1
|
Yu L, Cai S, Guo X. m6A RNA methylation modification is involved in the disease course of heart failure. Biotechnol Genet Eng Rev 2024; 40:961-975. [PMID: 36943073 DOI: 10.1080/02648725.2023.2191086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 03/08/2023] [Indexed: 03/23/2023]
Abstract
We explored N6-methyladenosine (m6A) RNA methylation as one of the gene regulatory mechanisms in heart failure (HF) biology. Understanding the different physiological mechanisms will facilitate the prevention and individualized treatment of HF. The Gene Expression Omnibus (GEO) database served as the source of the data. In GSE116250, differential analysis between ischemic cardiomyopathy (ICM), dilated cardiomyopathy (DCM) and controls yielded differentially expressed m6A regulators. Differential analysis between HF and controls in GSE131296 identifies m6A-modified genes and then performs enrichment analysis. Protein-protein interaction (PPI) network analysis was performed for the differentially expressed ICM- or DCM-associated genes in GSE116250 and GSE55296, respectively. Finally, the diagnostic genes for ICM and DCM were predicted using receiver operating characteristic (ROC) curve. YTHDC1, HNRNPC and HNRNPA2B1 were significantly downregulated in GSE116250 in DCM and ICM compared with controls. A total of 195 genes were identified in GSE131296 as subject to m6A alteration. These genes may play a role in HF through the MAPK signaling pathway and p53 signaling pathway. PPI network analysis identified CCL5, CXCR4 and CCL2 as key genes for ICM and IL-6 as a key gene for DCM. Through ROC curves, we identified m6A-modified APLP1, KLF2 as potential diagnostic genes for ICM, and m6A-modified FGF7, FREM1 and C14orf132 as potential diagnostic genes for DCM. Our findings support m6A modifying mechanisms in HF etiology that contribute to the treatment of HF. Thus, our data suggest that m6A methylation may be an interesting target for therapeutic intervention.
Collapse
Affiliation(s)
- Liyan Yu
- Department of gerontology, Yantaishan Hospital, Yantai, Shandong, China
| | - Shuxia Cai
- Department of gerontology, Yantaishan Hospital, Yantai, Shandong, China
| | - Xiuli Guo
- Department of gerontology, Yantaishan Hospital, Yantai, Shandong, China
| |
Collapse
|
2
|
Lee W, Lin SL, Chiang CS, Chen JY, Chieng WW, Huang SR, Chang TY, Linju Yen B, Hung MC, Chang KC, Lee HT, Jeng LB, Shyu WC. Role of HIF-1α-Activated IL-22/IL-22R1/Bmi1 Signaling Modulates the Self-Renewal of Cardiac Stem Cells in Acute Myocardial Ischemia. Stem Cell Rev Rep 2024:10.1007/s12015-024-10774-8. [PMID: 39264501 DOI: 10.1007/s12015-024-10774-8] [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] [Accepted: 08/06/2024] [Indexed: 09/13/2024]
Abstract
Impaired tissue regeneration negatively impacts on left ventricular (LV) function and remodeling after acute myocardial infarction (AMI). Little is known about the intrinsic regulatory machinery of ischemia-induced endogenous cardiac stem cells (eCSCs) self-renewing divisions after AMI. The interleukin 22 (IL-22)/IL-22 receptor 1 (IL-22R1) pathway has emerged as an important regulator of several cellular processes, including the self-renewal and proliferation of stem cells. However, whether the hypoxic environment could trigger the self-renewal of eCSCs via IL-22/IL-22R1 activation remains unknown. In this study, the upregulation of IL-22R1 occurred due to activation of hypoxia-inducible factor-1α (HIF-1α) under hypoxic and ischemic conditions. Systemic IL-22 administration not only attenuated cardiac remodeling, inflammatory responses, but also promoted eCSC-mediated cardiac repair after AMI. Unbiased RNA microarray analysis showed that the downstream mediator Bmi1 regulated the activation of CSCs. Therefore, the HIF-1α-induced IL-22/IL-22R1/Bmi1 cascade can modulate the proliferation and activation of eCSCs in vitro and in vivo. Collectively, investigating the HIF-1α-activated IL-22/IL-22R1/Bmi1 signaling pathway might offer a new therapeutic strategy for AMI via eCSC-induced cardiac repair.
Collapse
Affiliation(s)
- Wei Lee
- Cell Therapy Center, China Medical University Hospital (CMUH), Taichung, 404, Taiwan
| | - Syuan-Ling Lin
- Translational Medicine Research Center, CMUH, Taichung, 404, Taiwan
| | - Chih-Sheng Chiang
- Cell Therapy Center, China Medical University Hospital (CMUH), Taichung, 404, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University (CMU), Taichung, 404, Taiwan
- Neuroscience and Brain Disease Center and New Drug Development Center, CMU, Taichung, 404, Taiwan
| | - Jui-Yu Chen
- Translational Medicine Research Center, CMUH, Taichung, 404, Taiwan
| | - Wee-Wei Chieng
- Translational Medicine Research Center, CMUH, Taichung, 404, Taiwan
| | - Shu-Rou Huang
- Translational Medicine Research Center, CMUH, Taichung, 404, Taiwan
| | - Ting-Yu Chang
- Cell Therapy Center, China Medical University Hospital (CMUH), Taichung, 404, Taiwan
| | - B Linju Yen
- Regenerative Medicine Research Group, Institute of Cellular and System Medicine, National Health Research Institutes (NHRI), Zhunan, 350, Taiwan
| | - Mien-Chie Hung
- Graduate Institute of Biomedical Sciences and Research Centers for Cancer Biology and Molecular Medicine, CMU, Taichung, 404, Taiwan
| | - Kuan-Cheng Chang
- Division of Cardiovascular Medicine, Department of Medicine, CMUH, Taichung, 404, Taiwan
- School of Medicine, CMU, Taichung, 404, Taiwan
| | - Hsu-Tung Lee
- Department of Neurosurgery, Taichung Veterans General Hospital, Taichung, 404, Taiwan
| | - Long-Bin Jeng
- Cell Therapy Center, China Medical University Hospital (CMUH), Taichung, 404, Taiwan
- Organ Transplantation Center, CMUH, Taichung, 404, Taiwan
| | - Woei-Cherng Shyu
- Translational Medicine Research Center, CMUH, Taichung, 404, Taiwan.
- Graduate Institute of Biomedical Sciences, China Medical University (CMU), Taichung, 404, Taiwan.
- Neuroscience and Brain Disease Center and New Drug Development Center, CMU, Taichung, 404, Taiwan.
- Department of Neurology, CMUH, Taichung, 404, Taiwan.
- Department of Occupational Therapy, Asia University, No. 2, Yude Rd., North Dist, Taichung City, 404332, Taiwan.
| |
Collapse
|
3
|
Hilgendorf I, Frantz S, Frangogiannis NG. Repair of the Infarcted Heart: Cellular Effectors, Molecular Mechanisms and Therapeutic Opportunities. Circ Res 2024; 134:1718-1751. [PMID: 38843294 PMCID: PMC11164543 DOI: 10.1161/circresaha.124.323658] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 05/08/2024] [Indexed: 06/12/2024]
Abstract
The adult mammalian heart has limited endogenous regenerative capacity and heals through the activation of inflammatory and fibrogenic cascades that ultimately result in the formation of a scar. After infarction, massive cardiomyocyte death releases a broad range of damage-associated molecular patterns that initiate both myocardial and systemic inflammatory responses. TLRs (toll-like receptors) and NLRs (NOD-like receptors) recognize damage-associated molecular patterns (DAMPs) and transduce downstream proinflammatory signals, leading to upregulation of cytokines (such as interleukin-1, TNF-α [tumor necrosis factor-α], and interleukin-6) and chemokines (such as CCL2 [CC chemokine ligand 2]) and recruitment of neutrophils, monocytes, and lymphocytes. Expansion and diversification of cardiac macrophages in the infarcted heart play a major role in the clearance of the infarct from dead cells and the subsequent stimulation of reparative pathways. Efferocytosis triggers the induction and release of anti-inflammatory mediators that restrain the inflammatory reaction and set the stage for the activation of reparative fibroblasts and vascular cells. Growth factor-mediated pathways, neurohumoral cascades, and matricellular proteins deposited in the provisional matrix stimulate fibroblast activation and proliferation and myofibroblast conversion. Deposition of a well-organized collagen-based extracellular matrix network protects the heart from catastrophic rupture and attenuates ventricular dilation. Scar maturation requires stimulation of endogenous signals that inhibit fibroblast activity and prevent excessive fibrosis. Moreover, in the mature scar, infarct neovessels acquire a mural cell coat that contributes to the stabilization of the microvascular network. Excessive, prolonged, or dysregulated inflammatory or fibrogenic cascades accentuate adverse remodeling and dysfunction. Moreover, inflammatory leukocytes and fibroblasts can contribute to arrhythmogenesis. Inflammatory and fibrogenic pathways may be promising therapeutic targets to attenuate heart failure progression and inhibit arrhythmia generation in patients surviving myocardial infarction.
Collapse
Affiliation(s)
- Ingo Hilgendorf
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen and Faculty of Medicine at the University of Freiburg, Freiburg, Germany
| | - Stefan Frantz
- Medizinische Klinik und Poliklinik I, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Nikolaos G Frangogiannis
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx NY
| |
Collapse
|
4
|
Bhave S, Swain L, Qiao X, Martin G, Aryaputra T, Everett K, Kapur NK. ALK1 Deficiency Impairs the Wound-Healing Process and Increases Mortality in Murine Model of Myocardial Infarction. J Cardiovasc Transl Res 2024; 17:496-504. [PMID: 38064044 DOI: 10.1007/s12265-023-10471-w] [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: 07/31/2023] [Accepted: 11/24/2023] [Indexed: 07/03/2024]
Abstract
The functional role of TGFβ type I receptor, activin-like kinase (ALK)-1 in post-myocardial infarction (MI) cardiac remodeling is unknown. We hypothesize that reduced ALK1 activity reduces survival and promotes cardiac fibrosis after MI. MI was induced in wild-type (WT), and ALK+/- mice by left coronary ligation. After 14 days ALK1+/- mice had reduced survival with a higher rate of cardiac rupture compared to WT mice. ALK1+/- left ventricles (LVs) had increased volumes at the end of systole and at the end of diastole. After MI ALK1+/- LVs had increased profibrotic SMAD3 signaling, type 1 collagen, and fibrosis as well as increased levels of TGFβ1 co-receptor, endoglin, VEGF, and ALK1 ligands BMP9 and BMP10. ALK1+/- LVs had decreased levels of stromal-derived factor 1α. These data identify the critical role of ALK1 in post-MI survival and cardiac remodeling and implicate ALK1 as a potential therapeutic target to improve survival after MI.
Collapse
Affiliation(s)
- Shreyas Bhave
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, 02111, USA
| | - Lija Swain
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, 02111, USA
| | - Xiaoying Qiao
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, 02111, USA
| | - Gregory Martin
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, 02111, USA
| | - Tejasvi Aryaputra
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, 02111, USA
| | - Kay Everett
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, 02111, USA
| | - Navin K Kapur
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, 02111, USA.
| |
Collapse
|
5
|
Chikata Y, Iwata H, Miyosawa K, Naito R, Koike T, Moriya S, Yasuda H, Funamizu T, Doi S, Endo H, Wada H, Ogita M, Dohi T, Kasai T, Isoda K, Okazaki S, Miyauchi K, Minamino T. Elevated levels of plasma inactive stromal cell derived factor-1α predict poor long-term outcomes in diabetic patients following percutaneous coronary intervention. Cardiovasc Diabetol 2024; 23:114. [PMID: 38555431 PMCID: PMC10981820 DOI: 10.1186/s12933-024-02197-z] [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: 10/26/2023] [Accepted: 03/12/2024] [Indexed: 04/02/2024] Open
Abstract
BACKGROUND Since the complication of diabetes mellitus (DM) is a risk for adverse cardiovascular outcomes in patients with coronary artery disease (CAD), appropriate risk estimation is needed in diabetic patients following percutaneous coronary intervention (PCI). However, there is no useful biomarker to predict outcomes in this population. Although stromal cell derived factor-1α (SDF-1α), a circulating chemokine, was shown to have cardioprotective roles, the prognostic impact of SDF-1α in diabetic patients with CAD is yet to be fully elucidated. Moreover, roles of SDF-1α isoforms in outcome prediction remain unclear. Therefore, this study aimed to assess the prognostic implication of three forms of SDF-1α including total, active, and inactive forms of SDF-1α in patients with DM and after PCI. METHODS This single-center retrospective analysis involved consecutive patients with diabetes who underwent PCI for the first time between 2008 and 2018 (n = 849). Primary and secondary outcome measures were all-cause death and the composite of cardiovascular death, non-fatal myocardial infarction, and ischemic stroke (3P-MACE), respectively. For determining plasma levels of SDF-1α, we measured not only total, but also the active type of SDF-1α by ELISA. Inactive isoform of the SDF-1α was calculated by subtracting the active isoform from total SDF-1α. RESULTS Unadjusted Kaplan-Meier analyses revealed increased risk of both all-cause death and 3P-MACE in patients with elevated levels of inactive SDF-1α. However, plasma levels of total and active SDF-1α were not associated with cumulative incidences of outcome measures. Multivariate Cox hazard analyses repeatedly indicated the 1 higher log-transformed inactive SDF-1α was significantly associated with increased risk of all-cause death (hazard ratio (HR): 2.64, 95% confidence interval (CI): 1.28-5.34, p = 0.008) and 3P-MACE (HR: 2.51, 95% CI: 1.12-5.46, p = 0.02). Moreover, the predictive performance of inactive SDF-1α was higher than that of total SDF-1α (C-statistics of inactive and total SDF-1α for all-cause death: 0.631 vs 0.554, for 3P-MACE: 0.623 vs 0.524, respectively). CONCLUSION The study results indicate that elevated levels of plasma inactive SDF-1α might be a useful indicator of poor long-term outcomes in diabetic patients following PCI. TRIAL REGISTRATION This study describes a retrospective analysis of a prospective registry database of patients who underwent PCI at Juntendo University Hospital, Tokyo, Japan (Juntendo Physicians' Alliance for Clinical Trials, J-PACT), which is publicly registered (University Medical Information Network Japan-Clinical Trials Registry, UMIN-CTR 000035587).
Collapse
Affiliation(s)
- Yuichi Chikata
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo, Japan
| | - Hiroshi Iwata
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo, Japan.
| | | | - Ryo Naito
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo, Japan
| | - Takuma Koike
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo, Japan
| | - Soshi Moriya
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo, Japan
| | - Hidetoshi Yasuda
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo, Japan
| | - Takehiro Funamizu
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo, Japan
| | - Shinichiro Doi
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo, Japan
| | - Hirohisa Endo
- Department of Cardiology, Juntendo University Shizuoka Hospital, Shizuoka, Japan
| | - Hideki Wada
- Department of Cardiology, Juntendo University Shizuoka Hospital, Shizuoka, Japan
| | - Manabu Ogita
- Department of Cardiology, Juntendo University Shizuoka Hospital, Shizuoka, Japan
| | - Tomotaka Dohi
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo, Japan
| | - Takatoshi Kasai
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo, Japan
| | - Kikuo Isoda
- Department of Cardiology, Juntendo University Nerima Hospital, Tokyo, Japan
| | - Shinya Okazaki
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo, Japan
| | - Katsumi Miyauchi
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo, Japan
| | - Tohru Minamino
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo, Japan
| |
Collapse
|
6
|
Wang Y, Liu Y, Li X, Yao L, Mbadhi M, Chen S, Lv Y, Bao X, Chen L, Chen S, Zhang J, Wu Y, Lv J, Shi L, Tang J. Vagus nerve stimulation-induced stromal cell-derived factor-l alpha participates in angiogenesis and repair of infarcted hearts. ESC Heart Fail 2023; 10:3311-3329. [PMID: 37641543 PMCID: PMC10682864 DOI: 10.1002/ehf2.14475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 06/02/2023] [Accepted: 07/02/2023] [Indexed: 08/31/2023] Open
Abstract
AIMS We aim to explore the role and mechanism of vagus nerve stimulation (VNS) in coronary endothelial cells and angiogenesis in infarcted hearts. METHODS AND RESULTS Seven days after rat myocardial infarction (MI) was prepared by ligation of the left anterior descending coronary artery, the left cervical vagus nerve was treated with electrical stimulation 1 h after intraperitoneal administration of the α7-nicotinic acetylcholine inhibitor mecamylamine or the mAChR inhibitor atropine or 3 days after local injection of Ad-shSDF-1α into the infarcted heart. Cardiac tissue acetylcholine (ACh) and serum ACh, tumour necrosis factor α (TNF-α), interleukin 1β (IL-1β) and interleukin 6 (IL-6) levels were detected by ELISA to determine whether VNS was successful. An inflammatory injury model in human coronary artery endothelial cells (HCAECs) was established by lipopolysaccharide and identified by evaluating TNF-α, IL-1β and IL-6 levels and tube formation. Immunohistochemistry staining was performed to evaluate CD31-positive vessel density and stromal cell-derived factor-l alpha (SDF-1α) expression in the MI heart in vivo and the expression and distribution of SDF-1α, C-X-C motif chemokine receptor 4 and CXCR7 in HCAECs in vitro. Western blotting was used to detect the levels of SDF-1α, V-akt murine thymoma viral oncogene homolog (AKT), phosphorylated AKT (pAKT), specificity protein 1 (Sp1) and phosphorylation of Sp1 in HCAECs. Left ventricular performance, including left ventricular systolic pressure, left ventricular end-diastolic pressure and rate of the rise and fall of ventricular pressure, should be evaluated 28 days after VNS treatment. VNS was successfully established for MI therapy with decreases in serum TNF-α, IL-1β and IL-6 levels and increases in cardiac tissue and serum ACh levels, leading to increased SDF-1α expression in coronary endothelial cells of MI hearts, triggering angiogenesis of MI hearts with increased CD31-positive vessel density, which was abolished by the m/nAChR inhibitors mecamylamine and atropine or knockdown of SDF-1α by shRNA. ACh promoted SDF-1α expression and its distribution along with the branch of the formed tube in HCAECs, resulting in an increase in the number of tubes formed in HCAECs. ACh increased the levels of pAKT and phosphorylation of Sp1 in HCAECs, resulting in inducing SDF-1α expression, and the specific effects could be abolished by mecamylamine, atropine, the PI3K/AKT blocker wortmannin or the Sp1 blocker mithramycin. Functionally, VNS improved left ventricular performance, which could be abolished by Ad-shSDF-1α. CONCLUSIONS VNS promoted angiogenesis to repair the infarcted heart by inducing SDF-1α expression and redistribution along new branches during angiogenesis, which was associated with the m/nAChR-AKT-Sp1 signalling pathway.
Collapse
Affiliation(s)
- Yan Wang
- Department of Physiology, Faculty of Basic Medical Sciences, Hubei Key Laboratory of Embryonic Stem Cell ResearchHubei University of MedicineShiyanPR China
- Department of Pathology, Renmin HospitalHubei University of MedicineShiyanPR China
| | - Yun Liu
- Department of Physiology, Faculty of Basic Medical Sciences, Hubei Key Laboratory of Embryonic Stem Cell ResearchHubei University of MedicineShiyanPR China
| | - Xing‐yuan Li
- Department of Physiology, Faculty of Basic Medical Sciences, Hubei Key Laboratory of Embryonic Stem Cell ResearchHubei University of MedicineShiyanPR China
| | - Lu‐yuan Yao
- Department of Physiology, Faculty of Basic Medical Sciences, Hubei Key Laboratory of Embryonic Stem Cell ResearchHubei University of MedicineShiyanPR China
- Department of Anesthesiology, Institute of Anesthesiology, Taihe HospitalHubei University of MedicineShiyanPR China
| | - MagdaleenaNaemi Mbadhi
- Department of Physiology, Faculty of Basic Medical Sciences, Hubei Key Laboratory of Embryonic Stem Cell ResearchHubei University of MedicineShiyanPR China
| | - Shao‐Juan Chen
- Department of Physiology, Faculty of Basic Medical Sciences, Hubei Key Laboratory of Embryonic Stem Cell ResearchHubei University of MedicineShiyanPR China
- Department of Stomatology, Taihe HospitalHubei University of MedicineShiyanPR China
| | - Yan‐xia Lv
- Department of Physiology, Faculty of Basic Medical Sciences, Hubei Key Laboratory of Embryonic Stem Cell ResearchHubei University of MedicineShiyanPR China
| | - Xin Bao
- Department of Physiology, Faculty of Basic Medical Sciences, Hubei Key Laboratory of Embryonic Stem Cell ResearchHubei University of MedicineShiyanPR China
- Experimental Medical Center, Guoyao‐Dong Feng HospitalHubei University of MedicineShiyanPR China
| | - Long Chen
- Experimental Medical Center, Guoyao‐Dong Feng HospitalHubei University of MedicineShiyanPR China
| | - Shi‐You Chen
- Department of SurgeryUniversity of MissouriColumbiaMissouriUSA
| | - Jing‐xuan Zhang
- Department of Physiology, Faculty of Basic Medical Sciences, Hubei Key Laboratory of Embryonic Stem Cell ResearchHubei University of MedicineShiyanPR China
- Institute of Basic Medical Sciences, Institute of BiomedicineHubei University of MedicineShiyanPR China
| | - Yan Wu
- Department of Physiology, Faculty of Basic Medical Sciences, Hubei Key Laboratory of Embryonic Stem Cell ResearchHubei University of MedicineShiyanPR China
- Institute of Basic Medical Sciences, Institute of BiomedicineHubei University of MedicineShiyanPR China
| | - Jing Lv
- Department of Anesthesiology, Institute of Anesthesiology, Taihe HospitalHubei University of MedicineShiyanPR China
| | - Liu‐liu Shi
- Department of Physiology, Faculty of Basic Medical Sciences, Hubei Key Laboratory of Embryonic Stem Cell ResearchHubei University of MedicineShiyanPR China
- Institute of Basic Medical Sciences, Institute of BiomedicineHubei University of MedicineShiyanPR China
| | - Jun‐ming Tang
- Department of Physiology, Faculty of Basic Medical Sciences, Hubei Key Laboratory of Embryonic Stem Cell ResearchHubei University of MedicineShiyanPR China
- Institute of Basic Medical Sciences, Institute of BiomedicineHubei University of MedicineShiyanPR China
| |
Collapse
|
7
|
Wang Z, Xu H, Chen M, Lu Y, Zheng L, Ma L. CCL24/CCR3 axis plays a central role in angiotensin II-induced heart failure by stimulating M2 macrophage polarization and fibroblast activation. Cell Biol Toxicol 2023; 39:1413-1431. [PMID: 36131165 PMCID: PMC10425496 DOI: 10.1007/s10565-022-09767-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 08/26/2022] [Indexed: 11/02/2022]
Abstract
AIMS We aimed to investigate the effect and mechanism of pleiotropic chemokine CCL24 in heart failure. METHODS AND RESULTS Compared with normal donators, the expression of CCL24 and number of cardiac M2 macrophages in heart were higher in heart failure patients, the same as plasma CCL24. Treatment with CCL24 antibody hindered Ang II (1500 ng/kg/min)-induced cardiac adverse remodeling through preventing cardiac hypertrophy and fibrosis. RNA-seq showed that CCL24/CCR3 axis was involved in immune and inflammatory responses. Single-cell analysis of cytometry by time of flight (CyTOF) revealed that CCL24 antibody decreased the M2 macrophage and monocyte polarization during Ang II stimulation. Immunofluorescence co-localization analysis confirmed the expression of CCR3 in macrophage and fibroblasts. Then, in vitro experiments confirmed that CCL24/CCR3 axis was also involved in cardiac primary fibroblast activation through its G protein-coupled receptor function. CONCLUSION CCL24/CCR3 axis plays a crucial part in cardiac remodeling by stimulating M2 macrophage polarization and cardiac fibroblast activation. Cardiac M2 macrophages, CCL24 and circulation CCL24 increased in heart failure patients. Treatment with CCL24 Ab hindered Ang II induced cardiac structural dysfunction and electrical remodeling. In CCL24 Ab group RNA-seq found that it was related to immune responses and hypertrophic cardiomyopathy, CytoF revealed M2 macrophages and monocytes decreased obviously. In vitro,CCL24 promoted activation and migration of cardiac fibroblast.
Collapse
Affiliation(s)
- Zhen Wang
- Department of Cardiothoracic Surgery, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang, China
| | - Hongfei Xu
- Department of Cardiothoracic Surgery, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang, China
| | - Miao Chen
- Department of Cardiothoracic Surgery, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang, China
| | - Yunlong Lu
- Department of Cardiology, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang, China
| | - Liangrong Zheng
- Department of Cardiology, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang, China.
| | - Liang Ma
- Department of Cardiothoracic Surgery, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang, China.
| |
Collapse
|
8
|
Yu B, Li H, Zhang Z, Chen P, Wang L, Fan X, Ning X, Pan Y, Zhou F, Hu X, Chang J, Ou C. Extracellular vesicles engineering by silicates-activated endothelial progenitor cells for myocardial infarction treatment in male mice. Nat Commun 2023; 14:2094. [PMID: 37055411 PMCID: PMC10102163 DOI: 10.1038/s41467-023-37832-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 04/03/2023] [Indexed: 04/15/2023] Open
Abstract
Extracellular vesicles have shown good potential in disease treatments including ischemic injury such as myocardial infarction. However, the efficient production of highly active extracellular vesicles is one of the critical limitations for their clinical applications. Here, we demonstrate a biomaterial-based approach to prepare high amounts of extracellular vesicles with high bioactivity from endothelial progenitor cells (EPCs) by stimulation with silicate ions derived from bioactive silicate ceramics. We further show that hydrogel microspheres containing engineered extracellular vesicles are highly effective in the treatment of myocardial infarction in male mice by significantly enhancing angiogenesis. This therapeutic effect is attributed to significantly enhanced revascularization by the high content of miR-126a-3p and angiogenic factors such as VEGF and SDF-1, CXCR4 and eNOS in engineered extracellular vesicles, which not only activate endothelial cells but also recruit EPCs from the circulatory system.
Collapse
Affiliation(s)
- Bin Yu
- The 10th Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Southern Medical University, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, 510280, Guangzhou, China
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, 510280, Guangzhou, China
| | - Hekai Li
- Department of Cardiology and Laboratory of Heart Center, Zhujiang Hospital, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Southern Medical University, 510515, Guangzhou, China
| | - Zhaowenbin Zhang
- Wenzhou Institute, Zhejiang Engineering Research Center for Tissue Repair Materials, University of Chinese Academy of Sciences, 325000, Wenzhou, China
- State Key Laboratory of High-Performance Ceramics and Super fine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, 200050, Shanghai, People's Republic of China
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, 325000, Wenzhou, China
| | - Peier Chen
- Department of Cardiology and Laboratory of Heart Center, Zhujiang Hospital, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Southern Medical University, 510515, Guangzhou, China
| | - Ling Wang
- School of Biomedical Engineering, Biomaterials Research Center, Southern Medical University, 510515, Guangzhou, People's Republic of China
| | - Xianglin Fan
- Department of Cardiology and Laboratory of Heart Center, Zhujiang Hospital, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Southern Medical University, 510515, Guangzhou, China
| | - Xiaodong Ning
- The 10th Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Southern Medical University, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, 510280, Guangzhou, China
| | - Yuxuan Pan
- The 10th Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Southern Medical University, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, 510280, Guangzhou, China
| | - Feiran Zhou
- Department of Cardiology and Laboratory of Heart Center, Zhujiang Hospital, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Southern Medical University, 510515, Guangzhou, China
| | - Xinyi Hu
- Department of Cardiology and Laboratory of Heart Center, Zhujiang Hospital, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Southern Medical University, 510515, Guangzhou, China
| | - Jiang Chang
- The 10th Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Southern Medical University, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, 510280, Guangzhou, China.
- Wenzhou Institute, Zhejiang Engineering Research Center for Tissue Repair Materials, University of Chinese Academy of Sciences, 325000, Wenzhou, China.
- State Key Laboratory of High-Performance Ceramics and Super fine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, 200050, Shanghai, People's Republic of China.
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, 325000, Wenzhou, China.
| | - Caiwen Ou
- The 10th Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Southern Medical University, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, 510280, Guangzhou, China.
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, 510280, Guangzhou, China.
- Department of Cardiology and Laboratory of Heart Center, Zhujiang Hospital, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Southern Medical University, 510515, Guangzhou, China.
| |
Collapse
|
9
|
Romeo FJ, Mavropoulos SA, Ishikawa K. Progress in Clinical Gene Therapy for Cardiac Disorders. Mol Diagn Ther 2023; 27:179-191. [PMID: 36641770 PMCID: PMC10023344 DOI: 10.1007/s40291-022-00632-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/04/2022] [Indexed: 01/16/2023]
Abstract
Despite significant advances in novel treatments and approaches, cardiovascular disease remains the leading cause of death globally. Gene therapy is a promising option for many diseases, including cardiovascular diseases. In the last 30 years, gene therapy has slowly proceeded towards clinical translation and recently reached US Food and Drug Administration approval for several diseases such as Leber congenital amaurosis and spinal muscular atrophy, among others. Previous attempts at developing gene therapies for cardiovascular diseases have yielded promising results in preclinical studies and early-phase clinical trials. However, larger trials failed to demonstrate consistent benefits in patients with ischemic heart disease and heart failure. In this review, we summarize the history and current status of clinical cardiac gene therapy. Starting with angiogenic gene therapy, we also cover more recent gene therapy trials for heart failure and cardiomyopathies. New programs are actively vying to be the first to get Food and Drug Administration approval for a cardiac gene therapy product by taking advantage of novel techniques.
Collapse
Affiliation(s)
- Francisco J Romeo
- Icahn School of Medicine at Mount Sinai, Cardiovascular Research Institute, 1 Gustave L. Levy Place, Box 1014, New York, NY, 10029, USA
| | - Spyros A Mavropoulos
- Icahn School of Medicine at Mount Sinai, Cardiovascular Research Institute, 1 Gustave L. Levy Place, Box 1014, New York, NY, 10029, USA
| | - Kiyotake Ishikawa
- Icahn School of Medicine at Mount Sinai, Cardiovascular Research Institute, 1 Gustave L. Levy Place, Box 1014, New York, NY, 10029, USA.
| |
Collapse
|
10
|
SDF-1α-Releasing Microspheres Effectively Extend Stem Cell Homing after Myocardial Infarction. Biomedicines 2023; 11:biomedicines11020343. [PMID: 36830880 PMCID: PMC9953248 DOI: 10.3390/biomedicines11020343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 01/20/2023] [Accepted: 01/24/2023] [Indexed: 01/27/2023] Open
Abstract
Ischemic heart disease (IHD) is one of the main focuses in today's healthcare due to its implications and complications, and it is predicted to be increasing in prevalence due to the ageing population. Although the conventional pharmacological and interventional methods for the treatment of IHD presents with success in the clinical setting, the long-term complications of cardiac insufficiency are on a continual incline as a result of post-infarction remodeling of the cardiac tissue. The migration and involvement of stem cells to the cardiac muscle, followed by differentiation into cardiac myocytes, has been proven to be the natural process, though at a slow rate. SDF-1α is a novel candidate to mobilize stem cells homing to the ischemic heart. Endogenous SDF-1α levels are elevated after myocardial infarction, but their presence gradually decreases after approximately seven days. Additional administration of SDF-1α-releasing microspheres could be a tool for the extension of the time the stem cells are in the cardiac tissue after myocardial infarction. This, in turn, could constitute a novel therapy for more efficient regeneration of the heart muscle after injury. Through this practical study, it has been shown that the controlled release of SDF-1α from biodegradable microspheres into the pericardial sac fourteen days after myocardial infarction increases the concentration of exogenous SDF-1α, which persists in the tissue much longer than the level of endogenous SDF-1α. In addition, administration of SDF-1α-releasing microspheres increased the expression of the factors potentially involved in the involvement and retention of myocardial stem cells, which constitutes vascular endothelial growth factor A (VEGFA), stem cell factor (SCF), and vascular cell adhesion molecules (VCAMs) at the site of damaged tissue. This exhibits the possibility of combating the basic limitations of cell therapy, including ineffective stem cell implantation and the ability to induce the migration of endogenous stem cells to the ischemic cardiac tissue and promote heart repair.
Collapse
|
11
|
Xue K, Chen S, Chai J, Yan W, Zhu X, Ji D, Wu Y, Liu H, Wang W. Nitration of cAMP-Response Element Binding Protein Participates in Myocardial Infarction-Induced Myocardial Fibrosis via Accelerating Transcription of Col1a2 and Cxcl12. Antioxid Redox Signal 2023; 38:709-730. [PMID: 36324232 DOI: 10.1089/ars.2021.0273] [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] [Indexed: 11/06/2022]
Abstract
Aims: Myocardial fibrosis after myocardial infarction (MI) leads to heart failure. Nitration of protein can alter its function. cAMP-response element binding protein (CREB) is a key transcription factor involved in fibrosis. However, little is known about the role of nitrated CREB in MI-induced myocardial fibrosis. Meanwhile, downstream genes of transcription factor CREB in myocardial fibrosis have not been identified. This study aims to verify the hypothesis that nitrated CREB promotes MI-induced myocardial fibrosis via regulating the transcription of Col1a2 and Cxcl12. Results: Our study showed that (1) the level of nitrative stress was elevated and nitrated CREB was higher in the myocardium after MI. Tyr182, 307, and 336 were the nitration sites of CREB; (2) with the administration of peroxynitrite (ONOO-) scavengers, CREB phosphorylation, nuclear translocation, and binding activity to TORC2 (transducers of regulated CREB-2) were attenuated; (3) the expressions of extracellular matrix (ECM) proteins were upregulated and downregulated in accordance with the expression alteration of CREB both in vitro and in vivo; (4) CREB accelerated transcription of Col1a2 and Cxcl12 after MI directly. With the administration of ONOO- scavengers, ECM protein expressions were attenuated; meanwhile, the messenger RNA (mRNA) levels of Col1a2 and Cxcl12 were alleviated as well. Innovation and Conclusion: Nitration of transcription factor CREB participates in MI-induced myocardial fibrosis through enhancing its phosphorylation, nuclear translocation, and binding activity to TORCs, among which CREB transcripts Col1a2 and Cxcl12 directly. These data indicated that nitrated CREB might be a potential therapeutic target against MI-induced myocardial fibrosis.
Collapse
Affiliation(s)
- Ke Xue
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.,Beijing Key Laboratory for Metabolic Disorder-Related Cardiovascular Diseases, Beijing, China.,Department of Pathology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Shuai Chen
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.,Beijing Key Laboratory for Metabolic Disorder-Related Cardiovascular Diseases, Beijing, China
| | - Jiayin Chai
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.,Beijing Key Laboratory for Metabolic Disorder-Related Cardiovascular Diseases, Beijing, China
| | - Wenjing Yan
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.,Beijing Key Laboratory for Metabolic Disorder-Related Cardiovascular Diseases, Beijing, China
| | - Xinyu Zhu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.,Beijing Key Laboratory for Metabolic Disorder-Related Cardiovascular Diseases, Beijing, China
| | - Dengyu Ji
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.,Beijing Key Laboratory for Metabolic Disorder-Related Cardiovascular Diseases, Beijing, China
| | - Ye Wu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.,Beijing Key Laboratory for Metabolic Disorder-Related Cardiovascular Diseases, Beijing, China
| | - Huirong Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.,Beijing Key Laboratory for Metabolic Disorder-Related Cardiovascular Diseases, Beijing, China
| | - Wen Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.,Beijing Key Laboratory for Metabolic Disorder-Related Cardiovascular Diseases, Beijing, China
| |
Collapse
|
12
|
Cheng M, Chen C, Yu K, Lv X, Zeng Q, Dong N, Zhu F. Ablation of CXCR4 expression in cardiomyocytes exacerbates isoproterenol‑induced cell death and heart failure. Int J Mol Med 2022; 51:13. [PMID: 36579657 PMCID: PMC9869727 DOI: 10.3892/ijmm.2022.5216] [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: 07/11/2022] [Accepted: 11/21/2022] [Indexed: 12/30/2022] Open
Abstract
CXCR4 is a seven‑transmembrane‑spanning Gi‑coupled receptor for the SDF‑1 chemokine and plays a critical role in cardiovascular development and post‑injury repair. However, the specific role of CXCR4 in cardiomyocytes is incompletely understood. It was hypothesized that CXCR4 activation in cardiomyocytes antagonizes β‑adrenoceptor/Gs signaling‑induced cardiac dysfunction. Cardiomyocyte‑specific CXCR4 knockout (CXCR4‑CMKO) mice were generated by crossing CXCR4fl/fl and MHC‑Cre+/‑ mice. Their cardiac structure and function in the basal state are equivalent to that of the control MHC‑Cre+/‑ littermates until at least 4 months old. However, following continuous subcutaneous administration of isoproterenol (Iso) via an osmotic mini‑pump, the ventricular myocardial contractility, dilation, cardiomyocyte apoptosis, and interstitial fibrosis are worse in CXCR4‑CMKO mice than in MHC‑Cre+/‑ littermates. In the cultured H9C2 cardiomyocytes, SDF‑1 treatment markedly attenuated Iso‑induced apoptosis and reduction in phospho‑Akt, and this protective effect was lost by knockdown of CXCR4 or by co‑treatment with Gi inhibitors. In conclusion, CXCR4 promotes cardiomyocyte survival and heart function during β‑adrenergic stress.
Collapse
Affiliation(s)
- Min Cheng
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China,Correspondence to: Dr Min Cheng or Dr Feng Zhu, Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1377 Jiefang Avenue, Wuhan, Hubei 430022, P.R. China, E-mail: , E-mail:
| | - Can Chen
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Kunwu Yu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Xiao Lv
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Qiutang Zeng
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Nianguo Dong
- Department of Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Feng Zhu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China,Correspondence to: Dr Min Cheng or Dr Feng Zhu, Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1377 Jiefang Avenue, Wuhan, Hubei 430022, P.R. China, E-mail: , E-mail:
| |
Collapse
|
13
|
Xin L, Zheng X, Chen J, Hu S, Luo Y, Ge Q, Jin X, Ma L, Zhang S. An Acellular Scaffold Facilitates Endometrial Regeneration and Fertility Restoration via Recruiting Endogenous Mesenchymal Stem Cells. Adv Healthc Mater 2022; 11:e2201680. [PMID: 36049781 DOI: 10.1002/adhm.202201680] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 08/25/2022] [Indexed: 01/28/2023]
Abstract
Severe intrauterine adhesions (IUAs), characterized by inadequate endometrial repair and fibrosis, can lead to infertility. Stem cell-based therapies, which deliver mesenchymal stem cells (MSCs) to the wound site, hold a considerable promise for endometrium regeneration. However, some notable hurdles, such as stemness loss, immunogenicity, low retention and survival rate, limit their clinical application. Evidence shows a strategy of mobilizing endogenous MSCs recruitment can overcome the traditional limitations of exogenous stem cell-based therapies. Here, an acellular biomaterial named stromal derived factor-1 alpha (SDF-1α)/E7-modified collagen scaffold (CES) is explored. CES based on harnessing the innate regenerative potential of the body enables near-complete endometrium regeneration and fertility restoration both in a rat endometrium acute damage model and a rat IUA model. Mechanistically, the CES implantation promotes endogenous MSCs recruitment via a macrophage-coordinated strategy; then the homing MSCs exert the function of immunomodulation and altered local microenvironments toward regeneration. To conclude, CES, which can harness endogenous MSCs and overcome the traditional limitations of cell-based therapies, can serve as a clinically feasible and cell-free strategy with high therapeutic efficiency for IUA treatment.
Collapse
Affiliation(s)
- Liaobing Xin
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China.,Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province. No. 3 Qingchun East Road, Jianggan District, Hangzhou, 310016, China
| | - Xiaowen Zheng
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Jianmin Chen
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
| | - Sentao Hu
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Yilun Luo
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Qunzi Ge
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Xiaoying Jin
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China.,Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province. No. 3 Qingchun East Road, Jianggan District, Hangzhou, 310016, China
| | - Lie Ma
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province. No. 3 Qingchun East Road, Jianggan District, Hangzhou, 310016, China.,MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Songying Zhang
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China.,Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province. No. 3 Qingchun East Road, Jianggan District, Hangzhou, 310016, China
| |
Collapse
|
14
|
Zhang G, Dong D, Wan X, Zhang Y. Cardiomyocyte death in sepsis: Mechanisms and regulation (Review). Mol Med Rep 2022; 26:257. [PMID: 35703348 PMCID: PMC9218731 DOI: 10.3892/mmr.2022.12773] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Accepted: 05/20/2022] [Indexed: 11/06/2022] Open
Abstract
Sepsis‑induced cardiac dysfunction is one of the most common types of organ dysfunction in sepsis; its pathogenesis is highly complex and not yet fully understood. Cardiomyocytes serve a key role in the pathophysiology of cardiac function; due to the limited ability of cardiomyocytes to regenerate, their loss contributes to decreased cardiac function. The activation of inflammatory signalling pathways affects cardiomyocyte function and modes of cardiomyocyte death in sepsis. Prevention of cardiomyocyte death is an important therapeutic strategy for sepsis‑induced cardiac dysfunction. Thus, understanding the signalling pathways that activate cardiomyocyte death and cross‑regulation between death modes are key to finding therapeutic targets. The present review focused on advances in understanding of sepsis‑induced cardiomyocyte death pathways, including apoptosis, necroptosis, mitochondria‑mediated necrosis, pyroptosis, ferroptosis and autophagy. The present review summarizes the effect of inflammatory activation on cardiomyocyte death mechanisms, the diversity of regulatory mechanisms and cross‑regulation between death modes and the effect on cardiac function in sepsis to provide a theoretical basis for treatment of sepsis‑induced cardiac dysfunction.
Collapse
Affiliation(s)
- Geping Zhang
- Department of Critical Care Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Dan Dong
- Department of Critical Care Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Xianyao Wan
- Department of Critical Care Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Yongli Zhang
- Department of Critical Care Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| |
Collapse
|
15
|
Rosellini E, Cascone MG. Biomimetic Strategies to Develop Bioactive Scaffolds for Myocardial Tissue Engineering. Open Biomed Eng J 2022. [DOI: 10.2174/18741207-v16-e2205090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The aim of this paper is to provide an overview of the results of the research activity carried out in our laboratories, over the last 10 years, in relation to the development of strategies for the production of biomimetic and bioactive scaffolds for myocardial tissue engineering. Biomimetic and bioactive polymeric scaffolds for cardiac regeneration were designed and manufactured in our laboratories and their morphological, physicochemical, mechanical and biological properties were investigated by different techniques, such as scanning electron microscopy, infrared chemical imaging, swelling test, in vitro degradation assessment, dynamic mechanical analysis, in vitro and in vivo biological tests. Biomimetic scaffolds, able to favor tissue regeneration by mimicking nature, were engineered by different strategies, comprising: (i) the imitation of the composition and interactions among components of the natural extracellular matrix (ECM), by mixing of proteins and polysaccharides; (ii) the material surface modification, using both traditional and innovative techniques, such as molecular imprinting; (iii) the incorporation and release of specific active agents and (iv) the production of scaffolds with a microarchitecture similar to that of native ECM. All the developed strategies were found to be effective in creating materials able to influence cellular behavior and therefore to favor the process of new tissue formation. In particular, the approach based on the combination of different strategies aimed at creating a system capable of communicating with the cells and promoting specific cellular responses, as the ECM does, has appeared particularly promising, in view to favor the formation of a tissue equivalent to the cardiac one.
Collapse
|
16
|
Ma Q, Zhang N, You Y, Zhu J, Yu Z, Chen H, Xie X, Yu H. CXCR4 blockade in macrophage promotes angiogenesis in ischemic hindlimb by modulating autophagy. J Mol Cell Cardiol 2022; 169:57-70. [PMID: 35597127 DOI: 10.1016/j.yjmcc.2022.05.002] [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: 08/15/2021] [Revised: 04/14/2022] [Accepted: 05/07/2022] [Indexed: 11/30/2022]
Abstract
Chemokine receptor CXCR4 plays a crucial role in leukocyte recruitment and inflammation regulation to influence tissue repair in ischemic diseases. Here we assessed the effect of CXCR4 expression in macrophages on angiogenesis in the ischemic hindlimb of a mouse. Inflammatory cells were increased in the ischemic muscles of hindlimb, and CXCR4 was highly expressed in the infiltrated macrophages but not in neutrophils. Myeloid-specific CXCR4 knockout attenuated macrophage infiltration and subsequent reduced inflammatory response in the ischemic hindlimb, accompanied with better blood reperfusion and higher capillary density as compared with that in LysM Cre+/- (Cre) mice. Similar outcomes were also observed in CRE mice whose bone marrow cells were replaced with those from CXCR4-deficient mice. Gene ontology cluster analysis reviewed that Decorin, a negative regulator of angiogenesis, was reduced in CXCR4-deficient macrophages. CXCR4-deficient macrophages were less inducible into M1 phase by lipopolysaccharide and more favorable for M2 polarization under oxygen/glucose deprivation condition. Enhanced autophagy was detected in CXCR4-deficient macrophages, which was associated with less expression of both Decorin and the inflammatory cytokines. In summary, myeloid-specific CXCR4 deficiency reduced monocyte infiltration and the secretion of inflammatory cytokines and Decorin from macrophages, thus blunting inflammation response and promoting angiogenesis in the ischemic hindlimb.
Collapse
Affiliation(s)
- Qunchao Ma
- Department of Cardiology, Second Affiliated Hospital, College of Medicine, Zhejiang University, 88 Jiefang Rd, Hangzhou, Zhejiang Province 310009, PR China; Cardiovascular Key Laboratory of Zhejiang Province, 88 Jiefang Rd, Hangzhou, Zhejiang Province 310009, PR China
| | - Ning Zhang
- Department of Cardiology, Second Affiliated Hospital, College of Medicine, Zhejiang University, 88 Jiefang Rd, Hangzhou, Zhejiang Province 310009, PR China; Cardiovascular Key Laboratory of Zhejiang Province, 88 Jiefang Rd, Hangzhou, Zhejiang Province 310009, PR China
| | - Yayu You
- Department of Cardiology, Second Affiliated Hospital, College of Medicine, Zhejiang University, 88 Jiefang Rd, Hangzhou, Zhejiang Province 310009, PR China; Cardiovascular Key Laboratory of Zhejiang Province, 88 Jiefang Rd, Hangzhou, Zhejiang Province 310009, PR China
| | - Jinyun Zhu
- Department of Cardiology, Second Affiliated Hospital, College of Medicine, Zhejiang University, 88 Jiefang Rd, Hangzhou, Zhejiang Province 310009, PR China; Cardiovascular Key Laboratory of Zhejiang Province, 88 Jiefang Rd, Hangzhou, Zhejiang Province 310009, PR China
| | - Zhaosheng Yu
- State Key Laboratory of Fluid Power and Mechatronic Systems, Department of Mechanics, Zhejiang University, Hangzhou 310027, China
| | - Haibo Chen
- Department of Cardiology, Second Affiliated Hospital, College of Medicine, Zhejiang University, 88 Jiefang Rd, Hangzhou, Zhejiang Province 310009, PR China; Cardiovascular Key Laboratory of Zhejiang Province, 88 Jiefang Rd, Hangzhou, Zhejiang Province 310009, PR China
| | - Xiaojie Xie
- Department of Cardiology, Second Affiliated Hospital, College of Medicine, Zhejiang University, 88 Jiefang Rd, Hangzhou, Zhejiang Province 310009, PR China; Cardiovascular Key Laboratory of Zhejiang Province, 88 Jiefang Rd, Hangzhou, Zhejiang Province 310009, PR China.
| | - Hong Yu
- Department of Cardiology, Second Affiliated Hospital, College of Medicine, Zhejiang University, 88 Jiefang Rd, Hangzhou, Zhejiang Province 310009, PR China; Cardiovascular Key Laboratory of Zhejiang Province, 88 Jiefang Rd, Hangzhou, Zhejiang Province 310009, PR China.
| |
Collapse
|
17
|
An Overview of the Molecular Mechanisms Associated with Myocardial Ischemic Injury: State of the Art and Translational Perspectives. Cells 2022; 11:cells11071165. [PMID: 35406729 PMCID: PMC8998015 DOI: 10.3390/cells11071165] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 03/22/2022] [Accepted: 03/24/2022] [Indexed: 02/06/2023] Open
Abstract
Cardiovascular disease is the leading cause of death in western countries. Among cardiovascular diseases, myocardial infarction represents a life-threatening condition predisposing to the development of heart failure. In recent decades, much effort has been invested in studying the molecular mechanisms underlying the development and progression of ischemia/reperfusion (I/R) injury and post-ischemic cardiac remodeling. These mechanisms include metabolic alterations, ROS overproduction, inflammation, autophagy deregulation and mitochondrial dysfunction. This review article discusses the most recent evidence regarding the molecular basis of myocardial ischemic injury and the new potential therapeutic interventions for boosting cardioprotection and attenuating cardiac remodeling.
Collapse
|
18
|
Wang R, Wei W, Rong S, Wang T, Li B. Intravenous injection of SDF-1α-overexpressing bone marrow mesenchymal stem cells has a potential protective effect on myocardial ischemia in mice. Curr Stem Cell Res Ther 2022; 17:348-360. [PMID: 35306996 DOI: 10.2174/1574888x17666220318144608] [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: 11/15/2021] [Revised: 02/01/2022] [Accepted: 03/01/2022] [Indexed: 11/22/2022]
Abstract
Background Neutrophils are involved in the injury of myocytes during myocardial ischemia (MI). Stem cells migrate to the site of myocardial injury under homing signals and play a protective role, such as inhibiting inflammation. Chemokine SDF-1α and its related receptor CXCR4 are upregulated after myocardial infarction, which may play an important role in stem cell homing. Objectives This study aimed to explore the potential therapeutic effect of SDF-1α-modified bone marrow mesenchymal stem cells on myocardial ischemia/reperfusion (I/R) injury. Methods We explored the role of SDF-1α modified bone marrow mesenchymal stem cells in vivo and in vitro. SDF-1α and CXCR4 expression was detected under hypoxia/reoxygenation (H/R) condition. Cell migration was detected by the transwell method. The levels of SDF-1α and IL-1β, IL-6, IL-10, and TNF-α were detected in different groups. Results In vivo, SDF-1α was mainly upregulated and secreted by cardiomyocytes, and cardiomyocytes recruited stem cells through the SDF-1/CXCR4 pathway to reduce the damage of polymorphic mononuclear neutrophils to cardiomyocytes under H/R. Upregulation of SDF-1α increased the migration ability of BMSC Stem Cells to H/R-induced cardiomyocytes. In vitro, intravenous injection of SDF-1α gene-modified BMSC Stem Cells reduced inflammatory infiltration in the injured area as well as the level of systemic inflammatory factors. Conclusions SDF-1α-overexpressing BMSC Stem Cells protected the heart function of mice and significantly reduced I/R-induced myocardial injury, which has a potential protective effect on MI.
Collapse
Affiliation(s)
- Ruihua Wang
- Shanxi Medical University, Taiyuan, Shanxi 030001, PR China
- Department of Cardiology, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China;
- Department of Cardiology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Wen Wei
- The Affiliated Cardiovascular Hospital of Shanxi Medical University, Taiyuan, Shanxi 030024, P.R. China
| | - Shuling Rong
- Department of Cardiology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Ting Wang
- Shanxi Medical University, Taiyuan, Shanxi 030001, PR China
| | - Bao Li
- Shanxi Medical University, Taiyuan, Shanxi 030001, PR China;
- Department of Cardiology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| |
Collapse
|
19
|
Song B, Chen D, Liu Z, Cheng Y, Zhang Z, Han W, Zhang R, Gong Y. Stromal cell-derived factor-1 exerts opposing roles through CXCR4 and CXCR7 in angiotensin II-induced adventitial remodeling. Biochem Biophys Res Commun 2022; 594:38-45. [PMID: 35066378 DOI: 10.1016/j.bbrc.2022.01.030] [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: 11/09/2021] [Revised: 12/29/2021] [Accepted: 01/09/2022] [Indexed: 11/02/2022]
Abstract
Recent studies have emphasized the role of vascular adventitia inflammation and immune response in hypertension. It has been reported that stromal cell-derived factor-1 (SDF-1) plays various biological functions through its receptors C-X-C motif chemokine receptor 4 (CXCR4) and CXCR7 in tumor growth and tissue repair. However, it is unclear that whether SDF-1/CXCR4/CXCR7 axis is involved in hypertensive vascular remodeling. In the present study, the involvement of SDF-1/CXCR4/CXCR7 axis was evaluated with lentivirus-mediated shRNA of SDF-1 and CXCR7, CXCR4 antagonist AMD3100 and CXCR7 agonist VUF11207 in angiotensin II (AngII)-induced hypertensive mice and in cultured adventitial fibroblasts (AFs). Results showed that AngII infusion markedly increased SDF-1 expressed in vascular adventitia, but not in media and endothelium. Importantly, blockade of SDF-1/CXCR4 axis strikingly potentiated AngII-induced adventitial thickening and fibrosis, as indicated by enhanced collagen I deposition. In contrast, CXCR7 shRNA largely attenuated AngII-induced adventitial thickness and fibrosis, whereas CXCR7 activation with VUF11207 significantly potentiated AngII-induced adventitial thickening and fibrosis. In consistent with these in vivo study, CXCR4 inhibition with AMD3100 and CXCR7 activation with VUF11207 aggravated AngII-induced inflammation, proliferation and migration in cultured AFs. In summary, these results suggested that SDF-1 exerted opposing effects through CXCR4 and CXCR7 in AngII-induced vascular adventitial remodeling.
Collapse
Affiliation(s)
- Bei Song
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Department of General Practice, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dongrui Chen
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zixiong Liu
- Department of General Practice, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuwen Cheng
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zebei Zhang
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weiqing Han
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ruiyan Zhang
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Yanchun Gong
- Department of General Practice, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
20
|
Xiong Y, Tang R, Xu J, Jiang W, Gong Z, Zhang L, Li X, Ning Y, Huang P, Xu J, Chen G, Jin C, Li X, Qian H, Yang Y. Sequential transplantation of exosomes and mesenchymal stem cells pretreated with a combination of hypoxia and Tongxinluo efficiently facilitates cardiac repair. Stem Cell Res Ther 2022; 13:63. [PMID: 35130979 PMCID: PMC8822662 DOI: 10.1186/s13287-022-02736-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Accepted: 12/17/2021] [Indexed: 02/07/2023] Open
Abstract
Background Bone marrow-derived mesenchymal stem cells (MSCs), which possess immunomodulatory characteristic, are promising candidates for the treatment of acute myocardial infarction (AMI). However, the low retention and survival rate of MSCs in the ischemic heart limit their therapeutic efficacy. Strategies either modifying MSCs or alleviating the inflammatory environment, which facilitates the recruitment and survival of the engrafted MSCs, may solve the problem. Thus, we aimed to explore the therapeutic efficacy of sequential transplantation of exosomes and combinatorial pretreated MSCs in the treatment of AMI. Methods Exosomes derived from MSCs were delivered to infarcted hearts through intramyocardial injection followed by the intravenous infusion of differentially pretreated MSCs on Day 3 post-AMI. Enzyme linked immunosorbent assay (ELISA) was performed to evaluate the inflammation level as well as the SDF-1 levels in the infarcted border zone of the heart. Echocardiography and histological analysis were performed to assess cardiac function, infarct size, collagen area and angiogenesis. Results Sequential transplantation of exosomes and the combinatorial pretreated MSCs significantly facilitated cardiac repair compared to AMI rats treated with exosomes alone. Notably, compared to the other three methods of cotransplantation, combinatorial pretreatment with hypoxia and Tongxinluo (TXL) markedly enhanced the CXCR4 level of MSCs and promoted recruitment, which resulted in better cardiac function, smaller infarct size and enhanced angiogenesis. We further demonstrated that exosomes effectively reduced apoptosis in MSCs in vitro. Conclusion Sequential delivery of exosomes and pretreated MSCs facilitated cardiac repair post-AMI, and combined pretreatment with hypoxia and TXL better enhanced the cardioprotective effects. This method provides new insight into the clinical translation of stem cell-based therapy for AMI.
Collapse
|
21
|
Foglio E, Pellegrini L, Russo MA, Limana F. HMGB1-Mediated Activation of the Inflammatory-Reparative Response Following Myocardial Infarction. Cells 2022; 11:cells11020216. [PMID: 35053332 PMCID: PMC8773872 DOI: 10.3390/cells11020216] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/04/2022] [Accepted: 01/08/2022] [Indexed: 02/06/2023] Open
Abstract
Different cell types belonging to the innate and adaptive immune system play mutually non-exclusive roles during the different phases of the inflammatory-reparative response that occurs following myocardial infarction. A timely and finely regulation of their action is fundamental for the process to properly proceed. The high-mobility group box 1 (HMGB1), a highly conserved nuclear protein that in the extracellular space can act as a damage-associated molecular pattern (DAMP) involved in a large variety of different processes, such as inflammation, migration, invasion, proliferation, differentiation, and tissue regeneration, has recently emerged as a possible regulator of the activity of different immune cell types in the distinct phases of the inflammatory reparative process. Moreover, by activating endogenous stem cells, inducing endothelial cells, and by modulating cardiac fibroblast activity, HMGB1 could represent a master regulator of the inflammatory and reparative responses following MI. In this review, we will provide an overview of cellular effectors involved in these processes and how HMGB1 intervenes in regulating each of them. Moreover, we will summarize HMGB1 roles in regulating other cell types that are involved in the different phases of the inflammatory-reparative response, discussing how its redox status could affect its activity.
Collapse
Affiliation(s)
- Eleonora Foglio
- Technoscience, Parco Scientifico e Tecnologico Pontino, 04100 Latina, Italy;
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy;
| | - Laura Pellegrini
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy;
| | - Matteo Antonio Russo
- IRCCS San Raffaele Roma and MEBIC Consortium, 00166 Rome, Italy;
- San Raffaele University of Rome, 00166 Rome, Italy
| | - Federica Limana
- San Raffaele University of Rome, 00166 Rome, Italy
- Laboratory of Cellular and Molecular Pathology, IRCCS San Raffaele Roma, 00166 Rome, Italy
- Correspondence:
| |
Collapse
|
22
|
Atypical Roles of the Chemokine Receptor ACKR3/CXCR7 in Platelet Pathophysiology. Cells 2022; 11:cells11020213. [PMID: 35053329 PMCID: PMC8773869 DOI: 10.3390/cells11020213] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/06/2022] [Accepted: 01/07/2022] [Indexed: 12/23/2022] Open
Abstract
The manifold actions of the pro-inflammatory and regenerative chemokine CXCL12/SDF-1α are executed through the canonical GProteinCoupledReceptor CXCR4, and the non-canonical ACKR3/CXCR7. Platelets express CXCR4, ACKR3/CXCR7, and are a vital source of CXCL12/SDF-1α themselves. In recent years, a regulatory impact of the CXCL12-CXCR4-CXCR7 axis on platelet biogenesis, i.e., megakaryopoiesis, thrombotic and thrombo-inflammatory actions have been revealed through experimental and clinical studies. Platelet surface expression of ACKR3/CXCR7 is significantly enhanced following myocardial infarction (MI) in acute coronary syndrome (ACS) patients, and is also associated with improved functional recovery and prognosis. The therapeutic implications of ACKR3/CXCR7 in myocardial regeneration and improved recovery following an ischemic episode, are well documented. Cardiomyocytes, cardiac-fibroblasts, endothelial lining of the blood vessels perfusing the heart, besides infiltrating platelets and monocytes, all express ACKR3/CXCR7. This review recapitulates ligand induced differential trafficking of platelet CXCR4-ACKR3/CXCR7 affecting their surface availability, and in regulating thrombo-inflammatory platelet functions and survival through CXCR4 or ACKR3/CXCR7. It emphasizes the pro-thrombotic influence of CXCL12/SDF-1α exerted through CXCR4, as opposed to the anti-thrombotic impact of ACKR3/CXCR7. Offering an innovative translational perspective, this review also discusses the advantages and challenges of utilizing ACKR3/CXCR7 as a potential anti-thrombotic strategy in platelet-associated cardiovascular disorders, particularly in coronary artery disease (CAD) patients post-MI.
Collapse
|
23
|
Nasiri Boroujeni S, Chehelcheraghi F, Khaksarian M, Sedighi M, Ghorbanzadeh V, Nazari A. Applying Vasopressin-Pre-Conditioned Human Adipose Mesenchymal Stem Cells Improves Heart Condition after Transplantation into Infarcted Myocardium. INTERNATIONAL JOURNAL OF MOLECULAR AND CELLULAR MEDICINE 2022; 11:207-222. [PMID: 37605740 PMCID: PMC10440004 DOI: 10.22088/ijmcm.bums.11.3.207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 04/03/2023] [Accepted: 04/24/2023] [Indexed: 08/23/2023]
Abstract
Transplantation of H-AdMSCs may improve heart function after MI. AVP is a neurohypophyseal hormone that reduces cardiovascular damage. This study investigated the role of AVP preconditioning in the survival of MSCs and their effect on myocardial repair in the MI rats. H-AMSCs were isolated and incubated for 3 days. The expression of oxytocin and vasopressin receptors was evaluated by Real-time-PCR. Forty male Wistar rats were divided into 4 groups: control, sham, ASC and AVP-ASC. Ischemia was established by ligation of LAD coronary artery. Electrocardiography, fibrosis, angiogenesis, and apoptosis in myocardium were determined after 7 days. Results showed that preconditioned MSCs significantly increased cardiac function when compared with group that received non-preconditioned MSCs. This was associated with significantly reduced fibrosis, increased vascular density, and decreased resident myocyte apoptosis. Results indicate that AVP preconditioned MSCs can be consider a novel approach to management of MI.
Collapse
Affiliation(s)
| | - Farzaneh Chehelcheraghi
- Department of Anatomical Sciences, School of Medicine, Lorestan University of Medical Sciences, Khoramabad, Iran.
| | - Mojtaba Khaksarian
- Department of Physiology, Lorestan University of Medical Sciences, Khorramabad, Iran.
| | - Mehrnoosh Sedighi
- Cardiovascular Research Center, Shahid Rahimi Hospital, Lorestan University of Medical Sciences, Khoramabad, Iran.
- Razi Herbal Medicines Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran.
| | - Vajihe Ghorbanzadeh
- Cardiovascular Research Center, Shahid Rahimi Hospital, Lorestan University of Medical Sciences, Khoramabad, Iran.
| | - Afshin Nazari
- Cardiovascular Research Center, Shahid Rahimi Hospital, Lorestan University of Medical Sciences, Khoramabad, Iran.
| |
Collapse
|
24
|
Hsu Y, Huang K, Cheng K. Resuscitating the Field of Cardiac Regeneration: Seeking Answers from Basic Biology. Adv Biol (Weinh) 2021; 6:e2101133. [PMID: 34939372 DOI: 10.1002/adbi.202101133] [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: 08/11/2021] [Revised: 12/02/2021] [Indexed: 11/09/2022]
Abstract
Heart failure (HF) is one of the leading causes for hospital admissions worldwide. HF patients are classified based on the chronic changes in left ventricular ejection fraction (LVEF) as preserved (LVEF ≥ 50%), reduced (LVEF ≤ 40%), or mid-ranged (40% < LVEF < 50%) HFs. Treatments nowadays can prevent HFrEF progress, whereas only a few of the treatments have been proven to be effective in improving the survival of HFpEF. In this review, numerous mediators involved in the pathogenesis of HF are summarized. The regional upstream signaling and their diagnostic and therapeutic potential are also discussed. Additionally, the recent challenges and development in cardiac regenerative therapy that hold opportunities for future research and clinical translation are discussed.
Collapse
Affiliation(s)
- Yaching Hsu
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC, 27607, USA.,Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill & North Carolina State University, Raleigh, NC, 27607, USA
| | - Ke Huang
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC, 27607, USA.,Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill & North Carolina State University, Raleigh, NC, 27607, USA
| | - Ke Cheng
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC, 27607, USA.,Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill & North Carolina State University, Raleigh, NC, 27607, USA
| |
Collapse
|
25
|
Platelet ACKR3/CXCR7 Favors Anti-Platelet Lipids over an Atherothrombotic Lipidome and Regulates Thrombo-inflammation. Blood 2021; 139:1722-1742. [PMID: 34905596 DOI: 10.1182/blood.2021013097] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 11/30/2021] [Indexed: 11/20/2022] Open
Abstract
Platelet ACKR3/CXCR7 surface expression is enhanced and influences prognosis in coronary artery disease-(CAD) patients, who exhibit a distinct atherothrombotic platelet lipidome. Current investigation validates the potential of ACKR3/CXCR7 in regulating thrombo-inflammatory response, through its impact on the platelet lipidome. CAD patients-(n=230) with enhanced platelet-ACKR3/CXCR7 expression exhibited reduced aggregation. Pharmacological CXCR7-agonist-(VUF11207) significantly reduced pro-thrombotic platelet response in blood from ACS patients-(n=11) ex vivo. CXCR7-agonist administration reduced thrombotic functions and thrombo-inflammatory platelet-leukocyte interactions post myocardial infarction-(MI) and arterial injury in vivo. ACKR3/CXCR7-ligation did not affect surface availability of GPIbα, GPV, GPVI, GPIX, αv-integrin, β3-integrin, coagulation profile-(APTT, PT), bleeding time, plasma-dependent thrombin generation-(thrombinoscopy) or clot formation-(thromboelastography), but counteracted activation-induced phosphatidylserine exposure and procoagulant platelet-assisted thrombin generation. Targeted-(micro-UHPLC-ESI-QTrap-MS/MS) and untargeted-(UHPLC-ESI-QTOF-MS/MS) lipidomics analysis revealed that ACKR3/CXCR7-ligation favored generation of anti-thrombotic lipids-(dihomo-γ-linolenic acid-DGLA, 12-hydroxyeicosatrienoic acid-12-HETrE) over cyclooxygenase-COX-1-(thromboxane-TxA2), or 12-lipoxygenase-LOX-(12-HETE) metabolized pro-thrombotic, and phospholipase derived atherogenic-(lysophosphatidylcholine-LPC) lipids, in healthy subjects and CAD patients, contrary to anti-platelet therapy. Through 12-HETrE, ACKR3/CXCR7-ligation coordinated with Gαs-coupled prostacyclin receptor-(IP) to trigger cAMP-PKA mediated platelet inhibition. ACKR3/CXCR7-ligation reduced generation of lipid agonists-(arachidonic acid-AA,TxA2), lipid signaling intermediates-(lyophosphatidylinositol-LPI, diacylglycerol-DG), which affected calcium mobilization, intracellular signaling, consequently platelet interaction with physiological matrices and thrombo-inflammatory secretion-(IL1β,IFN-γ,TGF-β,IL-8), emphasizing its functional dichotomy from pro-thrombotic CXCR4. Moreover, CXCR7-agonist regulated heparin-induced thrombocytopenia-(HIT)-sera/IgG-induced platelet and neutrophil activation, heparin induced platelet aggregation-(HIPA), generation of COX-1-(TxA2), 12-LOX-(12-HETE) derived thrombo-inflammatory lipids, platelet-neutrophil aggregate formation, and thrombo-inflammatory secretion (sCD40L, IL-1β, IFN-γ, TNF-α, sP-selectin, IL-8, tissue factor-TF) ex vivo. Therefore, ACKR3/CXCR7 may offer a novel therapeutic strategy in acute/chronic thrombo-inflammation exaggerated cardiovascular pathologies, and CAD.
Collapse
|
26
|
Abbasi-Habashi S, Jickling GC, Winship IR. Immune Modulation as a Key Mechanism for the Protective Effects of Remote Ischemic Conditioning After Stroke. Front Neurol 2021; 12:746486. [PMID: 34956045 PMCID: PMC8695500 DOI: 10.3389/fneur.2021.746486] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 11/09/2021] [Indexed: 12/12/2022] Open
Abstract
Remote ischemic conditioning (RIC), which involves a series of short cycles of ischemia in an organ remote to the brain (typically the limbs), has been shown to protect the ischemic penumbra after stroke and reduce ischemia/reperfusion (IR) injury. Although the exact mechanism by which this protective signal is transferred from the remote site to the brain remains unclear, preclinical studies suggest that the mechanisms of RIC involve a combination of circulating humoral factors and neuronal signals. An improved understanding of these mechanisms will facilitate translation to more effective treatment strategies in clinical settings. In this review, we will discuss potential protective mechanisms in the brain and cerebral vasculature associated with RIC. We will discuss a putative role of the immune system and circulating mediators of inflammation in these protective processes, including the expression of pro-and anti-inflammatory genes in peripheral immune cells that may influence the outcome. We will also review the potential role of extracellular vesicles (EVs), biological vectors capable of delivering cell-specific cargo such as proteins and miRNAs to cells, in modulating the protective effects of RIC in the brain and vasculature.
Collapse
Affiliation(s)
- Sima Abbasi-Habashi
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| | - Glen C Jickling
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
- Division of Neurology, Faculty of Medicine, University of Alberta, Edmonton, AB, Canada
| | - Ian R Winship
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
- Neurochemical Research Unit, Department of Psychiatry, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
27
|
Oprescu N, Micheu MM, Scafa-Udriste A, Popa-Fotea NM, Dorobantu M. Inflammatory markers in acute myocardial infarction and the correlation with the severity of coronary heart disease. Ann Med 2021; 53:1041-1047. [PMID: 34180324 PMCID: PMC8245096 DOI: 10.1080/07853890.2021.1916070] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 04/07/2021] [Indexed: 11/15/2022] Open
Abstract
INTRODUCTION The inflammatory hypothesis of atherosclerosis is appealing in acute coronary syndromes, but the dynamics and precise role are not established. OBJECTIVES The study investigates the levels of C reactive protein (CRP), interleukin 1β (IL-1β) and stromal-derived factor 1α (SDF-1α) at the time of acute myocardial infarction (AMI) and at 1 and 6 months afterwards, compared with a control group. RESULTS In the acute phase of AMI, CRP and SDF-1α were significantly higher, while IL-1β showed lower levels compared with controls. CRP positively correlated with coronary stenosis severity (rho = 0.3, p=.05) and negatively related with left ventricle ejection fraction (LVEF) at 1 month (rho= -0.43, p=.05). IL-1β weakly correlated with the severity of coronary lesions (rho =0.29, p=.02) and strongly with LVEF (rho= -0.8, p=.05). SDF-1α, slightly correlated with LVEF at 1 month (rho = 0.22, p=.01) and with the severity of coronary atherosclerosis (rho= -0.41, p=.003). CONCLUSIONS CRP, IL-1β and SDF-1α have important dynamic in the first 6 months after AMI and CRP and SDF-1α levels correlated with the severity of coronary lesions and LVEF at 1 month after the acute ischaemic event.
Collapse
Affiliation(s)
- Nicoleta Oprescu
- Department of Cardiology, Clinical Emergency Hospital of Bucharest, Romania
| | | | - Alexandru Scafa-Udriste
- Department of Cardiology, Clinical Emergency Hospital of Bucharest, Romania
- Cardio-thoracic Department, University of Medicine and Pharmacy “Carol Davila”, Bucharest, Romania
| | - Nicoleta-Monica Popa-Fotea
- Department of Cardiology, Clinical Emergency Hospital of Bucharest, Romania
- Cardio-thoracic Department, University of Medicine and Pharmacy “Carol Davila”, Bucharest, Romania
| | - Maria Dorobantu
- Department of Cardiology, Clinical Emergency Hospital of Bucharest, Romania
- Cardio-thoracic Department, University of Medicine and Pharmacy “Carol Davila”, Bucharest, Romania
| |
Collapse
|
28
|
Raynor WY, Park PSU, Borja AJ, Sun Y, Werner TJ, Ng SJ, Lau HC, Høilund-Carlsen PF, Alavi A, Revheim ME. PET-Based Imaging with 18F-FDG and 18F-NaF to Assess Inflammation and Microcalcification in Atherosclerosis and Other Vascular and Thrombotic Disorders. Diagnostics (Basel) 2021; 11:diagnostics11122234. [PMID: 34943473 PMCID: PMC8700072 DOI: 10.3390/diagnostics11122234] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 11/19/2021] [Accepted: 11/23/2021] [Indexed: 01/13/2023] Open
Abstract
Positron emission tomography (PET) imaging with 18F-fluorodeoxyglucose (FDG) represents a method of detecting and characterizing arterial wall inflammation, with potential applications in the early assessment of vascular disorders such as atherosclerosis. By portraying early-stage molecular changes, FDG-PET findings have previously been shown to correlate with atherosclerosis progression. In addition, recent studies have suggested that microcalcification revealed by 18F-sodium fluoride (NaF) may be more sensitive at detecting atherogenic changes compared to FDG-PET. In this review, we summarize the roles of FDG and NaF in the assessment of atherosclerosis and discuss the role of global assessment in quantification of the vascular disease burden. Furthermore, we will review the emerging applications of FDG-PET in various vascular disorders, including pulmonary embolism, as well as inflammatory and infectious vascular diseases.
Collapse
Affiliation(s)
- William Y. Raynor
- Department of Radiology, Hospital of the University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA; (W.Y.R.); (P.S.U.P.); (A.J.B.); (T.J.W.); (A.A.)
| | - Peter Sang Uk Park
- Department of Radiology, Hospital of the University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA; (W.Y.R.); (P.S.U.P.); (A.J.B.); (T.J.W.); (A.A.)
- Perelman School of Medicine at the University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, PA 19104, USA;
| | - Austin J. Borja
- Department of Radiology, Hospital of the University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA; (W.Y.R.); (P.S.U.P.); (A.J.B.); (T.J.W.); (A.A.)
- Perelman School of Medicine at the University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, PA 19104, USA;
| | - Yusha Sun
- Perelman School of Medicine at the University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, PA 19104, USA;
| | - Thomas J. Werner
- Department of Radiology, Hospital of the University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA; (W.Y.R.); (P.S.U.P.); (A.J.B.); (T.J.W.); (A.A.)
| | - Sze Jia Ng
- Department of Medicine, Crozer-Chester Medical Center, Upland, PA 19013, USA; (S.J.N.); (H.C.L.)
| | - Hui Chong Lau
- Department of Medicine, Crozer-Chester Medical Center, Upland, PA 19013, USA; (S.J.N.); (H.C.L.)
| | - Poul Flemming Høilund-Carlsen
- Department of Nuclear Medicine, Odense University Hospital, 5000 Odense C, Denmark;
- Department of Clinical Research, University of Southern Denmark, 5000 Odense C, Denmark
| | - Abass Alavi
- Department of Radiology, Hospital of the University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA; (W.Y.R.); (P.S.U.P.); (A.J.B.); (T.J.W.); (A.A.)
| | - Mona-Elisabeth Revheim
- Department of Radiology, Hospital of the University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA; (W.Y.R.); (P.S.U.P.); (A.J.B.); (T.J.W.); (A.A.)
- Division of Radiology and Nuclear Medicine, Oslo University Hospital, Sognsvannsveien 20, 0372 Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Problemveien 7, 0315 Oslo, Norway
- Correspondence: or
| |
Collapse
|
29
|
Salybekov AA, Salybekova A, Sheng Y, Shinozaki Y, Yokoyama K, Kobayashi S, Asahara T. Extracellular Vesicles Derived From Regeneration Associated Cells Preserve Heart Function After Ischemia-Induced Injury. Front Cardiovasc Med 2021; 8:754254. [PMID: 34746267 PMCID: PMC8564358 DOI: 10.3389/fcvm.2021.754254] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 09/22/2021] [Indexed: 12/20/2022] Open
Abstract
Under vasculogenic conditioning, pro-inflammatory cell subsets of peripheral blood mononuclear cells (PBMCs) shift their phenotype to pro-regenerative cells such as vasculogenic endothelial progenitor cells, M2 macrophages, and regulatory T cells, collectively designated as regeneration-associated cells (RACs). In this study, we evaluated the therapeutic efficacy of RAC-derived extracellular vesicles (RACev) compared to mesenchymal stem cell-derived EVs (MSCev) in the context of myocardial ischemia reperfusion injury (M-IRI). Human PBMCs were cultured with defined growth factors for seven days to harvest RACs. RACev and MSCev were isolated via serial centrifugation and ultracentrifugation. EV quantity and size were characterized by nanoparticle tracking analysis. In vitro, RACev markedly enhanced the viability, and proliferation of human umbilical vein endothelial cells in a dose-dependent manner compared to MSCev. Notably, systemic injection of RACev improved cardiac functions at 4 weeks, such as fractional shortening, and protection from mitral regurgitation than the MSCev-treated group. Histologically, the RACev-transplanted group showed less interstitial fibrosis and enhanced capillary densities compared to the MSCev group. These beneficial effects were coupled with significant expression of angiogenesis, anti-fibrosis, anti-inflammatory, and cardiomyogenesis-related miRs in RACev, while modestly in MSCev. In vivo bioluminescence analysis showed preferential accumulation of RACev in the IR-injured myocardium, while MSCev accumulation was limited. Immune phenotyping analysis confirmed the immunomodulatory effect of MSCev and RACev. Overall, repetitive systemic transplantation of RACev is superior to MSCev in terms of cardiac function enhancements via crucial angiogenesis, anti-fibrosis, anti-inflammation miR delivery to the ischemic tissue.
Collapse
Affiliation(s)
- Amankeldi A Salybekov
- Kidney Disease and Transplant Center, Shonan Kamakura General Hospital, Kamakura, Japan.,Shonan Research Institute of Innovative Medicine, Shonan Kamakura General Hospital, Kamakura, Japan.,Division of Regenerative Medicine, Department of Center for Clinical and Translational Science, Shonan Kamakura General Hospital, Kamakura, Japan.,Department of Advanced Medicine Science, Tokai University School of Medicine, Isehara, Japan
| | - Ainur Salybekova
- Department of Advanced Medicine Science, Tokai University School of Medicine, Isehara, Japan
| | - Yin Sheng
- Department of Advanced Medicine Science, Tokai University School of Medicine, Isehara, Japan
| | - Yoshiko Shinozaki
- Teaching and Research Support Core Center, Tokai University School of Medicine, Isehara, Japan
| | - Keiko Yokoyama
- Teaching and Research Support Core Center, Tokai University School of Medicine, Isehara, Japan
| | - Shuzo Kobayashi
- Shonan Research Institute of Innovative Medicine, Shonan Kamakura General Hospital, Kamakura, Japan.,Division of Regenerative Medicine, Department of Center for Clinical and Translational Science, Shonan Kamakura General Hospital, Kamakura, Japan
| | - Takayuki Asahara
- Shonan Research Institute of Innovative Medicine, Shonan Kamakura General Hospital, Kamakura, Japan.,Department of Advanced Medicine Science, Tokai University School of Medicine, Isehara, Japan
| |
Collapse
|
30
|
Exosomal microRNA-98-5p from hypoxic bone marrow mesenchymal stem cells inhibits myocardial ischemia-reperfusion injury by reducing TLR4 and activating the PI3K/Akt signaling pathway. Int Immunopharmacol 2021; 101:107592. [PMID: 34715573 DOI: 10.1016/j.intimp.2021.107592] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 03/10/2021] [Accepted: 03/11/2021] [Indexed: 12/16/2022]
Abstract
OBJECTIVE MicroRNAs (miRNAs) are essential biomarkers during development of human diseases. We aimed to explore the role of hypoxia-induced bone marrow mesenchymal stem cells (BMSCs)-derived exosomal miR-98-5p in myocardial ischemia-reperfusion injury (MI/RI). METHODS BMSCs were isolated, cultured, stimulated by hypoxia and transfected with adenovirus expressing miR-98-5p. The exosomes were extracted from BMSCs and named as BMSC-exos. The rat MI/RI models were established by ligation of left anterior descending artery and were respectively injected. Then, hemodynamic indices, myocardial enzymes, oxidative stress factors, inflammatory factors, macrophage infiltration and infarct size in these rats were determined. Expression of miR-98-5p, toll-like receptor 4 (TLR4) and the phosphatidylinositol-3-kinase/protein kinase B (PI3K/Akt) signaling pathway-related proteins was assessed. The target relation between miR-98-5p and TLR4 was confirmed by bioinformatic method and dual luciferase report gene assay. RESULTS MiR-98-5p was downregulated, TLR4 was upregulated and the PI3K/Akt signaling pathway was inactivated in MI/RI rat myocardial tissues. Exosomal miR-98-5p from hypoxic BMSCs promoted cardiac function and suppressed myocardial enzyme levels, oxidative stress, inflammation response, macrophage infiltration and infarct size in I/R myocardial tissues. Moreover, TRL4 was targeted by miR-98-5p and miR-98-5p activated PI3K/Akt signaling pathway. CONCLUSION Hypoxia-induced BMSC-exos elevated miR-98-5p to protect against MI/RI. This study may be helpful for treatment of MI/RI.
Collapse
|
31
|
Yu X, Newland SA, Zhao TX, Lu Y, Sage AS, Sun Y, Sriranjan RS, Ma MKL, Lam BYH, Nus M, Harrison JE, Bond SJ, Cheng X, Silvestre JS, Rudd JHF, Cheriyan J, Mallat Z. Innate Lymphoid Cells Promote Recovery of Ventricular Function After Myocardial Infarction. J Am Coll Cardiol 2021; 78:1127-1142. [PMID: 34503682 PMCID: PMC8434674 DOI: 10.1016/j.jacc.2021.07.018] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 07/06/2021] [Accepted: 07/13/2021] [Indexed: 12/25/2022]
Abstract
BACKGROUND Innate lymphoid cells type 2 (ILC2s) play critical homeostatic functions in peripheral tissues. ILC2s reside in perivascular niches and limit atherosclerosis development. OBJECTIVES ILC2s also reside in the pericardium but their role in postischemic injury is unknown. METHODS We examined the role of ILC2 in a mouse model of myocardial infarction (MI), and compared mice with or without genetic deletion of ILC2. We determined infarct size using histology and heart function using echocardiography. We assessed cardiac ILC2 using flow cytometry and RNA sequencing. Based on these data, we devised a therapeutic strategy to activate ILC2 in mice with acute MI, using exogenous interleukin (IL)-2. We also assessed the ability of low-dose IL-2 to activate ILC2 in a double-blind randomized clinical trial of patients with acute coronary syndromes (ACS). RESULTS We found that ILC2 levels were increased in pericardial adipose tissue after experimental MI, and genetic ablation of ILC2 impeded the recovery of heart function. RNA sequencing revealed distinct transcript signatures in ILC2, and pointed to IL-2 axis as a major upstream regulator. Treatment of T-cell-deficient mice with IL-2 (to activate ILC2) significantly improved the recovery of heart function post-MI. Administration of low-dose IL-2 to patients with ACS led to activation of circulating ILC2, with significant increase in circulating IL-5, a prototypic ILC2-derived cytokine. CONCLUSIONS ILC2s promote cardiac healing and improve the recovery of heart function after MI in mice. Activation of ILC2 using low-dose IL-2 could be a novel therapeutic strategy to promote a reparative response after MI.
Collapse
Affiliation(s)
- Xian Yu
- Department of Medicine, Division of Cardiovascular Medicine, University of Cambridge, Cambridge, United Kingdom; Department of Cardiology, Union Hospital, Tongji, Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Stephen A Newland
- Department of Medicine, Division of Cardiovascular Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Tian X Zhao
- Department of Medicine, Division of Cardiovascular Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Yuning Lu
- Department of Medicine, Division of Cardiovascular Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Andrew S Sage
- Department of Medicine, Division of Cardiovascular Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Yanyi Sun
- Université de Paris, PARCC, INSERM, F-75015 Paris, France
| | - Rouchelle S Sriranjan
- Department of Medicine, Division of Cardiovascular Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Marcella K L Ma
- The Wellcome Trust-MRC Institute of Metabolic Science-Metabolic Research Laboratories, University of Cambridge, Cambridge, United Kingdom
| | - Brian Y H Lam
- The Wellcome Trust-MRC Institute of Metabolic Science-Metabolic Research Laboratories, University of Cambridge, Cambridge, United Kingdom
| | - Meritxell Nus
- Department of Medicine, Division of Cardiovascular Medicine, University of Cambridge, Cambridge, United Kingdom
| | - James E Harrison
- Department of Medicine, Division of Cardiovascular Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Simon J Bond
- Division of Experimental Medicine and Immunotherapeutics, University of Cambridge, Cambridge, United Kingdom
| | - Xiang Cheng
- Department of Cardiology, Union Hospital, Tongji, Medical College, Huazhong University of Science and Technology, Wuhan, China
| | | | - James H F Rudd
- Department of Medicine, Division of Cardiovascular Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Joseph Cheriyan
- Division of Experimental Medicine and Immunotherapeutics, University of Cambridge, Cambridge, United Kingdom; Cambridge Clinical Trials Unit, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Ziad Mallat
- Department of Medicine, Division of Cardiovascular Medicine, University of Cambridge, Cambridge, United Kingdom; Université de Paris, PARCC, INSERM, F-75015 Paris, France.
| |
Collapse
|
32
|
Sedighi M, Ghorbanzadeh V, Abaszadeh S, Karimi A, Cheraghi M, Rafieian-Kopaei M, Moghimian M, Mohammadi A, Veiskarami S, Mokhayeri Y, Nazari A. Up-regulation of chemokine receptor type 4 expression in the ischemic reperfused heart by alcoholic extract of Cichorium intybus rescue the heart from ischemia injury in male rat. J Pharm Pharmacol 2021; 73:1351-1360. [PMID: 34076244 DOI: 10.1093/jpp/rgab076] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 05/11/2021] [Indexed: 12/14/2022]
Abstract
OBJECTIVES Cichorium intybus is used in traditional medicine for various diseases including heart disease. This study aimed at evaluating the chemokine receptor type 4 up-regulation and cardioprotective effects of hydroalcoholic extract of C. intybus in a rat model of ischemic reperfusion. METHODS Animals in four groups of eight rats each received vehicle or one of three doses of C. intybus (50, 100 or 200 mg/kg/d) for 14 days. Then they were subjected to 30 min of ischemia followed by 7 days of reperfusion. At the end of the experiment, blood specimens were prepared for serum assays. The level of myocardium chemokine receptor type 4 was also measured using RT-PCR. KEY FINDINGS Cichorium intybus (CI-50) improved infarct size, episodes of the ventricular ectopic beat, ventricular tachycardia, and duration of ventricular tachycardia, QTc shortening. It also stabilized the ST segment changes and increased heart rate during ischemia. The blood pressure decreased in CI-50 group in comparison to the control and CI-200 group. C. intybus increased serum superoxide dismutase and reduced lactate dehydrogenase activity, Cardiac Troponin I and malondialdehyde levels. C. intybus led to an increase in the expression of chemokine receptor type 4. CONCLUSIONS These findings suggest that C. intybus administration before ischemia is able to induce cardioprotective effect against ischemic reperfusion injury, probably through chemokine receptor type 4 over-expression and antioxidant activity.
Collapse
Affiliation(s)
- Mehrnoosh Sedighi
- Cardiovascular Research Center, Shahid Rahimi Hospital, Lorestan University of Medical Sciences, Khoramabad, Iran
| | - Vajihe Ghorbanzadeh
- Razi Herbal Medicines Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Saber Abaszadeh
- Department of clinical biochemistry, Lorestan University of Medical Science, Khorramabad, Iran
| | - Arash Karimi
- Department of Anesthesiology, Anesthesiologist, Faculty of Medicine, Lorestan University of Medical Science, Khorramabad, Iran
| | - Mostafa Cheraghi
- Cardiovascular Research Center, Shahid Rahimi Hospital, Lorestan University of Medical Sciences, Khoramabad, Iran
| | - Mahmoud Rafieian-Kopaei
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Siences, Shahrekord, Iran
| | - Maryam Moghimian
- Department of Physiology, Gonabad University of Medical Science, Gonabad, Iran
| | - Asghar Mohammadi
- Cardiovascular Research Center, Shahid Rahimi Hospital, Lorestan University of Medical Sciences, Khoramabad, Iran
| | - Saeid Veiskarami
- Lorestan Agricultural and Natural Resources Research and Education Center, Department of animal science, Iran
| | - Yaser Mokhayeri
- Cardiovascular Research Center, Shahid Rahimi Hospital, Lorestan University of Medical Sciences, Khoramabad, Iran
| | - Afshin Nazari
- Department of Physiology, Lorestan University of Medical Science, Khorramabad, Iran
| |
Collapse
|
33
|
Role of Stromal Cell-Derived Factor-1 in Endothelial Progenitor Cell-Mediated Vascular Repair and Regeneration. Tissue Eng Regen Med 2021; 18:747-758. [PMID: 34449064 PMCID: PMC8440704 DOI: 10.1007/s13770-021-00366-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 06/07/2021] [Accepted: 06/15/2021] [Indexed: 12/13/2022] Open
Abstract
Endothelial progenitor cells (EPCs) are immature endothelial cells that participate in vascular repair and postnatal neovascularization and provide a novel and promising therapy for the treatment of vascular disease. Studies in different animal models have shown that EPC mobilization through pharmacological agents and autologous EPC transplantation contribute to restoring blood supply and tissue regeneration after ischemic injury. However, these effects of the progenitor cells in clinical studies exhibit mixed results. The therapeutic efficacy of EPCs is closely associated with the number of the progenitor cells recruited into ischemic regions and their functional abilities and survival in injury tissues. In this review, we discussed the regulating role of stromal cell-derived factor-1 (also known CXCL12, SDF-1) in EPC mobilization, recruitment, homing, vascular repair and neovascularization, and analyzed the underlying machemisms of these functions. Application of SDF-1 to improve the regenerative function of EPCs following vascular injury was also discussed. SDF-1 plays a crucial role in mobilizing EPC from bone marrow into peripheral circulation, recruiting the progenitor cells to target tissue and protecting against cell death under pathological conditions; thus improve EPC regenerative capacity. SDF-1 are crucial for regulating EPC regenerative function, and provide a potential target for improve therapeutic efficacy of the progenitor cells in treatment of vascular disease.
Collapse
|
34
|
Liu P, Sun H, Zhou X, Wang Q, Gao F, Fu Y, Li T, Wang Y, Li Y, Fan B, Li X, Jiang T, Qin X, Zheng Q. CXCL12/CXCR4 axis as a key mediator in atrial fibrillation via bioinformatics analysis and functional identification. Cell Death Dis 2021; 12:813. [PMID: 34453039 PMCID: PMC8397768 DOI: 10.1038/s41419-021-04109-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 06/23/2021] [Accepted: 06/24/2021] [Indexed: 02/06/2023]
Abstract
Atrial fibrillation (AF) is an increasingly prevalent arrhythmia with significant health and socioeconomic impact. The underlying mechanism of AF is still not well understood. In this study, we sought to identify hub genes involved in AF, and explored their functions and underlying mechanisms based on bioinformatics analysis. Five microarray datasets in GEO were used to identify the differentially expressed genes (DEGs) by Robust Rank Aggregation (RRA), and hub genes were screened out using protein-protein interaction (PPI) network. AF model was established using a mixture of acetylcholine and calcium chloride (Ach-CaCl2) by tail vein injection. We totally got 35 robust DEGs that mainly involve in extracellular matrix formation, leukocyte transendothelial migration, and chemokine signaling pathway. Among these DEGs, we identified three hub genes involved in AF, of which CXCL12/CXCR4 axis significantly upregulated in AF patients stands out as one of the most potent targets for AF prevention, and its effect on AF pathogenesis and underlying mechanisms were investigated in vivo subsequently with the specific CXCR4 antagonist AMD3100 (6 mg/kg). Our results demonstrated an elevated transcription and translation of CXCL12/CXCR4 axis in AF patients and mice, accompanied with the anabatic atrial inflammation and fibrosis, thereby providing the substrate for AF maintenance. Blocking its signaling via AMD3100 administration in AF model mice reduced AF inducibility and duration, partly ascribed to decreased atrial inflammation and structural remodeling. Mechanistically, these effects were achieved by reducing the recruitment of CD3+ T lymphocytes and F4/80+ macrophages, and suppressing the hyperactivation of ERK1/2 and AKT/mTOR signaling in atria of AF model mice. In conclusion, this study provides new evidence that antagonizing CXCR4 prevents the development of AF, and suggests that CXCL12/CXCR4 axis may be a potential therapeutic target for AF.
Collapse
Affiliation(s)
- Peng Liu
- Department of Cardiology, The Second Affiliate Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Hongke Sun
- Department of Cardiology, The Second Affiliate Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xin Zhou
- Department of Cardiology, The First Affiliate Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Qiaozhu Wang
- Department of Cardiology, The Second Affiliate Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Feng Gao
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Yuping Fu
- Department of Cardiology, The Second Affiliate Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Tong Li
- Department of Cardiology, The Second Affiliate Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yixin Wang
- Department of Cardiology, The Second Affiliate Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yingqi Li
- Department of Cardiology, The Second Affiliate Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Boyuan Fan
- Department of Cardiology, The Second Affiliate Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xiaoli Li
- Department of Cardiology, The Second Affiliate Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Tiannan Jiang
- Department of Internal Medicine, Health Care Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Xinghua Qin
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.
| | - Qiangsun Zheng
- Department of Cardiology, The Second Affiliate Hospital of Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
35
|
KLF15 negatively regulates cardiac fibrosis by which SDF-1β attenuates cardiac fibrosis in type 2 diabetic mice. Toxicol Appl Pharmacol 2021; 427:115654. [PMID: 34310909 DOI: 10.1016/j.taap.2021.115654] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 07/12/2021] [Accepted: 07/19/2021] [Indexed: 02/08/2023]
Abstract
Diabetic cardiomyopathy (DCM) is a serious diabetic complication that lacks effective preventive or therapeutic approaches. Wild-type and Klf15 knockout (Klf15-KO) mice were fed with either high fat diet (HFD, 60% kcal from fat) or normal diet (ND, 10% kcal from fat) for 3 months and then injected with streptozotocin or vehicle, to induce type 2 diabetes (T2D). All T2D and age-matched control mice were treated with or without SDF-1β at 5 mg/kg body-weight twice a week and also continually received HFD or ND for 3 months. At the end of 6-month study, after cardiac functions were measured, mice were euthanized to collect heart tissue. For in vitro mechanistic study, H9c2 cells were exposed to palmitate to mimic in vivo condition of T2D. SDF-1β prevented T2D-induced cardiac dysfunction and fibrosis and T2D-down-regulated KLF15 expression in wild-type diabetic heart tissue. However, the preventive effects of SDF-1β on both KLF15 expression and fibrosis was abolished, with partial cardiac protection in Klf15-KO/T2D mice. These results demonstrate partial KLF15-dependence for SDF-1β's cardiac fibrotic protection from T2D, but not on SDF-1β's protective effects on T2D-induced cardiac dysfunction. Further study showed that SDF-1β inhibited palmitate-induced cardiomyocyte fibrosis through its receptor CXCR7-mediated activation of p38β MAPK signaling pathway.
Collapse
|
36
|
Gao L, Wang L, Wei Y, Krishnamurthy P, Walcott GP, Menasché P, Zhang J. Exosomes secreted by hiPSC-derived cardiac cells improve recovery from myocardial infarction in swine. Sci Transl Med 2021; 12:12/561/eaay1318. [PMID: 32938792 DOI: 10.1126/scitranslmed.aay1318] [Citation(s) in RCA: 111] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 04/13/2020] [Accepted: 08/26/2020] [Indexed: 12/11/2022]
Abstract
Cell therapy treatment of myocardial infarction (MI) is mediated, in part, by exosomes secreted from transplanted cells. Thus, we compared the efficacy of treatment with a mixture of cardiomyocytes (CMs; 10 million), endothelial cells (ECs; 5 million), and smooth muscle cells (SMCs; 5 million) derived from human induced pluripotent stem cells (hiPSCs), or with exosomes extracted from the three cell types, in pigs after MI. Female pigs received sham surgery; infarction without treatment (MI group); or infarction and treatment with hiPSC-CMs, hiPSC-ECs, and hiPSC-SMCs (MI + Cell group); with homogenized fragments from the same dose of cells administered to the MI + Cell group (MI + Fra group); or with exosomes (7.5 mg) extracted from a 2:1:1 mixture of hiPSC-CMs:hiPSC-ECs:hiPSC-SMCs (MI + Exo group). Cells and exosomes were injected into the injured myocardium. In vitro, exosomes promoted EC tube formation and microvessel sprouting from mouse aortic rings and protected hiPSC-CMs by reducing apoptosis, maintaining intracellular calcium homeostasis, and increasing adenosine 5'-triphosphate. In vivo, measurements of left ventricular ejection fraction, wall stress, myocardial bioenergetics, cardiac hypertrophy, scar size, cell apoptosis, and angiogenesis in the infarcted region were better in the MI + Cell, MI + Fra, and MI + Exo groups than in the MI group 4 weeks after infarction. The frequencies of arrhythmic events in animals from the MI, MI + Cell, and MI + Exo groups were similar. Thus, exosomes secreted by hiPSC-derived cardiac cells improved myocardial recovery without increasing the frequency of arrhythmogenic complications and may provide an acellular therapeutic option for myocardial injury.
Collapse
Affiliation(s)
- Ling Gao
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL 35233, USA. .,Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200123, P.R. China
| | - Lu Wang
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Yuhua Wei
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Prasanna Krishnamurthy
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Gregory P Walcott
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Philippe Menasché
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL 35233, USA.,Department of Cardiovascular Surgery, Hôpital Européen Georges Pompidou, Université de Paris, PARCC, INSERM, F-75015 Paris, France
| | - Jianyi Zhang
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL 35233, USA.
| |
Collapse
|
37
|
Ghadge SK, Messner M, Seiringer H, Maurer T, Staggl S, Zeller T, Müller C, Börnigen D, Weninger WJ, Geyer SH, Sopper S, Krogsdam A, Pölzl G, Bauer A, Zaruba MM. Smooth Muscle Specific Ablation of CXCL12 in Mice Downregulates CXCR7 Associated with Defective Coronary Arteries and Cardiac Hypertrophy. Int J Mol Sci 2021; 22:ijms22115908. [PMID: 34072818 PMCID: PMC8198701 DOI: 10.3390/ijms22115908] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 05/26/2021] [Accepted: 05/28/2021] [Indexed: 12/27/2022] Open
Abstract
The chemokine CXCL12 plays a fundamental role in cardiovascular development, cell trafficking, and myocardial repair. Human genome-wide association studies even have identified novel loci downstream of the CXCL12 gene locus associated with coronary artery disease and myocardial infarction. Nevertheless, cell and tissue specific effects of CXCL12 are barely understood. Since we detected high expression of CXCL12 in smooth muscle (SM) cells, we generated a SM22-alpha-Cre driven mouse model to ablate CXCL12 (SM-CXCL12−/−). SM-CXCL12−/− mice revealed high embryonic lethality (50%) with developmental defects, including aberrant topology of coronary arteries. Postnatally, SM-CXCL12−/− mice developed severe cardiac hypertrophy associated with fibrosis, apoptotic cell death, impaired heart function, and severe coronary vascular defects characterized by thinned and dilated arteries. Transcriptome analyses showed specific upregulation of pathways associated with hypertrophic cardiomyopathy, collagen protein network, heart-related proteoglycans, and downregulation of the M2 macrophage modulators. CXCL12 mutants showed endothelial downregulation of the CXCL12 co-receptor CXCR7. Treatment of SM-CXCL12−/− mice with the CXCR7 agonist TC14012 attenuated cardiac hypertrophy associated with increased pERK signaling. Our data suggest a critical role of smooth muscle-specific CXCL12 in arterial development, vessel maturation, and cardiac hypertrophy. Pharmacological stimulation of CXCR7 might be a promising target to attenuate adverse hypertrophic remodeling.
Collapse
Affiliation(s)
- Santhosh Kumar Ghadge
- Department of Internal Medicine III, Cardiology and Angiology, Medical University Innsbruck, 6020 Innsbruck, Austria; (S.K.G.); (M.M.); (H.S.); (T.M.); (S.S.); (G.P.); (A.B.)
- Department of Medical Biochemistry, Max F. Perutz Laboratories (MFPL), Medical University of Vienna, 1090 Vienna, Austria
| | - Moritz Messner
- Department of Internal Medicine III, Cardiology and Angiology, Medical University Innsbruck, 6020 Innsbruck, Austria; (S.K.G.); (M.M.); (H.S.); (T.M.); (S.S.); (G.P.); (A.B.)
| | - Herbert Seiringer
- Department of Internal Medicine III, Cardiology and Angiology, Medical University Innsbruck, 6020 Innsbruck, Austria; (S.K.G.); (M.M.); (H.S.); (T.M.); (S.S.); (G.P.); (A.B.)
| | - Thomas Maurer
- Department of Internal Medicine III, Cardiology and Angiology, Medical University Innsbruck, 6020 Innsbruck, Austria; (S.K.G.); (M.M.); (H.S.); (T.M.); (S.S.); (G.P.); (A.B.)
| | - Simon Staggl
- Department of Internal Medicine III, Cardiology and Angiology, Medical University Innsbruck, 6020 Innsbruck, Austria; (S.K.G.); (M.M.); (H.S.); (T.M.); (S.S.); (G.P.); (A.B.)
| | - Tanja Zeller
- Clinic for Cardiology, Medical University Center Hamburg-Eppendorf, University Heart and Vascular Center Hamburg, 20251 Hamburg, Germany; (T.Z.); (C.M.); (D.B.)
| | - Christian Müller
- Clinic for Cardiology, Medical University Center Hamburg-Eppendorf, University Heart and Vascular Center Hamburg, 20251 Hamburg, Germany; (T.Z.); (C.M.); (D.B.)
| | - Daniela Börnigen
- Clinic for Cardiology, Medical University Center Hamburg-Eppendorf, University Heart and Vascular Center Hamburg, 20251 Hamburg, Germany; (T.Z.); (C.M.); (D.B.)
| | - Wolfgang J. Weninger
- Division of Anatomy & MIC, Medical University of Vienna, 1090 Vienna, Austria; (W.J.W.); (S.H.G.)
| | - Stefan H. Geyer
- Division of Anatomy & MIC, Medical University of Vienna, 1090 Vienna, Austria; (W.J.W.); (S.H.G.)
| | - Sieghart Sopper
- Department of Internal Medicine V, Hematology and Oncology, Medical University Innsbruck, 6020 Innsbruck, Austria;
| | - Anne Krogsdam
- Division of Bioinformatics, Medical University Innsbruck, Biocenter, 6020 Innsbruck, Austria;
| | - Gerhard Pölzl
- Department of Internal Medicine III, Cardiology and Angiology, Medical University Innsbruck, 6020 Innsbruck, Austria; (S.K.G.); (M.M.); (H.S.); (T.M.); (S.S.); (G.P.); (A.B.)
| | - Axel Bauer
- Department of Internal Medicine III, Cardiology and Angiology, Medical University Innsbruck, 6020 Innsbruck, Austria; (S.K.G.); (M.M.); (H.S.); (T.M.); (S.S.); (G.P.); (A.B.)
| | - Marc-Michael Zaruba
- Department of Internal Medicine III, Cardiology and Angiology, Medical University Innsbruck, 6020 Innsbruck, Austria; (S.K.G.); (M.M.); (H.S.); (T.M.); (S.S.); (G.P.); (A.B.)
- Correspondence:
| |
Collapse
|
38
|
Pluijmert NJ, Atsma DE, Quax PHA. Post-ischemic Myocardial Inflammatory Response: A Complex and Dynamic Process Susceptible to Immunomodulatory Therapies. Front Cardiovasc Med 2021; 8:647785. [PMID: 33996944 PMCID: PMC8113407 DOI: 10.3389/fcvm.2021.647785] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 03/02/2021] [Indexed: 01/04/2023] Open
Abstract
Following acute occlusion of a coronary artery causing myocardial ischemia and implementing first-line treatment involving rapid reperfusion, a dynamic and balanced inflammatory response is initiated to repair and remove damaged cells. Paradoxically, restoration of myocardial blood flow exacerbates cell damage as a result of myocardial ischemia-reperfusion (MI-R) injury, which eventually provokes accelerated apoptosis. In the end, the infarct size still corresponds to the subsequent risk of developing heart failure. Therefore, true understanding of the mechanisms regarding MI-R injury, and its contribution to cell damage and cell death, are of the utmost importance in the search for successful therapeutic interventions to finally prevent the onset of heart failure. This review focuses on the role of innate immunity, chemokines, cytokines, and inflammatory cells in all three overlapping phases following experimental, mainly murine, MI-R injury known as the inflammatory, reparative, and maturation phase. It provides a complete state-of-the-art overview including most current research of all post-ischemic processes and phases and additionally summarizes the use of immunomodulatory therapies translated into clinical practice.
Collapse
Affiliation(s)
- Niek J Pluijmert
- Department of Cardiology, Leiden University Medical Center, Leiden, Netherlands
| | - Douwe E Atsma
- Department of Cardiology, Leiden University Medical Center, Leiden, Netherlands
| | - Paul H A Quax
- Department of Surgery, Leiden University Medical Center, Leiden, Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
39
|
Zhu D, Cheng K. Cardiac Cell Therapy for Heart Repair: Should the Cells Be Left Out? Cells 2021; 10:641. [PMID: 33805763 PMCID: PMC7999733 DOI: 10.3390/cells10030641] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 02/25/2021] [Accepted: 03/10/2021] [Indexed: 12/11/2022] Open
Abstract
Cardiovascular disease (CVD) is still the leading cause of death worldwide. Coronary artery occlusion, or myocardial infarction (MI) causes massive loss of cardiomyocytes. The ischemia area is eventually replaced by a fibrotic scar. From the mechanical dysfunctions of the scar in electronic transduction, contraction and compliance, pathological cardiac dilation and heart failure develops. Once end-stage heart failure occurs, the only option is to perform heart transplantation. The sequential changes are termed cardiac remodeling, and are due to the lack of endogenous regenerative actions in the adult human heart. Regenerative medicine and biomedical engineering strategies have been pursued to repair the damaged heart and to restore normal cardiac function. Such strategies include both cellular and acellular products, in combination with biomaterials. In addition, substantial progress has been made to elucidate the molecular and cellular mechanisms underlying heart repair and regeneration. In this review, we summarize and discuss current therapeutic approaches for cardiac repair and provide a perspective on novel strategies that holding potential opportunities for future research and clinical translation.
Collapse
Affiliation(s)
- Dashuai Zhu
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC 27607, USA;
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill & North Carolina State University, Raleigh, NC 27607, USA
| | - Ke Cheng
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC 27607, USA;
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill & North Carolina State University, Raleigh, NC 27607, USA
| |
Collapse
|
40
|
Bennington J, Lankford S, Magalhaes RS, Shankle D, Fanning J, Kartini C, Suparto I, Kusumawardhani W, Putra MA, Mariya S, Badlani G, Williams JK. Chemokine Therapy in Cats With Experimental Renal Fibrosis and in a Kidney Disease Pilot Study. Front Vet Sci 2021; 8:646087. [PMID: 33748219 PMCID: PMC7969654 DOI: 10.3389/fvets.2021.646087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Accepted: 02/12/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Chronic tubulointerstitial fibrosis is a common final pathway leading to end stage kidney disease in cats and has no effective treatment. The use of cell-based molecules to treat kidney fibrosis may be a promising approach. The objectives were to test the effects of intra-renal chemokine CXCL12 injection in a pre-clinical cat model of unilateral ischemia/reperfusion (I/R)-induced kidney fibrosis and then, within a clinical pilot study, test the safety/feasibility of CXCL12 injection in cats that might have early chronic kidney disease (CKD). Methods: Pre-clinical: Thirty cats received intra-renal injection of 100, 200, or 400 ng of recombinant human CXCL12, or sterile saline, into the I/R kidney 70 days post-injury, or were non-injured, non-injected controls (n = 6/group). Kidney collagen content was quantified 4 months post-treatment using Masson's Trichrome and Picrosirius Red (PSR) stained tissues. In a separate study (n = 2) exploring short-term effects of CXCL12, 200 ng CXCL12 was injected into I/R kidneys and then harvested either 30 min (n = 1) or 1 month (n = 1) post-injection. Kidney concentrations of CXCL12, matrix metalloproteinase 1 (MMP-1), and lysyl oxidase-like enzyme 2 (LOXL-2) were quantified via ELISA. Clinical Pilot: 14 client-owned cats with potential early kidney disease received a single-treatment, bilateral intra-renal injection of 200 ng CXCL12 (n = 7), or received no injection (n = 7). Blood/urine samples were collected monthly for 9 months to assess renal function and CKD staging. Results: Pre-clinical: I/R increased the affected kidney collagen content, which both mid and high doses of CXCL12 restored to normal (ps < 0.05 vs. untreated). I/R increased collagen fiber width, which both mid and high doses of CXCL12 restored to normal (p < 0.001 vs. untreated). Early changes in kidney MMP-1, associated with collagen breakdown, and subsequent decreases in LOXL-2, associated with collagen cross-linking, in response to CXCL12 treatment may contribute to these findings. Clinical Pilot: Bilateral intra-renal injection of CXCL12 using ultrasound guidance in cats with CKD was feasible and safe in a general practice clinical setting with no obvious side effects noted during the 9-month follow-up period. Conclusions: Intra-renal injection of CXCL12 may prove to be an effective treatment for kidney fibrosis in cats with CKD. Additional mechanistic and clinical evaluations are needed.
Collapse
Affiliation(s)
- Julie Bennington
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC, United States
| | - Shannon Lankford
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC, United States
| | - Renata S. Magalhaes
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC, United States
| | - Douglas Shankle
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC, United States
| | - Jason Fanning
- Department of Health and Exercise Science, Wake Forest University, Winston-Salem, NC, United States
| | - Cucu Kartini
- Praktek Dokter Hewan Bersama Joint Veterinary Practice, Sunter, Indonesia
| | - Irma Suparto
- Primate Research Center, Institut Pertanian Bogor, Bogor Agricultural University, Bogor, Indonesia
| | | | - M. ArRaniri Putra
- Praktek Dokter Hewan Bersama Joint Veterinary Practice, Sunter, Indonesia
| | - Silmi Mariya
- Primate Research Center, Institut Pertanian Bogor, Bogor Agricultural University, Bogor, Indonesia
| | - Gopal Badlani
- Department of Urology, Wake Forest Baptist Health, Winston-Salem, NC, United States
| | - J. Koudy Williams
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC, United States
| |
Collapse
|
41
|
CXCR7 ameliorates myocardial infarction as a β-arrestin-biased receptor. Sci Rep 2021; 11:3426. [PMID: 33564089 PMCID: PMC7873251 DOI: 10.1038/s41598-021-83022-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 01/28/2021] [Indexed: 02/08/2023] Open
Abstract
Most seven transmembrane receptors (7TMRs) are G protein-coupled receptors; however, some 7TMRs evoke intracellular signals through β-arrestin as a biased receptor. As several β-arrestin-biased agonists have been reported to be cardioprotective, we examined the role of the chemokine receptor CXCR7 as a β-arrestin-biased receptor in the heart. Among 510 7TMR genes examined, Cxcr7 was the most abundantly expressed in the murine heart. Single-cell RNA-sequencing analysis revealed that Cxcr7 was abundantly expressed in cardiomyocytes and fibroblasts. Cardiomyocyte-specific Cxcr7 null mice showed more prominent cardiac dilatation and dysfunction than control mice 4 weeks after myocardial infarction. In contrast, there was no difference in cardiac phenotypes between fibroblast-specific Cxcr7-knockout mice and control mice even after myocardial infarction. TC14012, a specific agonist of CXCR7, significantly recruited β-arrestin to CXCR7 in CXCR7-expressing cells and activated extracellular signal-regulated kinase (ERK) in neonatal rat cardiomyocytes. Cxcr7 expression was significantly increased and ERK was activated in the border zone of the heart in control, but not Cxcr7 null mice. These results indicate that the abundantly expressed CXCR7 in cardiomyocytes may play a protective role in the heart as a β-arrestin-biased receptor and that CXCR7 may be a novel therapeutic target for myocardial infarction.
Collapse
|
42
|
Chen T, Qiao X, Cheng L, Liu M, Deng Y, Zhuo X. LGR4 silence aggravates ischemic injury by modulating mitochondrial function and oxidative stress via ERK signaling pathway in H9c2 cells. J Mol Histol 2021; 52:363-371. [PMID: 33559814 DOI: 10.1007/s10735-021-09957-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 01/18/2021] [Indexed: 11/29/2022]
Abstract
It is reported that LGR4 (leucine-rich repeat domain containing G protein-coupled receptor 4) plays a crucial role in the physiological function of many organs. However, few data are available on the function and mechanism of LGR4 in myocardial ischemia-reperfusion (I/R) injury. The aim of this study was to explore the function and mechanism of LGR4 in I/R injury. We incubated H9c2 cells in simulating ischemia buffer and then re-incubated them in normal culture medium to establish a model of I/R injury in vitro. The expression of LGR4 was evaluated by RT-PCR and western blot. Besides, the cell apoptosis was evaluated by flow cytometric analysis and the content of ROS, SOD, MDA, LDH, CK, ATP, cyt c were detected by special commercial kits. The expression of mitochondrial function-related proteins were detected by western blot. Then, the roles of ERK signaling pathway was determined with TBHQ (ERK activator) treatment. Our data have demonstrated that I/R boosted the expression of LGR4 in H9c2 cells. Knockdown of LGR4 increased the apoptosis rate of H9c2 cells and led to excessed oxidant stress and impaired mitochondrial function by increasing the levels of ROS, MDA, LDH, CK and cyt c and inhibiting SOD activity, ATP production. In addition, LGR4 silence inhibited the activation of ERK pathway. And TBHQ partially reversed the effects of LGR4 knockdown on H9c2 cells. To conclude, our study indicated that LGR4 regulated mitochondrial dysfunction and oxidative stress by ERK signaling pathways, which provides a potential cardiac protective target against I/R.
Collapse
Affiliation(s)
- Tao Chen
- Department of Cardiovascular Medicine, First Affiliated Hospital of Medical School, Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, Shaanxi, China
| | - Xiangrui Qiao
- Department of Cardiovascular Medicine, First Affiliated Hospital of Medical School, Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, Shaanxi, China
| | - Lele Cheng
- Department of Cardiovascular Medicine, First Affiliated Hospital of Medical School, Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, Shaanxi, China
| | - Mengping Liu
- Department of Cardiovascular Medicine, First Affiliated Hospital of Medical School, Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, Shaanxi, China
| | - Yangyang Deng
- Department of Cardiovascular Medicine, First Affiliated Hospital of Medical School, Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, Shaanxi, China
| | - Xiaozhen Zhuo
- Department of Cardiovascular Medicine, First Affiliated Hospital of Medical School, Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, Shaanxi, China.
| |
Collapse
|
43
|
Kontos C, El Bounkari O, Krammer C, Sinitski D, Hille K, Zan C, Yan G, Wang S, Gao Y, Brandhofer M, Megens RTA, Hoffmann A, Pauli J, Asare Y, Gerra S, Bourilhon P, Leng L, Eckstein HH, Kempf WE, Pelisek J, Gokce O, Maegdefessel L, Bucala R, Dichgans M, Weber C, Kapurniotu A, Bernhagen J. Designed CXCR4 mimic acts as a soluble chemokine receptor that blocks atherogenic inflammation by agonist-specific targeting. Nat Commun 2020; 11:5981. [PMID: 33239628 PMCID: PMC7689490 DOI: 10.1038/s41467-020-19764-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 10/28/2020] [Indexed: 11/22/2022] Open
Abstract
Targeting a specific chemokine/receptor axis in atherosclerosis remains challenging. Soluble receptor-based strategies are not established for chemokine receptors due to their discontinuous architecture. Macrophage migration-inhibitory factor (MIF) is an atypical chemokine that promotes atherosclerosis through CXC-motif chemokine receptor-4 (CXCR4). However, CXCR4/CXCL12 interactions also mediate atheroprotection. Here, we show that constrained 31-residue-peptides ('msR4Ms') designed to mimic the CXCR4-binding site to MIF, selectively bind MIF with nanomolar affinity and block MIF/CXCR4 without affecting CXCL12/CXCR4. We identify msR4M-L1, which blocks MIF- but not CXCL12-elicited CXCR4 vascular cell activities. Its potency compares well with established MIF inhibitors, whereas msR4M-L1 does not interfere with cardioprotective MIF/CD74 signaling. In vivo-administered msR4M-L1 enriches in atherosclerotic plaques, blocks arterial leukocyte adhesion, and inhibits atherosclerosis and inflammation in hyperlipidemic Apoe-/- mice in vivo. Finally, msR4M-L1 binds to MIF in plaques from human carotid-endarterectomy specimens. Together, we establish an engineered GPCR-ectodomain-based mimicry principle that differentiates between disease-exacerbating and -protective pathways and chemokine-selectively interferes with atherosclerosis.
Collapse
MESH Headings
- Aged
- Animals
- Antigens, CD/metabolism
- Atherosclerosis/drug therapy
- Atherosclerosis/genetics
- Atherosclerosis/pathology
- Atherosclerosis/surgery
- Binding Sites
- Carotid Artery, Common/pathology
- Carotid Artery, Common/surgery
- Chemokine CXCL12/metabolism
- Crystallography, X-Ray
- Disease Models, Animal
- Drug Design
- Drug Evaluation, Preclinical
- Endarterectomy, Carotid
- Female
- Humans
- Intramolecular Oxidoreductases/antagonists & inhibitors
- Intramolecular Oxidoreductases/metabolism
- Macrophage Migration-Inhibitory Factors/antagonists & inhibitors
- Macrophage Migration-Inhibitory Factors/metabolism
- Male
- Mice
- Mice, Knockout, ApoE
- Middle Aged
- Peptide Fragments/pharmacology
- Peptide Fragments/therapeutic use
- Receptors, CXCR4/chemistry
- Receptors, CXCR4/metabolism
- Receptors, CXCR4/ultrastructure
- Sialyltransferases/metabolism
- Signal Transduction/drug effects
Collapse
Affiliation(s)
- Christos Kontos
- Division of Peptide Biochemistry, TUM School of Life Sciences, Technische Universität München (TUM), 85354, Freising, Germany
| | - Omar El Bounkari
- Institute for Stroke and Dementia Research (ISD), Klinikum der Universität München, Ludwig-Maximilians-Universität (LMU) München, 81377, Munich, Germany
| | - Christine Krammer
- Institute for Stroke and Dementia Research (ISD), Klinikum der Universität München, Ludwig-Maximilians-Universität (LMU) München, 81377, Munich, Germany
| | - Dzmitry Sinitski
- Institute for Stroke and Dementia Research (ISD), Klinikum der Universität München, Ludwig-Maximilians-Universität (LMU) München, 81377, Munich, Germany
| | - Kathleen Hille
- Division of Peptide Biochemistry, TUM School of Life Sciences, Technische Universität München (TUM), 85354, Freising, Germany
| | - Chunfang Zan
- Institute for Stroke and Dementia Research (ISD), Klinikum der Universität München, Ludwig-Maximilians-Universität (LMU) München, 81377, Munich, Germany
| | - Guangyao Yan
- Institute for Stroke and Dementia Research (ISD), Klinikum der Universität München, Ludwig-Maximilians-Universität (LMU) München, 81377, Munich, Germany
| | - Sijia Wang
- Institute for Stroke and Dementia Research (ISD), Klinikum der Universität München, Ludwig-Maximilians-Universität (LMU) München, 81377, Munich, Germany
| | - Ying Gao
- Institute for Stroke and Dementia Research (ISD), Klinikum der Universität München, Ludwig-Maximilians-Universität (LMU) München, 81377, Munich, Germany
| | - Markus Brandhofer
- Institute for Stroke and Dementia Research (ISD), Klinikum der Universität München, Ludwig-Maximilians-Universität (LMU) München, 81377, Munich, Germany
| | - Remco T A Megens
- Institute for Cardiovascular Prevention, Klinikum der Universität München, Ludwig-Maximilians-Universität (LMU) München, 80336, Munich, Germany
| | - Adrian Hoffmann
- Institute for Stroke and Dementia Research (ISD), Klinikum der Universität München, Ludwig-Maximilians-Universität (LMU) München, 81377, Munich, Germany
- Department of Anaesthesiology, Klinikum der Universität München, Ludwig-Maximilians-Universität (LMU) München, 81377, Munich, Germany
| | - Jessica Pauli
- Department for Vascular and Endovascular Surgery, Klinikum rechts der Isar, Technische Universität München (TUM), 81675, Munich, Germany
| | - Yaw Asare
- Institute for Stroke and Dementia Research (ISD), Klinikum der Universität München, Ludwig-Maximilians-Universität (LMU) München, 81377, Munich, Germany
| | - Simona Gerra
- Institute for Stroke and Dementia Research (ISD), Klinikum der Universität München, Ludwig-Maximilians-Universität (LMU) München, 81377, Munich, Germany
| | - Priscila Bourilhon
- Institute for Stroke and Dementia Research (ISD), Klinikum der Universität München, Ludwig-Maximilians-Universität (LMU) München, 81377, Munich, Germany
| | - Lin Leng
- Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Hans-Henning Eckstein
- Department for Vascular and Endovascular Surgery, Klinikum rechts der Isar, Technische Universität München (TUM), 81675, Munich, Germany
| | - Wolfgang E Kempf
- Department for Vascular and Endovascular Surgery, Klinikum rechts der Isar, Technische Universität München (TUM), 81675, Munich, Germany
| | - Jaroslav Pelisek
- Department for Vascular and Endovascular Surgery, Klinikum rechts der Isar, Technische Universität München (TUM), 81675, Munich, Germany
- Department of Vascular Surgery, University Hospital Zurich, 8091, Zurich, Switzerland
| | - Ozgun Gokce
- Institute for Stroke and Dementia Research (ISD), Klinikum der Universität München, Ludwig-Maximilians-Universität (LMU) München, 81377, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377, Munich, Germany
| | - Lars Maegdefessel
- Department for Vascular and Endovascular Surgery, Klinikum rechts der Isar, Technische Universität München (TUM), 81675, Munich, Germany
| | - Richard Bucala
- Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Martin Dichgans
- Institute for Stroke and Dementia Research (ISD), Klinikum der Universität München, Ludwig-Maximilians-Universität (LMU) München, 81377, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377, Munich, Germany
| | - Christian Weber
- Institute for Cardiovascular Prevention, Klinikum der Universität München, Ludwig-Maximilians-Universität (LMU) München, 80336, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377, Munich, Germany
- Munich Heart Alliance, 80802, Munich, Germany
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229, Maastricht, The Netherlands
| | - Aphrodite Kapurniotu
- Division of Peptide Biochemistry, TUM School of Life Sciences, Technische Universität München (TUM), 85354, Freising, Germany.
| | - Jürgen Bernhagen
- Institute for Stroke and Dementia Research (ISD), Klinikum der Universität München, Ludwig-Maximilians-Universität (LMU) München, 81377, Munich, Germany.
- Munich Cluster for Systems Neurology (SyNergy), 81377, Munich, Germany.
- Munich Heart Alliance, 80802, Munich, Germany.
| |
Collapse
|
44
|
Fernández AR, Sánchez-Tarjuelo R, Cravedi P, Ochando J, López-Hoyos M. Review: Ischemia Reperfusion Injury-A Translational Perspective in Organ Transplantation. Int J Mol Sci 2020; 21:ijms21228549. [PMID: 33202744 PMCID: PMC7696417 DOI: 10.3390/ijms21228549] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 11/10/2020] [Accepted: 11/11/2020] [Indexed: 02/07/2023] Open
Abstract
Thanks to the development of new, more potent and selective immunosuppressive drugs together with advances in surgical techniques, organ transplantation has emerged from an experimental surgery over fifty years ago to being the treatment of choice for many end-stage organ diseases, with over 139,000 organ transplants performed worldwide in 2019. Inherent to the transplantation procedure is the fact that the donor organ is subjected to blood flow cessation and ischemia during harvesting, which is followed by preservation and reperfusion of the organ once transplanted into the recipient. Consequently, ischemia/reperfusion induces a significant injury to the graft with activation of the immune response in the recipient and deleterious effect on the graft. The purpose of this review is to discuss and shed new light on the pathways involved in ischemia/reperfusion injury (IRI) that act at different stages during the donation process, surgery, and immediate post-transplant period. Here, we present strategies that combine various treatments targeted at different mechanistic pathways during several time points to prevent graft loss secondary to the inflammation caused by IRI.
Collapse
Affiliation(s)
- André Renaldo Fernández
- Immunology, Universitary Hospital Marqués de Valdecilla- Research Institute IDIVAL Santander, 390008 Santander, Spain;
| | - Rodrigo Sánchez-Tarjuelo
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (R.S.-T.); (J.O.)
- Immunología de Trasplantes, Centro Nacional de Microbiología, Instituto de Salud Carlos III, 28220 Majadahonda (Madrid), Spain
| | - Paolo Cravedi
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
| | - Jordi Ochando
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (R.S.-T.); (J.O.)
- Immunología de Trasplantes, Centro Nacional de Microbiología, Instituto de Salud Carlos III, 28220 Majadahonda (Madrid), Spain
| | - Marcos López-Hoyos
- Immunology, Universitary Hospital Marqués de Valdecilla- Research Institute IDIVAL Santander, 390008 Santander, Spain;
- Red de Investigación Renal (REDINREN), 28040 Madrid, Spain
- Correspondence: ; Tel.: +34-942-292759
| |
Collapse
|
45
|
Hess A, Thackeray JT, Wollert KC, Bengel FM. Radionuclide Image-Guided Repair of the Heart. JACC Cardiovasc Imaging 2020; 13:2415-2429. [DOI: 10.1016/j.jcmg.2019.11.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 10/23/2019] [Accepted: 11/05/2019] [Indexed: 01/12/2023]
|
46
|
Ji Y, Yao J, He Y. Extracellular ubiquitin protects cardiomyocytes during ischemia/hypoxia by inhibiting mitochondrial apoptosis pathway through CXCR4. Biomed Pharmacother 2020; 131:110787. [PMID: 33152945 DOI: 10.1016/j.biopha.2020.110787] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 09/13/2020] [Accepted: 09/18/2020] [Indexed: 10/23/2022] Open
Abstract
AIM Acute myocardial infarction (AMI) is one of the deadliest diseases worldwide. The search for countermeasures to reduce cardiomyocytes death in the infarcted area has always been the focus of research. Ubiquitin (UB) is a small polypeptide mainly involved in proteasome-mediated protein degradation in cells, whereas extracellular UB in body fluids can also function through its receptor CXC chemokine receptor type 4 (CXCR4). This study aimed to explore the functional roles of extracellular UB in cardiomyocytes during ischemia/hypoxia (I/H). METHODS H9C2 cells were subjected to I/H treatment and cell injury was evaluated by cell viability, morphology changes and apoptosis rate. UB expression and levels of ubiquitinated proteins after I/H injury were measured. The effects of extracellular UB on I/H-induced cardiomyocytes apoptosis and the possible underlying mechanisms were studied. RESULTS I/H injury induced the decrease of cell viability as well as enhanced impaired cell morphology and apoptosis rate in H9C2 cells. Levels of UB mRNA and ubiquitinated proteins were significantly up-regulated after I/H treatment, whereas the concentration of extracellular UB in the conditioned media did not show significant change and the intracellular mono-UB levels in cells were down-regulated. Extracellular UB treatment protected cardiomyocytes from I/H injury by inhibiting the overactivation of mitochondria-dependent apoptosis pathway and up-regulating autophagy level. Inhibition of CXCR4 receptor using AMD3100 abolished cardioprotective effects of extracellular UB. CONCLUSION The up-regulation of UB was suggested to be an adaptive response to resist I/H-induced cardiomyocytes apoptosis, and additional extracellular UB treatment might serve as a new potential therapeutic drug for AMI.
Collapse
Affiliation(s)
- Yiqun Ji
- MOE Engineering Center of Hematological Disease, MOH Key Lab of Thrombosis and Hemostasis, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Jialu Yao
- Department of Cardiology, Suzhou Municipal Hospital, Suzhou, Jiangsu Province, China
| | - Yang He
- MOE Engineering Center of Hematological Disease, MOH Key Lab of Thrombosis and Hemostasis, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China.
| |
Collapse
|
47
|
Tehrani BN, Basir MB, Kapur NK. Acute myocardial infarction and cardiogenic shock: Should we unload the ventricle before percutaneous coronary intervention? Prog Cardiovasc Dis 2020; 63:607-622. [PMID: 32920027 DOI: 10.1016/j.pcad.2020.09.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 09/03/2020] [Indexed: 12/22/2022]
Abstract
Despite early reperfusion and coordinated systems of care, cardiogenic shock (CS) remains the number one cause of morbidity and in-hospital mortality following acute myocardial infarction (AMI). CS is a complex clinical syndrome that begins with hemodynamic instability and can progress to multi-organ failure and profound hemo-metabolic compromise. To improve outcomes, a clear understanding of the treatment objectives in CS and developing time-sensitive management strategies aimed at stabilizing hemodynamics and restoring myocardial perfusion are critical. Left ventricular (LV) load has been identified as an independent predictor of heart failure and mortality following AMI. Decades of preclinical and clinical research have identified several effective LV unloading strategies. Recent initiatives from single and multi-center registries and more recently the Door to Unload (DTU)-STEMI pilot study have provided valuable insight to developing a standardized treatment approach to AMI, based on early invasive hemodynamics and tailored circulatory support to unload the LV. To follow is a review of the pathophysiology and prevalence of shock, limitations of current therapies, and the pre-clinical and translational basis for incorporating LV unloading into contemporary AMI and shock care.
Collapse
Affiliation(s)
- Behnam N Tehrani
- Inova Heart and Vascular Institute, Falls Church, VA, United States of America
| | - Mir B Basir
- Henry Ford Medical Center, Detroit, MI, United States of America
| | - Navin K Kapur
- The CardioVascular Center, Tufts Medical Center, Boston, MA, United States of America.
| |
Collapse
|
48
|
Murphy JW, Rajasekaran D, Merkel J, Skeens E, Keeler C, Hodsdon ME, Lisi GP, Lolis E. High-Throughput Screening of a Functional Human CXCL12-CXCR4 Signaling Axis in a Genetically Modified S. cerevisiae: Discovery of a Novel Up-Regulator of CXCR4 Activity. Front Mol Biosci 2020; 7:164. [PMID: 32766282 PMCID: PMC7378375 DOI: 10.3389/fmolb.2020.00164] [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: 03/17/2020] [Accepted: 06/25/2020] [Indexed: 12/03/2022] Open
Abstract
CXCL12 activates CXCR4 and is involved in embryogenesis, hematopoiesis, and angiogenesis. It has pathological roles in HIV-1, WHIM disease, cancer, and autoimmune diseases. An antagonist, AMD3100, is used for the release of CD34+ hematopoietic stem cells from the bone marrow for autologous transplantation for lymphoma or multiple myeloma patients. Adverse effects are tolerated due to its short-term treatment, but AMD3100 is cardiotoxic in clinical studies for HIV-1. In an effort to determine whether Saccharomyces cerevisiae expressing a functional human CXCR4 could be used as a platform for identifying a ligand from a library of less ∼1,000 compounds, a high-throughput screening was developed. We report that 2-carboxyphenyl phosphate (fosfosal) up-regulates CXCR4 activation only in the presence of CXCL12. This is the first identification of a compound that increases CXCR4 activity by any mechanism. We mapped the fosfosal binding site on CXCL12, described its mechanism of action, and studied its chemical components, salicylate and phosphate, to conclude that they synergize to achieve the functional effect.
Collapse
Affiliation(s)
- James W Murphy
- Department of Pharmacology, Yale School of Medicine, Yale University, New Haven, CT, United States
| | - Deepa Rajasekaran
- Department of Pharmacology, Yale School of Medicine, Yale University, New Haven, CT, United States
| | - Janie Merkel
- Yale Center for Molecular Discovery, Yale School of Medicine, Yale University, New Haven, CT, United States
| | - Erin Skeens
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, United States
| | - Camille Keeler
- Department of Laboratory Medicine, Yale School of Medicine, Yale University, New Haven, CT, United States
| | - Michael E Hodsdon
- Department of Laboratory Medicine, Yale School of Medicine, Yale University, New Haven, CT, United States.,Yale Cancer Center, New Haven, CT, United States
| | - George P Lisi
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, United States
| | - Elias Lolis
- Department of Pharmacology, Yale School of Medicine, Yale University, New Haven, CT, United States.,Yale Cancer Center, New Haven, CT, United States
| |
Collapse
|
49
|
Marín-Juez R, El-Sammak H, Helker CSM, Kamezaki A, Mullapuli ST, Bibli SI, Foglia MJ, Fleming I, Poss KD, Stainier DYR. Coronary Revascularization During Heart Regeneration Is Regulated by Epicardial and Endocardial Cues and Forms a Scaffold for Cardiomyocyte Repopulation. Dev Cell 2020; 51:503-515.e4. [PMID: 31743664 DOI: 10.1016/j.devcel.2019.10.019] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 07/14/2019] [Accepted: 10/16/2019] [Indexed: 12/21/2022]
Abstract
Defective coronary network function and insufficient blood supply are both cause and consequence of myocardial infarction. Efficient revascularization after infarction is essential to support tissue repair and function. Zebrafish hearts exhibit a remarkable ability to regenerate, and coronary revascularization initiates within hours of injury, but how this process is regulated remains unknown. Here, we show that revascularization requires a coordinated multi-tissue response culminating with the formation of a complex vascular network available as a scaffold for cardiomyocyte repopulation. During a process we term "coronary-endocardial anchoring," new coronaries respond by sprouting (1) superficially within the regenerating epicardium and (2) intra-ventricularly toward the activated endocardium. Mechanistically, superficial revascularization is guided by epicardial Cxcl12-Cxcr4 signaling and intra-ventricular sprouting by endocardial Vegfa signaling. Our findings indicate that the injury-activated epicardium and endocardium support cardiomyocyte replenishment initially through the guidance of coronary sprouting. Simulating this process in the injured mammalian heart should help its healing.
Collapse
Affiliation(s)
- Rubén Marín-Juez
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany; German Center of Cardiovascular Research (DZHK), Partner site RheinMain, 60590 Frankfurt am Main, Germany.
| | - Hadil El-Sammak
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany; German Center of Cardiovascular Research (DZHK), Partner site RheinMain, 60590 Frankfurt am Main, Germany
| | - Christian S M Helker
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany; German Center of Cardiovascular Research (DZHK), Partner site RheinMain, 60590 Frankfurt am Main, Germany
| | - Aosa Kamezaki
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany; German Center of Cardiovascular Research (DZHK), Partner site RheinMain, 60590 Frankfurt am Main, Germany
| | - Sri Teja Mullapuli
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany; German Center of Cardiovascular Research (DZHK), Partner site RheinMain, 60590 Frankfurt am Main, Germany
| | - Sofia-Iris Bibli
- German Center of Cardiovascular Research (DZHK), Partner site RheinMain, 60590 Frankfurt am Main, Germany; Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, 60590 Frankfurt am Main, Germany
| | - Matthew J Foglia
- Regeneration Next, Duke University, Durham, NC 27710, USA; Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Ingrid Fleming
- German Center of Cardiovascular Research (DZHK), Partner site RheinMain, 60590 Frankfurt am Main, Germany; Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, 60590 Frankfurt am Main, Germany
| | - Kenneth D Poss
- Regeneration Next, Duke University, Durham, NC 27710, USA; Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Didier Y R Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany; German Center of Cardiovascular Research (DZHK), Partner site RheinMain, 60590 Frankfurt am Main, Germany.
| |
Collapse
|
50
|
Clinical relevance of single nucleotide polymorphisms in the CXCL1 and CXCL12 genes in patients with major trauma. J Trauma Acute Care Surg 2020; 86:440-447. [PMID: 30489503 DOI: 10.1097/ta.0000000000002141] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
BACKGROUND Genetic backgrounds have been recognized as significant determinants of susceptibility to sepsis. CXC chemokines play a significant role in innate immunity against infectious diseases. Genetic polymorphisms of CXC chemokine genes have been widely studied in inflammatory and infectious diseases but not in sepsis. Thus, we aimed to investigate the clinical relevance of CXC chemokine gene polymorphisms and susceptibility to sepsis in a traumatically injured population. METHODS Thirteen tag single nucleotide polymorphisms were selected from CXC chemokine genes using a multimarker tagging algorithm in the Tagger software. Three independent cohorts of injured patients (n = 1700) were prospectively recruited. Selected single nucleotide polymorphisms were genotyped using an improved multiplex ligation detection reaction method. Cytokine production in lipopolysaccharide-stimulated whole blood was measured using an enzyme-linked immunosorbent assay. RESULTS Among the 13 tag single nucleotide polymorphisms, four single nucleotide polymorphisms (rs1429638, rs266087, rs2297630, and rs2839693) were significantly associated with the susceptibility to sepsis, and three (rs3117604, rs1429638, and rs4074) were significantly associated with an increased multiple organ dysfunction score in the derivation cohort. However, only the clinical relevance of rs1429638 and rs266087 was confirmed in the validation cohorts. In addition, rs2297630 was significantly associated with interleukin 6 production. CONCLUSION The rs1429638 polymorphism in the CXCL1 gene and the rs2297630 polymorphism in the CXCL12 gene were associated with altered susceptibility to sepsis and might be used as important genetic markers to assess the risks of sepsis in trauma patients. LEVEL OF EVIDENCE Prognostic and epidemiologic study, level II.
Collapse
|