tapas K Nayak
- Aging + Cardiovascular Discovery Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Dev Parasania
- Aging + Cardiovascular Discovery Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Douglas G Tilley
- Aging + Cardiovascular Discovery Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA.
| |
Collapse
|
10
|
Achilli F, Maggiolini S, Madotto F, Bassetti B, Gentile F, Maggioni AP, Colombo GI, Pompilio G. Granulocyte colony-stimulating factor for stem cell mobilisation in acute myocardial infarction: a randomised controlled trial. Heart 2024; 110:1316-1326. [PMID: 39401872 DOI: 10.1136/heartjnl-2024-323926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 09/02/2024] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND To determine whether granulocyte colony-stimulating factor (G-CSF) improves clinical outcomes after large ST-elevation myocardial infarction (STEMI) when administered early in patients with left ventricular (LV) dysfunction after successful percutaneous coronary intervention (PCI). METHODS STEM-AMI OUTCOME was designed as a prospective, multicentre, nationwide, randomised, open-label, phase III trial (ClinicalTrials.gov ID: NCT01969890) to demonstrate the efficacy and safety of early G-CSF administration in reducing 2-year cardiac mortality and morbidity in patients with STEMI with LV ejection fraction ≤45% after PCI. The primary outcome was a composite of all-cause death, recurrence of myocardial infarction and hospitalisation for heart failure. Due to low recruitment and event rates, the study was discontinued and did not achieve adequate statistical power to verify the hypothesis. RESULTS Patients were randomly allocated to G-CSF (n=260) or standard of care (SOC; n=261). No difference was found in the composite primary outcome between study groups (HR 1.20; 95% CI 0.63 to 2.28). The 2-year mortality was 2.31% in the G-CSF and 2.68% in the control group (HR 0.88; 95% CI 0.29 to 2.60). Adverse events did not differ between the G-CSF (n=65) and SOC groups (n=58; OR 1.17; 95% CI 0.78 to 1.75). In post hoc analyses on the intervention group, we observed a trend towards fewer composite primary outcomes in patients with low bone marrow (BM) cell mobilisation (n=108) versus those with high mobilisation (n=152, with peak leucocyte count >50×109/L; HR 2.86; 95% CI 0.96 to 8.56). Primary outcomes were lower in patients with severe LV systolic dysfunction at discharge treated with G-CSF than in controls (interaction β±SE, -0.08±0.04; p=0.034). CONCLUSIONS Although inconclusive, this is the largest trial in the field of cell-based cardiac repair after STEMI providing evidence of the tolerability and long-term safety of G-CSF treatment. The results prompt further studies to understand which patient can benefit most from BM cell mobilisation. TRIAL REGISTRATION NUMBER NCT01969890.
Collapse
Affiliation(s)
| | | | - Fabiana Madotto
- Dipartimento Area Emergenza-Urgenza, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Beatrice Bassetti
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino Istituto di Ricovero e Cura a Carattere Scientifico, Milano, Italy
| | - Francesco Gentile
- Department of Cardiology, Hospital Bassini, Cinisello Balsamo, Italy
| | | | - Gualtiero I Colombo
- Unit of Immunology and Functional Genomics, Centro Cardiologico Monzino Istituto di Ricovero e Cura a Carattere Scientifico, Milano, Italy
| | - Giulio Pompilio
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino Istituto di Ricovero e Cura a Carattere Scientifico, Milano, Italy
- Department of Biomedical, Surgical and Dental Sciences, Università degli Studi di Milano, Milano, Italy
| |
Collapse
|
11
|
Lin L, Chu J, An S, Liu X, Tan R. The Biological Mechanisms and Clinical Roles of RNA-Binding Proteins in Cardiovascular Diseases. Biomolecules 2024; 14:1056. [PMID: 39334823 PMCID: PMC11430443 DOI: 10.3390/biom14091056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 08/22/2024] [Accepted: 08/23/2024] [Indexed: 09/30/2024] Open
Abstract
RNA-binding proteins (RBPs) have pivotal roles in cardiovascular biology, influencing various molecular mechanisms underlying cardiovascular diseases (CVDs). This review explores the significant roles of RBPs, focusing on their regulation of RNA alternative splicing, polyadenylation, and RNA editing, and their impact on CVD pathogenesis. For instance, RBPs are crucial in myocardial injury, contributing to disease progression and repair mechanisms. This review systematically analyzes the roles of RBPs in myocardial injury, arrhythmias, myocardial infarction, and heart failure, revealing intricate interactions that influence disease outcomes. Furthermore, the potential of RBPs as therapeutic targets for cardiovascular dysfunction is explored, highlighting the advances in drug development and clinical research. This review also discusses the emerging role of RBPs as biomarkers for cardiovascular diseases, offering insights into their diagnostic and prognostic potential. Despite significant progress, current research faces several limitations, which are critically examined. Finally, this review identifies the major challenges and outlines future research directions to advance the understanding and application of RBPs in cardiovascular medicine.
Collapse
Affiliation(s)
- Lizhu Lin
- Department of Anaesthesiology, The First People’s Hospital of Qinzhou, The Tenth Affiliated Hospital of Guangxi Medical University, Qinzhou 535000, China;
| | - Jiemei Chu
- Life Sciences Institute, Guangxi Medical University, Nanning 530021, China; (J.C.); (S.A.)
| | - Sanqi An
- Life Sciences Institute, Guangxi Medical University, Nanning 530021, China; (J.C.); (S.A.)
| | - Xinli Liu
- Life Sciences Institute, Guangxi Medical University, Nanning 530021, China; (J.C.); (S.A.)
| | - Runxian Tan
- Department of Laboratory Medicine, The First People’s Hospital of Qinzhou, The Tenth Affiliated Hospital of Guangxi Medical University, Qinzhou 535000, China
| |
Collapse
|
12
|
Wen J, Guan Y, Niu H, Dang Y, Guan J. Targeting cardiac resident CCR2+ macrophage-secreted MCP-1 to attenuate inflammation after myocardial infarction. Acta Biomater 2024:S1742-7061(24)00469-0. [PMID: 39182804 PMCID: PMC11846964 DOI: 10.1016/j.actbio.2024.08.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 07/26/2024] [Accepted: 08/16/2024] [Indexed: 08/27/2024]
Abstract
After myocardial infarction (MI), cardiac resident CCR2+ macrophages release various cytokines and chemokines, notably monocyte chemoattractant protein-1 (MCP-1). MCP-1 is instrumental in recruiting CCR2+ monocytes to the damaged region. The excessive arrival of these monocytes, which then become macrophages, perpetuates inflammation at the site of injury. This continuous inflammation leads to adverse tissue remodeling and compromises cardiac function over time. We hypothesized that neutralizing the MCP-1 secreted by cardiac resident CCR2+ macrophages can mitigate post-MI inflammation by curtailing the recruitment of monocytes and their differentiation into macrophages. In this work, we developed nanoparticles that target the infarcted heart, specifically accumulating in the damaged area after intravenous (IV) administration, and docking onto CCR2+ macrophages. These nanoparticles were designed to slowly release an MCP-1 binding peptide, HSWRHFHTLGGG (HSW), which neutralizes the upregulated MCP-1. We showed that the HSW reduced monocyte migration, inhibited pro-inflammatory cytokine upregulation, and suppressed myofibroblast differentiation in vitro. After IV delivery, the released HSW significantly decreased monocyte recruitment and pro-inflammatory macrophage density, increased cardiac cell survival, attenuated cardiac fibrosis, and improved cardiac function. Taken together, our findings support the strategy of MCP-1 neutralization at the acute phase of MI as a promising way to alleviate post-MI inflammation. STATEMENT OF SIGNIFICANCE: After a myocardial infarction (MI), CCR2+ macrophages resident in the heart release various cytokines and chemokines, notably monocyte chemoattractant protein-1 (MCP-1). MCP-1 is instrumental in attracting CCR2+ monocytes to the damaged region. The excessive arrival of these monocytes, which then become macrophages, perpetuates inflammation at the site of injury. This continuous inflammation leads to adverse tissue remodeling and compromises cardiac function over time. In this work, we tested the hypothesis that neutralizing the MCP-1 secreted by cardiac CCR2+ macrophages can mitigate post-MI inflammation by curtailing the recruitment of monocytes.
Collapse
Affiliation(s)
- Jiaxing Wen
- Institute of Materials Science and Engineering, Washington University in St. Louis. St. Louis, MO 63130, USA
| | - Ya Guan
- Institute of Materials Science and Engineering, Washington University in St. Louis. St. Louis, MO 63130, USA
| | - Hong Niu
- Department of Mechanical Engineering and Materials Science, Washington University in St. Louis. St. Louis, MO 63130, USA
| | - Yu Dang
- Institute of Materials Science and Engineering, Washington University in St. Louis. St. Louis, MO 63130, USA
| | - Jianjun Guan
- Institute of Materials Science and Engineering, Washington University in St. Louis. St. Louis, MO 63130, USA; Department of Mechanical Engineering and Materials Science, Washington University in St. Louis. St. Louis, MO 63130, USA; Department of Biomedical Engineering, Washington University in St. Louis. St. Louis, MO 63130, USA.
| |
Collapse
|
13
|
Azul L, Leandro A, Seiça R, Sena CM. Propagermanium as a Novel Therapeutic Approach for the Treatment of Endothelial Dysfunction in Type 2 Diabetes. Int J Mol Sci 2024; 25:8328. [PMID: 39125901 PMCID: PMC11312737 DOI: 10.3390/ijms25158328] [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: 06/03/2024] [Revised: 07/18/2024] [Accepted: 07/19/2024] [Indexed: 08/12/2024] Open
Abstract
Propagermanium (PG) has immune modulating activity and anti-inflammatory properties. This work aimed to study the therapeutic efficacy of PG on endothelial and perivascular dysfunction associated with type 2 diabetes. Non-obese type 2 diabetic Goto-Kakizaki (GK) rats were divided into four groups: (1) the control group; (2) the group treated with 50 mg/kg PG; (3) the group fed a high-fat diet (GKHFD); and (4) the group of GKHFD treated with 50 mg/kg PG. PG was given orally for 3 months. Several in vivo parameters and endothelial function were studied in aortas with perivascular adipose tissue PVAT (+) or without PVAT (-). We also determined the vascular inflammation and levels of CD36 in PVAT. In diabetic GK rats, PG did not affect the lipid profile or the results of the intraperitoneal glucose tolerance test. Instead, it improved the fasting glucose levels (18%, p < 0.01), insulin resistance (32%, p < 0.05), endothelial function (33 and 25% in aortas mounted with (+) or without PVAT (-), p < 0.05), and restored the anticontractile effect of the perivascular adipose tissue by reducing its inflammation (56%, p < 0.05) and oxidative stress profile (55%, p < 0.05). Due to its anti-inflammatory characteristics, PG likely improved endothelial dysfunction and restored the perivascular adipose tissue's anticontractile properties.
Collapse
Affiliation(s)
| | | | | | - Cristina M. Sena
- Institute of Physiology, iCBR, Faculty of Medicine, University of Coimbra, Subunit 1, Polo 3, Azinhaga de Santa Comba, Celas, 3000-548 Coimbra, Portugal
| |
Collapse
|
14
|
Kanuri B, Sreejit G, Biswas P, Murphy AJ, Nagareddy PR. Macrophage heterogeneity in myocardial infarction: Evolution and implications for diverse therapeutic approaches. iScience 2024; 27:110274. [PMID: 39040061 PMCID: PMC11261154 DOI: 10.1016/j.isci.2024.110274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/24/2024] Open
Abstract
Given the extensive participation of myeloid cells (especially monocytes and macrophages) in both inflammation and resolution phases post-myocardial infarction (MI) owing to their biphasic role, these cells are considered as crucial players in the disease pathogenesis. Multiple studies have agreed on the significant contribution of macrophage polarization theory (M2 vs. M1) while determining the underlying reasons behind the observed biphasic effects; nevertheless, this simplistic classification attracts severe drawbacks. The advent of multiple advanced technologies based on OMICS platforms facilitated a successful path to explore comprehensive cellular signatures that could expedite our understanding of macrophage heterogeneity and plasticity. While providing an overall basis behind the MI disease pathogenesis, this review delves into the literature to discuss the current knowledge on multiple macrophage clusters, including the future directions in this research arena. In the end, our focus will be on outlining the possible therapeutic implications based on the emerging observations.
Collapse
Affiliation(s)
- Babunageswararao Kanuri
- Department of Internal Medicine, Section of Cardiovascular Diseases, University of Oklahoma Health Sciences Center (OUHSC), Oklahoma City, OK, USA
| | - Gopalkrishna Sreejit
- Department of Pathology, New York University Grossman School of Medicine, New York City, NY, USA
| | - Priosmita Biswas
- Department of Molecular and Cell Biology, University of California Merced, Merced, CA, USA
| | - Andrew J. Murphy
- Baker Heart and Diabetes Institute, Division of Immunometabolism, Melbourne, VIC, Australia
| | - Prabhakara R. Nagareddy
- Department of Internal Medicine, Section of Cardiovascular Diseases, University of Oklahoma Health Sciences Center (OUHSC), Oklahoma City, OK, USA
| |
Collapse
|
15
|
Abdul-Rahman T, Lizano-Jubert I, Bliss ZSB, Garg N, Meale E, Roy P, Crino SA, Deepak BL, Miteu GD, Wireko AA, Qadeer A, Condurat A, Tanasa AD, Pyrpyris N, Sikora K, Horbas V, Sood A, Gupta R, Lavie CJ. RNA in cardiovascular disease: A new frontier of personalized medicine. Prog Cardiovasc Dis 2024; 85:93-102. [PMID: 38253161 DOI: 10.1016/j.pcad.2024.01.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Accepted: 01/14/2024] [Indexed: 01/24/2024]
Abstract
Personalized medicine has witnessed remarkable progress with the emergence of RNA therapy, offering new possibilities for the treatment of various diseases, and in particular in the context of cardiovascular disease (CVD). The ability to target the human genome through RNA manipulation offers great potential not only in the treatment of cardiac pathologies but also in their diagnosis and prevention, notably in cases of hyperlipidemia and myocardial infarctions. While only a few RNA-based treatments have entered clinical trials or obtained approval from the US Food and Drug Administration, the growing body of research on this subject is promising. However, the development of RNA therapies faces several challenges that must be overcome. These include the efficient delivery of drugs into cells, the potential for immunogenic responses, and safety. Resolving these obstacles is crucial to advance the development of RNA therapies. This review explores the newest developments in medical studies, treatment plans, and results related to RNA therapies for heart disease. Furthermore, it discusses the exciting possibilities and difficulties in this innovative area of research.
Collapse
Affiliation(s)
| | | | | | - Neil Garg
- Rowan-Virtua School of osteopathic medicine, Stratford, NJ, USA
| | - Emily Meale
- Rowan-Virtua School of osteopathic medicine, Stratford, NJ, USA
| | - Poulami Roy
- Department of Medicine, North Bengal Medical College and Hospital, Siliguri, India
| | | | | | - Goshen David Miteu
- School of Biosciences, University of Nottingham, Nottingham, England, United Kingdom
| | | | - Abdul Qadeer
- Hospital Internal Medicine Department, Scottsdale Campus, Mayo Clinic, AZ, USA
| | | | | | - Nikolaos Pyrpyris
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration General Hospital, Athens, Greece
| | | | | | - Aayushi Sood
- Department of Medicine, The Wright Center for Graduate Medical Education, Scranton, PA, USA
| | - Rahul Gupta
- Lehigh Valley Heart and Vascular Institute, Lehigh Valley Health Network, Allentown, PA, USA.
| | - Carl J Lavie
- Department of Cardiology, Ochsner Clinic Foundation, New Orleans, LA, United States; The University of Queensland Medical School, Ochsner Clinical School, New Orleans, LA, United States
| |
Collapse
|
16
|
Yang B, Hang S, Xu S, Gao Y, Yu W, Zang G, Zhang L, Wang Z. Macrophage polarisation and inflammatory mechanisms in atherosclerosis: Implications for prevention and treatment. Heliyon 2024; 10:e32073. [PMID: 38873669 PMCID: PMC11170185 DOI: 10.1016/j.heliyon.2024.e32073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 05/11/2024] [Accepted: 05/28/2024] [Indexed: 06/15/2024] Open
Abstract
Atherosclerosis is a chronic inflammatory disease characterised by plaque accumulation in the arteries. Macrophages are immune cells that are crucial in the development of atherosclerosis. Macrophages can adopt different phenotypes, with the M1 phenotype promoting inflammation while the M2 phenotype counteracting it. This review focuses on the factors that drive the polarisation of M1 macrophages towards a pro-inflammatory phenotype during AS. Additionally, we explored metabolic reprogramming mechanisms and cytokines secretion by M1 macrophages. Hyperlipidaemia is widely recognised as a major risk factor for atherosclerosis. Modified lipoproteins released in the presence of hyperlipidaemia can trigger the release of cytokines and recruit circulating monocytes, which adhere to the damaged endothelium and differentiate into macrophages. Macrophages engulf lipids, leading to the formation of foam cells. As atherosclerosis progresses, foam cells become the necrotic core within the atherosclerotic plaques, destabilising them and triggering ischaemic disease. Furthermore, we discuss recent research focusing on targeting macrophages or inflammatory pathways for preventive or therapeutic purposes. These include statins, PCSK9 inhibitors, and promising nanotargeted drugs. These new developments hold the potential for the prevention and treatment of atherosclerosis and its related complications.
Collapse
Affiliation(s)
- Bo Yang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Sanhua Hang
- Department of Hematology, Affiliated Danyang Hospital of Nantong University, Danyang, 212300, China
| | - Siting Xu
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Yun Gao
- Department of Pathology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Wenhua Yu
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Guangyao Zang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Lili Zhang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Zhongqun Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| |
Collapse
|
17
|
Pozzi S, Satchi-Fainaro R. The role of CCL2/CCR2 axis in cancer and inflammation: The next frontier in nanomedicine. Adv Drug Deliv Rev 2024; 209:115318. [PMID: 38643840 DOI: 10.1016/j.addr.2024.115318] [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: 01/14/2024] [Revised: 04/17/2024] [Accepted: 04/18/2024] [Indexed: 04/23/2024]
Abstract
The communication between cells and their microenvironment represents an intrinsic and essential attribute that takes place in several biological processes, including tissue homeostasis and tissue repair. Among these interactions, inflammation is certainly a central biological response that occurs through cytokines and the crosstalk with their respective receptors. In particular, the interaction between CCL2 and its main receptor, CCR2, plays a pivotal role in both harmful and protective inflammatory states, including cancer-mediated inflammation. The activation of the CCL2/CCR2 axis was shown to dictate the migration of macrophages with immune-suppressive phenotype and to aggravate the progression of different cancer types. In addition, this interaction mediates metastasis formation, further limiting the potential therapeutic outcome of anti-cancer drugs. Attempts to inhibit pharmacologically the CCL2/CCR2 axis have yet to show its anti-cancer efficacy as a single agent, but it sheds light on its role as a powerful tool to selectively alleviate pro-tumorigenic and anti-repair inflammation. In this review, we will elucidate the role of CCL2/CCR2 axis in promoting cancer inflammation by activating the host pro-tumorigenic phenotype. Moreover, we will provide some insight into the potential therapeutic benefit of targeting the CCL2/CCR2 axis for cancer and inflammation using novel delivery systems, aiming to sensitize non-responders to currently approved immunotherapies and offer new combinatory approaches.
Collapse
Affiliation(s)
- Sabina Pozzi
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Ronit Satchi-Fainaro
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel; Sagol School of Neurosciences, Tel Aviv University, Tel Aviv 6997801, Israel.
| |
Collapse
|
18
|
Wu Y, Ma Y. CCL2-CCR2 signaling axis in obesity and metabolic diseases. J Cell Physiol 2024; 239:e31192. [PMID: 38284280 DOI: 10.1002/jcp.31192] [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: 10/14/2023] [Revised: 12/10/2023] [Accepted: 12/29/2023] [Indexed: 01/30/2024]
Abstract
Obesity and metabolic diseases, such as insulin resistance, type 2 diabetes, nonalcoholic fatty liver disease, and cardiovascular ailments, represent formidable global health challenges, bearing considerable implications for both morbidity and mortality rates. It has become increasingly evident that chronic, low-grade inflammation plays a pivotal role in the genesis and advancement of these conditions. The involvement of C-C chemokine ligand 2 (CCL2) and its corresponding receptor, C-C chemokine receptor 2 (CCR2), has been extensively documented in numerous inflammatory maladies. Recent evidence indicates that the CCL2/CCR2 pathway extends beyond immune cell recruitment and inflammation, exerting a notable influence on the genesis and progression of metabolic syndrome. The present review seeks to furnish a comprehensive exposition of the CCL2-CCR2 signaling axis within the context of obesity and metabolic disorders, elucidating its molecular mechanisms, functional roles, and therapeutic implications.
Collapse
Affiliation(s)
- Yue Wu
- Shandong Provincial Key Laboratory of Animal Resistance Biology, Center for Cell Structure and Function, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, China
| | - Yanchun Ma
- Shandong Provincial Key Laboratory of Animal Resistance Biology, Center for Cell Structure and Function, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, China
| |
Collapse
|
19
|
Xu S, Xu C, Xu J, Zhang K, Zhang H. Macrophage Heterogeneity and Its Impact on Myocardial Ischemia-Reperfusion Injury: An Integrative Review. J Inflamm Res 2023; 16:5971-5987. [PMID: 38088942 PMCID: PMC10712254 DOI: 10.2147/jir.s436560] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 11/30/2023] [Indexed: 10/21/2024] Open
Abstract
The coronary reperfusion following acute myocardial infarction can paradoxically trigger myocardial ischemia-reperfusion (IR) injury. This complex phenomenon involves the intricate interplay of different subsets of macrophages. These macrophages are crucial players in the post-infarction inflammatory response and subsequent myocardial anti-inflammatory repair. However, their diverse functions can lead to both beneficial and detrimental effects. On one hand, these macrophages play a crucial role in orchestrating the inflammatory response, aiding in the clearance of cellular debris and initiating tissue repair mechanisms. On the other hand, their excessive infiltration and activation can contribute to the perpetuation of the inflammatory cascade, leading to additional myocardial injury and adverse cardiac remodeling. Multiple mechanisms contribute to the IR injury mediated by macrophages, including oxidative stress, apoptosis, and autophagy. These processes further exacerbate the damage to the already vulnerable myocardial tissue. To address this delicate balance, therapeutic strategies aiming to target and modulate macrophage polarization and function are being explored. By fine-tuning the immune inflammatory response, such interventions hold promise in mitigating post-infarction myocardial injury and fostering a more favorable environment for myocardial healing and recovery. Through advancements in this area of research, potential anti-inflammatory interventions may pave the way for improved clinical outcomes and better management of patients after acute myocardial infarction.
Collapse
Affiliation(s)
- Shuwan Xu
- Cardiovascular Department, the Eighth Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, Guangdong, People’s Republic of China
- Department of Cardiology, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Guangzhou, Guangdong, People’s Republic of China
| | - Cong Xu
- Cardiovascular Department, the Eighth Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, Guangdong, People’s Republic of China
| | - Jiahua Xu
- Cardiovascular Department, the Eighth Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, Guangdong, People’s Republic of China
| | - Kun Zhang
- Department of Cardiology, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Guangzhou, Guangdong, People’s Republic of China
| | - Huanji Zhang
- Cardiovascular Department, the Eighth Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, Guangdong, People’s Republic of China
| |
Collapse
|
20
|
Amador-Martínez I, Aparicio-Trejo OE, Bernabe-Yepes B, Aranda-Rivera AK, Cruz-Gregorio A, Sánchez-Lozada LG, Pedraza-Chaverri J, Tapia E. Mitochondrial Impairment: A Link for Inflammatory Responses Activation in the Cardiorenal Syndrome Type 4. Int J Mol Sci 2023; 24:15875. [PMID: 37958859 PMCID: PMC10650149 DOI: 10.3390/ijms242115875] [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: 09/26/2023] [Revised: 10/25/2023] [Accepted: 10/30/2023] [Indexed: 11/15/2023] Open
Abstract
Cardiorenal syndrome type 4 (CRS type 4) occurs when chronic kidney disease (CKD) leads to cardiovascular damage, resulting in high morbidity and mortality rates. Mitochondria, vital organelles responsible for essential cellular functions, can become dysfunctional in CKD. This dysfunction can trigger inflammatory responses in distant organs by releasing Damage-associated molecular patterns (DAMPs). These DAMPs are recognized by immune receptors within cells, including Toll-like receptors (TLR) like TLR2, TLR4, and TLR9, the nucleotide-binding domain, leucine-rich-containing family pyrin domain-containing-3 (NLRP3) inflammasome, and the cyclic guanosine monophosphate (cGMP)-adenosine monophosphate (AMP) synthase (cGAS)-stimulator of interferon genes (cGAS-STING) pathway. Activation of these immune receptors leads to the increased expression of cytokines and chemokines. Excessive chemokine stimulation results in the recruitment of inflammatory cells into tissues, causing chronic damage. Experimental studies have demonstrated that chemokines are upregulated in the heart during CKD, contributing to CRS type 4. Conversely, chemokine inhibitors have been shown to reduce chronic inflammation and prevent cardiorenal impairment. However, the molecular connection between mitochondrial DAMPs and inflammatory pathways responsible for chemokine overactivation in CRS type 4 has not been explored. In this review, we delve into mechanistic insights and discuss how various mitochondrial DAMPs released by the kidney during CKD can activate TLRs, NLRP3, and cGAS-STING immune pathways in the heart. This activation leads to the upregulation of chemokines, ultimately culminating in the establishment of CRS type 4. Furthermore, we propose using chemokine inhibitors as potential strategies for preventing CRS type 4.
Collapse
Affiliation(s)
- Isabel Amador-Martínez
- Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, Mexico City 04510, Mexico; (I.A.-M.); (A.K.A.-R.)
- Departamento de Fisiopatología Cardio-Renal, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico; (O.E.A.-T.); (L.G.S.-L.)
| | - Omar Emiliano Aparicio-Trejo
- Departamento de Fisiopatología Cardio-Renal, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico; (O.E.A.-T.); (L.G.S.-L.)
| | - Bismarck Bernabe-Yepes
- Departamento de Biomedicina Cardiovascular, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico;
| | - Ana Karina Aranda-Rivera
- Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, Mexico City 04510, Mexico; (I.A.-M.); (A.K.A.-R.)
- Laboratorio F-315, Departamento de Biología, Facultad de Química, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico;
| | - Alfredo Cruz-Gregorio
- Departamento de Fisiología, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico;
| | - Laura Gabriela Sánchez-Lozada
- Departamento de Fisiopatología Cardio-Renal, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico; (O.E.A.-T.); (L.G.S.-L.)
| | - José Pedraza-Chaverri
- Laboratorio F-315, Departamento de Biología, Facultad de Química, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico;
| | - Edilia Tapia
- Departamento de Fisiopatología Cardio-Renal, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico; (O.E.A.-T.); (L.G.S.-L.)
| |
Collapse
|
21
|
Francisco J, Del Re DP. Inflammation in Myocardial Ischemia/Reperfusion Injury: Underlying Mechanisms and Therapeutic Potential. Antioxidants (Basel) 2023; 12:1944. [PMID: 38001797 PMCID: PMC10669026 DOI: 10.3390/antiox12111944] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 10/23/2023] [Accepted: 10/27/2023] [Indexed: 11/26/2023] Open
Abstract
Acute myocardial infarction (MI) occurs when blood flow to the myocardium is restricted, leading to cardiac damage and massive loss of viable cardiomyocytes. Timely restoration of coronary flow is considered the gold standard treatment for MI patients and limits infarct size; however, this intervention, known as reperfusion, initiates a complex pathological process that somewhat paradoxically also contributes to cardiac injury. Despite being a sterile environment, ischemia/reperfusion (I/R) injury triggers inflammation, which contributes to infarct expansion and subsequent cardiac remodeling and wound healing. The immune response is comprised of subsets of both myeloid and lymphoid-derived cells that act in concert to modulate the pathogenesis and resolution of I/R injury. Multiple mechanisms, including altered metabolic status, regulate immune cell activation and function in the setting of acute MI, yet our understanding remains incomplete. While numerous studies demonstrated cardiac benefit following strategies that target inflammation in preclinical models, therapeutic attempts to mitigate I/R injury in patients were less successful. Therefore, further investigation leveraging emerging technologies is needed to better characterize this intricate inflammatory response and elucidate its influence on cardiac injury and the progression to heart failure.
Collapse
Affiliation(s)
| | - Dominic P. Del Re
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| |
Collapse
|
22
|
Liu Y, Shao YH, Zhang JM, Wang Y, Zhou M, Li HQ, Zhang CC, Yu PJ, Gao SJ, Wang XR, Jia LX, Piao CM, Du J, Li YL. Macrophage CARD9 mediates cardiac injury following myocardial infarction through regulation of lipocalin 2 expression. Signal Transduct Target Ther 2023; 8:394. [PMID: 37828006 PMCID: PMC10570328 DOI: 10.1038/s41392-023-01635-w] [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: 02/16/2023] [Revised: 08/15/2023] [Accepted: 08/31/2023] [Indexed: 10/14/2023] Open
Abstract
Immune cell infiltration in response to myocyte death regulates extracellular matrix remodeling and scar formation after myocardial infarction (MI). Caspase-recruitment domain family member 9 (CARD9) acts as an adapter that mediates the transduction of pro-inflammatory signaling cascades in innate immunity; however, its role in cardiac injury and repair post-MI remains unclear. We found that Card9 was one of the most upregulated Card genes in the ischemic myocardium of mice. CARD9 expression increased considerably 1 day post-MI and declined by day 7 post-MI. Moreover, CARD9 was mainly expressed in F4/80-positive macrophages. Card9 knockout (KO) led to left ventricular function improvement and infarct scar size reduction in mice 28 days post-MI. Additionally, Card9 KO suppressed cardiomyocyte apoptosis in the border region and attenuated matrix metalloproteinase (MMP) expression. RNA sequencing revealed that Card9 KO significantly suppressed lipocalin 2 (Lcn2) expression post-MI. Both LCN2 and the receptor solute carrier family 22 member 17 (SL22A17) were detected in macrophages. Subsequently, we demonstrated that Card9 overexpression increased LCN2 expression, while Card9 KO inhibited necrotic cell-induced LCN2 upregulation in macrophages, likely through NF-κB. Lcn2 KO showed beneficial effects post-MI, and recombinant LCN2 diminished the protective effects of Card9 KO in vivo. Lcn2 KO reduced MMP9 post-MI, and Lcn2 overexpression increased Mmp9 expression in macrophages. Slc22a17 knockdown in macrophages reduced MMP9 release with recombinant LCN2 treatment. In conclusion, our results demonstrate that macrophage CARD9 mediates the deterioration of cardiac function and adverse remodeling post-MI via LCN2.
Collapse
Affiliation(s)
- Yan Liu
- Beijing Anzhen Hospital, Capital Medical University; The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education; Beijing Collaborative Innovative Research Center for Cardiovascular Diseases; Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, 100029, China
| | - Yi-Hui Shao
- Beijing Anzhen Hospital, Capital Medical University; The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education; Beijing Collaborative Innovative Research Center for Cardiovascular Diseases; Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, 100029, China
| | - Jun-Meng Zhang
- Beijing Anzhen Hospital, Capital Medical University; The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education; Beijing Collaborative Innovative Research Center for Cardiovascular Diseases; Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, 100029, China
| | - Ying Wang
- Beijing Anzhen Hospital, Capital Medical University; The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education; Beijing Collaborative Innovative Research Center for Cardiovascular Diseases; Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, 100029, China
| | - Mei Zhou
- Beijing Anzhen Hospital, Capital Medical University; The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education; Beijing Collaborative Innovative Research Center for Cardiovascular Diseases; Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, 100029, China
| | - Hui-Qin Li
- Beijing Anzhen Hospital, Capital Medical University; The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education; Beijing Collaborative Innovative Research Center for Cardiovascular Diseases; Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, 100029, China
| | - Cong-Cong Zhang
- Beijing Anzhen Hospital, Capital Medical University; The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education; Beijing Collaborative Innovative Research Center for Cardiovascular Diseases; Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, 100029, China
| | - Pei-Jie Yu
- Beijing Anzhen Hospital, Capital Medical University; The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education; Beijing Collaborative Innovative Research Center for Cardiovascular Diseases; Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, 100029, China
| | - Shi-Juan Gao
- Beijing Anzhen Hospital, Capital Medical University; The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education; Beijing Collaborative Innovative Research Center for Cardiovascular Diseases; Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, 100029, China
| | - Xue-Rui Wang
- Beijing Anzhen Hospital, Capital Medical University; The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education; Beijing Collaborative Innovative Research Center for Cardiovascular Diseases; Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, 100029, China
| | - Li-Xin Jia
- Beijing Anzhen Hospital, Capital Medical University; The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education; Beijing Collaborative Innovative Research Center for Cardiovascular Diseases; Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, 100029, China
| | - Chun-Mei Piao
- Beijing Anzhen Hospital, Capital Medical University; The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education; Beijing Collaborative Innovative Research Center for Cardiovascular Diseases; Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, 100029, China
| | - Jie Du
- Beijing Anzhen Hospital, Capital Medical University; The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education; Beijing Collaborative Innovative Research Center for Cardiovascular Diseases; Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, 100029, China
| | - Yu-Lin Li
- Beijing Anzhen Hospital, Capital Medical University; The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education; Beijing Collaborative Innovative Research Center for Cardiovascular Diseases; Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, 100029, China.
| |
Collapse
|
23
|
Lavine KJ, Sultan D, Luehmann H, Detering L, Zhang X, Heo GS, Zhang X, Hoelscher M, Harrison K, Combadière C, Laforest R, Kreisel D, Woodard PK, Brody SL, Gropler RJ, Liu Y. CCR2 Imaging in Human ST-Segment Elevation Myocardial Infarction. NATURE CARDIOVASCULAR RESEARCH 2023; 2:874-880. [PMID: 38947883 PMCID: PMC11210834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Among the diverse populations of myeloid cells that reside within the healthy and diseased heart, C-C chemokine receptor 2 (CCR2) is specifically expressed on inflammatory populations of monocytes and macrophages that contribute to the development and progression of heart failure1-4. Here, we evaluated a peptide-based imaging probe (64Cu-DOTA-ECL1i) that specifically recognizes CCR2+ monocytes and macrophages for human cardiac imaging. Compared to healthy controls, 64Cu-DOTA-ECL1i heart uptake was increased in subjects following acute myocardial infarction, predominately localized within the infarct area, and was associated with impaired myocardial wall motion. These findings establish the feasibility of molecular imaging of CCR2 expression to visualize inflammatory monocytes and macrophages in the injured human heart.
Collapse
Affiliation(s)
- Kory J Lavine
- Department of Medicine, Washington University School of Medicine, St. Louis, MO USA
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO USA
- Department of Immunology and Pathology, Washington University School of Medicine, St. Louis, MO USA
| | - Deborah Sultan
- Department of Radiology, Washington University School of Medicine, St. Louis, MO USA
| | - Hannah Luehmann
- Department of Radiology, Washington University School of Medicine, St. Louis, MO USA
| | - Lisa Detering
- Department of Radiology, Washington University School of Medicine, St. Louis, MO USA
| | - Xiaohui Zhang
- Department of Radiology, Washington University School of Medicine, St. Louis, MO USA
| | - Gyu Seong Heo
- Department of Radiology, Washington University School of Medicine, St. Louis, MO USA
| | - Xiuli Zhang
- Department of Radiology, Washington University School of Medicine, St. Louis, MO USA
| | - Michelle Hoelscher
- Department of Radiology, Washington University School of Medicine, St. Louis, MO USA
| | - Kitty Harrison
- Department of Radiology, Washington University School of Medicine, St. Louis, MO USA
| | - Christophe Combadière
- Sorbonne Université, INSERM, CNRS, Centre d’Immunologie et des Maladies Infectieuses, Cimi-Paris, F-75013 Paris, France
| | - Richard Laforest
- Department of Radiology, Washington University School of Medicine, St. Louis, MO USA
| | - Daniel Kreisel
- Department of Immunology and Pathology, Washington University School of Medicine, St. Louis, MO USA
- Department of Surgery, Washington University School of Medicine, St. Louis, MO USA
| | - Pamela K. Woodard
- Department of Radiology, Washington University School of Medicine, St. Louis, MO USA
| | - Steven L. Brody
- Department of Medicine, Washington University School of Medicine, St. Louis, MO USA
- Department of Radiology, Washington University School of Medicine, St. Louis, MO USA
| | - Robert J Gropler
- Department of Medicine, Washington University School of Medicine, St. Louis, MO USA
- Department of Radiology, Washington University School of Medicine, St. Louis, MO USA
| | - Yongjian Liu
- Department of Radiology, Washington University School of Medicine, St. Louis, MO USA
| |
Collapse
|
24
|
Francisco J, Guan J, Zhang Y, Nakada Y, Mareedu S, Sung EA, Hu CM, Oka S, Zhai P, Sadoshima J, Del Re DP. Suppression of myeloid YAP antagonizes adverse cardiac remodeling during pressure overload stress. J Mol Cell Cardiol 2023; 181:1-14. [PMID: 37235928 PMCID: PMC10524516 DOI: 10.1016/j.yjmcc.2023.05.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 05/05/2023] [Accepted: 05/16/2023] [Indexed: 05/28/2023]
Abstract
Inflammation is an integral component of cardiovascular disease and is thought to contribute to cardiac dysfunction and heart failure. While ischemia-induced inflammation has been extensively studied in the heart, relatively less is known regarding cardiac inflammation during non-ischemic stress. Recent work has implicated a role for Yes-associated protein (YAP) in modulating inflammation in response to ischemic injury; however, whether YAP influences inflammation in the heart during non-ischemic stress is not described. We hypothesized that YAP mediates a pro-inflammatory response during pressure overload (PO)-induced non-ischemic injury, and that targeted YAP inhibition in the myeloid compartment is cardioprotective. In mice, PO elicited myeloid YAP activation, and myeloid-specific YAP knockout mice (YAPF/F;LysMCre) subjected to PO stress had better systolic function, and attenuated pathological remodeling compared to control mice. Inflammatory indicators were also significantly attenuated, while pro-resolving genes including Vegfa were enhanced, in the myocardium, and in isolated macrophages, of myeloid YAP KO mice after PO. Experiments using bone marrow-derived macrophages (BMDMs) from YAP KO and control mice demonstrated that YAP suppression shifted polarization toward a resolving phenotype. We also observed attenuated NLRP3 inflammasome priming and function in YAP deficient BMDMs, as well as in myeloid YAP KO hearts following PO, indicating disruption of inflammasome induction. Finally, we leveraged nanoparticle-mediated delivery of the YAP inhibitor verteporfin and observed attenuated PO-induced pathological remodeling compared to DMSO nanoparticle control treatment. These data implicate myeloid YAP as an important molecular nodal point that facilitates cardiac inflammation and fibrosis during PO stress and suggest that selective inhibition of YAP may prove a novel therapeutic target in non-ischemic heart disease.
Collapse
Affiliation(s)
- Jamie Francisco
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Jin Guan
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Yu Zhang
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Yasuki Nakada
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Satvik Mareedu
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Eun-Ah Sung
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Che-Ming Hu
- Institute of Biomedical Sciences, Academia Sinica, Taiwan
| | - Shinichi Oka
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Peiyong Zhai
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Junichi Sadoshima
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Dominic P Del Re
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, NJ, USA.
| |
Collapse
|
25
|
Chen C, Wang J, Liu C, Hu J. Cardiac resident macrophages: key regulatory mediators in the aftermath of myocardial infarction. Front Immunol 2023; 14:1207100. [PMID: 37457720 PMCID: PMC10348646 DOI: 10.3389/fimmu.2023.1207100] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 06/13/2023] [Indexed: 07/18/2023] Open
Abstract
Acute myocardial infarction (MI) is a prevalent and highly fatal global disease. Despite significant reduction in mortality rates with standard treatment regimens, the risk of heart failure (HF) remains high, necessitating innovative approaches to protect cardiac function and prevent HF progression. Cardiac resident macrophages (cMacs) have emerged as key regulators of the pathophysiology following MI. cMacs are a heterogeneous population composed of subsets with different lineage origins and gene expression profiles. Several critical aspects of post-MI pathophysiology have been shown to be regulated by cMacs, including recruitment of peripheral immune cells, clearance and replacement of damaged myocardial cells. Furthermore, cMacs play a crucial role in regulating cardiac fibrosis, risk of arrhythmia, energy metabolism, as well as vascular and lymphatic remodeling. Given the multifaceted roles of cMacs in post-MI pathophysiology, targeting cMacs represents a promising therapeutic strategy. Finally, we discuss novel treatment strategies, including using nanocarriers to deliver drugs to cMacs or using cell therapies to introduce exogenous protective cMacs into the heart.
Collapse
|
26
|
Sachse M, Tual-Chalot S, Ciliberti G, Amponsah-Offeh M, Stamatelopoulos K, Gatsiou A, Stellos K. RNA-binding proteins in vascular inflammation and atherosclerosis. Atherosclerosis 2023; 374:55-73. [PMID: 36759270 DOI: 10.1016/j.atherosclerosis.2023.01.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 12/01/2022] [Accepted: 01/12/2023] [Indexed: 01/19/2023]
Abstract
Atherosclerotic cardiovascular disease (ASCVD) remains the major cause of premature death and disability worldwide, even when patients with an established manifestation of atherosclerotic heart disease are optimally treated according to the clinical guidelines. Apart from the epigenetic control of transcription of the genetic information to messenger RNAs (mRNAs), gene expression is tightly controlled at the post-transcriptional level before the initiation of translation. Although mRNAs are traditionally perceived as the messenger molecules that bring genetic information from the nuclear DNA to the cytoplasmic ribosomes for protein synthesis, emerging evidence suggests that processes controlling RNA metabolism, driven by RNA-binding proteins (RBPs), affect cellular function in health and disease. Over the recent years, vascular endothelial cell, smooth muscle cell and immune cell RBPs have emerged as key co- or post-transcriptional regulators of several genes related to vascular inflammation and atherosclerosis. In this review, we provide an overview of cell-specific function of RNA-binding proteins involved in all stages of ASCVD and how this knowledge may be used for the development of novel precision medicine therapeutics.
Collapse
Affiliation(s)
- Marco Sachse
- Department of Cardiovascular Research, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Department of Cardiovascular Surgery, University Heart Center, University Hospital Hamburg Eppendorf, Hamburg, Germany
| | - Simon Tual-Chalot
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK.
| | - Giorgia Ciliberti
- Department of Cardiovascular Research, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; German Centre for Cardiovascular Research (Deutsches Zentrum für Herz-Kreislauf-Forschung, DZHK), Heidelberg/Mannheim Partner Site, Mannheim, Germany
| | - Michael Amponsah-Offeh
- Department of Cardiovascular Research, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; German Centre for Cardiovascular Research (Deutsches Zentrum für Herz-Kreislauf-Forschung, DZHK), Heidelberg/Mannheim Partner Site, Mannheim, Germany
| | - Kimon Stamatelopoulos
- Department of Clinical Therapeutics, Alexandra Hospital, National and Kapodistrian University of Athens School of Medicine, Athens, Greece
| | - Aikaterini Gatsiou
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK
| | - Konstantinos Stellos
- Department of Cardiovascular Research, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK; German Centre for Cardiovascular Research (Deutsches Zentrum für Herz-Kreislauf-Forschung, DZHK), Heidelberg/Mannheim Partner Site, Mannheim, Germany; Department of Cardiology, University Hospital Mannheim, Heidelberg University, Manheim, Germany.
| |
Collapse
|
27
|
Yap J, Irei J, Lozano-Gerona J, Vanapruks S, Bishop T, Boisvert WA. Macrophages in cardiac remodelling after myocardial infarction. Nat Rev Cardiol 2023; 20:373-385. [PMID: 36627513 DOI: 10.1038/s41569-022-00823-5] [Citation(s) in RCA: 127] [Impact Index Per Article: 63.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/25/2022] [Indexed: 01/12/2023]
Abstract
Myocardial infarction (MI), as a result of thrombosis or vascular occlusion, is the most prevalent cause of morbidity and mortality among all cardiovascular diseases. The devastating consequences of MI are compounded by the complexities of cellular functions involved in the initiation and resolution of early-onset inflammation and the longer-term effects related to scar formation. The resultant tissue damage can occur as early as 1 h after MI and activates inflammatory signalling pathways to elicit an immune response. Macrophages are one of the most active cell types during all stages after MI, including the cardioprotective, inflammatory and tissue repair phases. In this Review, we describe the phenotypes of cardiac macrophage involved in MI and their cardioprotective functions. A specific subset of macrophages called resident cardiac macrophages (RCMs) are derived from yolk sac progenitor cells and are maintained as a self-renewing population, although their numbers decrease with age. We explore sophisticated sequencing techniques that demonstrate the cardioprotective properties of this cardiac macrophage phenotype. Furthermore, we discuss the interactions between cardiac macrophages and other important cell types involved in the pathology and resolution of inflammation after MI. We summarize new and promising therapeutic approaches that target macrophage-mediated inflammation and the cardioprotective properties of RCMs after MI. Finally, we discuss future directions for the study of RCMs in MI and cardiovascular health in general.
Collapse
Affiliation(s)
- Jonathan Yap
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, USA
| | - Jason Irei
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, USA
| | - Javier Lozano-Gerona
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, USA
| | - Selena Vanapruks
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, USA
| | - Tianmai Bishop
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, USA
| | - William A Boisvert
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, USA.
| |
Collapse
|
28
|
Smith S, Ascione R. Targeting neuro-immune systems to achieve cardiac tissue repair following myocardial infarction: A review of therapeutic approaches from in-vivo preclinical to clinical studies. Pharmacol Ther 2023; 245:108397. [PMID: 36996910 PMCID: PMC7616359 DOI: 10.1016/j.pharmthera.2023.108397] [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: 11/25/2022] [Revised: 03/12/2023] [Accepted: 03/27/2023] [Indexed: 03/30/2023]
Abstract
Myocardial healing following myocardial infarction (MI) toward either functional tissue repair or excessive scarring/heart failure, may depend on a complex interplay between nervous and immune system responses, myocardial ischemia/reperfusion injury factors, as well as genetic and epidemiological factors. Hence, enhancing cardiac repair post MI may require a more patient-specific approach targeting this complex interplay and not just the heart, bearing in mind that the dysregulation or modulation of just one of these systems or some of their mechanisms may determine the outcome either toward functional repair or toward heart failure. In this review we have elected to focus on existing preclinical and clinical in-vivo studies aimed at testing novel therapeutic approaches targeting the nervous and immune systems to trigger myocardial healing toward functional tissue repair. To this end, we have only selected clinical and preclinical in-vivo studies reporting on novel treatments targeting neuro-immune systems to ultimately treat MI. Next, we have grouped and reported treatments under each neuro-immune system. Finally, for each treatment we have assessed and reported the results of each clinical/preclinical study and then discussed their results collectively. This structured approach has been followed for each treatment discussed. To keep this review focused, we have deliberately omitted to cover other important and related research areas such as myocardial ischemia/reperfusion injury, cell and gene therapies as well as any ex-vivo and in-vitro studies. The review indicates that some of the treatments targeting the neuro-immune/inflammatory systems appear to induce beneficial effects remotely on the healing heart post MI, warranting further validation. These remote effects on the heart also indicates the presence of an overarching synergic response occurring across the nervous and immune systems in response to acute MI, which appear to influence cardiac tissue repair in different ways depending on age and timing of treatment delivery following MI. The cumulative evidence arising from this review allows also to make informed considerations on safe as opposed to detrimental treatments, and within the safe treatments to ascertain those associated with conflicting or supporting preclinical data, and those warranting further validation.
Collapse
Affiliation(s)
- Sarah Smith
- Bristol Heart Institute and Translational Biomedical Research Centre, Faculty of Health Science, University of Bristol, Bristol, UK
| | - Raimondo Ascione
- Bristol Heart Institute and Translational Biomedical Research Centre, Faculty of Health Science, University of Bristol, Bristol, UK.
| |
Collapse
|
29
|
Fan YN, Zhao G, Zhang Y, Ye QN, Sun YQ, Shen S, Liu Y, Xu CF, Wang J. Progress in nanoparticle-based regulation of immune cells. MEDICAL REVIEW (2021) 2023; 3:152-179. [PMID: 37724086 PMCID: PMC10471115 DOI: 10.1515/mr-2022-0047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Accepted: 03/03/2023] [Indexed: 09/20/2023]
Abstract
Immune cells are indispensable defenders of the human body, clearing exogenous pathogens and toxicities or endogenous malignant and aging cells. Immune cell dysfunction can cause an inability to recognize, react, and remove these hazards, resulting in cancers, inflammatory diseases, autoimmune diseases, and infections. Immune cells regulation has shown great promise in treating disease, and immune agonists are usually used to treat cancers and infections caused by immune suppression. In contrast, immunosuppressants are used to treat inflammatory and autoimmune diseases. However, the key to maintaining health is to restore balance to the immune system, as excessive activation or inhibition of immune cells is a common complication of immunotherapy. Nanoparticles are efficient drug delivery systems widely used to deliver small molecule inhibitors, nucleic acid, and proteins. Using nanoparticles for the targeted delivery of drugs to immune cells provides opportunities to regulate immune cell function. In this review, we summarize the current progress of nanoparticle-based strategies for regulating immune function and discuss the prospects of future nanoparticle design to improve immunotherapy.
Collapse
Affiliation(s)
- Ya-Nan Fan
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong Province, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, Guangdong Province, China
| | - Gui Zhao
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong Province, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, Guangdong Province, China
| | - Yue Zhang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong Province, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, Guangdong Province, China
| | - Qian-Ni Ye
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong Province, China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, Guangdong Province, China
| | - Yi-Qun Sun
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong Province, China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, Guangdong Province, China
| | - Song Shen
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong Province, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, Guangdong Province, China
| | - Yang Liu
- Department of Biomedical Engineering, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Cong-Fei Xu
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong Province, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, Guangdong Province, China
| | - Jun Wang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong Province, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, Guangdong Province, China
| |
Collapse
|
30
|
Lugrin J, Parapanov R, Milano G, Cavin S, Debonneville A, Krueger T, Liaudet L. The systemic deletion of interleukin-1α reduces myocardial inflammation and attenuates ventricular remodeling in murine myocardial infarction. Sci Rep 2023; 13:4006. [PMID: 36899010 PMCID: PMC10006084 DOI: 10.1038/s41598-023-30662-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 02/27/2023] [Indexed: 03/12/2023] Open
Abstract
Myocardial inflammation following myocardial infarction (MI) is crucial for proper myocardial healing, yet, dysregulated inflammation may promote adverse ventricular remodeling and heart failure. IL-1 signaling contributes to these processes, as shown by dampened inflammation by inhibition of IL-1β or the IL-1 receptor. In contrast, the potential role of IL-1α in these mechanisms has received much less attention. Previously described as a myocardial-derived alarmin, IL-1α may also act as a systemically released inflammatory cytokine. We therefore investigated the effect of IL-1α deficiency on post-MI inflammation and ventricular remodeling in a murine model of permanent coronary occlusion. In the first week post-MI, global IL-1α deficiency (IL-1α KO mice) led to decreased myocardial expression of IL-6, MCP-1, VCAM-1, hypertrophic and pro-fibrotic genes, and reduced infiltration with inflammatory monocytes. These early changes were associated with an attenuation of delayed left ventricle (LV) remodeling and systolic dysfunction after extensive MI. In contrast to systemic Il1a-KO, conditional cardiomyocyte deletion of Il1a (CmIl1a-KO) did not reduce delayed LV remodeling and systolic dysfunction. In conclusion, systemic Il1a-KO, but not Cml1a-KO, protects against adverse cardiac remodeling after MI due to permanent coronary occlusion. Hence, anti-IL-1α therapies could be useful to attenuate the detrimental consequences of post-MI myocardial inflammation.
Collapse
Affiliation(s)
- J Lugrin
- Service of Adult Intensive Care Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland.
- Service of Thoracic Surgery, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland.
- Laboratoire de Chirurgie Thoracique, Centre des Laboratoires d'Epalinges, Chemin des Boveresses 155, 1066, Epalinges, Switzerland.
| | - R Parapanov
- Service of Adult Intensive Care Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Service of Thoracic Surgery, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - G Milano
- Department Coeur-Vaisseaux, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - S Cavin
- Service of Thoracic Surgery, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - A Debonneville
- Service of Thoracic Surgery, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - T Krueger
- Service of Thoracic Surgery, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - L Liaudet
- Service of Adult Intensive Care Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
31
|
Germano DB, Oliveira SB, Bachi ALL, Juliano Y, Novo NF, Bussador do Amaral J, França CN. Monocyte chemokine receptors as therapeutic targets in cardiovascular diseases. Immunol Lett 2023; 256-257:1-8. [PMID: 36893859 DOI: 10.1016/j.imlet.2023.03.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 02/27/2023] [Accepted: 03/03/2023] [Indexed: 03/09/2023]
Abstract
Chemokine receptors are fundamental in many processes related to cardiovascular diseases, such as monocyte migration to vessel walls, cell adhesion, and angiogenesis, among others. Even though many experimental studies have shown the utility of blocking these receptors or their ligands in the treatment of atherosclerosis, the findings in clinical research are still poor. Thus, in the current review we aimed to describe some promising results concerning the blockade of chemokine receptors as therapeutic targets in the treatment of cardiovascular diseases and also to discuss some challenges that need to be overcome before using these strategies in clinical practice.
Collapse
Affiliation(s)
| | | | | | - Yára Juliano
- Post Graduation Program in Health Sciences, Santo Amaro University, Sao Paulo, Brazil
| | - Neil Ferreira Novo
- Post Graduation Program in Health Sciences, Santo Amaro University, Sao Paulo, Brazil
| | - Jônatas Bussador do Amaral
- ENT Research Laboratory, Otorhinolaryngology -Head and Neck Surgery Department, Federal University of Sao Paulo, Sao Paulo, Brazil
| | - Carolina Nunes França
- Post Graduation Program in Health Sciences, Santo Amaro University, Sao Paulo, Brazil.
| |
Collapse
|
32
|
Jan N, Madni A, Khan S, Shah H, Akram F, Khan A, Ertas D, Bostanudin MF, Contag CH, Ashammakhi N, Ertas YN. Biomimetic cell membrane-coated poly(lactic- co-glycolic acid) nanoparticles for biomedical applications. Bioeng Transl Med 2023; 8:e10441. [PMID: 36925703 PMCID: PMC10013795 DOI: 10.1002/btm2.10441] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 10/10/2022] [Accepted: 10/20/2022] [Indexed: 12/27/2022] Open
Abstract
Poly(lactic-co-glycolic acid) (PLGA) nanoparticles (NPs) are commonly used for drug delivery because of their favored biocompatibility and suitability for sustained and controlled drug release. To prolong NP circulation time, enable target-specific drug delivery and overcome physiological barriers, NPs camouflaged in cell membranes have been developed and evaluated to improve drug delivery. Here, we discuss recent advances in cell membrane-coated PLGA NPs, their preparation methods, and their application to cancer therapy, management of inflammation, treatment of cardiovascular disease and control of infection. We address the current challenges and highlight future research directions needed for effective use of cell membrane-camouflaged NPs.
Collapse
Affiliation(s)
- Nasrullah Jan
- Akson College of PharmacyMirpur University of Science and Technology (MUST)MirpurPakistan
- Department of Pharmaceutics, Faculty of PharmacyThe Islamia University of BahawalpurBahawalpurPakistan
| | - Asadullah Madni
- Department of Pharmaceutics, Faculty of PharmacyThe Islamia University of BahawalpurBahawalpurPakistan
| | - Safiullah Khan
- Department of Pharmaceutics, Faculty of PharmacyThe Islamia University of BahawalpurBahawalpurPakistan
| | - Hassan Shah
- Department of Pharmaceutics, Faculty of PharmacyThe Islamia University of BahawalpurBahawalpurPakistan
| | - Faizan Akram
- Department of Pharmaceutics, Faculty of PharmacyThe Islamia University of BahawalpurBahawalpurPakistan
| | - Arshad Khan
- Department of Pharmaceutics, Faculty of PharmacyThe Islamia University of BahawalpurBahawalpurPakistan
| | - Derya Ertas
- Department of Biomedical EngineeringErciyes UniversityKayseriTurkey
| | - Mohammad F. Bostanudin
- College of PharmacyAl Ain UniversityAbu DhabiUnited Arab Emirates
- AAU Health and Biomedical Research CenterAl Ain UniversityAbu DhabiUnited Arab Emirates
| | - Christopher H. Contag
- Department of Microbiology and Molecular GeneticsMichigan State UniversityEast LansingMichiganUSA
- Institute for Quantitative Health Science and Engineering (IQ) and Department of Biomedical Engineering (BME)Michigan State UniversityEast LansingMichiganUSA
| | - Nureddin Ashammakhi
- Institute for Quantitative Health Science and Engineering (IQ) and Department of Biomedical Engineering (BME)Michigan State UniversityEast LansingMichiganUSA
- Department of BioengineeringUniversity of California, Los AngelesLos AngelesCaliforniaUSA
| | - Yavuz Nuri Ertas
- Department of Biomedical EngineeringErciyes UniversityKayseriTurkey
- ERNAM–Nanotechnology Research and Application CenterErciyes UniversityKayseriTurkey
- UNAM–National Nanotechnology Research CenterBilkent UniversityAnkaraTurkey
| |
Collapse
|
33
|
Li T, Yan Z, Fan Y, Fan X, Li A, Qi Z, Zhang J. Cardiac repair after myocardial infarction: A two-sided role of inflammation-mediated. Front Cardiovasc Med 2023; 9:1077290. [PMID: 36698953 PMCID: PMC9868426 DOI: 10.3389/fcvm.2022.1077290] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Accepted: 12/19/2022] [Indexed: 01/11/2023] Open
Abstract
Myocardial infarction is the leading cause of death and disability worldwide, and the development of new treatments can help reduce the size of myocardial infarction and prevent adverse cardiovascular events. Cardiac repair after myocardial infarction can effectively remove necrotic tissue, induce neovascularization, and ultimately replace granulation tissue. Cardiac inflammation is the primary determinant of whether beneficial cardiac repair occurs after myocardial infarction. Immune cells mediate inflammatory responses and play a dual role in injury and protection during cardiac repair. After myocardial infarction, genetic ablation or blocking of anti-inflammatory pathways is often harmful. However, enhancing endogenous anti-inflammatory pathways or blocking endogenous pro-inflammatory pathways may improve cardiac repair after myocardial infarction. A deficiency of neutrophils or monocytes does not improve overall cardiac function after myocardial infarction but worsens it and aggravates cardiac fibrosis. Several factors are critical in regulating inflammatory genes and immune cells' phenotypes, including DNA methylation, histone modifications, and non-coding RNAs. Therefore, strict control and timely suppression of the inflammatory response, finding a balance between inflammatory cells, preventing excessive tissue degradation, and avoiding infarct expansion can effectively reduce the occurrence of adverse cardiovascular events after myocardial infarction. This article reviews the involvement of neutrophils, monocytes, macrophages, and regulatory T cells in cardiac repair after myocardial infarction. After myocardial infarction, neutrophils are the first to be recruited to the damaged site to engulf necrotic cell debris and secrete chemokines that enhance monocyte recruitment. Monocytes then infiltrate the infarct site and differentiate into macrophages and they release proteases and cytokines that are harmful to surviving myocardial cells in the pre-infarct period. As time progresses, apoptotic neutrophils are cleared, the recruitment of anti-inflammatory monocyte subsets, the polarization of macrophages toward the repair phenotype, and infiltration of regulatory T cells, which secrete anti-inflammatory factors that stimulate angiogenesis and granulation tissue formation for cardiac repair. We also explored how epigenetic modifications regulate the phenotype of inflammatory genes and immune cells to promote cardiac repair after myocardial infarction. This paper also elucidates the roles of alarmin S100A8/A9, secreted frizzled-related protein 1, and podoplanin in the inflammatory response and cardiac repair after myocardial infarction.
Collapse
Affiliation(s)
- Tingting Li
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Zhipeng Yan
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yajie Fan
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xinbiao Fan
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Aolin Li
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zhongwen Qi
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China,*Correspondence: Zhongwen Qi,
| | - Junping Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China,Junping Zhang,
| |
Collapse
|
34
|
Isidoro CA, Deniset JF. The role of macrophage subsets in and around the heart in modulating cardiac homeostasis and pathophysiology. Front Immunol 2023; 14:1111819. [PMID: 36926341 PMCID: PMC10011174 DOI: 10.3389/fimmu.2023.1111819] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 02/08/2023] [Indexed: 03/08/2023] Open
Abstract
Cardiac and pericardial macrophages contribute to both homeostatic and pathophysiological processes. Recent advances have identified a vast repertoire of these macrophage populations in and around the heart - broadly categorized into a CCR2+/CCR2- dichotomy. While these unique populations can be further distinguished by origin, localization, and other cell surface markers, further exploration into the role of cardiac and pericardial macrophage subpopulations in disease contributes an additional layer of complexity. As such, novel transgenic models and exogenous targeting techniques have been employed to evaluate these macrophages. In this review, we highlight known cardiac and pericardial macrophage populations, their functions, and the experimental tools used to bolster our knowledge of these cells in the cardiac context.
Collapse
Affiliation(s)
- Carmina Albertine Isidoro
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada.,Libin Cardiovascular Institute, Cumming School of Medicine, Calgary, AB, Canada
| | - Justin F Deniset
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada.,Libin Cardiovascular Institute, Cumming School of Medicine, Calgary, AB, Canada.,Department of Cardiac Sciences, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
35
|
Krychtiuk KA, Ahrens I, Drexel H, Halvorsen S, Hassager C, Huber K, Kurpas D, Niessner A, Schiele F, Semb AG, Sionis A, Claeys MJ, Barrabes J, Montero S, Sinnaeve P, Pedretti R, Catapano A. Acute LDL-C reduction post ACS: strike early and strike strong: from evidence to clinical practice. A clinical consensus statement of the Association for Acute CardioVascular Care (ACVC), in collaboration with the European Association of Preventive Cardiology (EAPC) and the European Society of Cardiology Working Group on Cardiovascular Pharmacotherapy. EUROPEAN HEART JOURNAL. ACUTE CARDIOVASCULAR CARE 2022; 11:939-949. [PMID: 36574353 DOI: 10.1093/ehjacc/zuac123] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 10/02/2022] [Indexed: 12/28/2022]
Abstract
After experiencing an acute coronary syndrome (ACS), patients are at a high risk of suffering from recurrent ischaemic cardiovascular events, especially in the very early phase. Low density lipoprotein-cholesterol (LDL-C) is causally involved in atherosclerosis and a clear, monotonic relationship between pharmacologic LDL-C lowering and a reduction in cardiovascular events post-ACS has been shown, a concept termed 'the lower, the better'. Current ESC guidelines suggest an LDL-C guided, step-wise initiation and escalation of lipid-lowering therapy (LLT). Observational studies consistently show low rates of guideline-recommended LLT adaptions and concomitant low rates of LDL-C target goal achievement, leaving patients at residual risk, especially in the vulnerable post-ACS phase. In addition to the well-established 'the lower, the better' approach, a 'strike early and strike strong' approach in the early post-ACS phase with upfront initiation of a combined lipid-lowering approach using high-intensity statins and ezetimibe seems reasonable. We discuss the rationale, clinical trial evidence and experience for such an approach and highlight existing knowledge gaps. In addition, the concept of acute initiation of PCSK9 inhibition in the early phase is reviewed. Ultimately, we focus on hurdles and solutions to provide high-quality, evidence-based follow-up care in post-ACS patients.
Collapse
Affiliation(s)
- Konstantin A Krychtiuk
- Department of Internal Medicine II-Division of Cardiology, Medical University of Vienna, 1180 Vienna, Austria.,Duke Clinical Research Institute, Durham, NC 27701, USA
| | - Ingo Ahrens
- Department of Cardiology and Medical Intensive Care, Augustinerinnen Hospital Cologne, Academic Teaching Hospital University of Cologne, 50678 Cologne, Germany
| | - Heinz Drexel
- Vorarlberg Institute for Vascular Investigation and Treatment (VIVIT), Carinagasse 47, AT-6800 Feldkirch, Austria.,Private University of the Principality of Liechtenstein, Dorfstrasse 24, FL-9495 Triesen, Liechtenstein.,Department of Medicine I, Academic Teaching Hospital Feldkirch, Carinagasse 47, AT-6800 Feldkirch, Austria
| | - Sigrun Halvorsen
- Institute of Clinical Medicine, University of Oslo, 0372 Oslo, Norway.,Department of Cardiology, Oslo University Hospital Ullevål, Oslo, Norway
| | - Christian Hassager
- Department of Cardiology, Rigshospitalet and Department of Clinical Medicine, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Kurt Huber
- 3rd Department of Internal Medicine, Cardiology and Intensive Care Unit, Wilhelminenhospital, 1160 Vienna, Austria.,Ludwig Boltzmann Institute for Cardiovascular Research, 1090 Vienna, Austria.,Medical School, Sigmund Freud University, 1020 Vienna, Austria
| | - Donata Kurpas
- Family Medicine Department, Wroclaw Medical University, 50-367 Wroclaw, Poland
| | - Alexander Niessner
- Department of Internal Medicine II-Division of Cardiology, Medical University of Vienna, 1180 Vienna, Austria
| | - Francois Schiele
- Department of Cardiology, University Hospital Besancon, University of Franche-Comté, France and EA3920, Besancon, France
| | - Anne Grete Semb
- Preventive Cardio-Rheuma Clinic, Department of Rheumatology, Division of Innovation and Research, Center for Treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway
| | - Alessandro Sionis
- Intensive Cardiac Care Unit, Cardiology Department, Hospital de Sant Pau, IIB-Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain.,CIBER-CV, Centro de investigación Biomédica en Red de Enfermedades Cardiovasculares, 28029 Madrid, Spain
| | - Marc J Claeys
- Department of Cardiology, Antwerp University Hospital, 2650 Edegem, Belgium
| | - José Barrabes
- Acute Cardiac Care Unit, Cardiology Service, Vall d'Hebron Hospital Universitari, Universitat Autònoma de Barcelona, Barcelona, Spain.,CIBERC-V, Centro de investigación Biomédica en Red de Enfermedades Cardiovasculares, Madrid, Spain
| | - Santiago Montero
- Acute Cardiovascular Care Unit, Cardiology, Hospital Germans Trias i Pujol. Departament de Medicina, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Peter Sinnaeve
- Department of Cardiology, University Hospital Leuven, Leuven, Belgium
| | - Roberto Pedretti
- Director of Cardiovascular Department, Head of Cardiology Unit, IRCCS MultiMedica, Milan, Italy
| | - Alberico Catapano
- Professor of Pharmacology, Director Center of Epidemiology and Preventive Pharmacology, Director Laboratory of Lipoproteins, Immunity and Atherosclerosis Department of Pharmacological and Biomolecular Sciences Director Center for the Study of Atherosclerosis at Bassini Hospital University of Milan, Milan, Italy
| |
Collapse
|
36
|
Tu S, He W, Han J, Wu A, Ren W. Advances in imaging and treatment of atherosclerosis based on organic nanoparticles. APL Bioeng 2022; 6:041501. [PMCID: PMC9726224 DOI: 10.1063/5.0127835] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Accepted: 10/31/2022] [Indexed: 12/09/2022] Open
Abstract
Atherosclerosis, a systemic chronic inflammatory disease, can lead to thrombosis and vascular occlusion, thereby inducing a series of serious vascular diseases. Currently, distinguishing unstable plaques early and achieving more effective treatment are the two main clinical concerns in atherosclerosis. Organic nanoparticles have great potential in atherosclerotic imaging and treatment, showing superior biocompatibility, drug-loading capacity, and synthesis. This article illustrates the process of atherosclerosis onset and the key targeted cells, then systematically summarizes recent progress made in organic nanoparticle-based imaging of different types of targeted cells and therapeutic methods for atherosclerosis, including optical and acoustic-induced therapy, drug delivery, gene therapy, and immunotherapy. Finally, we discuss the major impediments that need to be addressed in future clinical practice. We believe this article will help readers to develop a comprehensive and in-depth understanding of organic nanoparticle-based atherosclerotic imaging and treatment, thus advancing further development of anti-atherosclerosis therapies.
Collapse
Affiliation(s)
| | - Wenming He
- Department of Cardiology, The Affiliated Hospital of Medical School, Ningbo University, 247 Renmin Road, Jiangbei District, Ningbo, Zhejiang Province 315020, China,Authors to whom correspondence should be addressed:; ; and
| | | | - Aiguo Wu
- Authors to whom correspondence should be addressed:; ; and
| | - Wenzhi Ren
- Authors to whom correspondence should be addressed:; ; and
| |
Collapse
|
37
|
Abstract
Monocytes/macrophages are key components of the body's innate ability to restore tissue function after injury. In most tissues, both embryo-derived tissue-resident macrophages and recruited blood monocyte-derived macrophages contribute to the injury response. The developmental origin of injury-associated macrophages has a major impact on the outcome of the healing process. Macrophages are abundant at all stages of repair and coordinate the progression through the different phases of healing. They are highly plastic cells that continuously adapt to their environment and acquire phase-specific activation phenotypes. Advanced omics methodologies have revealed a vast heterogeneity of macrophage activation phenotypes and metabolic status at injury sites in different organs. In this review, we highlight the role of the developmental origin, the link between the wound phase-specific activation state and metabolic reprogramming as well as the fate of macrophages during the resolution of the wounding response.
Collapse
Affiliation(s)
| | - Louise Injarabian
- Department of Dermatology, University of Cologne, 50937 Cologne, Germany
| | - Sabine A Eming
- Department of Dermatology, University of Cologne, 50937 Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany
- Institute of Zoology, Developmental Biology Unit, University of Cologne, 50674 Cologne, Germany
| |
Collapse
|
38
|
Kumric M, Urlic H, Bilalic A, Rezic-Muzinic N, Mastelic A, Markotic A, Rusic D, Borovac JA, Duplancic D, Luetic M, Covic I, Ticinovic Kurir T, Bozic J. Dynamic of Circulating DNAM-1+ Monocytes and NK Cells in Patients with STEMI Following Primary Percutaneous Coronary Intervention. J Cardiovasc Dev Dis 2022; 9:jcdd9110395. [PMID: 36421930 PMCID: PMC9693248 DOI: 10.3390/jcdd9110395] [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: 10/01/2022] [Revised: 11/10/2022] [Accepted: 11/14/2022] [Indexed: 11/18/2022] Open
Abstract
Although the role of inflammation and adverse cardiac remodeling in myocardial infarction (MI) have been extensively explored, gaps in knowledge on the complex interaction between these processes still exist. Data suggest that DNAX accessory molecule-1 (DNAM-1), an activating receptor implicated in NK cell education, may be involved in cardiac remodeling following coronary artery occlusion. In the present study, we aimed to explore the dynamic of DNAM-1+ monocytes and NK cells in peripheral blood in the early phase following reperfusion in patients with ST-elevation MI (STEMI). The study enrolled 49 patients older than 18 years of age diagnosed with STEMI, referred to primary percutaneous coronary intervention (pPCI). Blood samples were obtained at three distinct points (at admission, 3 h, and 24 h after pPCI) and analyzed using flow cytometry. The number of circulating DNAM-1+ monocytes (CD16++ and CD14++) and CD56dimCD16++NK cells was significantly reduced 3 h after pPCI and subsequently returned to initial levels 24 h after procedure (p = 0.003, p < 0.001, and p = 0.002, respectively). Notably, such dynamic was dependent on age of patients. A positive correlation between high sensitivity troponin I levels and number of CD16++DNAM-1+ monocytes in peripheral blood 3 h after pPCI was observed (r = 0.431, p = 0.003). In conclusion, in the present study we delineated the post-reperfusion dynamic of DNAM-1-expresing leukocytes. Additionally, we demonstrated that the number of CD16++ DNAM-1+ monocytes correlate with the extent of myocardial injury.
Collapse
Affiliation(s)
- Marko Kumric
- Department of Pathophysiology, University of Split School of Medicine, 21000 Split, Croatia
| | - Hrvoje Urlic
- Department of Pathophysiology, University of Split School of Medicine, 21000 Split, Croatia
| | - Admira Bilalic
- Department of Cardiology, University Hospital of Split, 21000 Split, Croatia
| | - Nikolina Rezic-Muzinic
- Department of Medical Chemistry and Biochemistry, University of Split School of Medicine, 21000 Split, Croatia
| | - Angela Mastelic
- Department of Medical Chemistry and Biochemistry, University of Split School of Medicine, 21000 Split, Croatia
| | - Anita Markotic
- Department of Medical Chemistry and Biochemistry, University of Split School of Medicine, 21000 Split, Croatia
| | - Doris Rusic
- Department of Pharmacy, University of Split School of Medicine, 21000 Split, Croatia
| | - Josip A. Borovac
- Department of Cardiology, University Hospital of Split, 21000 Split, Croatia
- Department of Health Studies, University of Split, 21000 Split, Croatia
| | - Darko Duplancic
- Department of Cardiology, University Hospital of Split, 21000 Split, Croatia
- Department of Internal Medicine, University of Split School of Medicine, 21000 Split, Croatia
| | - Marina Luetic
- Department of Pathophysiology, University of Split School of Medicine, 21000 Split, Croatia
| | - Ivan Covic
- Department of Pathophysiology, University of Split School of Medicine, 21000 Split, Croatia
| | - Tina Ticinovic Kurir
- Department of Pathophysiology, University of Split School of Medicine, 21000 Split, Croatia
- Department of Endocrinology, Diabetes and Metabolic Diseases, University Hospital of Split, 21000 Split, Croatia
| | - Josko Bozic
- Department of Pathophysiology, University of Split School of Medicine, 21000 Split, Croatia
- Correspondence:
| |
Collapse
|
39
|
Xu N, Yutzey KE. Therapeutic CCR2 Blockade Prevents Inflammation and Alleviates Myxomatous Valve Disease in Marfan Syndrome. JACC Basic Transl Sci 2022; 7:1143-1157. [PMID: 36687269 PMCID: PMC9849467 DOI: 10.1016/j.jacbts.2022.06.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 06/08/2022] [Accepted: 06/08/2022] [Indexed: 11/05/2022]
Abstract
Myxomatous valve disease (MVD) can lead to cardiac dysfunction and heart failure, yet medical therapies are lacking. C-C chemokine receptor type 2 (CCR2)+ immune cell infiltration promotes mitral valve inflammation in a Marfan syndrome (MFS) mouse model. The CCR2 genetic knockout reduces inflammation with downregulated proteases and improved extracellular matrix integrity. Pharmacological inhibition of CCR2+ cell infiltration by RS504393 prevents the initiation and progression of MVD, indicated by restored protease expression, improved extracellular matrix organization, and reduced valve leaflet thickness in MFS mice. Thus, the CCR2 antagonist RS504393 is a promising therapy for the treatment of MVD in MFS.
Collapse
Affiliation(s)
- Na Xu
- Division of Molecular Cardiovascular Biology, the Heart Institute, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, USA
| | - Katherine E. Yutzey
- Division of Molecular Cardiovascular Biology, the Heart Institute, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, USA
| |
Collapse
|
40
|
Zhang H, Yang K, Chen F, Liu Q, Ni J, Cao W, Hua Y, He F, Liu Z, Li L, Fan G. Role of the CCL2-CCR2 axis in cardiovascular disease: Pathogenesis and clinical implications. Front Immunol 2022; 13:975367. [PMID: 36110847 PMCID: PMC9470149 DOI: 10.3389/fimmu.2022.975367] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 08/08/2022] [Indexed: 11/16/2022] Open
Abstract
The CCL2-CCR2 axis is one of the major chemokine signaling pathways that has received special attention because of its function in the development and progression of cardiovascular disease. Numerous investigations have been performed over the past decades to explore the function of the CCL2-CCR2 signaling axis in cardiovascular disease. Laboratory data on the CCL2-CCR2 axis for cardiovascular disease have shown satisfactory outcomes, yet its clinical translation remains challenging. In this article, we describe the mechanisms of action of the CCL2-CCR2 axis in the development and evolution of cardiovascular diseases including heart failure, atherosclerosis and coronary atherosclerotic heart disease, hypertension and myocardial disease. Laboratory and clinical data on the use of the CCL2-CCR2 pathway as a targeted therapy for cardiovascular diseases are summarized. The potential of the CCL2-CCR2 axis in the treatment of cardiovascular diseases is explored.
Collapse
Affiliation(s)
- Haixia Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Hebei Key Laboratory of Integrated Traditional Chinese and Western Medicine for Diabetes and Its Complications, College of Traditional Chinese Medicine, North China University of Science and Technology, Tangshan, China
| | - Ke Yang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Feng Chen
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Qianqian Liu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Jingyu Ni
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Weilong Cao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Yunqing Hua
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Feng He
- Hubei Key Laboratory of Economic Forest Germplasm Improvement and Resources Comprehensive Utilization, Huanggang Normal University, Huanggang, China
| | - Zhihao Liu
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lan Li
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- *Correspondence: Lan Li, ; Guanwei Fan,
| | - Guanwei Fan
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Hubei Key Laboratory of Economic Forest Germplasm Improvement and Resources Comprehensive Utilization, Huanggang Normal University, Huanggang, China
- *Correspondence: Lan Li, ; Guanwei Fan,
| |
Collapse
|
41
|
Zhao T, Wang X, Liu Q, Yang T, Qu H, Zhou H. Ginsenoside Rd Promotes Cardiac Repair After Myocardial Infarction by Modulating Monocytes/Macrophages Subsets Conversion. Drug Des Devel Ther 2022; 16:2767-2782. [PMID: 36033133 PMCID: PMC9416535 DOI: 10.2147/dddt.s377624] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 08/12/2022] [Indexed: 11/23/2022] Open
Abstract
Purpose This study aimed to elucidate the potential molecular mechanisms by which GSRd improves cardiac inflammation and immune environment after MI. Materials and Methods The potential target genes of GSRd were predicted using the STITCH database. In vivo, MI mice models were established by left anterior descending ligation and were divided into the sham group, MI + Vehicle group, and MI + GSRd group. DMSO, DMSO, and GSRd 50 μL/day were intraperitoneally injected, respectively. After 28 days, echocardiography, Masson staining, immunofluorescence staining, flow cytometry, RT-PCR, and Western blot were performed. Mice peritoneal macrophages were extracted in vitro, and Western blot was performed after GSRd and/or Akt inhibitor MK2206 intervention. Results GSRd significantly improved mouse myocardial function, attenuated cardiac fibrosis, and inhibited inflammation and apoptosis in myocardial tissues after myocardial infarction. Meanwhile, GSRd increased non-classical Ly6Clow Mos/Mps while reduced of classical Ly6Chigh Mos/Mps at the same time in myocardial tissues. In addition, GSRd significantly reversed the activity of p-Akt and p-mTOR in the heart Mos/Mps after MI. In vitro studies showed that the activity of p-Akt and p-mTOR in peritoneal macrophages were significantly increased in a dose-dependent manner after GSRd treatment. Furthermore, the AKT inhibitor MK2206 was found to block the enhanced activity of p-Akt and p-mTOR induced by GSRd in peritoneal macrophages. Conclusion GSRd can enhance the transformation of Ly6Chigh Mos/Mps to Ly6Clow Mos/Mps in mice after MI by activating the Akt/mTOR signaling pathway, inhibiting cardiac dysfunction and promoting cardiac repair.
Collapse
Affiliation(s)
- Tingyao Zhao
- Department of Cardiovascular Disease, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Xinting Wang
- Institute of Cardiovascular Disease of Integrated Traditional Chinese and Western Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of 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, People’s Republic of China
| | - Tianshu Yang
- Department of Cardiovascular Disease, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Huiyan Qu
- Department of Cardiovascular Disease, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of 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, People’s Republic of China
- Correspondence: Hua Zhou; Huiyan Qu, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, No. 528, Zhangheng Road, Pudong New Area, Shanghai, 201203, People’s Republic of China, Email ;
| |
Collapse
|
42
|
Lv Z, Qiu L, Wang W, Liu Z, Liu Q, Wang L, Song L. RGD-Labeled Hemocytes With High Migration Activity Display a Potential Immunomodulatory Role in the Pacific Oyster Crassostrea gigas. Front Immunol 2022; 13:914899. [PMID: 35865522 PMCID: PMC9294365 DOI: 10.3389/fimmu.2022.914899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 06/03/2022] [Indexed: 11/13/2022] Open
Abstract
Immunocyte migration to infection sites is important for host cellular defense, but the main types of migrating hemocytes and their mechanisms against pathogen invasions are unclear in invertebrates. In the present study, a population of hemocytes in the Pacific oyster Crassostrea gigas labeled with a fluorescein isothiocyanate (FITC)-conjugated Arg-Gly-Asp (RGD)-containing peptide was sorted. RGD+ hemocytes were characterized by a smaller cell size and cytoplasmic-nucleo ratio, fewer cytoplasmic granules, and higher levels of myeloperoxidase, reactive oxygen species, and intracellular free calcium concentration. RGD+ hemocytes exhibited a high level of migration activity, which was further induced after V. splendidus infection. Transcriptome analysis revealed that RGD+ hemocytes highly expressed a series of migration-related genes, which together with migration-promoting genes were significantly upregulated after V. splendidus infection. The neuroendocrine system was also proven to regulate the migration activity of RGD+ hemocytes, especially with the excitatory neuroendocrine factor dopamine, which promoted migration activity as confirmed by receptor blocking assays. Meanwhile, RGD+ hemocytes could highly express immunomodulatory factor interleukin (IL)-17s and their receptor genes, which was positively related to the production of antimicrobial peptides in whole hemocytes after V. splendidus infection. Collectively, this study identified a specific hemocyte population, i.e., RGD+ hemocytes, that shows high migration activity in response to pathogen infection and exerts a potential immunomodulatory role by highly expressing IL-17s that might enhance the hemocytes’ antimicrobial peptide production in oysters.
Collapse
Affiliation(s)
- Zhao Lv
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, CAS Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, China
- Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, China
| | - Limei Qiu
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, CAS Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, China
- Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, China
- *Correspondence: Limei Qiu, ; Linsheng Song,
| | - Weilin Wang
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, China
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, China
| | - Zhaoqun Liu
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, China
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, China
| | - Qing Liu
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, CAS Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, China
- Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, China
| | - Lingling Wang
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, China
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, China
| | - Linsheng Song
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, China
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, China
- *Correspondence: Limei Qiu, ; Linsheng Song,
| |
Collapse
|
43
|
Elwazir MY, Bois JP, Chareonthaitawee P. Utilization of cardiac imaging in sarcoidosis. Expert Rev Cardiovasc Ther 2022; 20:253-266. [DOI: 10.1080/14779072.2022.2069560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Mohamed Y. Elwazir
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
- Department of Cardiology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - John P. Bois
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | | |
Collapse
|
44
|
Witten A, Martens L, Schäfer AC, Troidl C, Pankuweit S, Vlacil AK, Oberoi R, Schieffer B, Grote K, Stoll M, Markus B. Monocyte subpopulation profiling indicates CDK6-derived cell differentiation and identifies subpopulation-specific miRNA expression sets in acute and stable coronary artery disease. Sci Rep 2022; 12:5589. [PMID: 35379829 PMCID: PMC8979987 DOI: 10.1038/s41598-022-08600-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 03/09/2022] [Indexed: 12/20/2022] Open
Abstract
Coronary artery disease (CAD) is a long-lasting inflammatory disease characterized by monocyte migration into the vessel wall leading to clinical events like myocardial infarction (MI). However, the role of monocyte subsets, especially their miRNA-driven differentiation in this scenario is still in its infancy. Here, we characterized monocyte subsets in controls and disease phenotypes of CAD and MI patients using flow cytometry and miRNA and mRNA expression profiling using RNA sequencing. We observed major differences in the miRNA profiles between the classical (CD14++CD16−) and nonclassical (CD14+CD16++) monocyte subsets irrespective of the disease phenotype suggesting the Cyclin-dependent Kinase 6 (CDK6) to be an important player in monocyte maturation. Between control and MI patients, we found a set of miRNAs to be differentially expressed in the nonclassical monocytes and targeting CCND2 (Cyclin D2) that is able to enhance myocardial repair. Interestingly, miRNAs as miR-125b playing a role in vascular calcification were differentially expressed in the classical subset in patients suffering from CAD and not MI in comparison to control samples. In conclusion, our study describes specific peculiarities of monocyte subset miRNA expression in control and diseased samples and provides basis to further functional analysis and to identify new cardiovascular disease treatment targets.
Collapse
|
45
|
Yerra VG, Advani A. Role of CCR2-Positive Macrophages in Pathological Ventricular Remodelling. Biomedicines 2022; 10:661. [PMID: 35327464 PMCID: PMC8945438 DOI: 10.3390/biomedicines10030661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 03/01/2022] [Accepted: 03/08/2022] [Indexed: 12/10/2022] Open
Abstract
Even with recent advances in care, heart failure remains a major cause of morbidity and mortality, which urgently needs new treatments. One of the major antecedents of heart failure is pathological ventricular remodelling, the abnormal change in the size, shape, function or composition of the cardiac ventricles in response to load or injury. Accumulating immune cell subpopulations contribute to the change in cardiac cellular composition that occurs during ventricular remodelling, and these immune cells can facilitate heart failure development. Among cardiac immune cell subpopulations, macrophages that are recognized by their transcriptional or cell-surface expression of the chemokine receptor C-C chemokine receptor type 2 (CCR2), have emerged as playing an especially important role in adverse remodelling. Here, we assimilate the literature that has been generated over the past two decades describing the pathological roles that CCR2+ macrophages play in ventricular remodelling. The goal is to facilitate research and innovation efforts in heart failure therapeutics by drawing attention to the importance of studying the manner by which CCR2+ macrophages mediate their deleterious effects.
Collapse
Affiliation(s)
| | - Andrew Advani
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Toronto, ON M5B 1T8, Canada;
| |
Collapse
|
46
|
Oey O, Sunjaya AP. Applications of nanoparticles in cardiovascular imaging and therapeutics. Asian Cardiovasc Thorac Ann 2022; 30:653-660. [PMID: 35259973 DOI: 10.1177/02184923221087003] [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: 11/16/2022]
Abstract
Cardiovascular disease (CVD) is a major health problem afflicting millions of people worldwide. Early detection methods are lacking, and current therapies have significant limitations. Recently, there has been a surge in the number of studies investigating the utilisation of nanoparticles in cardiovascular imaging and therapy. With respect to cardiovascular imaging, previous studies have looked at the role of nanoparticles in thrombus formation, angiogenesis, blood pool and stem cell imaging. Whereas, with respect to therapy, nanoparticles have been studied for delivering drugs and nucleic acids, specifically to the site of interest; in the context of cardiac regeneration; and its potential in refining current therapy guidelines for CVD management. This review aims to extensively summarise the studies that have been conducted investigating the role of nanoparticles in different aspects of cardiovascular imaging and therapy.
Collapse
Affiliation(s)
- Oliver Oey
- 94920St John of God Midland Hospital, Perth, Australia.,85075Faculty of Medicine, University of Western Australia, Perth, Australia
| | - Anthony Paulo Sunjaya
- 98994Faculty of Medicine, University of New South Wales, Sydney, Australia.,98994The George Institute for Global Health, Sydney, Australia
| |
Collapse
|
47
|
Single-cell transcriptomics reveals cell-type-specific diversification in human heart failure. NATURE CARDIOVASCULAR RESEARCH 2022; 1:263-280. [PMID: 35959412 PMCID: PMC9364913 DOI: 10.1038/s44161-022-00028-6] [Citation(s) in RCA: 207] [Impact Index Per Article: 69.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Heart failure represents a major cause of morbidity and mortality worldwide. Single-cell transcriptomics have revolutionized our understanding of cell composition and associated gene expression. Through integrated analysis of single-cell and single-nucleus RNA-sequencing data generated from 27 healthy donors and 18 individuals with dilated cardiomyopathy, here we define the cell composition of the healthy and failing human heart. We identify cell-specific transcriptional signatures associated with age and heart failure and reveal the emergence of disease-associated cell states. Notably, cardiomyocytes converge toward common disease-associated cell states, whereas fibroblasts and myeloid cells undergo dramatic diversification. Endothelial cells and pericytes display global transcriptional shifts without changes in cell complexity. Collectively, our findings provide a comprehensive analysis of the cellular and transcriptomic landscape of human heart failure, identify cell type-specific transcriptional programs and disease-associated cell states and establish a valuable resource for the investigation of human heart failure.
Collapse
|
48
|
Aldarondo D, Wayne E. Monocytes as a convergent nanoparticle therapeutic target for cardiovascular diseases. Adv Drug Deliv Rev 2022; 182:114116. [PMID: 35085623 PMCID: PMC9359644 DOI: 10.1016/j.addr.2022.114116] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 01/10/2022] [Accepted: 01/13/2022] [Indexed: 12/17/2022]
Abstract
Due to the increasing population of individuals with cardiovascular diseases and related comorbidities, there is an increasing need for development of synergistic therapeutics. Monocytes are implicated in a broad spectrum of diseases and can serve as a focal point for therapeutic targeting. This review discusses the role of monocytes in cardiovascular diseases and highlights trends in monocyte targets nanoparticles in three cardiovascular-related diseases: Diabetes, Atherosclerosis, and HIV. Finally, the review offers perspectives on how to develop nanoparticle monocyte targeting strategies that can be beneficial for treating co-morbidities.
Collapse
Affiliation(s)
- Dasia Aldarondo
- Department of Chemical Engineering and Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Elizabeth Wayne
- Department of Chemical Engineering and Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA.
| |
Collapse
|
49
|
Abstract
Atherosclerosis is a chronic inflammatory disease of the arterial wall, characterized by the formation of plaques containing lipid, connective tissue and immune cells in the intima of large and medium-sized arteries. Over the past three decades, a substantial reduction in cardiovascular mortality has been achieved largely through LDL-cholesterol-lowering regimes and therapies targeting other traditional risk factors for cardiovascular disease, such as hypertension, smoking, diabetes mellitus and obesity. However, the overall benefits of targeting these risk factors have stagnated, and a huge global burden of cardiovascular disease remains. The indispensable role of immunological components in the establishment and chronicity of atherosclerosis has come to the forefront as a clinical target, with proof-of-principle studies demonstrating the benefit and challenges of targeting inflammation and the immune system in cardiovascular disease. In this Review, we provide an overview of the role of the immune system in atherosclerosis by discussing findings from preclinical research and clinical trials. We also identify important challenges that need to be addressed to advance the field and for successful clinical translation, including patient selection, identification of responders and non-responders to immunotherapies, implementation of patient immunophenotyping and potential surrogate end points for vascular inflammation. Finally, we provide strategic guidance for the translation of novel targets of immunotherapy into improvements in patient outcomes. In this Review, the authors provide an overview of the immune cells involved in atherosclerosis, discuss preclinical research and published and ongoing clinical trials assessing the therapeutic potential of targeting the immune system in atherosclerosis, highlight emerging therapeutic targets from preclinical studies and identify challenges for successful clinical translation. Inflammation is an important component of the pathophysiology of cardiovascular disease; an imbalance between pro-inflammatory and anti-inflammatory processes drives chronic inflammation and the formation of atherosclerotic plaques in the vessel wall. Clinical trials assessing canakinumab and colchicine therapies in atherosclerotic cardiovascular disease have provided proof-of-principle of the benefits associated with therapeutic targeting of the immune system in atherosclerosis. The immunosuppressive adverse effects associated with the systemic use of anti-inflammatory drugs can be minimized through targeted delivery of anti-inflammatory drugs to the atherosclerotic plaque, defining the window of opportunity for treatment and identifying more specific targets for cardiovascular inflammation. Implementing immunophenotyping in clinical trials in patients with atherosclerotic cardiovascular disease will allow the identification of immune signatures and the selection of patients with the highest probability of deriving benefit from a specific therapy. Clinical stratification via novel risk factors and discovery of new surrogate markers of vascular inflammation are crucial for identifying new immunotherapeutic targets and their successful translation into the clinic.
Collapse
|
50
|
Shabbir R, Raza A, Liaquat A, Shah SU, Saeed S, Sarwar U, Hamza M, Chudhary F, Hussain Z, Butt NM. Nanoparticles as a novel tool to inhibit inflammatory cytokines in human lymphocytes and macrophages of coronary artery disease. J Pharm Sci 2022; 111:1509-1521. [PMID: 34999090 DOI: 10.1016/j.xphs.2022.01.001] [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: 08/05/2021] [Revised: 12/28/2021] [Accepted: 01/04/2022] [Indexed: 11/29/2022]
Abstract
TNFα and NF-kB contribute in activation of pro-inflammatory signaling pathways and complications of coronary artery diseases (CAD). Current study highlights novel properties of Au (15 ± 2nm), ZnO (77± 45nm) and MgO (11± 4nm) nanoparticles (NPs) as possible anti-inflammatory agents with greater efficacy and lower toxicity. Decrease in TNFα and NF-kB levels in Single Vessel Disease (SVD), Double Vessel Disease (DVD) and Triple-Vessel coronary artery disease (TVD) macrophage and lymphocyte cultures at varying concentrations of NPs has been studied to find an effective therapeutic concentration (ETC). Au and MgO NPs exhibits 5µg/ml ETC compared to 1µg/ml ZnO in all three CAD categories with negligible toxicity. ZnO remains most statistically significant (p<0.001) in SVD and TVD cultures whereas MgO shows efficacy in DVD and TVD cultures with more than 50% reduction in TNFα and NF-kB levels at their respective ETCs. Au NPs exhibit prominent effect in DVD cultures. The mRNA expression results support the down-regulation of TNFα and NF-kB after NPs exposure in respective cultures. Findings of this prospective observational cohort study suggest use of NPs as an alternate anti-inflammatory agent in coronary artery and other diseases.
Collapse
Affiliation(s)
- Rabia Shabbir
- Preston Institute of Nanoscience and Technology (PINSAT), Preston University Kohat, Islamabad campus, Islamabad and Pakistan Academy of Sciences, Islamabad
| | - Abida Raza
- NILOP Nanomedicine Research Laboratories, National Institute of Lasers and Optronics College, PIEAS, Nilore, Islamabad.
| | | | | | - Sidra Saeed
- NILOP Nanomedicine Research Laboratories, National Institute of Lasers and Optronics College, PIEAS, Nilore, Islamabad
| | - Usama Sarwar
- NILOP Nanomedicine Research Laboratories, National Institute of Lasers and Optronics College, PIEAS, Nilore, Islamabad
| | - Muhammad Hamza
- Preston Institute of Nanoscience and Technology (PINSAT), Preston University Kohat, Islamabad campus, Islamabad and Pakistan Academy of Sciences, Islamabad
| | - Fayyaz Chudhary
- Preston Institute of Nanoscience and Technology (PINSAT), Preston University Kohat, Islamabad campus, Islamabad and Pakistan Academy of Sciences, Islamabad
| | | | - N M Butt
- Preston Institute of Nanoscience and Technology (PINSAT), Preston University Kohat, Islamabad campus, Islamabad and Pakistan Academy of Sciences, Islamabad.
| |
Collapse
|