1
|
Lee J, Lee SG, Kim BS, Park S, Sundaram MN, Kim BG, Kim CY, Hwang NS. Paintable Decellularized-ECM Hydrogel for Preventing Cardiac Tissue Damage. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2307353. [PMID: 38502886 DOI: 10.1002/advs.202307353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 02/07/2024] [Indexed: 03/21/2024]
Abstract
The tissue-specific heart decellularized extracellular matrix (hdECM) demonstrates a variety of therapeutic advantages, including fibrosis reduction and angiogenesis. Consequently, recent research for myocardial infarction (MI) therapy has utilized hdECM with various delivery techniques, such as injection or patch implantation. In this study, a novel approach for hdECM delivery using a wet adhesive paintable hydrogel is proposed. The hdECM-containing paintable hydrogel (pdHA_t) is simply applied, with no theoretical limit to the size or shape, making it highly beneficial for scale-up. Additionally, pdHA_t exhibits robust adhesion to the epicardium, with a minimal swelling ratio and sufficient adhesion strength for MI treatment when applied to the rat MI model. Moreover, the adhesiveness of pdHA_t can be easily washed off to prevent undesired adhesion with nearby organs, such as the rib cages and lungs, which can result in stenosis. During the 28 days of in vivo analysis, the pdHA_t not only facilitates functional regeneration by reducing ventricular wall thinning but also promotes neo-vascularization in the MI region. In conclusion, the pdHA_t presents a promising strategy for MI treatment and cardiac tissue regeneration, offering the potential for improved patient outcomes and enhanced cardiac function post-MI.
Collapse
Affiliation(s)
- Jaewoo Lee
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul, 151-742, Republic of Korea
| | - Seul-Gi Lee
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul, 143-701, Republic of Korea
| | - Beom-Seok Kim
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul, 151-742, Republic of Korea
- Research Division, EGC Therapeutics, Seoul, 08790, Republic of Korea
| | - Shinhye Park
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul, 143-701, Republic of Korea
| | - M Nivedhitha Sundaram
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul, 151-742, Republic of Korea
| | - Byung-Gee Kim
- Research Division, EGC Therapeutics, Seoul, 08790, Republic of Korea
- Institute of Molecular Biology and Genetics, Institute for Sustainable Development (ISD), Seoul National University, Seoul, 08826, Republic of Korea
- Bio-MAX/N-Bio, Institute of BioEngineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - C-Yoon Kim
- College of Veterinary Medicine, Konkuk University, Seoul, 05029, Republic of Korea
| | - Nathaniel S Hwang
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul, 151-742, Republic of Korea
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul, 151-742, Republic of Korea
- Bio-MAX/N-Bio, Institute of BioEngineering, Seoul National University, Seoul, 08826, Republic of Korea
| |
Collapse
|
2
|
Medina JP, Bermejo-Álvarez I, Pérez-Baos S, Yáñez R, Fernández-García M, García-Olmo D, Mediero A, Herrero-Beaumont G, Largo R. MSC therapy ameliorates experimental gouty arthritis hinting an early COX-2 induction. Front Immunol 2023; 14:1193179. [PMID: 37533852 PMCID: PMC10391650 DOI: 10.3389/fimmu.2023.1193179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 07/03/2023] [Indexed: 08/04/2023] Open
Abstract
Objective The specific effect of Adipose-Derived Mesenchymal Stem Cells (Ad-MSC) on acute joint inflammation, where the response mostly depends on innate immunity activation, remains elusive. The pathogenesis of gouty arthritis, characterized by the deposition of monosodium urate (MSU) crystals in the joints, associated to acute flares, has been associated to NLRP3 inflammasome activation and subsequent amplification of the inflammatory response. Our aim was to study the effect of human Ad-MSC administration in the clinical inflammatory response of rabbits after MSU injection, and the molecular mechanisms involved. Methods Ad-MSC were administered by intraarterial route shortly after intraarticular MSU crystal injections. Joint and systemic inflammation was sequentially studied, and the mechanisms involved in NLRP3 inflammasome activation, and the synthesis of inflammatory mediators were assessed in the synovial membranes 72h after insult. Ad-MSC and THP-1-derived macrophages stimulated with MSU were co-cultured in transwell system. Results A single systemic dose of Ad-MSC accelerated the resolution of local and systemic inflammatory response. In the synovial membrane, Ad-MSC promoted alternatively M2 macrophage presence, inhibiting NLRP3 inflammasome and inducing the production of anti-inflammatory cytokines, such as IL-10 or TGF-β, and decreasing nuclear factor-κB activity. Ad-MSC induced a net anti-inflammatory balance in MSU-stimulated THP-1 cells, with a higher increase in IL-10 and IDO expression than that observed for IL-1β and TNF. Conclusion Our in vivo and in vitro results showed that a single systemic dose of Ad-MSC decrease the intensity and duration of the inflammatory response by an early local COX-2 upregulation and PGE2 release. Ad-MSCs suppressed NF-kB activity, NLRP3 inflammasome, and promoted the presence of M2 alternative macrophages in the synovium. Therefore, this therapeutic approach could be considered as a pharmacological alternative in patients with comorbidities that preclude conventional treatment.
Collapse
Affiliation(s)
- Juan Pablo Medina
- Bone and Joint Research Unit, Rheumatology Dept, IIS-Fundación Jiménez Díaz Universidad Autonoma de Madrid (UAM), Madrid, Spain
| | - Ismael Bermejo-Álvarez
- Bone and Joint Research Unit, Rheumatology Dept, IIS-Fundación Jiménez Díaz Universidad Autonoma de Madrid (UAM), Madrid, Spain
| | - Sandra Pérez-Baos
- Bone and Joint Research Unit, Rheumatology Dept, IIS-Fundación Jiménez Díaz Universidad Autonoma de Madrid (UAM), Madrid, Spain
| | - Rosa Yáñez
- Hematopoietic Innovative Therapies Division, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT) and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBER-ER), Madrid, Spain
- Advanced Therapies Dept, IIS-Fundación Jiménez Díaz UAM, Madrid, Spain
| | - María Fernández-García
- Hematopoietic Innovative Therapies Division, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT) and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBER-ER), Madrid, Spain
- Advanced Therapies Dept, IIS-Fundación Jiménez Díaz UAM, Madrid, Spain
| | - Damián García-Olmo
- New Therapies Laboratory, IIS-Fundación Jiménez Díaz UAM, Madrid, Spain
- Department of Surgery, Fundación Jiménez Díaz University Hospital, Madrid, Spain
- Department of Surgery, School of Medicine UAM, Madrid, Spain
| | - Aránzazu Mediero
- Bone and Joint Research Unit, Rheumatology Dept, IIS-Fundación Jiménez Díaz Universidad Autonoma de Madrid (UAM), Madrid, Spain
| | - Gabriel Herrero-Beaumont
- Bone and Joint Research Unit, Rheumatology Dept, IIS-Fundación Jiménez Díaz Universidad Autonoma de Madrid (UAM), Madrid, Spain
| | - Raquel Largo
- Bone and Joint Research Unit, Rheumatology Dept, IIS-Fundación Jiménez Díaz Universidad Autonoma de Madrid (UAM), Madrid, Spain
| |
Collapse
|
3
|
Lang CI, Dahmen A, Vasudevan P, Lemcke H, Gäbel R, Öner A, Ince H, David R, Wolfien M. Cardiac cell therapies for the treatment of acute myocardial infarction in mice: systematic review and meta-analysis. Cytotherapy 2023; 25:640-652. [PMID: 36890093 DOI: 10.1016/j.jcyt.2023.01.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 01/22/2023] [Accepted: 01/24/2023] [Indexed: 03/08/2023]
Abstract
Backgound Aims: This meta-analysis aims at summarizing the whole body of research on cell therapies for acute myocardial infarction (MI) in the mouse model to bring forward ongoing research in this field of regenerative medicine. Despite rather modest effects in clinical trials, pre-clinical studies continue to report beneficial effects of cardiac cell therapies for cardiac repair following acute ischemic injury. Results: The authors' meta-analysis of data from 166 mouse studies comprising 257 experimental groups demonstrated a significant improvement in left ventricular ejection fraction of 10.21% after cell therapy compared with control animals. Subgroup analysis indicated that second-generation cell therapies such as cardiac progenitor cells and pluripotent stem cell derivatives had the highest therapeutic potential for minimizing myocardial damage post-MI. Conclusions: Whereas the vision of functional tissue replacement has been replaced by the concept of regional scar modulation in most of the investigated studies, rather basic methods for assessing cardiac function were most frequently used. Hence, future studies will highly benefit from integrating methods for assessment of regional wall properties to evolve a deeper understanding of how to modulate cardiac healing after acute MI.
Collapse
Affiliation(s)
| | - Anika Dahmen
- Department of Cardiac Surgery, Rostock University Medical Center, Rostock, Germany; Department of Life, Light and Matter, University of Rostock, Rostock, Germany
| | - Praveen Vasudevan
- Department of Cardiac Surgery, Rostock University Medical Center, Rostock, Germany; Department of Life, Light and Matter, University of Rostock, Rostock, Germany
| | - Heiko Lemcke
- Department of Cardiac Surgery, Rostock University Medical Center, Rostock, Germany; Department of Life, Light and Matter, University of Rostock, Rostock, Germany
| | - Ralf Gäbel
- Department of Cardiac Surgery, Rostock University Medical Center, Rostock, Germany; Department of Life, Light and Matter, University of Rostock, Rostock, Germany
| | - Alper Öner
- Department of Cardiology, Rostock University Medical Center, Rostock, Germany
| | - Hüseyin Ince
- Department of Cardiology, Rostock University Medical Center, Rostock, Germany
| | - Robert David
- Department of Cardiac Surgery, Rostock University Medical Center, Rostock, Germany; Department of Life, Light and Matter, University of Rostock, Rostock, Germany
| | - Markus Wolfien
- Institute of Medical Informatics and Biometry, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
4
|
In Search of the Holy Grail: Stem Cell Therapy as a Novel Treatment of Heart Failure with Preserved Ejection Fraction. Int J Mol Sci 2023; 24:ijms24054903. [PMID: 36902332 PMCID: PMC10003723 DOI: 10.3390/ijms24054903] [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: 12/29/2022] [Revised: 02/20/2023] [Accepted: 02/25/2023] [Indexed: 03/06/2023] Open
Abstract
Heart failure, a leading cause of hospitalizations and deaths, is a major clinical problem. In recent years, the increasing incidence of heart failure with preserved ejection fraction (HFpEF) has been observed. Despite extensive research, there is no efficient treatment for HFpEF available. However, a growing body of evidence suggests stem cell transplantation, due to its immunomodulatory effect, may decrease fibrosis and improve microcirculation and therefore, could be the first etiology-based therapy of the disease. In this review, we explain the complex pathogenesis of HFpEF, delineate the beneficial effects of stem cells in cardiovascular therapy, and summarize the current knowledge concerning cell therapy in diastolic dysfunction. Furthermore, we identify outstanding knowledge gaps that may indicate directions for future clinical studies.
Collapse
|
5
|
Feng Q, Li Q, Zhou H, Sun L, Lin C, Jin Y, Wang D, Guo G. The role of major immune cells in myocardial infarction. Front Immunol 2023; 13:1084460. [PMID: 36741418 PMCID: PMC9892933 DOI: 10.3389/fimmu.2022.1084460] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Accepted: 12/19/2022] [Indexed: 01/20/2023] Open
Abstract
Myocardial infarction (MI) is a cardiovascular disease (CVD) with high morbidity and mortality worldwide, often leading to adverse cardiac remodeling and heart failure, which is a serious threat to human life and health. The immune system makes an important contribution to the maintenance of normal cardiac function. In the disease process of MI, necrotic cardiomyocytes release signals that activate nonspecific immunity and trigger the action of specific immunity. Complex immune cells play an important role in all stages of MI progression by removing necrotic cardiomyocytes and tissue and promoting the healing of damaged tissue cells. With the development of biomaterials, cardiac patches have become an emerging method of repairing MI, and the development of engineered cardiac patches through the construction of multiple animal models of MI can help treat MI. This review introduces immune cells involved in the development of MI, summarizes the commonly used animal models of MI and the newly developed cardiac patch, so as to provide scientific reference for the accurate diagnosis and effective treatment of MI.
Collapse
Affiliation(s)
- Qiang Feng
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, China,Laboratory Animal Center, College of Animal Science, Jilin University, Changchun, China
| | - Qirong Li
- Laboratory Animal Center, College of Animal Science, Jilin University, Changchun, China
| | - Hengzong Zhou
- Laboratory Animal Center, College of Animal Science, Jilin University, Changchun, China
| | - Liqun Sun
- Department of Pathogenobiology, Jilin University Mycology Research Center, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Chao Lin
- School of Grain Science and Technology, Jilin Business and Technology College, Changchun, China
| | - Ye Jin
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
| | - Dongxu Wang
- Laboratory Animal Center, College of Animal Science, Jilin University, Changchun, China,*Correspondence: Gongliang Guo,
| | - Gongliang Guo
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, China,*Correspondence: Gongliang Guo,
| |
Collapse
|
6
|
Li Z, Ding Y, Peng Y, Yu J, Pan C, Cai Y, Dong Q, Zhong Y, Zhu R, Yu K, Zeng Q. Effects of IL-38 on Macrophages and Myocardial Ischemic Injury. Front Immunol 2022; 13:894002. [PMID: 35634320 PMCID: PMC9136064 DOI: 10.3389/fimmu.2022.894002] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 04/13/2022] [Indexed: 11/13/2022] Open
Abstract
Macrophages play an important role in clearing necrotic myocardial tissues, myocardial ischemia-reperfusion injury, and ventricular remodeling after myocardial infarction. M1 macrophages not only participate in the inflammatory response in myocardial tissues after infarction, which causes heart damage, but also exert a protective effect on the heart during ischemia. In contrast, M2 macrophages exhibit anti-inflammatory and tissue repair properties by inducing the production of high levels of anti-inflammatory cytokines and fibro-progenitor cells. Interleukin (IL)-38, a new member of the IL-1 family, has been reported to modulate the IL-36 signaling pathway by playing a role similar to that of the IL-36 receptor antagonist, which also affects the production and secretion of macrophage-related inflammatory factors that play an anti-inflammatory role. IL-38 can relieve myocardial ischemia-reperfusion injury by promoting the differentiation of M1 macrophages into M2 macrophages, inhibit the activation of NOD-like receptor thermal protein domain-associated protein 3 (NLRP3) inflammasome, and increase the secretion of anti-inflammatory cytokines, such as IL-10 and transforming growth factor-β. The intact recombinant IL-38 can also bind to interleukin 1 receptor accessory protein-like 1 (IL-1RAPL1) to activate the c-jun N-terminal kinase/activator protein 1 (JNK/AP1) pathway and increase the production of IL-6. In addition, IL-38 regulates dendritic cell-induced cardiac regulatory T cells, thereby regulating macrophage polarization and improving ventricular remodeling after myocardial infarction. Accordingly, we speculated that IL-38 and macrophage regulation may be therapeutic targets for ameliorating myocardial ischemic injury and ventricular remodeling after myocardial infarction. However, the specific mechanism of the IL-38 action warrants further investigation.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Kunwu Yu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qiutang Zeng
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
7
|
Xiang J, Zhou L, Xie Y, Zhu Y, Xiao L, Chen Y, Zhou W, Chen D, Wang M, Cai L, Guo L. Mesh-like electrospun membrane loaded with atorvastatin facilitates cutaneous wound healing by promoting the paracrine function of mesenchymal stem cells. Stem Cell Res Ther 2022; 13:190. [PMID: 35526075 PMCID: PMC9080129 DOI: 10.1186/s13287-022-02865-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 04/01/2022] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Functional electrospun membranes are promising dressings for promoting wound healing. However, their microstructure and drug loading capacity need further improvements. It is the first time to design a novel mesh-like electrospun fiber loaded with atorvastatin (ATV) and investigated its effects on paracrine secretion by bone marrow-derived mesenchymal stem cells (BMSCs) and wound healing in vivo. METHODS We fabricated a mesh-like electrospun membrane using a copper mesh receiver. The physical properties of the membranes were evaluated by SEM, FTIR spectroscopy, tensile strength analysis, and contrast angle test. Drug release was measured by plotting concentration as a function of time. We tested the effects of conditioned media (CM) derived from BMSCs on endothelial cell migration and angiogenesis. We used these BMSCs and performed RT-PCR and ELISA to evaluate the expressions of vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (b-FGF) genes and proteins, respectively. The involvement of FAK and AKT mechanotransduction pathways in the regulation of BMSC secretion by material surface topography was also investigated. Furthermore, we established a rat model of wound healing, applied ATV-loaded mesh-like membranes (PCL/MAT) seeded with BMSCs on wounds, and assessed their efficacy for promoting wound healing. RESULTS FTIR spectroscopy revealed successful ATV loading in PCL/MAT. Compared with random electrospun fibers (PCL/R) and mesh-like electrospun fibers without drug load (PCL/M), PCL/MAT induced maximum promotion of human umbilical vein endothelial cell (HUVEC) migration. In the PCL/MAT group, the cell sheet scratches were nearly closed after 24 h. However, the cell sheet scratches remained open in other treatments at the same time point. The PCL/MAT promoted angiogenesis and led to the generation of longer tubes than the other treatments. Finally, the PCL/MAT induced maximum gene expression and protein secretion of VEGF and b-FGF. As for material surface topography effect on BMSCs, FAK and AKT signaling pathways were shown to participate in the modulation of MSC morphology and its paracrine function. In vivo, PCL/MAT seeded with BMSCs significantly accelerated healing and improved neovascularization and collagen reconstruction in the wound area compared to the other treatments. CONCLUSIONS The mesh-like topography of fibrous scaffolds combined with ATV release creates a unique microenvironment that promotes paracrine secretion of BMSCs, thereby accelerating wound healing. Hence, drug-loaded mesh-like electrospun membranes may be highly efficacious for wound healing and as artificial skin. It is a promising approach to solve the traumatic skin defect and accelerate recovery, which is essential to developing functional materials for future regenerative medicine.
Collapse
Affiliation(s)
- Jieyu Xiang
- Department of Plastic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Ling Zhou
- Department of Plastic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Yuanlong Xie
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Yufan Zhu
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Lingfei Xiao
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Yan Chen
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Wei Zhou
- Department of Plastic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Danyang Chen
- Department of Plastic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Min Wang
- Department of Plastic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Lin Cai
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| | - Liang Guo
- Department of Plastic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
8
|
Migration and phenotype switching of macrophages at early-phase of bone-formation by secretomes from bone marrow derived mesenchymal stem cells using rat calvaria bone defect model. J Dent Sci 2022; 17:421-429. [PMID: 35028066 PMCID: PMC8739749 DOI: 10.1016/j.jds.2021.08.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 08/17/2021] [Indexed: 01/08/2023] Open
Abstract
Background/purpose Conditioned media of cultured mesenchymal stem cells (MSCs) contain numerous kinds of secretomes such as cytokines and chemokines. We previously reported that conditioned media of bone marrow-derived MSCs (MSC-CM) promote bone formation. Recently, macrophage phenotype switching from the pro-inflammatory M1 type to the anti-inflammatory M2 type has been reported to be an important phenomenon during tissue regeneration. Some studies reported that this phenotype switching is regulated by secretomes. In this study, macrophage phenotype during bone formation by MSC-CM was investigated. Materials and methods Human MSCs (hMSCs) were cultured in serum-free medium and the collected medium was defined as MSC-CM. Macrophage-related gene expressions in hMSCs cultured with MSC-CM were evaluated by quantitative real-time polymerase chain reaction. MSC-CM was implanted and the evaluations by micro-CT and immunohistochemistry were performed using a rat the calvaria bone defect model. Results Two and four weeks after implantation, the MSC-CM group demonstrated enhanced bone regeneration. Gene expressions of C–C motif chemokine 2 (CCL2), colony-stimulating factor 2 (CSF2) and CD163 was significantly upregulated in cells exposed to MSC-CM. Immunohistochemical staining revealed that iNOS-positive M1 macrophages were reduced, while CD204-positive M2 macrophages were increased in the MSC-CM group at 72 h after implantation, and the M2/M1 ratio increased only in the MSC-CM group. Conclusion MSC-CM enhances macrophage migration and induces M1 to M2 type macrophage switching at an early stage of osteogenesis. Such phenotype switching provides a favorable environment for angiogenesis, cellular migration, and osteogenesis and contributes to MSC-CM-induced early bone formation.
Collapse
|
9
|
Astaxanthin promotes M2 macrophages and attenuates cardiac remodeling after myocardial infarction by suppression inflammation in rats. Chin Med J (Engl) 2021; 133:1786-1797. [PMID: 32701588 PMCID: PMC7470000 DOI: 10.1097/cm9.0000000000000814] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background Cardiac remodeling after acute myocardial infarction (AMI) is an important process. The present study aimed to assess the protective effects of astaxanthin (ASX) on cardiac remodeling after AMI. Methods The study was conducted between April and September 2018. To create a rat AMI model, rats were anesthetized, and the left anterior descending coronary artery was ligated. The rats in the ASX group received 10 mg·kg−1·day−1 ASX by gavage for 28 days. On the 1st day after AMI, but before ASX administration, six rats from each group were sacrificed to evaluate changes in the heart function and peripheral blood (PB) levels of inflammatory factors. On the 7th day after AMI, eight rats from each group were sacrificed to evaluate the PB levels of inflammatory factors and the M2 macrophage count using both immunofluorescence (IF) and flow cytometry (FC). The remaining rats were observed for 28 days. Cardiac function was examined using echocardiography. The inflammatory factors, namely, tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), and IL-10, were assessed using enzyme-linked immunosorbent assay. The heart weight/body weight (BW), and lung weight (LW)/BW ratios were calculated, and myocardial fibrosis in the form of collagen volume fraction was measured using Masson trichrome staining. Hematoxylin and eosin (H&E) staining was used to determine the myocardial infarct size (MIS), and TdT-mediated dUTP nick-end labeling staining was used to analyze the myocardial apoptosis index. The levels of apoptosis-related protein, type I/III collagen, transforming growth factor β1 (TGF-β1), metalloproteinase 9 (MMP9), and caspase 3 were assessed by Western blotting. Unpaired t-test, one-way analysis of variance, and non-parametric Mann-Whitney test were used to analyze the data. Results On day 1, cardiac function was worse in the ASX group than in the sham group (left ventricular end-systolic diameter [LVIDs]: 0.72 ± 0.08 vs. 0.22 ± 0.06 cm, t = −11.38; left ventricular end-diastolic diameter [LVIDd]: 0.89 ± 0.09 vs. 0.48 ± 0.05 cm, t = −9.42; end-systolic volume [ESV]: 0.80 [0.62, 0.94] vs. 0.04 [0.03, 0.05] mL, Z = −2.89; end-diastolic volume [EDV]: 1.39 [1.03, 1.49] vs. 0.28 [0.22, 0.32] mL, Z = −2.88; ejection fraction [EF]: 0.40 ± 0.04 vs. 0.86 ± 0.05, t = 10.00; left ventricular fractional shortening [FS] rate: 0.19 [0.18, 0.20] %FS vs. 0.51 [0.44, 0.58] %FS, Z = −2.88, all P < 0.01; n = 6). The levels of inflammatory factors significantly increased (TNF-α: 197.60 [133.89, 237.94] vs. 50.48 [47.21 57.10] pg/mL, Z = −2.88; IL-1β: 175.23 [160.74, 215.09] vs. 17.78 [16.83, 19.56] pg/mL, Z = −2.88; IL-10: 67.64 [58.90, 71.46] vs. 12.33 [11.64, 13.98] pg/mL, Z = −2.88, all P < 0.01; n = 6). On day 7, the levels of TNF-α and IL-1β were markedly lower in the ASX group than in the AMI group (TNF-α: 71.70 [68.60, 76.00] vs. 118.07 [106.92, 169.08] pg/mL, F = 42.64; IL-1β: 59.90 [50.83, 73.78] vs. 151.60 [108.4, 198.36] pg/mL, F = 44.35, all P < 0.01, n = 8). Conversely, IL-10 levels significantly increased (141.84 [118.98, 158.36] vs. 52.96 [42.68, 74.52] pg/mL, F = 126.67, P < 0.01, n = 8). The M2 macrophage count significantly increased (2891.42 ± 211.29 vs. 1583.38 ± 162.22, F = 274.35, P < 0.01 by immunofluorescence test; 0.96 ± 0.18 vs. 0.36 ± 0.05, F = 46.24, P < 0.05 by flowcytometry test). On day 28, cardiac function was better in the ASX group than in the AMI group (LVIDs: 0.50 [0.41, 0.56] vs. 0.64 [0.56, 0.74] cm, Z = −3.60; LVIDd: 0.70 [0.60, 0.76] vs. 0.80 [0.74 0.88] cm, Z = −2.96; ESV: 0.24 [0.18, 0.45] vs. 0.58 [0.44, 0.89] mL, Z = −3.62; EDV: 0.76 [0.44, 1.04] vs. 1.25 [0.82, 1.46] mL, Z = −2.54; EF: 0.60 ± 0.08 vs. 0.50 ± 0.12, F = 160.48; %FS: 0.29 [0.24, 0.31] vs. 0.20 [0.17, 0.21], Z = −4.43, all P < 0.01; n = 16). The MIS and LW/BW ratio were markedly lower in the ASX group than in the AMI group (myocardial infarct size: 32.50 ± 1.37 vs. 50.90 ± 1.73, t = 23.63, P < 0.01, n = 8; LW/BW: 1.81 ± 0.15 vs. 2.17 ± 0.37, t = 3.66, P = 0.01, n = 16). The CVF was significantly lower in the ASX group than in the AMI group: 12.88 ± 2.53 vs. 28.92 ± 3.31, t = 10.89, P < 0.01, n = 8. The expression of caspase 3, TGF-β1, MMP9, and type I/III collagen was lower in the ASX group than in the AMI group (caspase 3: 0.38 ± 0.06 vs. 0.66 ± 0.04, t = 8.28; TGF-β1: 0.37 ± 0.04 vs. 0.62 ± 0.07, t = 6.39; MMP9: 0.20 ± 0.06 vs. 0.40 ± 0.06, t = 4.62; type I collagen: 0.42 ± 0.09 vs. 0.74 ± 0.07, t = 5.73; type III collagen: 0.13 ± 0.02 vs. 0.74 ± 0.07, t = 4.32, all P < 0.01; n = 4). Conclusions ASX treatment after AMI may promote M2 macrophages and effectively attenuate cardiac remodeling by inhibiting inflammation and reducing myocardial fibrosis.
Collapse
|
10
|
New Insights and Novel Therapeutic Potentials for Macrophages in Myocardial Infarction. Inflammation 2021; 44:1696-1712. [PMID: 33866463 PMCID: PMC8460536 DOI: 10.1007/s10753-021-01467-2] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 03/09/2021] [Accepted: 04/05/2021] [Indexed: 12/19/2022]
Abstract
Cardiovascular disease (CVD) has long been the leading cause of death worldwide, and myocardial infarction (MI) accounts for the greatest proportion of CVD. Recent research has revealed that inflammation plays a major role in the pathogenesis of CVD and other manifestations of atherosclerosis. Overwhelming evidence supports the view that macrophages, as the basic cell component of the innate immune system, play a pivotal role in atherosclerosis initiation and progression. Limited but indispensable resident macrophages have been detected in the healthy heart; however, the number of cardiac macrophages significantly increases during cardiac injury. In the early period of initial cardiac damage (e.g., MI), numerous classically activated macrophages (M1) originating from the bone marrow and spleen are rapidly recruited to damaged sites, where they are responsible for cardiac remodeling. After the inflammatory stage, the macrophages shift toward an alternatively activated phenotype (M2) that promotes cardiac repair. In addition, extensive studies have shown the therapeutic potential of macrophages as targets, especially for emerging nanoparticle-mediated drug delivery systems. In the present review, we focused on the role of macrophages in the development and progression of MI, factors regulating macrophage activation and function, and the therapeutic potential of macrophages in MI.
Collapse
|
11
|
Hasan AS, Luo L, Baba S, Li TS. Estrogen is required for maintaining the quality of cardiac stem cells. PLoS One 2021; 16:e0245166. [PMID: 33481861 PMCID: PMC7822545 DOI: 10.1371/journal.pone.0245166] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Accepted: 12/22/2020] [Indexed: 12/24/2022] Open
Abstract
Compared to the age-matched men, the incidence of cardiovascular diseases is lower in premenopausal but higher in postmenopausal women, suggesting the cardio-protective role of estrogen in females. Although cardiac stem cells (CSCs) express estrogen receptors, yet the effects of estrogen on CSCs remain unclear. In this study, we investigated the potential role of estrogen in maintaining the quality of CSCs by in vivo and in vitro experiments. For the in vivo study, estrogen deficiency was induced by ovariectomy in 6-weeks-old C57BL/6 female mice, and then randomly given 17β-estradiol (E2) replacements at a low dose (0.01 mg/60 days) and high dose (0.18 mg/60 days), or vehicle treatment. All mice were killed 2 months after treatments, and heart tissues were collected for ex vivo expansion of CSCs. Compared to age-matched healthy controls, estrogen deficiency slightly decreased the yield of CSCs with significantly lower telomerase activity and more DNA damage. Interestingly, E2 replacements at low and high doses significantly increased the yield of CSCs and reversed the quality impairment of CSCs following estrogen deficiency. For the in vitro study, twice-passaged CSCs from the hearts of adult healthy female mice were cultured with the supplement of 0.01, 0.1, and 1 μM E2 in the medium for 3 days. We found that E2 supplement increased c-kit expression, increased proliferative activity, improved telomerase activity, and reduced DNA damage of CSCs in a dose-dependent manner. Our data suggested the potential role of estrogen in maintaining the quality of CSCs, providing new insight into the cardio-protective effects of estrogen.
Collapse
Affiliation(s)
- Al Shaimaa Hasan
- Department of Stem Cell Biology, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki, Japan
- Department of Medical Pharmacology, Qena Faculty of Medicine, South Valley University, Qena, Egypt
| | - Lan Luo
- Department of Stem Cell Biology, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki, Japan
- School of Medical Technology, Xuzhou Medical University, Xuzhou City, Jiangsu Province, China
| | - Satoko Baba
- Department of Stem Cell Biology, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki, Japan
| | - Tao-Sheng Li
- Department of Stem Cell Biology, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki, Japan
- * E-mail:
| |
Collapse
|
12
|
Pharmacological Modulation of Cardiac Remodeling after Myocardial Infarction. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:8815349. [PMID: 33488934 PMCID: PMC7790555 DOI: 10.1155/2020/8815349] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 11/13/2020] [Accepted: 12/21/2020] [Indexed: 12/14/2022]
Abstract
Cardiac remodeling describes a series of structural and functional changes in the heart after myocardial infarction (MI). Adverse post-MI cardiac remodeling directly jeopardizes the recovery of cardiac functions and the survival rate in MI patients. Several classes of drugs are proven to be useful to reduce the mortality of MI patients. However, it is an ongoing challenge to prevent the adverse effects of cardiac remodeling. The present review aims to identify the pharmacological therapies from the existing clinical drugs for the treatment of adverse post-MI cardiac remodeling. Post-MI cardiac remodeling is a complex process involving ischemia/reperfusion, inflammation, cell death, and deposition of extracellular matrix (ECM). Thus, the present review included two parts: (1) to examine the basic pathophysiology in the cardiovascular system and the molecular basis of cardiac remodeling and (2) to identify the pathological aspects of cardiac remodeling and the potential of the existing pharmacotherapies. Ultimately, the present review highlights drug repositioning as a strategy to discover effective therapies from the existing drugs against post-MI cardiac remodeling.
Collapse
|
13
|
Dergilev KV, Vasilets ID, Tsokolaeva ZI, Zubkova ES, Parfenova EV. [Perspectives of cell therapy for myocardial infarction and heart failure based on cardiosphere cells]. TERAPEVT ARKH 2020; 92:111-120. [PMID: 32598708 DOI: 10.26442/00403660.2020.04.000634] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Indexed: 12/13/2022]
Abstract
Cardiovascular diseases are the leading cause of morbidity and mortality worldwide. In recent years, researchers are attracted to the use of cell therapy based on stem cell and progenitor cells, which has been a promising strategy for cardiac repair after injury. However, conducted research using intracoronary or intramyocardial transplantation of various types of stem/progenitor cells as a cell suspension showed modest efficiency. This is due to the low degree of integration and cell survival after transplantation. To overcome these limitations, the concept of the use of multicellular spheroids modeling the natural microenvironment of cells has been proposed, which allows maintaining their viability and therapeutic properties. It is of great interest to use so-called cardial spheroids (cardiospheres) spontaneously forming three-dimensional structures under low-adhesive conditions, consisting of a heterogeneous population of myocardial progenitor cells and extracellular matrix proteins. This review presents data on methods for creating cardiospheres, directed regulation of their properties and reparative potential, as well as the results of preclinical and clinical studies on their use for the treatment of heart diseases.
Collapse
Affiliation(s)
| | | | - Z I Tsokolaeva
- National Medical Research Center for Cardiology.,Negovsky Scientific Research Institute of General Reanimatology of the Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology
| | - E S Zubkova
- National Medical Research Center for Cardiology
| | - E V Parfenova
- National Medical Research Center for Cardiology.,Lomonosov Moscow State University
| |
Collapse
|
14
|
Mentkowski KI, Mursleen A, Snitzer JD, Euscher LM, Lang JK. CDC-derived extracellular vesicles reprogram inflammatory macrophages to an arginase 1-dependent proangiogenic phenotype. Am J Physiol Heart Circ Physiol 2020; 318:H1447-H1460. [PMID: 32330087 DOI: 10.1152/ajpheart.00155.2020] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Macrophages play a pivotal role in tissue repair following myocardial infarction (MI). In response to injury, they exist along a spectrum of activation states tightly regulated by their microenvironment. Cardiosphere-derived cells (CDCs) have been shown to mediate cardioprotection via modulation of the macrophage response. Our study was designed to gain mechanistic insight into the role of CDC-derived extracellular vesicles (EVs) in modulating macrophage phenotypes and operant signaling pathways to better understand their potential contribution to immunomodulatory cardioprotection. We found that CDC-derived EVs alter the functional phenotype of macrophages, modifying levels of phagocytosis and efferocytosis without changing viability or proliferation. Interestingly, extracellular vesicles differentially regulate several M1/M2 genes dependent on macrophage activation before EV treatment but consistently upregulate arginase 1 regardless of macrophage origin or polarization state. CDC-derived EVs polarize M1 macrophages to a proangiogenic phenotype dependent on arginase 1 upregulation and independent of VEGF-A. In addition, EV-dependent arginase 1 upregulation downregulates nitric oxide (NO) secretion in activated macrophages. These data suggest a novel urea-cycle-dependent mechanism in macrophages that promotes angiogenesis and provides additional mechanistic insight into the potential contribution of CDC-derived extracellular vesicles in immunomodulatory cardioprotection.NEW & NOTEWORTHY We hypothesized that in the window of therapeutic extracellular vesicle (EV) administration, inflammatory M1 macrophages are likely the primary target of cardiosphere-derived cell (CDC)-derived EVs. The effect of CDC-EVs on this population, however, is currently unknown. In this study, we demonstrate that CDC-derived EVs polarize M1 macrophages to a proangiogenic phenotype dependent on arginase 1 upregulation. These results provide insight into an immunomodulatory mechanism of CDC-EVs in a more physiologically relevant model of post-myocardial infarction (post-MI) macrophage polarization.
Collapse
Affiliation(s)
- Kyle I Mentkowski
- Department of Medicine, Division of Cardiology, Jacobs School of Medicine and Biomedical Sciences, Buffalo, New York.,Department of Biomedical Engineering, University at Buffalo, Buffalo, New York
| | - Asma Mursleen
- Department of Medicine, Division of Cardiology, Jacobs School of Medicine and Biomedical Sciences, Buffalo, New York
| | - Jonathan D Snitzer
- Department of Medicine, Division of Cardiology, Jacobs School of Medicine and Biomedical Sciences, Buffalo, New York
| | - Lindsey M Euscher
- Department of Medicine, Division of Cardiology, Jacobs School of Medicine and Biomedical Sciences, Buffalo, New York.,Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, Buffalo, New York
| | - Jennifer K Lang
- Department of Medicine, Division of Cardiology, Jacobs School of Medicine and Biomedical Sciences, Buffalo, New York.,Department of Biomedical Engineering, University at Buffalo, Buffalo, New York.,Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, Buffalo, New York.,Veterans Affairs Western New York Healthcare System, Buffalo, New York
| |
Collapse
|
15
|
18F-FDG PET-Based Imaging of Myocardial Inflammation Predicts a Functional Outcome Following Transplantation of mESC-Derived Cardiac Induced Cells in a Mouse Model of Myocardial Infarction. Cells 2019; 8:cells8121613. [PMID: 31835854 PMCID: PMC6952872 DOI: 10.3390/cells8121613] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 12/02/2019] [Accepted: 12/09/2019] [Indexed: 01/15/2023] Open
Abstract
Cellular inflammation following acute myocardial infarction has gained increasing importance as a target mechanism for therapeutic approaches. We sought to investigate the effect of syngeneic cardiac induced cells (CiC) on myocardial inflammation using 18F-FDG PET (Positron emission tomography)-based imaging and the resulting effect on cardiac pump function using cardiac magnetic resonance (CMR) imaging in a mouse model of myocardial infarction. Mice underwent permanent left anterior descending coronary artery (LAD) ligation inducing an acute inflammatory response. The therapy group received an intramyocardial injection of 106 CiC into the border zone of the infarction. Five days after myocardial infarction, 18F-FDG PET was performed under anaesthesia with ketamine and xylazine (KX) to image the inflammatory response in the heart. Flow cytometry of the mononuclear cells in the heart was performed to analyze the inflammatory response. The effect of CiC therapy on cardiac function was determined after three weeks by CMR. The 18F-FDG PET imaging of the heart five days after myocardial infarction (MI) revealed high focal tracer accumulation in the border zone of the infarcted myocardium, whereas no difference was observed in the tracer uptake between infarct and remote myocardium. The CiC transplantation induced a shift in 18F-FDG uptake pattern, leading to significantly higher 18F-FDG uptake in the whole heart, as well as the remote area of the heart. Correspondingly, high numbers of CD11+ cells could be measured by flow cytometry in this region. The CiC transplantation significantly improved the left ventricular ejection function (LVEF) three weeks after myocardial infarction. The CiC transplantation after myocardial infarction leads to an improvement in pump function through modulation of the cellular inflammatory response five days after myocardial infarction. By combining CiC transplantation and the cardiac glucose uptake suppression protocol with KX in a mouse model, we show for the first time, that imaging of cellular inflammation after myocardial infarction using 18F-FDG PET can be used as an early prognostic tool for assessing the efficacy of cardiac stem cell therapies.
Collapse
|
16
|
Kim D, Chung JJ, Jung Y, Kim SH. The effect of Substance P/Heparin conjugated PLCL polymer coating of bioinert ePTFE vascular grafts on the recruitment of both ECs and SMCs for accelerated regeneration. Sci Rep 2019; 9:17083. [PMID: 31745143 PMCID: PMC6863833 DOI: 10.1038/s41598-019-53514-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 10/24/2019] [Indexed: 12/11/2022] Open
Abstract
Artificial vascular grafts consisting of ePTFE have been mainly used in clinics for the treatment of cardiovascular disease. However, artificial grafts can become clogged after a long time due to thrombosis, as graft maturation by endothelialization is limited. The strategy introduced in this study is to induce graft remodeling through interaction between the bioinert graft and the body. The Substance P (SP) and heparin were covalently conjugated with PLCL, an elastic biocompatible copolymer and the Substance P-conjugated PLCL (SP-PLCL) and/or heparin-conjugated PLCL (Hep-PLCL) were vacuum-coated onto ePTFE vascular grafts. To assess the effectiveness of the coating, coated samples were evaluated by implanting them subcutaneously into SD-Rats. Coatings allow grafts to be remodeled by creating a microenvironment where cells can grow by infiltrating into the grafts while also greatly enhancing angiogenesis. In particular, a double coating of Hep-PLCL and SP-PLCL (Hep/SP-PLCL) at four weeks showed markedly improved vascular remodeling through the recruitment of mesenchymal stem cells (MSCs), vascular cells (ECs, SMCs) and M2 macrophages. Based on these results, it is expected that when the Hep/SP-PLCL-coated ePTFE vascular grafts are implanted in situ, long-term patency will be assured due to the appropriate formation of an endothelial layer and smooth muscle cells in the grafts like native vessels.
Collapse
Affiliation(s)
- Donghak Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea
- Center for Biomaterials, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Justin J Chung
- Center for Biomaterials, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Youngmee Jung
- Center for Biomaterials, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
- Department of Biomedical Engineering, Korea University of Science and Technology (UST), Daejeon, 305-350, Republic of Korea
| | - Soo Hyun Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea.
- Center for Biomaterials, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea.
- Department of Biomedical Engineering, Korea University of Science and Technology (UST), Daejeon, 305-350, Republic of Korea.
| |
Collapse
|
17
|
Wu JMF, Cheng YY, Tang TWH, Shih C, Chen JH, Hsieh PCH. Prostaglandin E 2 Receptor 2 Modulates Macrophage Activity for Cardiac Repair. J Am Heart Assoc 2019; 7:e009216. [PMID: 30371325 PMCID: PMC6404869 DOI: 10.1161/jaha.118.009216] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Background Prostaglandin E2 has long been known to be an immune modulator. It is released after tissue injury and plays a role in modulating macrophage activities, which are essential for tissue regeneration. However, the involvement of prostaglandin E2 receptor 2 (EP2)–dependent regulation of macrophages in postischemic heart is unclear. This study aims to evaluate the role of EP2 in damaged heart. Methods and Results The effect of EP2 in postischemic heart was evaluated using EP2‐deficient transgenic mice. We demonstrated that cardiac function was worse after myocardial injury on loss of EP2. Furthermore, EP2 deficiency also altered proinflammatory response and resulted in a defect in macrophage recruitment to the injured myocardium. Transcriptome analysis revealed that the expression of erythroid differentiation regulator 1 (Erdr1) was significantly induced in EP2‐deficient macrophages. Knocking down Erdr1 expression restored migration ability of EP2‐deficient cells both in vitro and in vivo. By using a genetic fate‐mapping approach, we showed that abolishment of EP2 expression effectively attenuated cell replenishment. Conclusions The EP2‐dependent signaling pathway plays a critical role in regulating macrophage recruitment to the injured myocardium, thereby exerting a function in modulating the inflammatory microenvironment for cardiac repair.
Collapse
Affiliation(s)
- Jasmine M F Wu
- 1 Institute of Basic Medical Sciences and Institute of Clinical Medicine National Cheng Kung University Tainan Taiwan.,2 Institute of Biomedical Sciences Academia Sinica Taipei Taiwan.,4 Leibniz Institute on Aging-Fritz Lipmann Institute Jena Germany
| | - Yuan-Yuan Cheng
- 2 Institute of Biomedical Sciences Academia Sinica Taipei Taiwan
| | - Tony W H Tang
- 2 Institute of Biomedical Sciences Academia Sinica Taipei Taiwan
| | - Crystal Shih
- 2 Institute of Biomedical Sciences Academia Sinica Taipei Taiwan
| | - Jyh-Hong Chen
- 3 Division of Cardiology Department of Medicine College of Medicine China Medical University Taichung Taiwan
| | - Patrick C H Hsieh
- 1 Institute of Basic Medical Sciences and Institute of Clinical Medicine National Cheng Kung University Tainan Taiwan.,2 Institute of Biomedical Sciences Academia Sinica Taipei Taiwan
| |
Collapse
|
18
|
Yap J, Cabrera-Fuentes HA, Irei J, Hausenloy DJ, Boisvert WA. Role of Macrophages in Cardioprotection. Int J Mol Sci 2019; 20:E2474. [PMID: 31109146 PMCID: PMC6566352 DOI: 10.3390/ijms20102474] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 05/14/2019] [Accepted: 05/17/2019] [Indexed: 12/21/2022] Open
Abstract
Cardiovascular diseases are the leading cause of mortality worldwide. It is widely known that non-resolving inflammation results in atherosclerotic conditions, which are responsible for a host of downstream pathologies including thrombosis, myocardial infarction (MI), and neurovascular events. Macrophages, as part of the innate immune response, are among the most important cell types in every stage of atherosclerosis. In this review we discuss the principles governing macrophage function in the healthy and infarcted heart. More specifically, how cardiac macrophages participate in myocardial infarction as well as cardiac repair and remodeling. The intricate balance between phenotypically heterogeneous populations of macrophages in the heart have profound and highly orchestrated effects during different phases of myocardial infarction. In the early "inflammatory" stage of MI, resident cardiac macrophages are replaced by classically activated macrophages derived from the bone marrow and spleen. And while the macrophage population shifts towards an alternatively activated phenotype, the inflammatory response subsides giving way to the "reparative/proliferative" phase. Lastly, we describe the therapeutic potential of cardiac macrophages in the context of cell-mediated cardio-protection. Promising results demonstrate innovative concepts; one employing a subset of yolk sac-derived, cardiac macrophages that have complete restorative capacity in the injured myocardium of neonatal mice, and in another example, post-conditioning of cardiac macrophages with cardiosphere-derived cells significantly improved patient's post-MI diagnoses.
Collapse
Affiliation(s)
- Jonathan Yap
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI 96813, USA.
| | - Hector A Cabrera-Fuentes
- Tecnologico de Monterrey, Centro de Biotecnologia-FEMSA, Monterrey, NL 264610, Mexico.
- National Heart Research Institute Singapore, National Heart Centre, Singapore 169609, Singapore.
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore 169857, Singapore.
- Institute of Biochemistry, Medical School, Justus-Liebig University, 35392 Giessen, Germany.
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, 420008 Kazan, Russia.
| | - Jason Irei
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI 96813, USA.
| | - Derek J Hausenloy
- National Heart Research Institute Singapore, National Heart Centre, Singapore 169609, Singapore.
- Institute of Biochemistry, Medical School, Justus-Liebig University, 35392 Giessen, Germany.
- Yong Loo Lin School of Medicine, National University Singapore, Singapore 117597, Singapore.
- The Hatter Cardiovascular Institute, University College London, London WC1E 6HX, UK.
- The National Institute of Health Research University College London Hospitals Biomedical Research Centre, Research & Development, London W1T 7DN, UK.
| | - William A Boisvert
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI 96813, USA.
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, 420008 Kazan, Russia.
| |
Collapse
|
19
|
Luo L, Li TS, Tang J, Cheng K. Response by Luo et al to Letter Regarding Article, "Fabrication of Synthetic Mesenchymal Stem Cells for the Treatment of Acute Myocardial Infarction in Mice". Circ Res 2019; 120:e48-e49. [PMID: 28546361 DOI: 10.1161/circresaha.117.311151] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Lan Luo
- Department of Stem Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, Japan
| | - Tao-Sheng Li
- Department of Stem Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, Japan
| | - Junnan Tang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, China
| | - Ke Cheng
- Department of Molecular Biomedical Sciences, NC State University, Department of Biomedical Engineering, UNC-Chapel Hill and NC state University, Division of Molecular Pharmaceutics, UNC-Chapel Hill
| |
Collapse
|
20
|
Potential Role of the Resident Mesenchymal Stem-Like Cells in Renal Fibrogenesis after Ureteral Obstruction. Stem Cells Int 2019; 2019:2543171. [PMID: 30949209 PMCID: PMC6425387 DOI: 10.1155/2019/2543171] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 01/03/2019] [Accepted: 01/15/2019] [Indexed: 01/06/2023] Open
Abstract
The mechanisms of renal fibrogenesis after ureteral obstruction remain unclear. We tried to primarily expand mesenchymal stem cells from renal tissues and then investigated their role in fibrogenesis after ureteral obstruction. Unilateral ureteral obstruction was induced by ligating the left ureteral duct of adult C57BL/6 mice. We collected the kidneys for experiments at 2, 7, and 14 days after operation. Histological analysis showed obviously fibrotic changes in the left kidney at 7 days and further increased at 14 days after ureteral obstruction. To expand mesenchymal stem cells, we minced the renal tissues into small explants (about 1 mm3) and cultured onto 10 cm dishes. Interestingly, the outgrowth of cells was observed significantly earlier from the explants of the obstructed left kidney than that of the unobstructed right kidney. These expanded cells showed the potency of adipogenic, osteogenic, and chondrogenic differentiations and positively expressed with CD44 and partly expressed with CD90, CD105, and CD106, but negatively expressed with CD34, CD45, and FSP1, suggesting the phenotype of mesenchymal stem-like cells (MSLCs). The mouse fibrosis RT2 profiler PCR array showed that many genes were changed over 2-fold in the MSLCs expanded from both kidneys at 2, 7, and 14 days after operation. Interestingly, profibrotic genes were prevalently enhanced in the left kidney with ureteral obstruction. Histological analysis also showed obviously infiltration of inflammatory cells in the left kidney at 14 days after operation. Our data indicate the potential role of resident MSLCs in renal fibrogenesis after ureteral obstruction, but further experiments are required to understand the relevant mechanisms.
Collapse
|
21
|
Liu YC, Hsiao YY, Ku KL, Liao HF, Chao WC. Mahonia oiwakensis Extract and Its Bioactive Compounds Exert Anti-Inflammatory Activities and VEGF Production Through M2-Macrophagic Polarization and STAT6 Activation. J Med Food 2018; 21:654-664. [PMID: 29652553 DOI: 10.1089/jmf.2017.4084] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Macrophages play an indispensable role in the host immune defense. Macrophages can undergo polarization into classically (M1) and alternatively (M2) activated macrophages. M1 macrophages activate immune and inflammatory response, while M2 macrophages are involved in tissue remodeling. Mahonia oiwakensis (Mo) is a herbal medicine in Asia used for its anti-inflammatory and analgesic properties; however, the mechanism is unclear. This study analyzed the effect of Mo extracts and its effects on the polarization of both macrophage RAW264.7 cells and mouse splenic macrophages. Water (Mo-W) and EtOH extracts (Mo-E) did not change the viability of RAW264.7 cells, whereas Mo-E inhibited nitric oxide (NO) production. The major compounds, berberine and palmatine, decreased the viability and NO levels of cells. The secretion of inflammatory cytokines CXCL16, IL-6, L-selectin, MCP1, RANTES, and sTNF-R1 was downregulated, whereas the production of vascular endothelial growth factor (VEGF) was upregulated by Mo-E, berberine, and palmatine treatments. Mo-E, berberine, and palmatine stimulated the expression of macrophage CD68 and M2-type CD204 markers, decreased M1-mediated p-STAT1 and NF-κB, and increased M2-mediated p-STAT6 expression. Similar effects on M2 polarization were also observed in splenic macrophages from mice. In conclusion, Mo-E, berberine, and palmatine modulated macrophages through the suppression of M1-mediated inflammation and the recruitment of M2-mediated VEGF secretion and STAT6 expression.
Collapse
Affiliation(s)
- Yea-Chen Liu
- 1 Department of Biological Resources, National Chiayi University , Chiayi, Taiwan
| | - Yu-Ying Hsiao
- 2 Department of Biochemical Science and Technology, National Chiayi University , Chiayi, Taiwan
| | - Kuo-Lung Ku
- 3 Department of Applied Chemistry, National Chiayi University , Chiayi, Taiwan
| | - Hui-Fen Liao
- 2 Department of Biochemical Science and Technology, National Chiayi University , Chiayi, Taiwan
| | - Wei-Chun Chao
- 4 Department of Forestry and Natural Resources, National Chiayi University , Chiayi, Taiwan
| |
Collapse
|
22
|
Alvarez P, Briasoulis A. Immune Modulation in Heart Failure: the Promise of Novel Biologics. CURRENT TREATMENT OPTIONS IN CARDIOVASCULAR MEDICINE 2018. [DOI: 10.1007/s11936-018-0617-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
23
|
Couret D, Bourane S, Catan A, Nativel B, Planesse C, Dorsemans AC, Ait-Arsa I, Cournot M, Rondeau P, Patche J, Tran-Dinh A, Lambert G, Diotel N, Meilhac O. A hemorrhagic transformation model of mechanical stroke therapy with acute hyperglycemia in mice. J Comp Neurol 2018; 526:1006-1016. [PMID: 29277912 DOI: 10.1002/cne.24386] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 12/07/2017] [Accepted: 12/07/2017] [Indexed: 12/25/2022]
Abstract
Clinical benefit for mechanical thrombectomy (MT) in stroke was recently demonstrated in multiple large prospective studies. Acute hyperglycemia (HG) is an important risk factor of poor outcome in stroke patients, including those that underwent MT. The aim of this therapy is to achieve a complete reperfusion in a short time, given that reperfusion damage is dependent on the duration of ischemia. Here, we investigated the effects of acute HG in a mouse model of ischemic stroke induced by middle cerebral artery occlusion (MCAO). Hyperglycemic (intraperitoneal [ip] injection of glucose) and control (ip saline injection) 10-week male C57BL6 mice were subjected to MCAO (30, 90, and 180 min) followed by reperfusion obtained by withdrawal of the monofilament. Infarct volume, hemorrhagic transformation (HT), neutrophil infiltration, and neurological scores were assessed at 24 hr by performing vital staining, ELISA immunofluorescence, and behavioral test, respectively. Glucose injection led to transient HG (blood glucose = 250-390 mg/dL) that significantly increased infarct volume, HT, and worsened neurological outcome. In addition, we report that HG promoted blood-brain barrier disruption as shown by hemoglobin accumulation in the brain parenchyma and tended to increase neutrophil extravasation within the infarcted area. Acute HG increased neurovascular damage for all MCAO durations tested. HTs were observed as early as 90 min after ischemia under hyperglycemic conditions. This model mimics MT ischemia/reperfusion and allows the exploration of brain injury in hyperglycemic conditions.
Collapse
Affiliation(s)
- David Couret
- Université de La Réunion, INSERM, UMR 1188 Diabète Athérothrombose Réunion Océan Indien (DéTROI), Saint-Denis de La Réunion, France.,CHU de La Réunion, Service de Neuroréanimation, Saint-Pierre de La Réunion, France
| | - Steeve Bourane
- Université de La Réunion, INSERM, UMR 1188 Diabète Athérothrombose Réunion Océan Indien (DéTROI), Saint-Denis de La Réunion, France
| | - Aurélie Catan
- Université de La Réunion, INSERM, UMR 1188 Diabète Athérothrombose Réunion Océan Indien (DéTROI), Saint-Denis de La Réunion, France
| | - Brice Nativel
- Université de La Réunion, INSERM, UMR 1188 Diabète Athérothrombose Réunion Océan Indien (DéTROI), Saint-Denis de La Réunion, France
| | - Cynthia Planesse
- Université de La Réunion, INSERM, UMR 1188 Diabète Athérothrombose Réunion Océan Indien (DéTROI), Saint-Denis de La Réunion, France
| | - Anne-Claire Dorsemans
- Université de La Réunion, INSERM, UMR 1188 Diabète Athérothrombose Réunion Océan Indien (DéTROI), Saint-Denis de La Réunion, France
| | - Imade Ait-Arsa
- Plateforme CYROI, Cyclotron Réunion Océan Indien, Sainte-Clotilde, France
| | - Maxime Cournot
- Université de La Réunion, INSERM, UMR 1188 Diabète Athérothrombose Réunion Océan Indien (DéTROI), Saint-Denis de La Réunion, France.,Hôpital Gabriel Martin, Service de Cardiologie, Saint-Paul de La Réunion, France.,CHU de La Réunion, Saint-Denis de La Réunion, France
| | - Philippe Rondeau
- Université de La Réunion, INSERM, UMR 1188 Diabète Athérothrombose Réunion Océan Indien (DéTROI), Saint-Denis de La Réunion, France
| | - Jessica Patche
- Université de La Réunion, INSERM, UMR 1188 Diabète Athérothrombose Réunion Océan Indien (DéTROI), Saint-Denis de La Réunion, France
| | | | - Gilles Lambert
- Université de La Réunion, INSERM, UMR 1188 Diabète Athérothrombose Réunion Océan Indien (DéTROI), Saint-Denis de La Réunion, France
| | - Nicolas Diotel
- Université de La Réunion, INSERM, UMR 1188 Diabète Athérothrombose Réunion Océan Indien (DéTROI), Saint-Denis de La Réunion, France
| | - Olivier Meilhac
- Université de La Réunion, INSERM, UMR 1188 Diabète Athérothrombose Réunion Océan Indien (DéTROI), Saint-Denis de La Réunion, France.,CHU de La Réunion, Service de Neuroréanimation, Saint-Pierre de La Réunion, France.,CHU de La Réunion, Saint-Denis de La Réunion, France
| |
Collapse
|
24
|
DeBerge M, Zhang S, Glinton K, Grigoryeva L, Hussein I, Vorovich E, Ho K, Luo X, Thorp EB. Efferocytosis and Outside-In Signaling by Cardiac Phagocytes. Links to Repair, Cellular Programming, and Intercellular Crosstalk in Heart. Front Immunol 2017; 8:1428. [PMID: 29163503 PMCID: PMC5671945 DOI: 10.3389/fimmu.2017.01428] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 10/13/2017] [Indexed: 12/24/2022] Open
Abstract
Phagocytic sensing and engulfment of dying cells and extracellular bodies initiate an intracellular signaling cascade within the phagocyte that can polarize cellular function and promote communication with neighboring non-phagocytes. Accumulating evidence links phagocytic signaling in the heart to cardiac development, adult myocardial homeostasis, and the resolution of cardiac inflammation of infectious, ischemic, and aging-associated etiology. Phagocytic clearance in the heart may be carried out by professional phagocytes, such as macrophages, and non-professional cells, including myofibrolasts and potentially epithelial cells. During cardiac development, phagocytosis initiates growth cues for early cardiac morphogenesis. In diseases of aging, including myocardial infarction, heightened levels of cell death require efficient phagocytic debridement to salvage further loss of terminally differentiated adult cardiomyocytes. Additional risk factors, including insulin resistance and other systemic risk factors, contribute to inefficient phagocytosis, altered phagocytic signaling, and delayed cardiac inflammation resolution. Under such conditions, inflammatory presentation of myocardial antigen may lead to autoimmunity and even possible rejection of transplanted heart allografts. Increased understanding of these basic mechanisms offers therapeutic opportunities.
Collapse
Affiliation(s)
- Matthew DeBerge
- Department of Pathology, Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Shuang Zhang
- Department of Pathology, Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Kristofor Glinton
- Department of Pathology, Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Luba Grigoryeva
- Department of Pathology, Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Islam Hussein
- Department of Pathology, Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Esther Vorovich
- Department of Pathology, Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Karen Ho
- Department of Pathology, Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Xunrong Luo
- Department of Pathology, Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Edward B Thorp
- Department of Pathology, Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| |
Collapse
|
25
|
Abstract
INTRODUCTION Over the past decade, it has become clear that long-term engraftment of any ex vivo expanded cell product transplanted into injured myocardium is modest and all therapeutic regeneration is mediated by stimulation of endogenous repair rather than differentiation of transplanted cells into working myocardium. Given that increasing the retention of transplanted cells boosts myocardial function, focus on the fundamental mechanisms limiting retention and survival of transplanted cells may enable strategies to help to restore normal cardiac function. Areas covered: This review outlines the challenges confronting cardiac engraftment of ex vivo expanded cells and explores means of enhancing cell-mediated repair of injured myocardium. Expert opinion: Stem cell therapy has already come a long way in terms of regenerating damaged hearts though the poor retention of transplanted cells limits the full potential of truly cardiotrophic cell products. Multifaceted strategies directed towards fundamental mechanisms limiting the long-term survival of transplanted cells will be needed to enhance transplanted cell retention and cell-mediated repair of damaged myocardium for cardiac cell therapy to reach its full potential.
Collapse
Affiliation(s)
| | - Darryl R Davis
- a University of Ottawa Heart Institute , Ottawa , ON , Canada
| |
Collapse
|
26
|
Lu J, Xie L, Liu C, Zhang Q, Sun S. PTEN/PI3k/AKT Regulates Macrophage Polarization in Emphysematous mice. Scand J Immunol 2017; 85:395-405. [PMID: 28273403 DOI: 10.1111/sji.12545] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 03/02/2017] [Indexed: 12/27/2022]
Affiliation(s)
- J. Lu
- Department of Respiratory Medicine; the Third XiangYa Hospital of Central South University; Changsha Hunan Province China
| | - L. Xie
- Department of Respiratory Medicine; the Third XiangYa Hospital of Central South University; Changsha Hunan Province China
| | - C. Liu
- Department of Respiratory Medicine; the Third XiangYa Hospital of Central South University; Changsha Hunan Province China
| | - Q. Zhang
- Department of Respiratory Medicine; the Third XiangYa Hospital of Central South University; Changsha Hunan Province China
| | - S. Sun
- Department of Respiratory Medicine; the Third XiangYa Hospital of Central South University; Changsha Hunan Province China
| |
Collapse
|
27
|
Jung M, Ma Y, Iyer RP, DeLeon-Pennell KY, Yabluchanskiy A, Garrett MR, Lindsey ML. IL-10 improves cardiac remodeling after myocardial infarction by stimulating M2 macrophage polarization and fibroblast activation. Basic Res Cardiol 2017; 112:33. [PMID: 28439731 DOI: 10.1007/s00395-017-0622-5] [Citation(s) in RCA: 309] [Impact Index Per Article: 38.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 04/13/2017] [Indexed: 01/26/2023]
Abstract
Inflammation resolution is important for scar formation following myocardial infarction (MI) and requires the coordinated actions of macrophages and fibroblasts. In this study, we hypothesized that exogenous interleukin-10 (IL-10), an anti-inflammatory cytokine, promotes post-MI repair through actions on these cardiac cell types. To test this hypothesis, C57BL/6J mice (male, 3- to 6-month old, n = 24/group) were treated with saline or IL-10 (50 μg/kg/day) by osmotic mini-pump infusion starting at day (d) 1 post-MI and sacrificed at d7 post-MI. IL-10 infusion doubled plasma IL-10 concentrations by d7 post-MI. Despite similar infarct areas and mortality rates, IL-10 treatment significantly decreased LV dilation (1.6-fold for end-systolic volume and 1.4-fold for end-diastolic volume) and improved ejection fraction 1.8-fold (both p < 0.05). IL-10 treatment attenuated inflammation at d7 post-MI, evidenced by decreased numbers of Mac-3-positive macrophages in the infarct (p < 0.05). LV macrophages isolated from d7 post-MI mice treated with IL-10 showed significantly elevated gene expression of M2 markers (Arg1, Ym1, and Tgfb1; all p < 0.05). We further performed RNA-seq analysis on post-MI cardiac macrophages and identified 410 significantly different genes (155 increased, 225 decreased by IL-10 treatment). By functional network analysis grouping, the majority of genes (133 out of 410) were part of the cellular assembly and repair functional group. Of these, hyaluronidase 3 (Hyal3) was the most important feature identified by p value. IL-10 treatment decreased Hyal3 by 28%, which reduced hyaluronan degradation and limited collagen deposition (all p < 0.05). In addition, in vivo IL-10 treatment increased fibroblast activation (proliferation, migration, and collagen production), an effect that was both directly and indirectly influenced by macrophage M2 polarization. Combined, our results indicate that in vivo infusion of IL-10 post-MI improves the LV microenvironment to dampen inflammation and facilitate cardiac wound healing.
Collapse
Affiliation(s)
- Mira Jung
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, 2500 North State St., Jackson, MS, 39216-4505, USA
| | - Yonggang Ma
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, 2500 North State St., Jackson, MS, 39216-4505, USA
| | - Rugmani Padmanabhan Iyer
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, 2500 North State St., Jackson, MS, 39216-4505, USA
| | - Kristine Y DeLeon-Pennell
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, 2500 North State St., Jackson, MS, 39216-4505, USA.,Research Service, G.V. (Sonny) Montgomery Veterans Affairs Medical Center, Jackson, MS, USA
| | - Andriy Yabluchanskiy
- Donald W. Reynolds Department of Geriatric Medicine, Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | | | - Merry L Lindsey
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, 2500 North State St., Jackson, MS, 39216-4505, USA. .,Research Service, G.V. (Sonny) Montgomery Veterans Affairs Medical Center, Jackson, MS, USA.
| |
Collapse
|
28
|
Gombozhapova A, Rogovskaya Y, Shurupov V, Rebenkova M, Kzhyshkowska J, Popov SV, Karpov RS, Ryabov V. Macrophage activation and polarization in post-infarction cardiac remodeling. J Biomed Sci 2017; 24:13. [PMID: 28173864 PMCID: PMC5297120 DOI: 10.1186/s12929-017-0322-3] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 01/31/2017] [Indexed: 02/07/2023] Open
Abstract
Adverse cardiac remodeling leads to impaired ventricular function and heart failure, remaining a major cause of mortality and morbidity in patients with acute myocardial infarction. It have been shown that, even if all the recommended therapies for ST-segment elevation myocardial infarction are performed, one third of patients undergoes progressive cardiac remodeling that represents morphological basis for following heart failure. The need to extend our knowledge about factors leading to different clinical scenarios of myocardial infarction and following complications has resulted in a research of immuno-inflammatory pathways and molecular activities as the basis for post-infarction remodeling. Recently, macrophages (cells of the innate immune system) have become a subject of scientific interest under both normal and pathological conditions. Macrophages, besides their role in host protection and tissue homeostasis, play an important role in pathophysiological processes induced by myocardial infarction. In this article we summarize data about the function of monocytes and macrophages plasticity in myocardial infarction and outline potential role of these cells as effective targets to control processes of inflammation, cardiac remodeling and healing following acute coronary event.
Collapse
Affiliation(s)
- Aleksandra Gombozhapova
- Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk 111a Kievskaya Street, 634012, Tomsk, Russian Federation. .,National Research Tomsk State University, 36 Lenin Avenue, 634050, Tomsk, Russian Federation.
| | - Yuliya Rogovskaya
- Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk 111a Kievskaya Street, 634012, Tomsk, Russian Federation.,National Research Tomsk State University, 36 Lenin Avenue, 634050, Tomsk, Russian Federation
| | - Vladimir Shurupov
- Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk 111a Kievskaya Street, 634012, Tomsk, Russian Federation
| | - Mariya Rebenkova
- Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk 111a Kievskaya Street, 634012, Tomsk, Russian Federation.,National Research Tomsk State University, 36 Lenin Avenue, 634050, Tomsk, Russian Federation
| | - Julia Kzhyshkowska
- National Research Tomsk State University, 36 Lenin Avenue, 634050, Tomsk, Russian Federation.,University of Heidelberg, 1-3 Theodor-Kutzer Ufer, 68167, Mannheim, Germany
| | - Sergey V Popov
- Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk 111a Kievskaya Street, 634012, Tomsk, Russian Federation
| | - Rostislav S Karpov
- Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk 111a Kievskaya Street, 634012, Tomsk, Russian Federation.,Siberian State Medical University, 2 Moscovsky trakt, 634055, Tomsk, Russian Federation
| | - Vyacheslav Ryabov
- Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk 111a Kievskaya Street, 634012, Tomsk, Russian Federation.,National Research Tomsk State University, 36 Lenin Avenue, 634050, Tomsk, Russian Federation.,Siberian State Medical University, 2 Moscovsky trakt, 634055, Tomsk, Russian Federation
| |
Collapse
|
29
|
Correction: Cardiosphere-Derived Cells Facilitate Heart Repair by Modulating M1/M2 Macrophage Polarization and Neutrophil Recruitment. PLoS One 2017; 12:e0171892. [PMID: 28166288 PMCID: PMC5293208 DOI: 10.1371/journal.pone.0171892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
|