1
|
Salybekov AA, Tashov K, Sheng Y, Salybekova A, Shinozaki Y, Asahara T, Kobayashi S. Cardioimmunology in Health and Diseases: Impairment of the Cardio-Spleno-Bone Marrow Axis Following Myocardial Infarction in Diabetes Mellitus. Int J Mol Sci 2024; 25:11833. [PMID: 39519382 PMCID: PMC11546687 DOI: 10.3390/ijms252111833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 10/31/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
A comprehensive understanding of the cardio-spleen-bone marrow immune cell axis is essential for elucidating the alterations occurring during the pathogenesis of diabetes mellitus (DM). This study investigates the dynamics of immune cell kinetics in DM after myocardial infarction (MI) over time. MI was induced in diabetic and healthy control groups using C57BL/N6 mice, with sacrifices occurring at days 1, 3, 7, and 28 post-MI to collect heart, peripheral blood (PB), spleen, and bone marrow (BM) samples. Cell suspensions from each organ were isolated and analyzed via flow cytometry. Additionally, the endothelial progenitor cell-colony-forming assay (EPC-CFA) was performed using mononuclear cells derived from BM, PB, and the spleen. The results indicated that, despite normal production in BM and the spleen, CD45+ cells were lower in the PB of DM mice at days 1 to 3. Further analysis revealed a reduction in total and pro-inflammatory neutrophils (N1s) in PB at days 1 to 3 and in the spleen at days 3 to 7 in DM mice, suggesting that DM-induced alterations in splenic neutrophils fail to meet the demand in PB and ischemic tissues. Infiltrating macrophages (total, M1, M2) were reduced at day 3 in the DM-ischemic heart, with total and M1 (days 1-3) and M2 (days 3-7) macrophages being significantly decreased in DM-PB compared to controls, indicating impaired macrophage recruitment and polarization in DM. Myeloid dendritic cells (mDCs) in the heart were higher from days 1 to 7, which corresponded with the enhanced recruitment of CD8+ cells from days 1 to 28 in the DM-infarcted myocardium. Total CD4+ cells decreased in DM-PB at days 1 to 3, suggesting a delayed adaptive immune response to MI. B cells were reduced in PB at days 1 to 3, in myocardium at day 3, and in the spleen at day 7, indicating compromised mobilization from BM. EPC-CFA results showed a marked decrease in definitive EPC colonies in the spleen and BM from days 1 to 28 in DM mice compared to controls in vitro, highlighting that DM severely impairs EPC colony-forming activity by limiting the differentiation of EPCs from primitive to definitive forms. Taking together, this study underscores significant disruptions in the cardio-spleen-bone marrow immune cell axis following MI in DM, revealing delayed innate and adaptive immune responses along with impaired EPC differentiation.
Collapse
Affiliation(s)
- Amankeldi A. Salybekov
- Kidney Disease and Transplant Center, Shonan Kamakura General Hospital, Kamakura 247-8533, Japan;
- Shonan Research Institute of Innovative Medicine, Shonan Kamakura General Hospital, Kamakura 247-8533, Japan
- Department of Advanced Medicine Science, Tokai University School of Medicine, Isehara 259-1193, Japan; (K.T.); (Y.S.)
| | - Kanat Tashov
- Department of Advanced Medicine Science, Tokai University School of Medicine, Isehara 259-1193, Japan; (K.T.); (Y.S.)
| | - Yin Sheng
- Department of Advanced Medicine Science, Tokai University School of Medicine, Isehara 259-1193, Japan; (K.T.); (Y.S.)
| | - Ainur Salybekova
- Department of Advanced Medicine Science, Tokai University School of Medicine, Isehara 259-1193, Japan; (K.T.); (Y.S.)
| | - Yoshiko Shinozaki
- Teaching and Research Support Core Center, Tokai University School of Medicine, Isehara 259-1193, Japan
| | - Takayuki Asahara
- Shonan Research Institute of Innovative Medicine, Shonan Kamakura General Hospital, Kamakura 247-8533, Japan
- Department of Advanced Medicine Science, Tokai University School of Medicine, Isehara 259-1193, Japan; (K.T.); (Y.S.)
| | - Shuzo Kobayashi
- Kidney Disease and Transplant Center, Shonan Kamakura General Hospital, Kamakura 247-8533, Japan;
- Shonan Research Institute of Innovative Medicine, Shonan Kamakura General Hospital, Kamakura 247-8533, Japan
| |
Collapse
|
2
|
Li Z, Wang S, Qin Y, Yang B, Wang C, Lu T, Xu J, Zhu L, Yuan C, Han W. Gabapentin attenuates cardiac remodeling after myocardial infarction by inhibiting M1 macrophage polarization through the peroxisome proliferator-activated receptor-γ pathway. Eur J Pharmacol 2024; 967:176398. [PMID: 38350591 DOI: 10.1016/j.ejphar.2024.176398] [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: 06/20/2023] [Revised: 01/16/2024] [Accepted: 02/05/2024] [Indexed: 02/15/2024]
Abstract
OBJECTIVES Inflammation regulates ventricular remodeling after myocardial infarction (MI), and gabapentin exerts anti-inflammatory effects. We investigated the anti-inflammatory role and mechanism of gabapentin after MI. METHODS Rats were divided into the sham group (n = 12), MI group (n = 20), and MI + gabapentin group (n = 16). MI was induced by left coronary artery ligation. The effects of gabapentin on THP-1-derived macrophages were examined in vitro. RESULTS In vivo, 1 week after MI, gabapentin significantly reduced inducible nitric oxide synthase (iNOS; M1 macrophage marker) expression and decreased pro-inflammatory factors (tumor necrosis factor [TNF]-α and interleukin [IL]-1β). Gabapentin upregulated the M2 macrophage marker arginase-1, as well as CD163 expression, and increased the expression of anti-inflammatory factors, including chitinase-like 3, IL-10, and transforming growth factor-β. Four weeks after MI, cardiac function, infarct size, and cardiac fibrosis improved after gabapentin treatment. Gabapentin inhibited sympathetic nerve activity and decreased ventricular electrical instability in rats after MI. Tyrosine hydroxylase and growth-associated protein 43 were suppressed after gabapentin treatment. Gabapentin downregulated nerve growth factor (NGF) and reduced pro-inflammatory factors (iNOS, TNF-α, and IL-1β). In vitro, gabapentin reduced NGF, iNOS, TNF-α, and IL-1β expression in lipopolysaccharide-stimulated macrophages. Mechanistic studies revealed that the peroxisome proliferator-activated receptor-γ antagonist GW9662 attenuated the effects of gabapentin. Moreover, gabapentin reduced α2δ1 expression in the macrophage plasma membrane and reduced the calcium content of macrophages. CONCLUSION Gabapentin attenuates cardiac remodeling by inhibiting inflammation via peroxisome proliferator-activated receptor-γ activation and preventing calcium overload.
Collapse
Affiliation(s)
- Zhenjun Li
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Shaoxian Wang
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Ying Qin
- College of Sports and Human Sciences, Harbin Sport University, Harbin, 150001, China
| | - Bo Yang
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Chengcheng Wang
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Tianyi Lu
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Jie Xu
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Lige Zhu
- Medical Department, The Second Affiliated Hospital of Hei Long Jiang University of Chinese Medicine, Harbin, 150001, China
| | - Chen Yuan
- School of Basic Medical Sciences, Harbin Medical University, Harbin, 150081, China
| | - Wei Han
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China; Department of Heart Failure, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China.
| |
Collapse
|
3
|
Flanagan T, Foster TP, Galbato TE, Lum PY, Louie B, Song G, Halberstadt AL, Billac GB, Nichols CD. Serotonin-2 Receptor Agonists Produce Anti-inflammatory Effects through Functionally Selective Mechanisms That Involve the Suppression of Disease-Induced Arginase 1 Expression. ACS Pharmacol Transl Sci 2024; 7:478-492. [PMID: 38357283 PMCID: PMC10863441 DOI: 10.1021/acsptsci.3c00297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 01/11/2024] [Accepted: 01/12/2024] [Indexed: 02/16/2024]
Abstract
Functional selectivity in the context of serotonin 2A (5-HT2A) receptor agonists is often described as differences psychedelic compounds have in the activation of Gq vs β-arrestin signaling in the brain and how that may relate to inducing psychoactive and hallucinatory properties with respect to each other. However, the presence of 5-HT2A receptors throughout the body in several cell types, including endothelial, endocrine, and immune-related tissues, suggests that functional selectivity may exist in the periphery as well. Here, we examine functional selectivity between two 5-HT2A receptor agonists of the phenylalkylamine class: (R)-2,5-dimethoxy-4-iodoamphetamine [(R)-DOI] and (R)-2,5-dimethoxy-4-trifluoromethylamphetamine [(R)-DOTFM]. Despite comparable in vitro activity at the 5-HT2A receptor as well as similar behavioral potency, (R)-DOTFM does not exhibit an ability to prevent inflammation or elevated airway hyperresponsiveness (AHR) in an acute murine ovalbumin-induced asthma model as does (R)-DOI. Furthermore, there are distinct differences between protein expression and inflammatory-related gene expression in pulmonary tissues between the two compounds. Using (R)-DOI and (R)-DOTFM as tools, we further elucidated the anti-inflammatory mechanisms underlying the powerful anti-inflammatory effects of certain psychedelics and identified key mechanistic components of the anti-inflammatory effects of psychedelics, including suppression of arginase 1 expression.
Collapse
Affiliation(s)
- Thomas
W. Flanagan
- Department
of Pharmacology and Experimental TherapeuticsLouisiana State University Health Sciences CenterNew Orleans, Louisiana70112, United States
| | - Timothy P. Foster
- Department
of Microbiology, Immunology, and ParasitologyLouisiana State University Health Sciences CenterNew Orleans, Louisiana70112, United States
| | - Thomas E. Galbato
- Department
of Microbiology, Immunology, and ParasitologyLouisiana State University Health Sciences CenterNew Orleans, Louisiana70112, United States
| | - Pek Yee Lum
- Auransa
Inc.Palo Alto, California94301, United States
| | - Brent Louie
- Auransa
Inc.Palo Alto, California94301, United States
| | - Gavin Song
- Auransa
Inc.Palo Alto, California94301, United States
| | - Adam L. Halberstadt
- Department
of PsychiatryUniversity of San Diego, California, San Diego, California92093, United States
| | - Gerald B. Billac
- Department
of Pharmacology and Experimental TherapeuticsLouisiana State University Health Sciences CenterNew Orleans, Louisiana70112, United States
| | - Charles D. Nichols
- Department
of Pharmacology and Experimental TherapeuticsLouisiana State University Health Sciences CenterNew Orleans, Louisiana70112, United States
| |
Collapse
|
4
|
Kanuri B, Biswas P, Dahdah A, Murphy AJ, Nagareddy PR. Impact of age and sex on myelopoiesis and inflammation during myocardial infarction. J Mol Cell Cardiol 2024; 187:80-89. [PMID: 38163742 PMCID: PMC10922716 DOI: 10.1016/j.yjmcc.2023.11.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 11/16/2023] [Accepted: 11/21/2023] [Indexed: 01/03/2024]
Abstract
Of all the different risk factors known to cause cardiovascular disease (CVD), age and sex are considered to play a crucial role. Aging follows a continuum from birth to death, and therefore it inevitably acts as a risk for CVD. Along with age, sex differences have also been shown to demonstrate variations in immune system responses to pathological insults. It has been widely perceived that females are protected against myocardial infarction (MI) and the protection is quite apparent in young vs. old women. Acute MI leads to changes in the population of myeloid and lymphoid cells at the injury site with myeloid bias being observed in the initial inflammation and the lymphoid in the late-resolution phases of the pathology. Multiple evidence demonstrates that aging enhances damage to various cellular processes through inflamm-aging, an inflammatory process identified to increase pro-inflammatory markers in circulation and tissues. Following MI, marked changes were observed in different sub-sets of major myeloid cell types viz., neutrophils, monocytes, and macrophages. There is a paucity of information regarding the tissue and site-specific functions of these sub-sets. In this review, we highlight the importance of age and sex as crucial risk factors by discussing their role during MI-induced myelopoiesis while emphasizing the current status of myeloid cell sub-sets. We further put forth the need for designing and executing age and sex interaction studies aimed to determine the appropriate age and sex to develop personalized therapeutic strategies post-MI.
Collapse
Affiliation(s)
- Babunageswararao Kanuri
- Department of Internal Medicine, Section of Cardiovascular Diseases, University of Oklahoma Health Sciences Center (OUHSC), Oklahoma City, OK, USA
| | - Priosmita Biswas
- Department of Internal Medicine, Section of Cardiovascular Diseases, University of Oklahoma Health Sciences Center (OUHSC), Oklahoma City, OK, USA
| | - Albert Dahdah
- Department of Internal Medicine, Section of Cardiovascular Diseases, University of Oklahoma Health Sciences Center (OUHSC), Oklahoma City, OK, USA
| | - Andrew J Murphy
- Baker Heart and Diabetes Institute, Division of Immunometabolism, Melbourne, Australia
| | - Prabhakara R Nagareddy
- Department of Internal Medicine, Section of Cardiovascular Diseases, University of Oklahoma Health Sciences Center (OUHSC), Oklahoma City, OK, USA.
| |
Collapse
|
5
|
Shen S, Zhang M, Wang X, Liu Q, Su H, Sun B, Guo Z, Tian B, Gan H, Gong C, Ma L. Single-cell RNA sequencing reveals S100a9 hi macrophages promote the transition from acute inflammation to fibrotic remodeling after myocardial ischemia‒reperfusion. Theranostics 2024; 14:1241-1259. [PMID: 38323308 PMCID: PMC10845204 DOI: 10.7150/thno.91180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 12/29/2023] [Indexed: 02/08/2024] Open
Abstract
Rationale: The transition from acute inflammation to fibrosis following myocardial ischemia‒reperfusion (MIR) significantly affects prognosis. Macrophages play a pivotal role in inflammatory damage and repair after MIR. However, the heterogeneity and transformation mechanisms of macrophages during this transition are not well understood. Methods: In this study, we used single-cell RNA sequencing (scRNA-seq) and mass cytometry to examine murine monocyte-derived macrophages after MIR to investigate macrophage subtypes and their roles in the MIR process. S100a9-/- mice were used to establish MIR model to clarify the mechanism of alleviating inflammation and fibrosis after MIR. Reinfusion of bone marrow-derived macrophages (BMDMs) after macrophage depletion (MD) in mice subjected to MIR were performed to further examine the role of S100a9hi macrophages in MIR. Results: We identified a unique subtype of S100a9hi macrophages that originate from monocytes and are involved in acute inflammation and fibrosis. These S100a9hi macrophages infiltrate the heart as early as 2 h post-reperfusion and activate the Myd88/NFκB/NLRP3 signaling pathway, amplifying inflammatory responses. As the tissue environment shifts from proinflammatory to reparative, S100a9 activates transforming growth factor-β (Tgf-β)/p-smad3 signaling. This activation not only induces the transformation of myocardial fibroblasts to myofibroblasts but also promotes fibrosis via the macrophage-to-myofibroblast transition (MMT). Targeting S100a9 with a specific inhibitor could effectively mitigate acute inflammatory damage and halt the progression of fibrosis, including MMT. Conclusion: S100a9hi macrophages are a promising therapeutic target for managing the transition from inflammation to fibrosis after MIR.
Collapse
Affiliation(s)
- Shichun Shen
- Department of Cardiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Meng Zhang
- Department of Cardiology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xiaohe Wang
- Department of Cardiology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Qiaoling Liu
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, United Kingdom
| | - Huimin Su
- Department of Cardiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Bingyi Sun
- The First Clinical Medical school of Anhui Medical university, Hefei, China
| | - Zhiqing Guo
- Department of Cardiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Beiduo Tian
- Department of Cardiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Hong Gan
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei 230032, Anhui, China
- MOE Key Laboratory of Population Health Across Life Cycle, Hefei, China
| | - Chen Gong
- Department of Pediatrics, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Likun Ma
- Department of Cardiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| |
Collapse
|
6
|
Sharma P, Venkatachalam K, Binesh A. Decades Long Involvement of THP-1 Cells as a Model for Macrophage Research: A Comprehensive Review. Antiinflamm Antiallergy Agents Med Chem 2024; 23:85-104. [PMID: 38676532 DOI: 10.2174/0118715230294413240415054610] [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: 01/03/2024] [Revised: 03/22/2024] [Accepted: 03/26/2024] [Indexed: 04/29/2024]
Abstract
Over the years, researchers have endeavored to identify dependable and reproducible in vitro models for examining macrophage behavior under controlled conditions. The THP-1 cell line has become a significant and widely employed tool in macrophage research within these models. Originating from the peripheral blood of individuals with acute monocytic leukemia, this human monocytic cell line can undergo transformation into macrophage-like cells, closely mirroring primary human macrophages when exposed to stimulants. Macrophages play a vital role in the innate immune system, actively regulating inflammation, responding to infections, and maintaining tissue homeostasis. A comprehensive understanding of macrophage biology and function is crucial for gaining insights into immunological responses, tissue healing, and the pathogenesis of diseases such as viral infections, autoimmune disorders, and neoplastic conditions. This review aims to thoroughly evaluate and emphasize the extensive history of THP-1 cells as a model for macrophage research. Additionally, it will delve into the significance of THP-1 cells in advancing our comprehension of macrophage biology and their invaluable contributions to diverse scientific domains.
Collapse
Affiliation(s)
- Prakhar Sharma
- Institute of Fisheries Post Graduate Studies, Tamil Nadu Dr. J. Jayalalithaa Fisheries University (TNJFU), OMR Campus, Vaniyanchavadi, Chennai, 603103, Tamil Nadu, India
| | - Kaliyamurthi Venkatachalam
- Institute of Fisheries Post Graduate Studies, Tamil Nadu Dr. J. Jayalalithaa Fisheries University (TNJFU), OMR Campus, Vaniyanchavadi, Chennai, 603103, Tamil Nadu, India
| | - Ambika Binesh
- Institute of Fisheries Post Graduate Studies, Tamil Nadu Dr. J. Jayalalithaa Fisheries University (TNJFU), OMR Campus, Vaniyanchavadi, Chennai, 603103, Tamil Nadu, India
| |
Collapse
|
7
|
Dai G, Li M, Xu H, Quan N. Status of Research on Sestrin2 and Prospects for its Application in Therapeutic Strategies Targeting Myocardial Aging. Curr Probl Cardiol 2023; 48:101910. [PMID: 37422038 DOI: 10.1016/j.cpcardiol.2023.101910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 06/27/2023] [Indexed: 07/10/2023]
Abstract
Cardiac aging is accompanied by changes in the heart at the cellular and molecular levels, leading to alterations in cardiac structure and function. Given today's increasingly aging population, the decline in cardiac function caused by cardiac aging has a significant impact on quality of life. Antiaging therapies to slow the aging process and attenuate changes in cardiac structure and function have become an important research topic. Treatment with drugs, including metformin, spermidine, rapamycin, resveratrol, astaxanthin, Huolisu oral liquid, and sulforaphane, has been demonstrated be effective in delaying cardiac aging by stimulating autophagy, delaying ventricular remodeling, and reducing oxidative stress and the inflammatory response. Furthermore, caloric restriction has been shown to play an important role in delaying aging of the heart. Many studies in cardiac aging and cardiac aging-related models have demonstrated that Sestrin2 has antioxidant and anti-inflammatory effects, stimulates autophagy, delays aging, regulates mitochondrial function, and inhibits myocardial remodeling by regulation of relevant signaling pathways. Therefore, Sestrin2 is likely to become an important target for antimyocardial aging therapy.
Collapse
Affiliation(s)
- Gaoying Dai
- Department of Cardiovascular Center, The First Hospital of Jilin University, Changchun, China
| | - Meina Li
- Department of Infection Control, The First Hospital of Jilin University, Changchun, China
| | - He Xu
- Department of Integrative Medicine, Lequn Branch, The First Hospital of Jilin University, Changchun, China
| | - Nanhu Quan
- Department of Cardiovascular Center, The First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
8
|
Yang Y, Johnson J, Troupes CD, Feldsott EA, Kraus L, Megill E, Bian Z, Asangwe N, Kino T, Eaton DM, Wang T, Wagner M, Ma L, Bryan C, Wallner M, Kubo H, Berretta RM, Khan M, Wang H, Kishore R, Houser SR, Mohsin S. miR-182/183-Rasa1 axis induced macrophage polarization and redox regulation promotes repair after ischemic cardiac injury. Redox Biol 2023; 67:102909. [PMID: 37801856 PMCID: PMC10570148 DOI: 10.1016/j.redox.2023.102909] [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] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 09/26/2023] [Indexed: 10/08/2023] Open
Abstract
Few therapies have produced significant improvement in cardiac structure and function after ischemic cardiac injury (ICI). Our possible explanation is activation of local inflammatory responses negatively impact the cardiac repair process following ischemic injury. Factors that can alter immune response, including significantly altered cytokine levels in plasma and polarization of macrophages and T cells towards a pro-reparative phenotype in the myocardium post-MI is a valid strategy for reducing infarct size and damage after myocardial injury. Our previous studies showed that cortical bone stem cells (CBSCs) possess reparative effects after ICI. In our current study, we have identified that the beneficial effects of CBSCs appear to be mediated by miRNA in their extracellular vesicles (CBSC-EV). Our studies showed that CBSC-EV treated animals demonstrated reduced scar size, attenuated structural remodeling, and improved cardiac function versus saline treated animals. These effects were linked to the alteration of immune response, with significantly altered cytokine levels in plasma, and polarization of macrophages and T cells towards a pro-reparative phenotype in the myocardium post-MI. Our detailed in vitro studies demonstrated that CBSC-EV are enriched in miR-182/183 that mediates the pro-reparative polarization and metabolic reprogramming in macrophages, including enhanced OXPHOS rate and reduced ROS, via Ras p21 protein activator 1 (RASA1) axis under Lipopolysaccharides (LPS) stimulation. In summary, CBSC-EV deliver unique molecular cargoes, such as enriched miR-182/183, that modulate the immune response after ICI by regulating macrophage polarization and metabolic reprogramming to enhance repair.
Collapse
Affiliation(s)
- Yijun Yang
- Cardiovascular Research Center (CVRC), Temple University Lewis Katz School of Medicine, PA, United States
| | - Jaslyn Johnson
- Cardiovascular Research Center (CVRC), Temple University Lewis Katz School of Medicine, PA, United States
| | - Constantine D Troupes
- Cardiovascular Research Center (CVRC), Temple University Lewis Katz School of Medicine, PA, United States
| | - Eric A Feldsott
- Cardiovascular Research Center (CVRC), Temple University Lewis Katz School of Medicine, PA, United States
| | - Lindsay Kraus
- Cardiovascular Research Center (CVRC), Temple University Lewis Katz School of Medicine, PA, United States
| | - Emily Megill
- Cardiovascular Research Center (CVRC), Temple University Lewis Katz School of Medicine, PA, United States
| | - Zilin Bian
- Tandon School of Engineering, New York University, NY, United States
| | - Ngefor Asangwe
- Cardiovascular Research Center (CVRC), Temple University Lewis Katz School of Medicine, PA, United States
| | - Tabito Kino
- Cardiovascular Research Center (CVRC), Temple University Lewis Katz School of Medicine, PA, United States
| | - Deborah M Eaton
- Cardiovascular Research Center (CVRC), Temple University Lewis Katz School of Medicine, PA, United States
| | - Tao Wang
- Cardiovascular Research Center (CVRC), Temple University Lewis Katz School of Medicine, PA, United States
| | - Marcus Wagner
- Cardiovascular Research Center (CVRC), Temple University Lewis Katz School of Medicine, PA, United States
| | - Lena Ma
- Cardiovascular Research Center (CVRC), Temple University Lewis Katz School of Medicine, PA, United States
| | - Christopher Bryan
- Cardiovascular Research Center (CVRC), Temple University Lewis Katz School of Medicine, PA, United States
| | - Markus Wallner
- Cardiovascular Research Center (CVRC), Temple University Lewis Katz School of Medicine, PA, United States; Division of Cardiology, Medical University of Graz, 8036, Graz, Austria
| | - Hajime Kubo
- Cardiovascular Research Center (CVRC), Temple University Lewis Katz School of Medicine, PA, United States
| | - Remus M Berretta
- Cardiovascular Research Center (CVRC), Temple University Lewis Katz School of Medicine, PA, United States
| | - Mohsin Khan
- Center for Metabolic Disease Research (CMDR), Temple University Lewis Katz School of Medicine, PA, United States
| | - Hong Wang
- Center for Metabolic Disease Research (CMDR), Temple University Lewis Katz School of Medicine, PA, United States
| | - Raj Kishore
- Center for Translational Medicine, Temple University Lewis Katz School of Medicine, PA, United States
| | - Steven R Houser
- Cardiovascular Research Center (CVRC), Temple University Lewis Katz School of Medicine, PA, United States
| | - Sadia Mohsin
- Cardiovascular Research Center (CVRC), Temple University Lewis Katz School of Medicine, PA, United States.
| |
Collapse
|
9
|
Markina YV, Kirichenko TV, Tolstik TV, Bogatyreva AI, Zotova US, Cherednichenko VR, Postnov AY, Markin AM. Target and Cell Therapy for Atherosclerosis and CVD. Int J Mol Sci 2023; 24:10308. [PMID: 37373454 DOI: 10.3390/ijms241210308] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 06/06/2023] [Accepted: 06/16/2023] [Indexed: 06/29/2023] Open
Abstract
Cardiovascular diseases (CVD) and, in particular, atherosclerosis, remain the main cause of death in the world today. Unfortunately, in most cases, CVD therapy begins after the onset of clinical symptoms and is aimed at eliminating them. In this regard, early pathogenetic therapy for CVD remains an urgent problem in modern science and healthcare. Cell therapy, aimed at eliminating tissue damage underlying the pathogenesis of some pathologies, including CVD, by replacing it with various cells, is of the greatest interest. Currently, cell therapy is the most actively developed and potentially the most effective treatment strategy for CVD associated with atherosclerosis. However, this type of therapy has some limitations. In this review, we have tried to summarize the main targets of cell therapy for CVD and atherosclerosis in particular based on the analysis using the PubMed and Scopus databases up to May 2023.
Collapse
Affiliation(s)
- Yuliya V Markina
- Petrovsky National Research Center of Surgery, Moscow 119991, Russia
| | | | - Taisiya V Tolstik
- Petrovsky National Research Center of Surgery, Moscow 119991, Russia
| | | | - Ulyana S Zotova
- Petrovsky National Research Center of Surgery, Moscow 119991, Russia
| | | | - Anton Yu Postnov
- Petrovsky National Research Center of Surgery, Moscow 119991, Russia
| | - Alexander M Markin
- Petrovsky National Research Center of Surgery, Moscow 119991, Russia
- Peoples' Friendship University of Russia named after Patrice Lumumba (RUDN University), Moscow 117198, Russia
| |
Collapse
|
10
|
Zhao P, Cai Z, Zhang X, Liu M, Xie F, Liu Z, Lu S, Ma X. Hydrogen Attenuates Inflammation by Inducing Early M2 Macrophage Polarization in Skin Wound Healing. Pharmaceuticals (Basel) 2023; 16:885. [PMID: 37375833 DOI: 10.3390/ph16060885] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2023] Open
Abstract
The heterogeneous and highly plastic cell populations of macrophages are important mediators of cellular responses during all stages of wound healing, especially in the inflammatory stage. Molecular hydrogen (H2), which has potent antioxidant and anti-inflammatory effects, has been shown to promote M2 polarization in injury and disease. However, more in vivo time series studies of the role of M1-to-M2 polarization in wound healing are needed. In the current study, we performed time series experiments on a dorsal full-thickness skin defect mouse model in the inflammatory stage to examine the effects of H2 inhalation. Our results revealed that H2 could promote very early M1-to-M2 polarization (on days 2-3 post wounding, 2-3 days earlier than in conventional wound healing), without disturbing the functions of the M1 phenotype. Time series analysis of the transcriptome, blood cell counts, and multiple cytokines further indicated that peripheral blood monocytes were a source of H2-induced M2 macrophages and that the functions of H2 in macrophage polarization were not only dependent on its antioxidant effects. Therefore, we believe that H2 could reduce inflammation in wound care by shifting early macrophage polarization in clinical settings.
Collapse
Affiliation(s)
- Pengxiang Zhao
- Faculty of Environment and Life, Beijing University of Technology, Beijing 100124, China
- Beijing Molecular Hydrogen Research Center, Beijing 100124, China
- Beijing International Science and Technology Cooperation Base of Antivirus Drug, Beijing 100124, China
| | - Zisong Cai
- Faculty of Environment and Life, Beijing University of Technology, Beijing 100124, China
- Beijing Molecular Hydrogen Research Center, Beijing 100124, China
- Beijing International Science and Technology Cooperation Base of Antivirus Drug, Beijing 100124, China
| | - Xujuan Zhang
- Faculty of Environment and Life, Beijing University of Technology, Beijing 100124, China
- Beijing Molecular Hydrogen Research Center, Beijing 100124, China
- Beijing International Science and Technology Cooperation Base of Antivirus Drug, Beijing 100124, China
| | - Mengyu Liu
- Faculty of Environment and Life, Beijing University of Technology, Beijing 100124, China
- Beijing Molecular Hydrogen Research Center, Beijing 100124, China
- Beijing International Science and Technology Cooperation Base of Antivirus Drug, Beijing 100124, China
| | - Fei Xie
- Faculty of Environment and Life, Beijing University of Technology, Beijing 100124, China
- Beijing Molecular Hydrogen Research Center, Beijing 100124, China
- Beijing International Science and Technology Cooperation Base of Antivirus Drug, Beijing 100124, China
| | - Ziyi Liu
- Faculty of Environment and Life, Beijing University of Technology, Beijing 100124, China
- Beijing Molecular Hydrogen Research Center, Beijing 100124, China
- Beijing International Science and Technology Cooperation Base of Antivirus Drug, Beijing 100124, China
| | - Shidong Lu
- Faculty of Environment and Life, Beijing University of Technology, Beijing 100124, China
- Beijing Molecular Hydrogen Research Center, Beijing 100124, China
- Beijing International Science and Technology Cooperation Base of Antivirus Drug, Beijing 100124, China
| | - Xuemei Ma
- Faculty of Environment and Life, Beijing University of Technology, Beijing 100124, China
- Beijing Molecular Hydrogen Research Center, Beijing 100124, China
- Beijing International Science and Technology Cooperation Base of Antivirus Drug, Beijing 100124, China
| |
Collapse
|
11
|
Fang W, Yang M, Liu M, Jin Y, Wang Y, Yang R, Wang Y, Zhang K, Fu Q. Review on Additives in Hydrogels for 3D Bioprinting of Regenerative Medicine: From Mechanism to Methodology. Pharmaceutics 2023; 15:1700. [PMID: 37376148 PMCID: PMC10302687 DOI: 10.3390/pharmaceutics15061700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 05/29/2023] [Accepted: 06/02/2023] [Indexed: 06/29/2023] Open
Abstract
The regeneration of biological tissues in medicine is challenging, and 3D bioprinting offers an innovative way to create functional multicellular tissues. One common way in bioprinting is bioink, which is one type of the cell-loaded hydrogel. For clinical application, however, the bioprinting still suffers from satisfactory performance, e.g., in vascularization, effective antibacterial, immunomodulation, and regulation of collagen deposition. Many studies incorporated different bioactive materials into the 3D-printed scaffolds to optimize the bioprinting. Here, we reviewed a variety of additives added to the 3D bioprinting hydrogel. The underlying mechanisms and methodology for biological regeneration are important and will provide a useful basis for future research.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Kaile Zhang
- Department of Urology, Affiliated Sixth People’s Hospital, Shanghai Jiaotong University, No. 600 Yi-Shan Road, Shanghai 200233, China; (W.F.); (M.Y.)
| | - Qiang Fu
- Department of Urology, Affiliated Sixth People’s Hospital, Shanghai Jiaotong University, No. 600 Yi-Shan Road, Shanghai 200233, China; (W.F.); (M.Y.)
| |
Collapse
|
12
|
Pearce DP, Nemcek MT, Witzenburg CM. Don't go breakin' my heart: cardioprotective alterations to the mechanical and structural properties of reperfused myocardium during post-infarction inflammation. Biophys Rev 2023; 15:329-353. [PMID: 37396449 PMCID: PMC10310682 DOI: 10.1007/s12551-023-01068-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 05/21/2023] [Indexed: 07/04/2023] Open
Abstract
Myocardial infarctions (MIs) kickstart an intense inflammatory response resulting in extracellular matrix (ECM) degradation, wall thinning, and chamber dilation that leaves the heart susceptible to rupture. Reperfusion therapy is one of the most effective strategies for limiting adverse effects of MIs, but is a challenge to administer in a timely manner. Late reperfusion therapy (LRT; 3 + hours post-MI) does not limit infarct size, but does reduce incidences of post-MI rupture and improves long-term patient outcomes. Foundational studies employing LRT in the mid-twentieth century revealed beneficial reductions in infarct expansion, aneurysm formation, and left ventricle dysfunction. The mechanism by which LRT acts, however, is undefined. Structural analyses, relying largely on one-dimensional estimates of ECM composition, have found few differences in collagen content between LRT and permanently occluded animal models when using homogeneous samples from infarct cores. Uniaxial testing, on the other hand, revealed slight reductions in stiffness early in inflammation, followed soon after by an enhanced resistance to failure for cases of LRT. The use of one-dimensional estimates of ECM organization and gross mechanical function have resulted in a poor understanding of the infarct's spatially variable mechanical and structural anisotropy. To resolve these gaps in literature, future work employing full-field mechanical, structural, and cellular analyses is needed to better define the spatiotemporal post-MI alterations occurring during the inflammatory phase of healing and how they are impacted following reperfusion therapy. In turn, these studies may reveal how LRT affects the likelihood of rupture and inspire novel approaches to guide scar formation.
Collapse
Affiliation(s)
- Daniel P. Pearce
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53706 USA
| | - Mark T. Nemcek
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53706 USA
| | - Colleen M. Witzenburg
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53706 USA
| |
Collapse
|
13
|
Han D, Wang F, Qiao Z, Wang B, Zhang Y, Jiang Q, Liu M, Zhuang Y, An Q, Bai Y, Shangguan J, Zhang J, Liang G, Shen D. Neutrophil membrane-camouflaged nanoparticles alleviate inflammation and promote angiogenesis in ischemic myocardial injury. Bioact Mater 2023; 23:369-382. [DOI: 10.1016/j.bioactmat.2022.11.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 11/07/2022] [Accepted: 11/22/2022] [Indexed: 11/29/2022] Open
|
14
|
Wilmes V, Kur IM, Weigert A, Verhoff MA, Gradhand E, Kauferstein S. iNOS expressing macrophages co-localize with nitrotyrosine staining after myocardial infarction in humans. Front Cardiovasc Med 2023; 10:1104019. [PMID: 37063955 PMCID: PMC10098457 DOI: 10.3389/fcvm.2023.1104019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 03/15/2023] [Indexed: 04/03/2023] Open
Abstract
IntroductionInducible nitric oxide synthase (iNOS) produces micromolar amounts of nitric oxide (NO) upon the right stimuli, whose further reactions can lead to oxidative stress. In murine models of myocardial infarction (MI), iNOS is known to be expressed in infiltrating macrophages, which at early onset enter the infarcted zone and are associated with inflammation. In contrast cardiac tissue resident macrophages are thought to enhance regeneration of tissue injury and re-establish homeostasis. Both detrimental and beneficial effects of iNOS have been described, still the role of iNOS in MI is not fully understood. Our aim was to examine cell expression patterns of iNOS and nitrotyrosine (NT) production in human MI.Material and MethodsWe examined in postmortem human MI hearts the iNOS mRNA expression by means of qPCR. Further we performed immunohistochemical stainings for cell type identification. Afterwards a distance analysis between iNOS and NT was carried out to determine causality between iNOS and NT production.ResultsiNOS mRNA expression was significantly increased in infarcted regions of human MI hearts and iNOS protein expression was detected in resident macrophages in infarcted human hearts as well as in controls hearts, being higher in resident macrophages in MI hearts compared to control. Furthermore in MI and in healthy human hearts cells showing signs of NT production peaked within 10–15 µm proximity of iNOS+ cells.DiscussionThese results indicate that, unexpectedly, resident macrophages are the main source of iNOS expression in postmortem human MI hearts. The peak of NT positive cells within 10–15 µm of iNOS+ cells suggest an iNOS dependent level of NT and therefore iNOS dependent oxidative stress. Our results contribute to understanding the role of iNOS in human MI.
Collapse
Affiliation(s)
- Verena Wilmes
- Institute of Legal Medicine, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
- Correspondence: Verena Wilmes
| | - Ivan M. Kur
- Institute of Biochemistry I, Faculty of Medicine, Goethe University, Frankfurt am Main, Germany
| | - Andreas Weigert
- Institute of Biochemistry I, Faculty of Medicine, Goethe University, Frankfurt am Main, Germany
- Cardio-Pulmonary Institute (CPI), Goethe University, Frankfurt am Main, Germany
| | - Marcel A. Verhoff
- Institute of Legal Medicine, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Elise Gradhand
- Dr. Senckenberg Institute of Pathology, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Silke Kauferstein
- Institute of Legal Medicine, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| |
Collapse
|
15
|
Li L, Ma Q, Wang M, Mou J, Han Y, Wang J, Ye J, Sun G. Single-cell transcriptome sequencing of macrophages in common cardiovascular diseases. J Leukoc Biol 2023; 113:139-148. [PMID: 36822177 DOI: 10.1093/jleuko/qiac014] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Indexed: 01/18/2023] Open
Abstract
Macrophages are strategically located throughout the body at key sites in the immune system. A key feature in atherosclerosis is the uptake and accumulation of lipoproteins by arterial macrophages, leading to the formation of foam cells. After myocardial infarction, macrophages derived from monocytes infiltrate the infarcted heart. Macrophages are also closely related to adverse remodeling after heart failure. An in-depth understanding of the functions and characteristics of macrophages is required to study heart health and pathophysiological processes; however, the heterogeneity and plasticity explained by the classic M1/M2 macrophage paradigm are too limited. Single-cell sequencing is a high-throughput sequencing technique that enables the sequencing of the genome or transcriptome of a single cell. It effectively complements the heterogeneity of gene expression in a single cell that is ignored by conventional sequencing and can give valuable insights into the development of complex diseases. In the present review, we summarize the available research on the application of single-cell transcriptome sequencing to study the changes in macrophages during common cardiovascular diseases, such as atherosclerosis, myocardial infarction, and heart failure. This article also discusses the contribution of this knowledge to understanding the pathogenesis, development, diagnosis, and treatment of heart diseases.
Collapse
Affiliation(s)
- Lanfang Li
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Malianwa Road, Haidian District, Beijing, China
| | - Qiuxiao Ma
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Xiyuan Playground, Haidian District, Beijing, China
| | - Min Wang
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Malianwa Road, Haidian District, Beijing, China
| | - Junyu Mou
- School of Pharmacy, Harbin University of Commerce, Xuehai Street, Songbei District, Harbin, China
| | - Yanwei Han
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Waihuan East Road, Panyu District, Guangzhou, China
| | - Jialu Wang
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Malianwa Road, Haidian District, Beijing, China
| | - Jingxue Ye
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Malianwa Road, Haidian District, Beijing, China
| | - Guibo Sun
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Malianwa Road, Haidian District, Beijing, China
| |
Collapse
|
16
|
Man Q, Gao Z, Chen K. Functional Potassium Channels in Macrophages. J Membr Biol 2023; 256:175-187. [PMID: 36622407 DOI: 10.1007/s00232-022-00276-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Accepted: 12/30/2022] [Indexed: 01/10/2023]
Abstract
Macrophages are the predominant component of innate immunity, which is an important protective barrier of our body. Macrophages are present in all organs and tissues of the body, their main functions include immune surveillance, bacterial killing, tissue remodeling and repair, and clearance of cell debris. In addition, macrophages can present antigens to T cells and facilitate inflammatory response by releasing cytokines. Macrophages are of high concern due to their crucial roles in multiple physiological processes. In recent years, new advances are emerging after great efforts have been made to explore the mechanisms of macrophage activation. Ion channel is a class of multimeric transmembrane protein that allows specific ions to go through cell membrane. The flow of ions through ion channel between inside and outside of cell membrane is required for maintaining cell morphology and intracellular signal transduction. Expressions of various ion channels in macrophages have been detected. The roles of ion channels in macrophage activation are gradually caught attention. K+ channels are the most studied channels in immune system. However, very few of published papers reviewed the studies of K+ channels on macrophages. Here, we will review the four types of K+ channels that are expressed in macrophages: voltage-gated K+ channel, calcium-activated K+ channel, inwardly rectifying K+ channel and two-pore domain K+ channel.
Collapse
Affiliation(s)
- Qiaoyan Man
- Department of Pharmacology, Ningbo University School of Medicine, A506, Wang Changlai Building818 Fenghua Rd, Ningbo, China
| | - Zhe Gao
- Ningbo Institute of Medical Sciences, 42 Yangshan Rd, Ningbo, China.
| | - Kuihao Chen
- Department of Pharmacology, Ningbo University School of Medicine, A506, Wang Changlai Building818 Fenghua Rd, Ningbo, China.
| |
Collapse
|
17
|
Zhang M, Wang C, Wang R, Xu J, Wang Z, Yan J, Cai Y, Li L, Huo Y, Dong S. Adenosine kinase promotes post-infarction cardiac repair by epigenetically maintaining reparative macrophage phenotype. J Mol Cell Cardiol 2023; 174:88-100. [PMID: 36473288 PMCID: PMC10420407 DOI: 10.1016/j.yjmcc.2022.11.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 11/19/2022] [Accepted: 11/23/2022] [Indexed: 12/07/2022]
Abstract
Pro-inflammatory and reparative macrophages are crucial in clearing necrotic myocardium and promoting cardiac repair after myocardial infarction (MI), respectively. Extracellular adenosine has been demonstrated to modulate macrophage polarization through adenosine receptors. However, the role of intracellular adenosine in macrophage polarization has not been explored and adenosine kinase (ADK) is a major enzyme regulating intracellular adenosine levels. Here, we aimed to elucidate the role of ADK in macrophage polarization and its subsequent impact on MI. We demonstrated that ADK was upregulated in bone marrow-derived macrophages (BMDMs) after IL-4 treatment and was highly expressed in the infarct area at day 7 post-MI, especially in macrophages. Compared with wild-type mice, myeloid-specific Adk knockout mice showed increased infarct size, limited myofibroblast differentiation, reduced collagen deposition and more severe cardiac dysfunction after MI, which was related to impaired reparative macrophage phenotype in MI tissue. We found that ADK deletion or inhibition significantly decreased the expression of reparative genes, such as Arg1, Ym1, Fizz1, and Cd206 in BMDMs after IL-4 treatment. The increased intracellular adenosine due to Adk deletion inhibited transmethylation reactions and decreased the trimethylation of H3K4 in BMDMs after IL-4 treatment. Mechanistically, we demonstrated that Adk deletion suppressed reparative macrophage phenotype through decreased IRF4 expression, which resulted from reduced levels of H3K4me3 on the Irf4 promotor. Together, our study reveals that ADK exerts a protective effect against MI by promoting reparative macrophage polarization through epigenetic mechanisms.
Collapse
Affiliation(s)
- Min Zhang
- Department of Cardiology, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen 518020, China; The First Affiliated Hospital, Jinan University, Guangzhou 510632, China
| | - Caiping Wang
- Department of Cardiology, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen 518020, China
| | - Rongning Wang
- Department of Cardiology, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen 518020, China
| | - Jiean Xu
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, China
| | - Zhefeng Wang
- Department of Cardiology, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen 518020, China; The First Affiliated Hospital, Jinan University, Guangzhou 510632, China
| | - Jianlong Yan
- Department of Cardiology, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen 518020, China
| | - Yongfeng Cai
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, China
| | - Liangping Li
- The First Affiliated Hospital, Jinan University, Guangzhou 510632, China; Institute of Clinical Oncology, The First Affiliated Hospital, Jinan University, Guangzhou 510632, China
| | - Yuqing Huo
- Vascular Biology Center, Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, GA 30912, United States
| | - Shaohong Dong
- Department of Cardiology, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen 518020, China.
| |
Collapse
|
18
|
Wu XT, Wang YX, Feng XM, Feng M, Sun HH. Update on the roles of macrophages in the degeneration and repair process of intervertebral discs. Joint Bone Spine 2022; 90:105514. [PMID: 36529418 DOI: 10.1016/j.jbspin.2022.105514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 11/25/2022] [Accepted: 11/29/2022] [Indexed: 12/23/2022]
Abstract
Intervertebral disc (IVD) degeneration is the common cause of lumbar degenerative diseases, causing severe social and economic burden. The process of IVD degeneration involves a complex of pathologic changes on both extracellular matrix degradation and resident cell apoptosis. In recent years, there is increasing evidence that macrophages play vital roles during the damage and repair process of IVD degeneration. Nevertheless, the interactions between macrophages and IVD are not well understood, even if the IVD has long been regarded as the immune privileged site. Therefore, this review mainly focuses on the progress and obstacles of studies investigating the blood supply, immune response and especially macrophages during the IVD degeneration process.
Collapse
Affiliation(s)
- Xiao-Tao Wu
- Spine department, Northern Jiangsu People's Hospital, Yangzhou City 225001, China; Spine Department, Zhongda Hospital, School of Medicine, Southeast University, Nanjing City 210009, Jiangsu, China
| | - Yong-Xiang Wang
- Spine department, Northern Jiangsu People's Hospital, Yangzhou City 225001, China
| | - Xin-Min Feng
- Spine department, Northern Jiangsu People's Hospital, Yangzhou City 225001, China
| | - Min Feng
- Day treatment ward, Northern Jiangsu People's Hospital, Yangzhou City 225001, China.
| | - Hui-Hui Sun
- Spine department, Northern Jiangsu People's Hospital, Yangzhou City 225001, China.
| |
Collapse
|
19
|
Targeted Phagocytosis Induction for Cancer Immunotherapy via Bispecific MerTK-Engaging Antibodies. Int J Mol Sci 2022; 23:ijms232415673. [PMID: 36555321 PMCID: PMC9779728 DOI: 10.3390/ijms232415673] [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: 10/31/2022] [Revised: 12/06/2022] [Accepted: 12/07/2022] [Indexed: 12/14/2022] Open
Abstract
The Tyro, Axl, and MerTK receptors (TAMRs) play a significant role in the clearance of apoptotic cells. In this work, the spotlight was set on MerTK, as it is one of the prominent TAMRs expressed on the surface of macrophages and dendritic cells. MerTK-specific antibodies were previously isolated from a transgenic rat-derived immune library with suitable biophysical properties. Further characterisation resulted in an agonistic MerTK antibody that led to phospho AKT activation in a dose-dependent manner. In this proof-of-concept study, a MerTK-specific antibody, MerK28, was combined with tandem, biparatopic EGFR-binding VHH camelid antibody domains (7D9G) in different architectures to generate bispecific antibodies with the capacity to bind EGFR and MerTK simultaneously. The bispecific molecules exhibited appropriate binding properties with regard to both targets in their soluble forms as well as to cells, which resulted in the engagement of macrophage-like THP-1 cells with epidermoid carcinoma A431 cells. Furthermore, targeted phagocytosis in co-culture experiments was observed only with the bispecific variants and not the parental MerTK-binding antibody. This work paves the way for the generation of bispecific macrophage-engaging antibodies for targeted phagocytosis harnessing the immune-modulating roles of MerTK in immunotherapy.
Collapse
|
20
|
Liu Z, Wang L, Xing Q, Liu X, Hu Y, Li W, Yan Q, Liu R, Huang N. Identification of GLS as a cuproptosis-related diagnosis gene in acute myocardial infarction. Front Cardiovasc Med 2022; 9:1016081. [PMID: 36440046 PMCID: PMC9691691 DOI: 10.3389/fcvm.2022.1016081] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 10/24/2022] [Indexed: 11/12/2022] Open
Abstract
Acute myocardial infarction (AMI) has the characteristics of sudden onset, rapid progression, poor prognosis, and so on. Therefore, it is urgent to identify diagnostic and prognostic biomarkers for it. Cuproptosis is a new form of mitochondrial respiratory-dependent cell death. However, studies are limited on the clinical significance of cuproptosis-related genes (CRGs) in AMI. In this study, we systematically assessed the genetic alterations of CRGs in AMI by bioinformatics approach. The results showed that six CRGs (LIAS, LIPT1, DLAT, PDHB, MTF1, and GLS) were markedly differentially expressed between stable coronary heart disease (stable_CAD) and AMI. Correlation analysis indicated that CRGs were closely correlated with N6-methyladenosine (m6A)-related genes through R language “corrplot” package, especially GLS was positively correlated with FMR1 and MTF1 was negatively correlated with HNRNPA2B1. Immune landscape analysis results revealed that CRGs were closely related to various immune cells, especially GLS was positively correlated with T cells CD4 memory resting and negatively correlated with monocytes. Kaplan–Meier analysis demonstrated that the group with high DLAT expression had a better prognosis. The area under curve (AUC) certified that GLS had good diagnostic value, in the training set (AUC = 0.87) and verification set (ACU = 0.99). Gene set enrichment analysis (GSEA) suggested that GLS was associated with immune- and hypoxia-related pathways. In addition, Gene Ontology (GO) analysis, Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis, competing endogenous RNA (ceRNA) analysis, transcription factor (TF), and compound prediction were performed to reveal the regulatory mechanism of CRGs in AMI. Overall, our study can provide additional information for understanding the role of CRGs in AMI, which may provide new insights into the identification of therapeutic targets for AMI.
Collapse
Affiliation(s)
- Zheng Liu
- Clinical Pharmacy, Xiangtan Center Hospital, Xiangtan, China
- Zhou Honghao Research Institute Xiangtan, Xiangtan, China
| | - Lei Wang
- Department of Cardiovascular Medicine, Xiangtan Center Hospital, Xiangtan, China
| | - Qichang Xing
- Clinical Pharmacy, Xiangtan Center Hospital, Xiangtan, China
- Zhou Honghao Research Institute Xiangtan, Xiangtan, China
| | - Xiang Liu
- Clinical Pharmacy, Xiangtan Center Hospital, Xiangtan, China
- Zhou Honghao Research Institute Xiangtan, Xiangtan, China
| | - Yixiang Hu
- Clinical Pharmacy, Xiangtan Center Hospital, Xiangtan, China
- Zhou Honghao Research Institute Xiangtan, Xiangtan, China
| | - Wencan Li
- Clinical Pharmacy, Xiangtan Center Hospital, Xiangtan, China
- Zhou Honghao Research Institute Xiangtan, Xiangtan, China
| | - Qingzi Yan
- Clinical Pharmacy, Xiangtan Center Hospital, Xiangtan, China
- Zhou Honghao Research Institute Xiangtan, Xiangtan, China
| | - Renzhu Liu
- Clinical Pharmacy, Xiangtan Center Hospital, Xiangtan, China
- Zhou Honghao Research Institute Xiangtan, Xiangtan, China
| | - Nan Huang
- Clinical Pharmacy, Xiangtan Center Hospital, Xiangtan, China
- *Correspondence: Nan Huang,
| |
Collapse
|
21
|
Bakhshian Nik A, Alvarez-Argote S, O'Meara CC. Interleukin 4/13 signaling in cardiac regeneration and repair. Am J Physiol Heart Circ Physiol 2022; 323:H833-H844. [PMID: 36149768 PMCID: PMC9602781 DOI: 10.1152/ajpheart.00310.2022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 09/13/2022] [Accepted: 09/13/2022] [Indexed: 12/14/2022]
Abstract
Interleukin 4 (IL4) and interleukin 13 (IL13) are closely related cytokines that have been classically attributed to type II immunity, namely, differentiation of T-helper 2 (TH2) cells and alternative activation of macrophages. Although the role of IL4/13 has been well described in various contexts such as defense against helminth parasites, pathogenesis of allergic disease, and several models of wound healing, relatively little is known about the role of IL4/13 in the heart following injury. Emerging literature has identified various roles for IL4/13 in animal models of cardiac regeneration as well as in the adult mammalian heart following myocardial injury. Notably, although IL4 and IL13 signal to hematopoietic cell types following myocardial infarction (MI) to promote wound healing phenotypes, there is substantial evidence that these cytokines can signal directly to non-hematopoietic cell types in the heart during development, homeostasis, and following injury. Comprehensive understanding of the molecular and cellular actions of IL4/13 in the heart is still lacking, but overall evidence to date suggests that activation of these cytokines results in beneficial outcomes with respect to cardiac repair. Here, we aim to comprehensively review the role of IL4 and IL13 and their prospective mechanisms in cardiac regeneration and repair.
Collapse
Affiliation(s)
- Amirala Bakhshian Nik
- Department of Physiology, Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Santiago Alvarez-Argote
- Department of Physiology, Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Caitlin C O'Meara
- Department of Physiology, Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| |
Collapse
|
22
|
Durda P, Raffield LM, Lange EM, Olson NC, Jenny NS, Cushman M, Deichgraeber P, Grarup N, Jonsson A, Hansen T, Mychaleckyj JC, Psaty BM, Reiner AP, Tracy RP, Lange LA. Circulating Soluble CD163, Associations With Cardiovascular Outcomes and Mortality, and Identification of Genetic Variants in Older Individuals: The Cardiovascular Health Study. J Am Heart Assoc 2022; 11:e024374. [PMID: 36314488 PMCID: PMC9673628 DOI: 10.1161/jaha.121.024374] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Background Monocytes/macrophages participate in cardiovascular disease. CD163 (cluster of differentiation 163) is a monocyte/macrophage receptor, and the shed sCD163 (soluble CD163) reflects monocyte/macrophage activation. We examined the association of sCD163 with incident cardiovascular disease events and performed a genome-wide association study to identify sCD163-associated variants. Methods and Results We measured plasma sCD163 in 5214 adults (aged ≥65 years, 58.7% women, 16.2% Black) of the CHS (Cardiovascular Health Study). We used Cox regression models (associations of sCD163 with incident events and mortality); median follow-up was 26 years. Genome-wide association study analyses were stratified on race. Adjusted for age, sex, and race and ethnicity, sCD163 levels were associated with all-cause mortality (hazard ratio [HR], 1.08 [95% CI, 1.04-1.12] per SD increase), cardiovascular disease mortality (HR, 1.15 [95% CI, 1.09-1.21]), incident coronary heart disease (HR, 1.10 [95% CI, 1.04-1.16]), and incident heart failure (HR, 1.18 [95% CI, 1.12-1.25]). When further adjusted (eg, cardiovascular disease risk factors), only incident coronary heart disease lost significance. In European American individuals, genome-wide association studies identified 38 variants on chromosome 2 near MGAT5 (top result rs62165726, P=3.3×10-18),19 variants near chromosome 17 gene ASGR1 (rs55714927, P=1.5×10-14), and 18 variants near chromosome 11 gene ST3GAL4. These regions replicated in the European ancestry ADDITION-PRO cohort, a longitudinal cohort study nested in the Danish arm of the Anglo-Danish-Dutch study of Intensive Treatment Intensive Treatment In peOple with screeNdetcted Diabetes in Primary Care. In Black individuals, we identified 9 variants on chromosome 6 (rs3129781 P=7.1×10-9) in the HLA region, and 3 variants (rs115391969 P=4.3×10-8) near the chromosome 16 gene MYLK3. Conclusions Monocyte function, as measured by sCD163, may be predictive of overall and cardiovascular-specific mortality and incident heart failure.
Collapse
Affiliation(s)
- Peter Durda
- Department of Pathology and Laboratory MedicineLarner College of Medicine, University of VermontBurlingtonVT
| | | | - Ethan M. Lange
- Division of Biomedical Informatics and Personalized Medicine, Department of MedicineUniversity of Colorado Anschutz Medical CampusAuroraCO
| | - Nels C. Olson
- Department of Pathology and Laboratory MedicineLarner College of Medicine, University of VermontBurlingtonVT
| | - Nancy Swords Jenny
- Department of Pathology and Laboratory MedicineLarner College of Medicine, University of VermontBurlingtonVT
| | - Mary Cushman
- Department of Pathology and Laboratory MedicineLarner College of Medicine, University of VermontBurlingtonVT,Department of MedicineLarner College of Medicine, University of VermontBurlingtonVT
| | - Pia Deichgraeber
- Steno Diabetes CenterAarhus University HospitalAarhusDenmark,Department of Endocrinology and Internal MedicineAarhus University HospitalAarhusDenmark
| | - Niels Grarup
- Novo Nordisk Foundation Center for Basic Metabolic ResearchCopenhagenDenmark
| | - Anna Jonsson
- Novo Nordisk Foundation Center for Basic Metabolic ResearchCopenhagenDenmark
| | - Torben Hansen
- Novo Nordisk Foundation Center for Basic Metabolic ResearchCopenhagenDenmark
| | | | - Bruce M. Psaty
- Cardiovascular Health Research Unit, Departments of Medicine, Epidemiology and Health ServicesUniversity of WashingtonSeattleWA
| | - Alex P. Reiner
- Department of EpidemiologyUniversity of WashingtonSeattleWA
| | - Russell P. Tracy
- Department of Pathology and Laboratory MedicineLarner College of Medicine, University of VermontBurlingtonVT,Department of BiochemistryLarner College of Medicine, University of VermontBurlingtonVT
| | - Leslie A. Lange
- Division of Biomedical Informatics and Personalized Medicine, Department of MedicineUniversity of Colorado Anschutz Medical CampusAuroraCO
| |
Collapse
|
23
|
Porcine Small Intestinal Submucosa Alters the Biochemical Properties of Wound Healing: A Narrative Review. Biomedicines 2022; 10:biomedicines10092213. [PMID: 36140314 PMCID: PMC9496019 DOI: 10.3390/biomedicines10092213] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 08/29/2022] [Accepted: 09/06/2022] [Indexed: 11/16/2022] Open
Abstract
Among the many biological scaffold materials currently available for clinical use, the small intestinal submucosa (SIS) is an effective material for wound healing. SIS contains numerous active forms of extracellular matrix that support angiogenesis, cell migration, and proliferation, providing growth factors involved in signaling for tissue formation and assisting wound healing. SIS not only serves as a bioscaffold for cell migration and differentiation, but also restores the impaired dynamic reciprocity between cells and the extracellular matrix, ultimately driving wound healing. Here, we review the evidence on how SIS can shift the biochemical balance in a wound from chronic to an acute state.
Collapse
|
24
|
Li XC, Luo SJ, Fan W, Zhou TL, Tan DQ, Tan RX, Xian QZ, Li J, Huang CM, Wang MS. Macrophage polarization regulates intervertebral disc degeneration by modulating cell proliferation, inflammation mediator secretion, and extracellular matrix metabolism. Front Immunol 2022; 13:922173. [PMID: 36059551 PMCID: PMC9433570 DOI: 10.3389/fimmu.2022.922173] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Accepted: 07/29/2022] [Indexed: 01/17/2023] Open
Abstract
Macrophage infiltration and polarization have been increasingly observed in intervertebral disc (IVD) degeneration (IDD). However, their biological roles in IDD are still unrevealed. We harvested conditioned media (CM) derived from a spectrum of macrophages induced from THP-1 cells, and examined how they affect nucleus pulposus cells (NPCs) in vitro, by studying cell proliferation, extracellular matrix (ECM) synthesis, and pro-inflammation expression; and in vivo by injection CM in a rat IDD model. Then, high-throughput sequencing was used to detect differentially expressed genes (DEGs). Gene Ontology (GO), the Kyoto Encyclopedia of Genes and Genomes (KEGG), and protein-protein interaction (PPI) networks were used to further analysis. Higher CCR7+ (M1 marker) and CD206+ (M2 marker) cell counts were found in the degenerated human IVD tissues as compared with the control. Furthermore, the cell co-culture model showed M1CM attenuated NPC proliferation, downregulated the expression of ECM anabolic genes encoding aggrecan and collagen IIα1, upregulated the expression of ECM catabolic genes encoding MMP-13, and inflammation-related genes encoding IL-1β, IL-6, and IL-12, while M2CM showed contrasting trends. In IDD model, higher histological scores and lower disc height index were found following M1CM treatment, while M2CM exhibited opposite results. M1CM injection decreased ECM anabolic and increased ECM catabolic, as well as the upregulation of inflammation-related genes after 8 weeks treatment, while M2CM slowed down these trends. Finally, a total of 637 upregulated and 655 downregulated genes were detected in M1CM treated NPCs, and 975 upregulated genes and 930 downregulated genes in the M2CM groups. The top 30 GO terms were shown and the most significant KEGG pathway was cell cycle in both groups. Based on the PPI analysis, the five most significant hub genes were PLK1, KIF20A, RRM2, CDC20, and UBE2C in the M1CM groups and RRM2, CCNB1, CDC20, PLK1, and UBE2C in the M2CM groups. In conclusion, macrophage polarization exhibited diverse roles in IDD progression, with M1CM exacerbating cell proliferation suppression and IVD degeneration, while M2CM attenuated IDD development. These findings may facilitate the further elucidation of the role of macrophage polarization in IDD, and provide novel insights into the therapeutic potential of macrophages.
Collapse
Affiliation(s)
- Xiao-Chuan Li
- Postdoctoral Innovation Practice Base of Gaozhou People’s Hospital, Gaozhou, China
- Department of Orthopedic Surgery, Gaozhou People’s Hospital, Gaozhou, China
- Central Laboratory of Orthopedics, Gaozhou People’s Hospital, Gaozhou, China
| | - Shao-Jian Luo
- Department of Orthopedic Surgery, Gaozhou People’s Hospital, Gaozhou, China
- Central Laboratory of Orthopedics, Gaozhou People’s Hospital, Gaozhou, China
| | - Wu Fan
- Department of Orthopedic Surgery, Gaozhou People’s Hospital, Gaozhou, China
| | - Tian-Li Zhou
- Central Laboratory of Orthopedics, Gaozhou People’s Hospital, Gaozhou, China
| | - Dan-Qin Tan
- Central Laboratory of Orthopedics, Gaozhou People’s Hospital, Gaozhou, China
| | - Rong-Xiong Tan
- Department of Orthopedic Surgery, Gaozhou People’s Hospital, Gaozhou, China
- Central Laboratory of Orthopedics, Gaozhou People’s Hospital, Gaozhou, China
| | - Qun-Ze Xian
- Department of Orthopedic Surgery, Gaozhou People’s Hospital, Gaozhou, China
- Central Laboratory of Orthopedics, Gaozhou People’s Hospital, Gaozhou, China
| | - Jian Li
- Department of Orthopedic Surgery, Gaozhou People’s Hospital, Gaozhou, China
- Central Laboratory of Orthopedics, Gaozhou People’s Hospital, Gaozhou, China
| | - Chun-Ming Huang
- Postdoctoral Innovation Practice Base of Gaozhou People’s Hospital, Gaozhou, China
- Department of Orthopedic Surgery, Gaozhou People’s Hospital, Gaozhou, China
- Central Laboratory of Orthopedics, Gaozhou People’s Hospital, Gaozhou, China
| | - Mao-Sheng Wang
- Postdoctoral Innovation Practice Base of Gaozhou People’s Hospital, Gaozhou, China
- *Correspondence: Mao-Sheng Wang,
| |
Collapse
|
25
|
Liu C, Xu Y, Lu Y, Du P, Li X, Wang C, Guo P, Diao L, Lu G. Mesenchymal stromal cells pretreated with proinflammatory cytokines enhance skin wound healing via IL-6-dependent M2 polarization. Stem Cell Res Ther 2022; 13:414. [PMID: 35964139 PMCID: PMC9375394 DOI: 10.1186/s13287-022-02934-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 05/23/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Numerous studies have shown that mesenchymal stromal cells (MSCs) promote cutaneous wound healing via paracrine signaling. Our previous study found that the secretome of MSCs was significantly amplified by treatment with IFN-γ and TNF-α (IT). It has been known that macrophages are involved in the initiation and termination of inflammation, secretion of growth factors, phagocytosis, cell proliferation, and collagen deposition in wound, which is the key factor during wound healing. In this study, we aim to test whether the supernatant of MSCs pretreated with IT (S-IT MSCs) possesses a more pronounced effect on improving wound healing and describe the interplay between S-IT MSCs and macrophages as well as the potential mechanism in skin wound healing. METHODS In the present study, we used a unique supernatant of MSCs from human umbilical cord-derived MSCs (UC-MSCs) pretreated with IT, designated S-IT MSCs, subcutaneously injected into a mice total skin excision. We evaluated the effect of S-IT MSCs on the speed and quality of wound repair via IT MSCs-derived IL-6-dependent M2 polarization in vivo by hematoxylin-eosin staining (H&E), immunohistochemistry (IHC), immunofluorescence (IF), Masson's trichrome staining, Sirius red staining, quantitative real-time PCR (qPCR). In addition, the effect of S-IT MSCs on the polarization of macrophages toward M2 phenotype and the potential mechanism of it were also investigated in vitro by flow cytometry (FCM), enzyme-linked immunosorbent assay (ELISA), tube formation assay, and western blot analysis. RESULTS Compared with control supernatant (S-MSCs), our H&E and IF results showed that S-IT MSCs were more effectively in promoting macrophages convert to the M2 phenotype and enhancing phagocytosis of M2 macrophages. Meanwhile, the results of tube formation assay, IHC, Masson's trichrome staining, Sirius red staining showed that the abilities of M2 phenotype to promote vascularization and collagen deposition were significantly enhanced by S-IT MSCs-treated, thereby accelerating higher quality wound healing. Further, our ELISA, FCM, qPCR and western blot results showed that IL-6 was highly enriched in S-IT MSCs and acted as a key regulator to induce macrophages convert to the M2 phenotype through IL-6-dependent signaling pathways, ultimately achieving the above function of promoting wound repair. CONCLUSIONS These findings provide the first evidence that the S-IT MSCs is more capable of eliciting M2 polarization of macrophages via IL-6-dependent signaling pathways and accelerating wound healing, which may represent a new strategy for optimizing the therapeutic effect of MSCs on wound healing.
Collapse
Affiliation(s)
- Chenyang Liu
- Nanjng University of Traditional Chinese Medcine, Nanjng, Jiangsu, China.,Engineering Research Center of the Ministry of Education for Wound Repair Technology, Jiangnan University, The Affiliated Hospital of Jiangnan University, Jiangsu, China
| | - Yan Xu
- Nanjng University of Traditional Chinese Medcine, Nanjng, Jiangsu, China.,Engineering Research Center of the Ministry of Education for Wound Repair Technology, Jiangnan University, The Affiliated Hospital of Jiangnan University, Jiangsu, China
| | - Yichi Lu
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Pan Du
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Xiaoxiao Li
- Nanjng University of Traditional Chinese Medcine, Nanjng, Jiangsu, China
| | - Chengchun Wang
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Peng Guo
- Nantong University, Nantong, Jiangsu, China
| | - Ling Diao
- Engineering Research Center of the Ministry of Education for Wound Repair Technology, Jiangnan University, The Affiliated Hospital of Jiangnan University, Jiangsu, China.
| | - Guozhong Lu
- Nanjng University of Traditional Chinese Medcine, Nanjng, Jiangsu, China. .,Engineering Research Center of the Ministry of Education for Wound Repair Technology, Jiangnan University, The Affiliated Hospital of Jiangnan University, Jiangsu, China.
| |
Collapse
|
26
|
Long C, Guo R, Han R, Li K, Wan Y, Xu J, Gong X, Zhao Y, Yao X, Liu J. Effects of macrophages on the proliferation and cardiac differentiation of human induced pluripotent stem cells. Cell Commun Signal 2022; 20:108. [PMID: 35850719 PMCID: PMC9290307 DOI: 10.1186/s12964-022-00916-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 06/09/2022] [Indexed: 12/03/2022] Open
Abstract
Background Macrophage phenotypes switch from proinflammatory (M1) to anti-inflammatory (M2) following myocardial injury. Implanted stem cells (e.g., induced pluripotent stem cells (iPSCs)) for cardiomyogenesis will inevitably contact the inflammatory environment at the myocardial infarction site. To understand how the macrophages affect the behavior of iPSCs, therefore, improve the therapeutic efficacy, we generated three macrophage subtypes and assessed their effects on the proliferation, cardiac differentiation, and maturation of iPSCs. Methods M0, M1, and M2 macrophages were polarized using cytokines, and their properties were confirmed by the expression of specific markers using reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and immunofluorescence. The effects of macrophages on iPSCs were studied using Transwell co-culture models. The proliferative ability of iPSCs was investigated by cell counting and CCK-8 assays. The cardiac differentiation ability of iPSCs was determined by the cardiomyocyte (CM) yield. The maturation of CM was analyzed by the expression of cardiac-specific genes using RT-qPCR, the sarcomere organization using immunofluorescence, and the mitochondrial function using oxidative respiration analysis. Results The data showed that the co-culture of iPSCs with M0, M1, or M2 macrophages significantly decreased iPSCs’ proliferative ability. M2 macrophages did not affect the CM yield during the cardiac differentiation of iPSCs. Still, they promoted the maturation of CM by improving sarcomeric structures, increasing contractile- and ion transport-associated gene expression, and enhancing mitochondrial respiration. M0 macrophages did not significantly affect the cardiomyogenesis ability of iPSCs during co-culture. In contrast, co-culture with M1 macrophages significantly reduced the cardiac differentiation and maturation of iPSCs. Conclusions M1- or M2-polarized macrophages play critical roles in the proliferation, cardiac differentiation, and maturation of iPSCs, providing knowledge to improve the outcomes of stem cell regeneration therapy. Video abstract
Supplementary Information The online version contains supplementary material available at 10.1186/s12964-022-00916-1.
Collapse
Affiliation(s)
- Canling Long
- Central Laboratory, The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen & Longgang District People's Hospital of Shenzhen, Shenzhen, 518172, Guangdong, China
| | - Rui Guo
- Central Laboratory, The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen & Longgang District People's Hospital of Shenzhen, Shenzhen, 518172, Guangdong, China
| | - Ruijuan Han
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen & Longgang District People's Hospital of Shenzhen, Shenzhen, 518172, Guangdong, China
| | - Kang Li
- Central Laboratory, The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen & Longgang District People's Hospital of Shenzhen, Shenzhen, 518172, Guangdong, China
| | - Yanbing Wan
- Central Laboratory, The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen & Longgang District People's Hospital of Shenzhen, Shenzhen, 518172, Guangdong, China
| | - Jiqing Xu
- Cardiothoracic Surgery Department, The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen & Longgang District People's Hospital of Shenzhen, Shenzhen, 518172, Guangdong, China
| | - Xiaoyu Gong
- Central Laboratory, The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen & Longgang District People's Hospital of Shenzhen, Shenzhen, 518172, Guangdong, China
| | - Yanqiu Zhao
- Central Laboratory, The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen & Longgang District People's Hospital of Shenzhen, Shenzhen, 518172, Guangdong, China
| | - Xinhuang Yao
- Central Laboratory, The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen & Longgang District People's Hospital of Shenzhen, Shenzhen, 518172, Guangdong, China
| | - Jia Liu
- Central Laboratory, The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen & Longgang District People's Hospital of Shenzhen, Shenzhen, 518172, Guangdong, China.
| |
Collapse
|
27
|
Yang M, Xiong J, Zou Q, Wang X, Hu K, Zhao Q. Sinapic Acid Attenuated Cardiac Remodeling After Myocardial Infarction by Promoting Macrophage M2 Polarization Through the PPARγ Pathway. Front Cardiovasc Med 2022; 9:915903. [PMID: 35898278 PMCID: PMC9309384 DOI: 10.3389/fcvm.2022.915903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 06/02/2022] [Indexed: 11/22/2022] Open
Abstract
Background Macrophage polarization is an important regulatory mechanism of ventricular remodeling. Studies have shown that sinapic acid (SA) exerts an anti-inflammatory effect. However, the effect of SA on macrophages is still unclear. Objectives The purpose of the study was to investigate the role of SA in macrophage polarization and ventricular remodeling after myocardial infarction (MI). Methods An MI model was established by ligating the left coronary artery. The rats with MI were treated with SA for 1 or 4 weeks after MI. The effect of SA on bone marrow-derived macrophages (BMDMs) was also observed in vitro. Results Cardiac systolic dysfunction was significantly improved after SA treatment. SA reduced MCP-1 and CCR2 expression and macrophage infiltration. SA decreased the levels of the inflammatory factors TNF-α, IL-1α, IL-1β, and iNOS and increased the levels of the M2 macrophage markers CD206, Arg-1, IL-10, Ym-1, Fizz-1, and TGF-β at 1 week after MI. SA significantly increased CD68+/CD206+ macrophage infiltration. Myocardial interstitial fibrosis and MMP-2 and MMP-9 levels were decreased, and the sympathetic nerve marker TH and nerve sprouting marker GAP43 were suppressed after SA treatment at 4 weeks after MI. The PPARγ level was notably upregulated after SA treatment. In vitro, SA also increased the expression of PPARγ mRNA in BMDMs and IL-4-treated BMDMs in a concentration-dependent manner. SA enhanced Arg1 and IL-10 expression in BMDMs, and the PPARγ antagonist GW9662 attenuated M2 macrophage marker expression. Conclusions Our results demonstrated that SA attenuated structural and neural remodeling by promoting macrophage M2 polarization via PPARγ activation after MI.
Collapse
Affiliation(s)
- Mei Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Jun Xiong
- Department of Emergency, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qiang Zou
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Xi Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Ke Hu
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, China
- *Correspondence: Qingyan Zhao
| | - Qingyan Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
- Ke Hu
| |
Collapse
|
28
|
Zarubova J, Hasani-Sadrabadi MM, Ardehali R, Li S. Immunoengineering strategies to enhance vascularization and tissue regeneration. Adv Drug Deliv Rev 2022; 184:114233. [PMID: 35304171 PMCID: PMC10726003 DOI: 10.1016/j.addr.2022.114233] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Revised: 03/07/2022] [Accepted: 03/11/2022] [Indexed: 12/11/2022]
Abstract
Immune cells have emerged as powerful regulators of regenerative as well as pathological processes. The vast majority of regenerative immunoengineering efforts have focused on macrophages; however, growing evidence suggests that other cells of both the innate and adaptive immune system are as important for successful revascularization and tissue repair. Moreover, spatiotemporal regulation of immune cells and their signaling have a significant impact on the regeneration speed and the extent of functional recovery. In this review, we summarize the contribution of different types of immune cells to the healing process and discuss ways to manipulate and control immune cells in favor of vascularization and tissue regeneration. In addition to cell delivery and cell-free therapies using extracellular vesicles, we discuss in situ strategies and engineering approaches to attract specific types of immune cells and modulate their phenotypes. This field is making advances to uncover the extraordinary potential of immune cells and their secretome in the regulation of vascularization and tissue remodeling. Understanding the principles of immunoregulation will help us design advanced immunoengineering platforms to harness their power for tissue regeneration.
Collapse
Affiliation(s)
- Jana Zarubova
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA; Department of Biomaterials and Tissue Engineering, Institute of Physiology of the Czech Academy of Sciences, Prague 14220, Czech Republic
| | | | - Reza Ardehali
- Division of Cardiology, Department of Internal Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA; Eli and Edythe Broad Stem Cell Research Center, University of California, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Song Li
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA; Eli and Edythe Broad Stem Cell Research Center, University of California, Los Angeles, CA 90095, USA; Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA.
| |
Collapse
|
29
|
Hasselbach L, Weidner J, Elsässer A, Theilmeier G. Heart Failure Relapses in Response to Acute Stresses - Role of Immunological and Inflammatory Pathways. Front Cardiovasc Med 2022; 9:809935. [PMID: 35548445 PMCID: PMC9081344 DOI: 10.3389/fcvm.2022.809935] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 03/23/2022] [Indexed: 12/15/2022] Open
Abstract
Cardiovascular diseases continue to be the most imminent health care problems in the western world, accounting for numerous deaths per year. Heart failure (HF), namely the reduction of left ventricular function, is one of the major cardiovascular disease entities. It is chronically progressing with relapsing acute decompensations and an overall grave prognosis that is little different if not worse than most malignant diseases. Interestingly acute metabolically and/or immunologically challenging events like infections or major surgical procedures will cause relapses in the course of preexisting chronic heart failure, decrease the patients wellbeing and worsen myocardial function. HF itself and or its progression has been demonstrated to be driven at least in part by inflammatory pathways that are similarly turned on by infectious or non-infectious stress responses. These thus add to HF progression or relapse. TNF-α plasma levels are associated with disease severity and progression in HF. In addition, several cytokines (e.g., IL-1β, IL-6) are involved in deteriorating left ventricular function. Those observations are based on clinical studies using inhibitors of cytokines or their receptors or they stem from animal studies examining the effect of cytokine mediated inflammation on myocardial remodeling in models of heart failure. This short review summarizes the known underlying immunological processes that are shared by and drive all: chronic heart failure, select infectious diseases, and inflammatory stress responses. In conclusion the text provides a brief summary of the current development in immunomodulatory therapies for HF and their overlap with treatments of other disease entities.
Collapse
Affiliation(s)
- Lisa Hasselbach
- Division of Cardiology and Division of Perioperative Inflammation and Infection, Department Human Medicine, University of Oldenburg, Oldenburg, Germany
| | - Johannes Weidner
- Division of Perioperative Inflammation and Infection, Department Human Medicine, University of Oldenburg, Oldenburg, Germany
| | - Albrecht Elsässer
- Division of Cardiology, Department Human Medicine, University of Oldenburg, Oldenburg, Germany
| | - Gregor Theilmeier
- Division of Perioperative Inflammation and Infection, Department Human Medicine, University of Oldenburg, Oldenburg, Germany
| |
Collapse
|
30
|
Qin YY, Huang XR, Zhang J, Wu W, Chen J, Wan S, Yu XY, Lan HY. Neuropeptide Y attenuates cardiac remodeling and deterioration of function following myocardial infarction. Mol Ther 2022; 30:881-897. [PMID: 34628054 PMCID: PMC8821956 DOI: 10.1016/j.ymthe.2021.10.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 04/29/2021] [Accepted: 09/30/2021] [Indexed: 02/04/2023] Open
Abstract
Plasma levels of neuropeptide Y (NPY) are elevated in patients with acute myocardial infarction (AMI), but its role in AMI remains unclear, which was examined here in NPY wild-type/knockout (WT/KO) mice treated with/without exogenous NPY and its Y1 receptor antagonist (Y1Ra) BIBP 3226. We found that AMI mice lacking NPY developed more severe AMI than WT mice with worse cardiac dysfunction, progressive cardiac inflammation and fibrosis, and excessive apoptosis but impairing angiogenesis. All of these changes were reversed when the NPY KO mice were treated with exogenous NPY in a dose-dependent manner. Interestingly, treatment with NPY also dose dependently attenuated AMI in WT mice, which was blocked by BIBP 3226. Phenotypically, cardiac NPY was de novo expressed by infiltrating macrophages during the repairing or fibrosing process in heart-failure patients and AMI mice. Mechanistically, NPY was induced by transforming growth factor (TGF)-β1 in bone marrow-derived macrophages and signaled through its Y1R to exert its pathophysiological activities by inhibiting p38/nuclear factor κB (NF-κB)-mediated M1 macrophage activation while promoting the reparative M2 phenotype in vivo and in vitro. In conclusion, NPY can attenuate AMI in mice. Inhibition of cardiac inflammation and fibrosis while enhancing angiogenesis but reducing apoptosis may be the underlying mechanisms through which NPY attenuates cardiac remodeling and deterioration of function following AMI.
Collapse
Affiliation(s)
- Yu-Yan Qin
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 511436, China; Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, Lui Che Woo Institute of Innovative Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, New Territories, Hong Kong, China
| | - Xiao-Ru Huang
- Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, Lui Che Woo Institute of Innovative Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, New Territories, Hong Kong, China; Guangdong-Hong Kong Joint Laboratory on Immunological and Genetic Kidney Diseases, Guangdong Academy of Medical Sciences, Guangdong Provincial People's Hospital, Guangzhou, China
| | - Jian Zhang
- Department of Cardiovascular Surgery, Shenyang Northern Hospital, No. 83, Wenhua Road, Shenhe District, Shenyang, Liaoning, China
| | - Wenjing Wu
- Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, Lui Che Woo Institute of Innovative Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, New Territories, Hong Kong, China
| | - Junzhe Chen
- Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, Lui Che Woo Institute of Innovative Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, New Territories, Hong Kong, China
| | - Song Wan
- Department of Surgery, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Xi-Yong Yu
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 511436, China.
| | - Hui-Yao Lan
- Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, Lui Che Woo Institute of Innovative Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, New Territories, Hong Kong, China; The Chinese University of Hong Kong (CUHK)-Guangdong Provincial People's Hospital Joint Research Laboratory on Immunological and Genetic Kidney Diseases, CUHK, Hong Kong, China.
| |
Collapse
|
31
|
Snarski P, Sukhanov S, Yoshida T, Higashi Y, Danchuk S, Chandrasekar B, Tian D, Rivera-Lopez V, Delafontaine P. Macrophage-Specific IGF-1 Overexpression Reduces CXCL12 Chemokine Levels and Suppresses Atherosclerotic Burden in Apoe-Deficient Mice. Arterioscler Thromb Vasc Biol 2022; 42:113-126. [PMID: 34852642 PMCID: PMC8792341 DOI: 10.1161/atvbaha.121.316090] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
OBJECTIVE IGF-1 (insulin-like growth factor 1) exerts pleiotropic effects including promotion of cellular growth, differentiation, survival, and anabolism. We have shown that systemic IGF-1 administration reduced atherosclerosis in Apoe-/- (apolipoprotein E deficient) mice, and this effect was associated with a reduction in lesional macrophages and a decreased number of foam cells in the plaque. Almost all cell types secrete IGF-1, but the effect of macrophage-derived IGF-1 on the pathogenesis of atherosclerosis is poorly understood. We hypothesized that macrophage-derived IGF-1 will reduce atherosclerosis. Approach and Results: We created macrophage-specific IGF-1 overexpressing mice on an Apoe-/- background. Macrophage-specific IGF-1 overexpression reduced plaque macrophages, foam cells, and atherosclerotic burden and promoted features of stable atherosclerotic plaque. Macrophage-specific IGF1 mice had a reduction in monocyte infiltration into plaque, decreased expression of CXCL12 (CXC chemokine ligand 12), and upregulation of ABCA1 (ATP-binding cassette transporter 1), a cholesterol efflux regulator, in atherosclerotic plaque and in peritoneal macrophages. IGF-1 prevented oxidized lipid-induced CXCL12 upregulation and foam cell formation in cultured THP-1 macrophages and increased lipid efflux. We also found an increase in cholesterol efflux in macrophage-specific IGF1-derived peritoneal macrophages. CONCLUSIONS Macrophage IGF-1 overexpression reduced atherosclerotic burden and increased features of plaque stability, likely via a reduction in CXCL12-mediated monocyte recruitment and an increase in ABCA1-dependent macrophage lipid efflux.
Collapse
Affiliation(s)
- Patricia Snarski
- Section of Cardiology, John W. Deming Department of Medicine, Tulane University School of Medicine, New Orleans, LA,Department of Physiology, Tulane University School of Medicine, New Orleans, LA
| | - Sergiy Sukhanov
- Section of Cardiology, John W. Deming Department of Medicine, Tulane University School of Medicine, New Orleans, LA,Department of Physiology, Tulane University School of Medicine, New Orleans, LA
| | - Tadashi Yoshida
- Section of Cardiology, John W. Deming Department of Medicine, Tulane University School of Medicine, New Orleans, LA,Department of Physiology, Tulane University School of Medicine, New Orleans, LA
| | - Yusuke Higashi
- Section of Cardiology, John W. Deming Department of Medicine, Tulane University School of Medicine, New Orleans, LA,Department of Physiology, Tulane University School of Medicine, New Orleans, LA
| | - Svitlana Danchuk
- Section of Cardiology, John W. Deming Department of Medicine, Tulane University School of Medicine, New Orleans, LA,Department of Physiology, Tulane University School of Medicine, New Orleans, LA
| | - Bysani Chandrasekar
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO,Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO
| | - Di Tian
- Department of Pathology, Tulane University School of Medicine, New Orleans, LA
| | | | - Patrick Delafontaine
- Section of Cardiology, John W. Deming Department of Medicine, Tulane University School of Medicine, New Orleans, LA,Department of Physiology, Tulane University School of Medicine, New Orleans, LA,Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA
| |
Collapse
|
32
|
Liu J, Wang H, Zhang L, Li X, Ding X, Ding G, Wei F. Periodontal ligament stem cells promote polarization of M2 macrophages. J Leukoc Biol 2022; 111:1185-1197. [PMID: 34982483 DOI: 10.1002/jlb.1ma1220-853rr] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Macrophages are widely distributed in a variety of tissues, and the different state of macrophages polarization is closely related to the occurrence, development, and prognosis of inflammation, including periodontitis, a chronic inflammatory disease leading to tooth loss worldwide. Periodontal ligament stem cells (PDLSCs) play a key role in immune regulation and periodontal tissues regeneration, contributing to cell-based therapy of periodontitis. However, the interactions between PDLSCs and macrophages are still elusive. The purpose of present study is to investigate the effect of PDLSCs conditioned medium (PDLSCs-CM) on the macrophage polarization and the possible mechanism. PDLSCs were isolated using tissue explant methods and characterized via multipotent differentiation test and examination of expression profiles of mesenchymal stem cells (MSCs) markers. The supernatant of PDLSCs was collected, centrifuged, filtered, and used as PDLSCs-CM. Then, PDLSCs-CM was cocultured with M0 macrophages or IL-4- and IL-13-induced M2 macrophages. The level of surface markers of M1/M2 macrophages and production of several proinflammatory or anti-inflammatory factors were evaluated by flow cytometric analysis and enzyme-linked immunosorbent assay, respectively. The associated genes and proteins involved in the JNK pathway were investigated to explore the potential mechanism that may regulate PDLSCs-CM-mediated macrophage polarization. PDLSCs expressed MSCs markers, including STRO-1, CD146, CD90, and CD73, and were negative for CD34 and CD45, could undergo osteogenic and adipogenic differentiation when cultured in defined medium. After incubation with PDLSCs-CM, no significant increase of CD80+ and HLA-DR+ M1 macrophages was shown while evaluated CD209+ and CD206+ M2 macrophages were observed. In addition, the levels of anti-inflammatory factors such as IL-10, TGF-β, and CCL18 were increased instead of proinflammatory factors such as IL-1β, TNF-α with PDLSC-CM treatment. There was a decrease of JNK expression on M0 macrophages by qRT-PCR analysis and an increase of protein phosphorylation on M0 macrophages after incubation with PDLSCs-CM. Furthermore, as for the enhancement of IL-4- and IL-13-mediated M2 polarization by PDLSCs-CM, the mRNA level of JNK decreased, and the protein phosphorylation level of JNK increased. In addition, the treatment of JNK pathway inhibitor, SP600125, could inhibit the expression and secretion level of anti-inflammatory factor such as IL-10 in M2 polarization induced by PDLSCs-CM. Collectively, PDLSCs were able to induce M2 macrophage polarization instead of M1 polarization, and capable of enhancing M2 macrophage polarization induced by IL-4 and IL-13. The JNK pathway was involved in the promotion of M2 macrophage polarization.
Collapse
Affiliation(s)
- Jiani Liu
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China
| | - Hong Wang
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China
| | - Ludan Zhang
- School of Stomatology, Weifang Medical University, Weifang, China
| | - Xiaoyu Li
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China
| | - Xiaoling Ding
- Clinical Competency Training Center, Weifang Medical University, Weifang, China
| | - Gang Ding
- School of Stomatology, Weifang Medical University, Weifang, China
| | - Fulan Wei
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China
| |
Collapse
|
33
|
Han H, Dai D, Du R, Hu J, Zhu Z, Lu L, Zhu J, Zhang R. Oncostatin M promotes infarct repair and improves cardiac function after myocardial infarction. Am J Transl Res 2021; 13:11329-11340. [PMID: 34786061 PMCID: PMC8581943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 08/07/2021] [Indexed: 06/13/2023]
Abstract
Myocardial infarction (MI) is one of the leading causes of morbidity and mortality worldwide. The immune response plays a central role in post-MI cardiac repair. A growing body of evidence suggests that oncostatin M (OSM), a pleiomorphic cytokine of the interleukin (IL)-6 family, participates in the cardiac healing and remodeling process. However, previous studies have shown inconsistent results, and the exact mechanisms underlying this process have not yet been fully elucidated. We verified whether OSM is involved in the healing process and cardiac remodeling after MI and sought to explore its potential mechanisms. Our data implied OSM's role in facilitating the post-MI healing process in mice, manifested by improved cardiac functional performance and a reduction in fibrotic changes. Furthermore, our flow cytometry analysis revealed that OSM influences the dynamics of cardiac monocytes and macrophages. In mice with a blunted C-X-C motif receptor (CCR)2 signaling pathway, OSM reserved its protective roles and polarized cardiac macrophages toward a reparative phenotype. Moreover, OSM reduced the number of matrix metalloproteinase (MMP)-9+ immune cells and increased the number of tissue inhibitor of metalloproteinase (TIMP)-1+ immune cells in the infarct area, mitigating the maladaptive remodeling following MI. These findings demonstrate that OSM favorably modulates cardiac remodeling, partially by accelerating the shift in the cardiac macrophage phenotype from M1 to M2 and by correcting the MMP-9 and TIMP-1 balance.
Collapse
Affiliation(s)
- Hui Han
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of MedicineShanghai 200025, P. R. China
- Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of MedicineShanghai 200025, P. R. China
| | - Daopeng Dai
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of MedicineShanghai 200025, P. R. China
- Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of MedicineShanghai 200025, P. R. China
| | - Run Du
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of MedicineShanghai 200025, P. R. China
| | - Jinquan Hu
- Department of Orthopaedics, Changzheng Hospital Affiliated with Second Military Medical UniversityShanghai 200003, P. R. China
| | - Zhengbin Zhu
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of MedicineShanghai 200025, P. R. China
| | - Lin Lu
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of MedicineShanghai 200025, P. R. China
- Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of MedicineShanghai 200025, P. R. China
| | - Jinzhou Zhu
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of MedicineShanghai 200025, P. R. China
| | - Ruiyan Zhang
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of MedicineShanghai 200025, P. R. China
- Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of MedicineShanghai 200025, P. R. China
| |
Collapse
|
34
|
Song J, Frieler RA, Vigil TM, Ma J, Brombacher F, Goonewardena SN, Goldstein DR, Mortensen RM. Inactivation of Interleukin-4 Receptor α Signaling in Myeloid Cells Protects Mice From Angiotensin II/High Salt-Induced Cardiovascular Dysfunction Through Suppression of Fibrotic Remodeling. J Am Heart Assoc 2021; 10:e017329. [PMID: 34132103 PMCID: PMC8403318 DOI: 10.1161/jaha.120.017329] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Background Hypertension‐induced cardiovascular remodeling is characterized by chronic low‐grade inflammation. Interleukin‐4 receptor α (IL‐4Rα) signaling is importantly involved in cardiovascular remodeling, however, the target cell type(s) is unclear. Here, we investigated the role of myeloid‐specific IL‐4Rα signaling in cardiovascular remodeling induced by angiotensin II and high salt. Methods and Results Myeloid IL‐4Rα deficiency suppressed both the in vitro and in vivo expression of alternatively activated macrophage markers including Arg1 (arginase 1), Ym1 (chitinase 3‐like 3), and Relmα/Fizz1 (resistin‐like molecule α). After angiotensin II and high salt treatment, myeloid‐specific IL‐4Rα deficiency did not change hypertrophic remodeling within the heart and aorta. However, myeloid IL‐4Rα deficiency resulted in a substantial reduction in fibrosis through the suppression of profibrotic pathways and the enhancement of antifibrotic signaling. Decreased fibrosis was associated with significant preservation of myocardial function in MyIL4RαKO mice and was mediated by attenuated alternative macrophage activation. Conclusions Myeloid IL‐4Rα signaling is substantially involved in fibrotic cardiovascular remodeling by controlling alternative macrophage activation and regulating fibrosis‐related signaling. Inhibiting myeloid IL‐4Rα signaling may be a potential strategy to prevent hypertensive cardiovascular diseases.
Collapse
Affiliation(s)
- Jianrui Song
- Department of Cell and Developmental Biology University of Michigan Medical School Ann Arbor MI.,Department of Molecular and Integrative Physiology University of Michigan Ann Arbor MI
| | - Ryan A Frieler
- Department of Molecular and Integrative Physiology University of Michigan Ann Arbor MI
| | - Thomas M Vigil
- Department of Molecular and Integrative Physiology University of Michigan Ann Arbor MI
| | - Jun Ma
- Department of Thoracic Surgery Shanxi Province People's Hospital Taiyuan P.R. China
| | - Frank Brombacher
- International Center for Genetic Engineering and Biotechnology University of Cape TownDivision of Immunology and South African Medical Research Council (SAMRC) Cape Town South Africa
| | - Sascha N Goonewardena
- Division of Cardiovascular Medicine Department of Internal Medicine University of Michigan Ann Arbor MI
| | - Daniel R Goldstein
- Division of Cardiovascular Medicine Department of Internal Medicine University of Michigan Ann Arbor MI.,Institute of Gerontology University of Michigan Ann Arbor MI.,Department of Microbiology and Immunology University of Michigan Ann Arbor MI
| | - Richard M Mortensen
- Department of Molecular and Integrative Physiology University of Michigan Ann Arbor MI.,Division of Metabolism, Endocrinology, and Diabetes Department of Internal Medicine University of Michigan Ann Arbor MI.,Department of Pharmacology University of Michigan Ann Arbor MI
| |
Collapse
|
35
|
Li L, Wei K, Ding Y, Ahati P, Xu H, Fang H, Wang H. M2a Macrophage-Secreted CHI3L1 Promotes Extracellular Matrix Metabolic Imbalances via Activation of IL-13Rα2/MAPK Pathway in Rat Intervertebral Disc Degeneration. Front Immunol 2021; 12:666361. [PMID: 34168643 PMCID: PMC8217759 DOI: 10.3389/fimmu.2021.666361] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 05/24/2021] [Indexed: 12/17/2022] Open
Abstract
The accumulation of macrophages in degenerated discs is a common phenomenon. However, the roles and mechanisms of M2a macrophages in intervertebral disc degeneration (IDD) have not been illuminated. This study investigated the expression of the M2a macrophage marker (CD206) in human and rat intervertebral disc tissues by immunohistochemistry. To explore the roles of M2a macrophages in IDD, nucleus pulposus (NP) cells were co-cultured with M2a macrophages in vitro. To clarify whether the CHI3L1 protein mediates the effect of M2a macrophages on NP cells, siRNA was used to knock down CHI3L1 transcription. To elucidate the underlying mechanisms, NP cells were incubated with recombinant CHI3L1 proteins, then subjected to western blotting analysis of the IL-13Rα2 receptor and MAPK pathway. CD206-positive cells were detected in degenerated human and rat intervertebral disc tissues. Notably, M2a macrophages promoted the expression of catabolism genes (MMP-3 and MMP-9) and suppressed the expression of anabolism genes (aggrecan and collagen II) in NP cells. These effects were abrogated by CHI3L1 knockdown in M2a macrophages. Exposure to recombinant CHI3L1 promoted an extracellular matrix metabolic imbalance in NP cells via the IL-13Rα2 receptor, along with activation of the ERK and JNK MAPK signaling pathways. This study elucidated the roles of M2a macrophages in IDD and identified potential mechanisms for these effects.
Collapse
Affiliation(s)
- Long Li
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kang Wei
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yifan Ding
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Paerxiati Ahati
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Haoran Xu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huang Fang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huan Wang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
36
|
Li Y, Dong M, Wang Q, Kumar S, Zhang R, Cheng W, Xiang J, Wang G, Ouyang K, Zhou R, Xie Y, Lu Y, Yi J, Duan H, Liu J. HIMF deletion ameliorates acute myocardial ischemic injury by promoting macrophage transformation to reparative subtype. Basic Res Cardiol 2021; 116:30. [PMID: 33893593 PMCID: PMC8064941 DOI: 10.1007/s00395-021-00867-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 03/31/2021] [Indexed: 12/12/2022]
Abstract
Appropriately manipulating macrophage M1/M2 phenotypic transition is a promising therapeutic strategy for tissue repair after myocardial infarction (MI). Here we showed that gene ablation of hypoxia-induced mitogenic factor (HIMF) in mice (Himf−/− and HIMFflox/flox;Lyz2-Cre) attenuated M1 macrophage-dominated inflammatory response and promoted M2 macrophage accumulation in infarcted hearts. This in turn reduced myocardial infarct size and improved cardiac function after MI. Correspondingly, expression of HIMF in macrophages induced expression of pro-inflammatory cytokines; the culturing medium of HIMF-overexpressing macrophages impaired the cardiac fibroblast viability and function. Furthermore, macrophage HIMF was found to up-regulate C/EBP-homologous protein (CHOP) expression, which exaggerated the release of pro-inflammatory cytokines via activating signal transducer of activator of transcription 1 (STAT1) and 3 (STAT3) signaling. Together these data suggested that HIMF promotes M1-type and prohibits M2-type macrophage polarization by activating the CHOP–STAT1/STAT3 signaling pathway to negatively regulate myocardial repair. HIMF might thus constitute a novel target to treat MI.
Collapse
Affiliation(s)
- Yanjiao Li
- Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Department of Pathophysiology, Shenzhen University Health Science Center, Shenzhen, 518060, China.,Guangdong Key Laboratory of Regional Immunity and Diseases, Department of Pathology, Shenzhen University Health Science Center, Shenzhen, 518060, China
| | - Min Dong
- Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Department of Pathophysiology, Shenzhen University Health Science Center, Shenzhen, 518060, China.,Guangdong Key Laboratory of Regional Immunity and Diseases, Department of Pathology, Shenzhen University Health Science Center, Shenzhen, 518060, China
| | - Qing Wang
- Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Department of Pathophysiology, Shenzhen University Health Science Center, Shenzhen, 518060, China.,Guangdong Key Laboratory of Regional Immunity and Diseases, Department of Pathology, Shenzhen University Health Science Center, Shenzhen, 518060, China
| | - Santosh Kumar
- Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Department of Pathophysiology, Shenzhen University Health Science Center, Shenzhen, 518060, China.,Guangdong Key Laboratory of Regional Immunity and Diseases, Department of Pathology, Shenzhen University Health Science Center, Shenzhen, 518060, China
| | - Rui Zhang
- Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Department of Pathophysiology, Shenzhen University Health Science Center, Shenzhen, 518060, China.,Guangdong Key Laboratory of Regional Immunity and Diseases, Department of Pathology, Shenzhen University Health Science Center, Shenzhen, 518060, China
| | - Wanwen Cheng
- Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Department of Pathophysiology, Shenzhen University Health Science Center, Shenzhen, 518060, China.,Guangdong Key Laboratory of Regional Immunity and Diseases, Department of Pathology, Shenzhen University Health Science Center, Shenzhen, 518060, China
| | - Jiaqing Xiang
- Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Department of Pathophysiology, Shenzhen University Health Science Center, Shenzhen, 518060, China.,Guangdong Key Laboratory of Regional Immunity and Diseases, Department of Pathology, Shenzhen University Health Science Center, Shenzhen, 518060, China
| | - Gang Wang
- Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Department of Pathophysiology, Shenzhen University Health Science Center, Shenzhen, 518060, China.,Guangdong Key Laboratory of Regional Immunity and Diseases, Department of Pathology, Shenzhen University Health Science Center, Shenzhen, 518060, China
| | - Kunfu Ouyang
- Drug Discovery Center, State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Shenzhen Graduate School, Peking University, Shenzhen, 51055, China
| | - Ruxing Zhou
- Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Department of Pathophysiology, Shenzhen University Health Science Center, Shenzhen, 518060, China.,Guangdong Key Laboratory of Regional Immunity and Diseases, Department of Pathology, Shenzhen University Health Science Center, Shenzhen, 518060, China
| | - Yaohong Xie
- Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Department of Pathophysiology, Shenzhen University Health Science Center, Shenzhen, 518060, China.,Guangdong Key Laboratory of Regional Immunity and Diseases, Department of Pathology, Shenzhen University Health Science Center, Shenzhen, 518060, China
| | - Yishen Lu
- Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Department of Pathophysiology, Shenzhen University Health Science Center, Shenzhen, 518060, China.,Guangdong Key Laboratory of Regional Immunity and Diseases, Department of Pathology, Shenzhen University Health Science Center, Shenzhen, 518060, China
| | - Jing Yi
- Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Department of Pathophysiology, Shenzhen University Health Science Center, Shenzhen, 518060, China.,Guangdong Key Laboratory of Regional Immunity and Diseases, Department of Pathology, Shenzhen University Health Science Center, Shenzhen, 518060, China
| | - Haixia Duan
- Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Department of Pathophysiology, Shenzhen University Health Science Center, Shenzhen, 518060, China.,Guangdong Key Laboratory of Regional Immunity and Diseases, Department of Pathology, Shenzhen University Health Science Center, Shenzhen, 518060, China
| | - Jie Liu
- Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Department of Pathophysiology, Shenzhen University Health Science Center, Shenzhen, 518060, China. .,Guangdong Key Laboratory of Regional Immunity and Diseases, Department of Pathology, Shenzhen University Health Science Center, Shenzhen, 518060, China.
| |
Collapse
|
37
|
Walravens AS, Smolgovsky S, Li L, Kelly L, Antes T, Peck K, Quon T, Ibrahim A, Marbán E, Berman B, Marbán L, R-Borlado L, de Couto G. Mechanistic and therapeutic distinctions between cardiosphere-derived cell and mesenchymal stem cell extracellular vesicle non-coding RNA. Sci Rep 2021; 11:8666. [PMID: 33883598 PMCID: PMC8060398 DOI: 10.1038/s41598-021-87939-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 03/30/2021] [Indexed: 02/06/2023] Open
Abstract
Cell therapy limits ischemic injury following myocardial infarction (MI) by preventing cell death, modulating the immune response, and promoting tissue regeneration. The therapeutic efficacy of cardiosphere-derived cells (CDCs) and mesenchymal stem cells (MSCs) is associated with extracellular vesicle (EV) release. Prior head-to-head comparisons have shown CDCs to be more effective than MSCs in MI models. Despite differences in cell origin, it is unclear why EVs from different adult stem cell populations elicit differences in therapeutic efficacy. Here, we compare EVs derived from multiple human MSC and CDC donors using diverse in vitro and in vivo assays. EV membrane protein and non-coding RNA composition are highly specific to the parent cell type; for example, miR-10b is enriched in MSC-EVs relative to CDC-EVs, while Y RNA fragments follow the opposite pattern. CDC-EVs enhance the Arg1/Nos2 ratio in macrophages in vitro and reduce MI size more than MSC-EVs and suppress inflammation during acute peritonitis in vivo. Thus, CDC-EVs are distinct from MSC-EVs, confer immunomodulation, and protect the host against ischemic myocardial injury and acute inflammation.
Collapse
Affiliation(s)
| | - Sasha Smolgovsky
- Capricor Therapeutics, Inc., 8840 Wilshire Blvd., Beverly Hills, CA, 90211, USA
| | - Liang Li
- Capricor Therapeutics, Inc., 8840 Wilshire Blvd., Beverly Hills, CA, 90211, USA
| | - Lauren Kelly
- Capricor Therapeutics, Inc., 8840 Wilshire Blvd., Beverly Hills, CA, 90211, USA
| | - Travis Antes
- Capricor Therapeutics, Inc., 8840 Wilshire Blvd., Beverly Hills, CA, 90211, USA
| | - Kiel Peck
- Capricor Therapeutics, Inc., 8840 Wilshire Blvd., Beverly Hills, CA, 90211, USA
| | - Tanner Quon
- Capricor Therapeutics, Inc., 8840 Wilshire Blvd., Beverly Hills, CA, 90211, USA
| | - Ahmed Ibrahim
- Smidt Heart Institute, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Los Angeles, CA, 90048, USA
| | - Eduardo Marbán
- Smidt Heart Institute, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Los Angeles, CA, 90048, USA
| | - Benjamin Berman
- Center for Bioinformatics and Functional Genomics, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Linda Marbán
- Capricor Therapeutics, Inc., 8840 Wilshire Blvd., Beverly Hills, CA, 90211, USA.
| | - Luis R-Borlado
- Capricor Therapeutics, Inc., 8840 Wilshire Blvd., Beverly Hills, CA, 90211, USA
| | - Geoffrey de Couto
- Capricor Therapeutics, Inc., 8840 Wilshire Blvd., Beverly Hills, CA, 90211, USA.
- Smidt Heart Institute, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Los Angeles, CA, 90048, USA.
| |
Collapse
|
38
|
Zhang J, Wang J, Wu Y, Li W, Gong K, Zhao P. Identification of SLED1 as a Potential Predictive Biomarker and Therapeutic Target of Post-Infarct Heart Failure by Bioinformatics Analyses. Int Heart J 2021; 62:23-32. [PMID: 33518662 DOI: 10.1536/ihj.20-439] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The aim of this study was to explore potential predictive biomarkers and therapeutic targets of post-infarct heart failure (HF) using bioinformatics analyses.CEL raw data of GSE59867 and GSE62646 were downloaded from the GEO database. Differentially expressed genes (DEGs) between patients with ST-segment elevation myocardial infarction (STEMI) and those with stable coronary artery disease (CAD) at admission and DEGs between admission and 6 months after myocardial infarction (MI) in patients with STEMI were analyzed. A gene ontology (GO) analysis and a gene set enrichment analysis (GSEA) were performed, and a protein-protein interaction network was constructed. Critical genes were further analyzed.In total, 147 DEGs were screened between STEMI and CAD at admission, and 62 DEGs were identified in patients with STEMI between admission and 6 months after MI. The results of GO and GSEA indicate that neutrophils, neutrophil-related immunity responses, and monocytes/macrophages play important roles in MI pathogenesis. SLED1 expression was higher in patients with HF than in those without HF at admission and 1 month after MI. GSEA indicates that mTORC1 activation, E2F targets, G2M checkpoint, and MYC targets v1 inhibition may play key roles in the development of post-infarct HF. Furthermore, SLED1 may be involved in the development of post-infarct HF by activating mTORC1 and inhibiting E2F targets, G2M checkpoint, and MYC targets v1.SLED1 may be a novel biomarker of post-infarct HF and may serve as a potential therapeutic target in this disease.
Collapse
Affiliation(s)
- Jiajia Zhang
- Department of Cardiology, The Affiliated Hospital of Yangzhou University, Yangzhou University
| | - Jun Wang
- Department of Cardiology, The Affiliated Hospital of Yangzhou University, Yangzhou University
| | - Yong Wu
- Department of Cardiology, The Affiliated Hospital of Yangzhou University, Yangzhou University
| | - Wei Li
- Department of Cardiology, The Affiliated Hospital of Yangzhou University, Yangzhou University
| | - Kaizheng Gong
- Department of Cardiology, The Affiliated Hospital of Yangzhou University, Yangzhou University
| | - Pei Zhao
- Department of Cardiology, The Affiliated Hospital of Yangzhou University, Yangzhou University
| |
Collapse
|
39
|
Cheong A, McGrath S, Robinson T, Maliki R, Spurling A, Lock P, Rephaeli A, Nudelman A, Parker BS, Pepe S, Cutts SM. A switch in mechanism of action prevents doxorubicin-mediated cardiac damage. Biochem Pharmacol 2021; 185:114410. [PMID: 33428897 DOI: 10.1016/j.bcp.2021.114410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 01/03/2021] [Accepted: 01/05/2021] [Indexed: 10/22/2022]
Abstract
Cancer patients treated with doxorubicin are at risk of congestive heart failure due to doxorubicin-mediated cardiotoxicity via topoisomerase IIβ poisoning. Acute cardiac muscle damage occurs in response to the very first dose of doxorubicin, however, cardioprotection has been reported after co-treatment of doxorubicin with acyloxyalkyl ester prodrugs. The aim of this study was to examine the role played by various forms of acute cardiac damage mediated by doxorubicin and determine a mechanism for the cardioprotective effect of formaldehyde-releasing prodrug AN-9 (pivaloyloxymethyl butyrate). Doxorubicin-induced cardiac damage in BALB/c mice bearing mammary tumours was established with a single dose of doxorubicin (4 or 16 mg/kg) administered alone or in combination with AN-9 (100 mg/kg). AN-9 protected the heart from doxorubicin-induced myocardial apoptosis and also significantly reduced dsDNA breaks, independent from the level of doxorubicin biodistribution to the heart. Covalent incorporation of [14C]doxorubicin into DNA showed that the combination treatment yielded significantly higher levels of formaldehyde-mediated doxorubicin-DNA adducts compared to doxorubicin alone, yet this form of damage was associated with cardioprotection from apoptosis. The cardiac transcriptomic analysis indicates that the combination treatment initiates inflammatory response signalling pathways. Doxorubicin and AN-9 combination treatments were cardioprotective, yet preserved doxorubicin-mediated anti-tumour proliferation and apoptosis in mammary tumours. This was associated with a switch in doxorubicin action from cardiac topoisomerase IIβ poisoning to covalent-DNA adduct formation. Co-administration of doxorubicin and formaldehyde-releasing prodrugs, such as AN-9, may be a promising cardioprotective therapy while maintaining doxorubicin activity in primary mammary tumours.
Collapse
Affiliation(s)
- Alison Cheong
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia
| | - Sean McGrath
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia
| | - Tina Robinson
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia
| | - Ruqaya Maliki
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia
| | - Alex Spurling
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia
| | - Peter Lock
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia
| | - Ada Rephaeli
- Laboratory for Pharmacology and Experimental Oncology, Felsenstein Medical Research Center, Petach Tikva, Israel; Sackler Faculty of Medicine, Tel Aviv University, 49100 Tel Aviv, Israel
| | - Abraham Nudelman
- Division of Medicinal Chemistry, Department of Chemistry, Bar-Ilan University, Ramat Gan 52900, Israel
| | - Belinda S Parker
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia
| | - Salvatore Pepe
- Murdoch Children's Research Institute, Department of Cardiology, Royal Children's Hospital, Australia; Department of Paediatrics, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Suzanne M Cutts
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia.
| |
Collapse
|
40
|
Lin Y, Ding S, Chen Y, Xiang M, Xie Y. Cardiac Adipose Tissue Contributes to Cardiac Repair: a Review. Stem Cell Rev Rep 2021; 17:1137-1153. [PMID: 33389679 DOI: 10.1007/s12015-020-10097-4] [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] [Accepted: 11/25/2020] [Indexed: 02/06/2023]
Abstract
Cardiac adipose tissue is a metabolically active adipose tissue in close proximity to heart. Recent studies emphasized the benefits of cardiac adipose tissue in heart remodeling, such as reducing infarction size, enhancing neovascularization and regulating immune response, through a series of cellular mechanisms. In the present manuscript, we provide a comprehensive review regarding the role of cardiac adipose tissue in cardiac repair. We focus on different cardiac adipose tissues according to their distinguished anatomical structures. This review summarizes the latest evidence on the relationship between cardiac adipose tissue and cardiac repair. Cardiac adipose tissues (CAT) were systematically reviewed in the current manuscript which focused on the components of CAT, debates about cardiac adipose stem cells and their effect on heart.
Collapse
Affiliation(s)
- Yan Lin
- Department of Cardiology, Cardiovascular Key Laboratory of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China
| | - Siyin Ding
- Department of Cardiology, Cardiovascular Key Laboratory of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China
| | - Yuwen Chen
- Department of Cardiology, Cardiovascular Key Laboratory of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China
| | - Meixiang Xiang
- Department of Cardiology, Cardiovascular Key Laboratory of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China.
| | - Yao Xie
- Department of Cardiology, Cardiovascular Key Laboratory of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China.
| |
Collapse
|
41
|
Song J, Frieler RA, Whitesall SE, Chung Y, Vigil TM, Muir LA, Ma J, Brombacher F, Goonewardena SN, Lumeng CN, Goldstein DR, Mortensen RM. Myeloid interleukin-4 receptor α is essential in postmyocardial infarction healing by regulating inflammation and fibrotic remodeling. Am J Physiol Heart Circ Physiol 2021; 320:H323-H337. [PMID: 33164548 PMCID: PMC7847075 DOI: 10.1152/ajpheart.00251.2020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 10/23/2020] [Accepted: 10/29/2020] [Indexed: 02/06/2023]
Abstract
Interleukin-4 receptor α (IL4Rα) signaling plays an important role in cardiac remodeling during myocardial infarction (MI). However, the target cell type(s) of IL4Rα signaling during this remodeling remains unclear. Here, we investigated the contribution of endogenous myeloid-specific IL4Rα signaling in cardiac remodeling post-MI. We established a murine myeloid-specific IL4Rα knockout (MyIL4RαKO) model with LysM promoter-driven Cre recombination. Macrophages from MyIL4RαKO mice showed significant downregulation of alternatively activated macrophage markers but an upregulation of classical activated macrophage markers both in vitro and in vivo, indicating the successful inactivation of IL4Rα signaling in macrophages. To examine the role of myeloid IL4Rα during MI, we subjected MyIL4RαKO and littermate floxed control (FC) mice to MI. We found that cardiac function was significantly impaired as a result of myeloid-specific IL4Rα deficiency. This deficiency resulted in a dysregulated inflammatory response consisting of decreased production of anti-inflammatory cytokines. Myeloid IL4Rα deficiency also led to reduced collagen 1 deposition and an imbalance of matrix metalloproteinases (MMPs)/tissue inhibitors of metalloproteinases (TIMPs), with upregulated MMPs and downregulated TIMPs, which resulted in insufficient fibrotic remodeling. In conclusion, this study identifies that myeloid-specific IL4Rα signaling regulates inflammation and fibrotic remodeling during MI. Therefore, myeloid-specific activation of IL4Rα signaling could offer protective benefits after MI.NEW & NOTEWORTHY This study showed, for the first time, the role of endogenous IL4Rα signaling in myeloid cells during cardiac remodeling and the underlying mechanisms. We identified myeloid cells are the critical target cell types of IL4Rα signaling during cardiac remodeling post-MI. Deficiency of myeloid IL4Rα signaling causes deteriorated cardiac function post-MI, due to dysregulated inflammation and insufficient fibrotic remodeling. This study sheds light on the potential of activating myeloid-specific IL4Rα signaling to modify remodeling post-MI. This brings hope to patients with MI and diminishes side effects by cell type-specific instead of whole body treatment.
Collapse
Affiliation(s)
- Jianrui Song
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan
| | - Ryan A Frieler
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Steven E Whitesall
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Yutein Chung
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Thomas M Vigil
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Lindsey A Muir
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, Michigan
| | - Jun Ma
- Department of Thoracic Surgery, Shanxi Province People's Hospital, Taiyuan, People's Republic of China
| | - Frank Brombacher
- International Center for Genetic Engineering and Biotechnology, University of Cape Town, Cape Town, South Africa
| | - Sascha N Goonewardena
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Carey N Lumeng
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, Michigan
| | - Daniel R Goldstein
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
- Institute of Gerontology, University of Michigan, Ann Arbor, Michigan
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan
| | - Richard M Mortensen
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
- Division of Metabolism, Endocrinology, and Diabetes, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
42
|
House A, Atalla I, Lee EJ, Guvendiren M. Designing Biomaterial Platforms for Cardiac Tissue and Disease Modeling. ADVANCED NANOBIOMED RESEARCH 2021; 1:2000022. [PMID: 33709087 PMCID: PMC7942203 DOI: 10.1002/anbr.202000022] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Heart disease is one of the leading causes of death in the world. There is a growing demand for in vitro cardiac models that can recapitulate the complex physiology of the cardiac tissue. These cardiac models can provide a platform to better understand the underlying mechanisms of cardiac development and disease and aid in developing novel treatment alternatives and platforms towards personalized medicine. In this review, a summary of engineered cardiac platforms is presented. Basic design considerations for replicating the heart's microenvironment are discussed considering the anatomy of the heart. This is followed by a detailed summary of the currently available biomaterial platforms for modeling the heart tissue in vitro. These in vitro models include 2D surface modified structures, 3D molded structures, porous scaffolds, electrospun scaffolds, bioprinted structures, and heart-on-a-chip devices. The challenges faced by current models and the future directions of in vitro cardiac models are also discussed. Engineered in vitro tissue models utilizing patients' own cells could potentially revolutionize the way we develop treatment and diagnostic alternatives.
Collapse
Affiliation(s)
- Andrew House
- Instructive Biomaterials and Additive Manufacturing Laboratory, Otto H. York Chemical and Materials Engineering, 138 York Center, University Heights, Newark, NJ 07102, USA
| | - Iren Atalla
- Instructive Biomaterials and Additive Manufacturing Laboratory, Otto H. York Chemical and Materials Engineering, 138 York Center, University Heights, Newark, NJ 07102, USA
| | - Eun Jung Lee
- Instructive Biomaterials and Additive Manufacturing Laboratory, Otto H. York Chemical and Materials Engineering, 138 York Center, University Heights, Newark, NJ 07102, USA
| | - Murat Guvendiren
- Instructive Biomaterials and Additive Manufacturing Laboratory, Otto H. York Chemical and Materials Engineering, 138 York Center, University Heights, Newark, NJ 07102, USA
| |
Collapse
|
43
|
Eosinophils improve cardiac function after myocardial infarction. Nat Commun 2020; 11:6396. [PMID: 33328477 PMCID: PMC7745020 DOI: 10.1038/s41467-020-19297-5] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 10/07/2020] [Indexed: 02/06/2023] Open
Abstract
Clinical studies reveal changes in blood eosinophil counts and eosinophil cationic proteins that may serve as risk factors for human coronary heart diseases. Here we report an increase of blood or heart eosinophil counts in humans and mice after myocardial infarction (MI), mostly in the infarct region. Genetic or inducible depletion of eosinophils exacerbates cardiac dysfunction, cell death, and fibrosis post-MI, with concurrent acute increase of heart and chronic increase of splenic neutrophils and monocytes. Mechanistic studies reveal roles of eosinophil IL4 and cationic protein mEar1 in blocking H2O2- and hypoxia-induced mouse and human cardiomyocyte death, TGF-β-induced cardiac fibroblast Smad2/3 activation, and TNF-α-induced neutrophil adhesion on the heart endothelial cell monolayer. In vitro-cultured eosinophils from WT mice or recombinant mEar1 protein, but not eosinophils from IL4-deficient mice, effectively correct exacerbated cardiac dysfunctions in eosinophil-deficient ∆dblGATA mice. This study establishes a cardioprotective role of eosinophils in post-MI hearts. Blood eosinophil (EOS) counts may serve as risk factors for human coronary heart diseases. Here the authors show that increased circulating and myocardial EOS after myocardial infarction play a cardioprotective role by reducing cardiomyocyte death, cardiac fibroblast activation and fibrosis, and endothelium activation-mediated inflammatory cell accumulation.
Collapse
|
44
|
McArthur S, Juban G, Gobbetti T, Desgeorges T, Theret M, Gondin J, Toller-Kawahisa JE, Reutelingsperger CP, Chazaud B, Perretti M, Mounier R. Annexin A1 drives macrophage skewing to accelerate muscle regeneration through AMPK activation. J Clin Invest 2020; 130:1156-1167. [PMID: 32015229 PMCID: PMC7269594 DOI: 10.1172/jci124635] [Citation(s) in RCA: 103] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 11/21/2019] [Indexed: 12/11/2022] Open
Abstract
Understanding the circuits that promote an efficient resolution of inflammation is crucial to deciphering the molecular and cellular processes required to promote tissue repair. Macrophages play a central role in the regulation of inflammation, resolution, and repair/regeneration. Using a model of skeletal muscle injury and repair, herein we identified annexin A1 (AnxA1) as the extracellular trigger of macrophage skewing toward a pro-reparative phenotype. Brought into the injured tissue initially by migrated neutrophils, and then overexpressed in infiltrating macrophages, AnxA1 activated FPR2/ALX receptors and the downstream AMPK signaling cascade, leading to macrophage skewing, dampening of inflammation, and regeneration of muscle fibers. Mice lacking AnxA1 in all cells or only in myeloid cells displayed a defect in this reparative process. In vitro experiments recapitulated these properties, with AMPK-null macrophages lacking AnxA1-mediated polarization. Collectively, these data identified the AnxA1/FPR2/AMPK axis as an important pathway in skeletal muscle injury regeneration.
Collapse
Affiliation(s)
- Simon McArthur
- Institute of Dentistry and.,William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Gaëtan Juban
- Université Claude Bernard Lyon 1, CNRS UMR-5310, INSERM U-1217, Institut NeuroMyoGène, Lyon, France
| | - Thomas Gobbetti
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Thibaut Desgeorges
- Université Claude Bernard Lyon 1, CNRS UMR-5310, INSERM U-1217, Institut NeuroMyoGène, Lyon, France
| | - Marine Theret
- Université Claude Bernard Lyon 1, CNRS UMR-5310, INSERM U-1217, Institut NeuroMyoGène, Lyon, France
| | - Julien Gondin
- Université Claude Bernard Lyon 1, CNRS UMR-5310, INSERM U-1217, Institut NeuroMyoGène, Lyon, France
| | - Juliana E Toller-Kawahisa
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom.,Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Chris P Reutelingsperger
- Department of Biochemistry and.,Cardiovascular Research Institute Maastricht, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, Netherlands
| | - Bénédicte Chazaud
- Université Claude Bernard Lyon 1, CNRS UMR-5310, INSERM U-1217, Institut NeuroMyoGène, Lyon, France
| | - Mauro Perretti
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom.,Centre for Inflammation and Therapeutic Innovation, Queen Mary University of London, London, United Kingdom
| | - Rémi Mounier
- Université Claude Bernard Lyon 1, CNRS UMR-5310, INSERM U-1217, Institut NeuroMyoGène, Lyon, France
| |
Collapse
|
45
|
Hitscherich P, Lee EJ. Crosstalk Between Cardiac Cells and Macrophages Postmyocardial Infarction: Insights from In Vitro Studies. TISSUE ENGINEERING PART B-REVIEWS 2020; 27:475-485. [PMID: 33096955 DOI: 10.1089/ten.teb.2020.0198] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Cardiovascular disease, including myocardial infarction (MI), is the leading cause of death in the western world. Following MI, a large number of cardiomyocytes are lost and inflammatory cells such as monocytes and macrophages migrate into the damaged region to remove dead cells and tissue. These inflammatory cells secrete growth factors to induce degradation of the extracellular matrix in the myocardium and recruit cardiac fibroblasts. However, the contribution of specific macrophage subsets on cardiac cell function and survival in the steady state as well as in the diseased state is not well known. There is an increasing demand for in vitro cardiac disease models to bridge the critical missing link in the existing experimental methods. In this review, studies using in vitro models to examine the interaction between macrophages and cardiac cells, including cardiomyocytes, endothelial cells, and fibroblasts, are summarized to better understand the complex inflammatory cascade post-MI. The current challenges and the future directions of in vitro cardiac models are also discussed. Detailed and more mechanistic insights into macrophages and cardiac cell interactions during the multiphase repair process could potentially revolutionize the development of treatments and diagnostic alternatives. Impact statement The inflammatory cascade postmyocardial infarction (MI) is very complex. In vitro cardiac disease model studies bridge the critical missing link in the existing experimental methods and provide insights, including multicellular interaction post-MI. Detailed and more mechanistic insights into macrophages and cardiac cell interactions during the multiphase repair process could potentially revolutionize in developing treatments and diagnostic alternatives.
Collapse
Affiliation(s)
- Pamela Hitscherich
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, New Jersey, USA
| | - Eun Jung Lee
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, New Jersey, USA
| |
Collapse
|
46
|
Tobin SW, Alibhai FJ, Weisel RD, Li RK. Considering Cause and Effect of Immune Cell Aging on Cardiac Repair after Myocardial Infarction. Cells 2020; 9:E1894. [PMID: 32823583 PMCID: PMC7465938 DOI: 10.3390/cells9081894] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 08/11/2020] [Accepted: 08/12/2020] [Indexed: 12/16/2022] Open
Abstract
The importance of the immune system for cardiac repair following myocardial infarction is undeniable; however, the complex nature of immune cell behavior has limited the ability to develop effective therapeutics. This limitation highlights the need for a better understanding of the function of each immune cell population during the inflammatory and resolution phases of cardiac repair. The development of reliable therapies is further complicated by aging, which is associated with a decline in cell and organ function and the onset of cardiovascular and immunological diseases. Aging of the immune system has important consequences on heart function as both chronic cardiac inflammation and an impaired immune response to cardiac injury are observed in older individuals. Several studies have suggested that rejuvenating the aged immune system may be a valid therapeutic candidate to prevent or treat heart disease. Here, we review the basic patterns of immune cell behavior after myocardial infarction and discuss the autonomous and nonautonomous manners of hematopoietic stem cell and immune cell aging. Lastly, we identify prospective therapies that may rejuvenate the aged immune system to improve heart function such as anti-inflammatory and senolytic therapies, bone marrow transplant, niche remodeling and regulation of immune cell differentiation.
Collapse
Affiliation(s)
- Stephanie W. Tobin
- Division of Cardiovascular Surgery, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5T 1P5, Canada; (S.W.T.); (F.J.A.); (R.D.W.)
| | - Faisal J. Alibhai
- Division of Cardiovascular Surgery, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5T 1P5, Canada; (S.W.T.); (F.J.A.); (R.D.W.)
| | - Richard D. Weisel
- Division of Cardiovascular Surgery, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5T 1P5, Canada; (S.W.T.); (F.J.A.); (R.D.W.)
- Division of Cardiac Surgery, Peter Munk Cardiac Centre, Toronto General Hospital and University of Toronto, Toronto, ON M5G 2N2, Canada
| | - Ren-Ke Li
- Division of Cardiovascular Surgery, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5T 1P5, Canada; (S.W.T.); (F.J.A.); (R.D.W.)
- Division of Cardiac Surgery, Peter Munk Cardiac Centre, Toronto General Hospital and University of Toronto, Toronto, ON M5G 2N2, Canada
| |
Collapse
|
47
|
Xia W, Zou C, Chen H, Xie C, Hou M. Immune checkpoint inhibitor induces cardiac injury through polarizing macrophages via modulating microRNA-34a/Kruppel-like factor 4 signaling. Cell Death Dis 2020; 11:575. [PMID: 32709878 PMCID: PMC7382486 DOI: 10.1038/s41419-020-02778-2] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 07/10/2020] [Accepted: 07/13/2020] [Indexed: 02/06/2023]
Abstract
Cancer immunotherapy has become a well-established treatment option for some cancers; however, its use is hampered by its cardiovascular adverse effects. Immune checkpoint inhibitors (ICIs)-related cardiac toxicity took place in kinds of different forms, such as myocarditis, acute coronary syndrome, and pericardial disease, with high mortality rates. This study aimed to investigate the roles of programmed death-1 (PD-1) inhibitor, one of widespread used ICIs, in the development of murine cardiac injury. PD-1 inhibitor is known to transduce immunoregulatory signals that modulate macrophages polarization to attack tumor cells. Hence, this study explored whether the cardiovascular adverse effects of PD-1 inhibitor were related to macrophage polarization. MicroRNA-34a (miR-34a), which appears to regulate the polarization of cultured macrophages to induce inflammation, is examined in cardiac injury and macrophage polarization induced by the PD-1 inhibitor. As a target of miR-34a, Krüppel-like factor 4 (KLF4) acted as an anti-inflammation effector to take cardiac protective effect. Further, it investigated whether modulating the miR-34a/KLF4-signaling pathway could influence macrophage polarization. The PD-1 inhibitor markedly induced M1 phenotype macrophage polarization with impaired cardiac function, whereas miR-34a inhibitor transfection treatment reversed M1 polarization and cardiac injury in vivo. In vitro, PD-1 inhibitor-induced M1 polarization was accompanied by an increase in the expression of miR-34a but a decrease in the expression of KLF4. TargetScan and luciferase assay showed that miR-34a targeted the KLF4 3′-untranslated region. Either miR-34a inhibition or KLF4 overexpression could abolish M1 polarization induced by the PD-1 inhibitor. The findings strongly suggested that the PD-1 inhibitor exerted its effect in promoting M1 polarization and cardiac injury by modulating the miR-34a/KLF4-signaling pathway and inducing myocardial inflammation. These findings might help us to understand the pathogenesis of cardiac injury during immunotherapy, and provide new targets in ameliorating cardiac injury in patients with cancer receiving PD-1 inhibitor treatment.
Collapse
Affiliation(s)
- Wenzheng Xia
- Department of Neurosurgery, First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China.,Department of Neurosurgery, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Changlin Zou
- Department of Radiation Oncology, First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Hanbin Chen
- Department of Radiation Oncology, First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Congying Xie
- Department of Radiation Oncology, First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China.
| | - Meng Hou
- Department of Radiation Oncology, First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
48
|
Zogbi C, Oliveira NC, Levy D, Bydlowski SP, Bassaneze V, Neri EA, Krieger JE. Beneficial effects of IL-4 and IL-6 on rat neonatal target cardiac cells. Sci Rep 2020; 10:12350. [PMID: 32704142 PMCID: PMC7378182 DOI: 10.1038/s41598-020-69413-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 06/23/2020] [Indexed: 02/06/2023] Open
Abstract
The nature of the early post-natal immune response in rodents appears to influence cardiac regeneration even though the underlying molecules remain poorly understood. Consistent with this idea, we show now significant changes in the expression of immune and cell movement gene pathways in heart samples from 1- and 7-day-old rats with ventricle resection. We then tested whether conditioned media from adult M2 anti-inflammatory macrophages target neonatal cardiac cells to a pro-regenerative like phenotype compared to the M1 pro-inflammatory macrophages. We found that M2 compared to M1 macrophage-conditioned media upregulates neonatal cardiomyocyte proliferation, suppresses myofibroblast-induced differentiation and stimulates endothelial cell tube formation. Using a cytokine array, we selected four candidate cytokine molecules uniquely expressed in M2 macrophage-conditioned media and showed that two of them (IL-4 and IL-6) induce endothelial cell proliferation whilst IL-4 promotes proliferation in neonatal cardiomyocytes and prevents myofibroblast-induced collagen type I secretion. Altogether, we provided evidence that adult M2 macrophage-conditioned media displays a paracrine beneficial pro-regenerative response in target cardiac cells and that IL-4 and IL-6 recapitulate, at least in part, these pleiotropic effects. Further characterization of macrophage immune phenotypes and their secreted molecules may give rise to novel therapeutic approaches for post-natal cardiac repair.
Collapse
Affiliation(s)
- Camila Zogbi
- Lab Genetics & Mol Cardiology/LIM 13, Heart Institute (InCor), University of São Paulo Medical School, Av Dr Eneas C Aguiar 44, Sao Paulo, SP, 05403-000, Brazil
| | - Nathalia C Oliveira
- Lab Genetics & Mol Cardiology/LIM 13, Heart Institute (InCor), University of São Paulo Medical School, Av Dr Eneas C Aguiar 44, Sao Paulo, SP, 05403-000, Brazil
| | - Débora Levy
- Lab Genetics & Mol Cardiology/LIM 13, Heart Institute (InCor), University of São Paulo Medical School, Av Dr Eneas C Aguiar 44, Sao Paulo, SP, 05403-000, Brazil
| | - Sergio P Bydlowski
- Lab Genetics & Mol Cardiology/LIM 13, Heart Institute (InCor), University of São Paulo Medical School, Av Dr Eneas C Aguiar 44, Sao Paulo, SP, 05403-000, Brazil
| | - Vinicius Bassaneze
- Lab Genetics & Mol Cardiology/LIM 13, Heart Institute (InCor), University of São Paulo Medical School, Av Dr Eneas C Aguiar 44, Sao Paulo, SP, 05403-000, Brazil
| | - Elida A Neri
- Lab Genetics & Mol Cardiology/LIM 13, Heart Institute (InCor), University of São Paulo Medical School, Av Dr Eneas C Aguiar 44, Sao Paulo, SP, 05403-000, Brazil
| | - Jose E Krieger
- Lab Genetics & Mol Cardiology/LIM 13, Heart Institute (InCor), University of São Paulo Medical School, Av Dr Eneas C Aguiar 44, Sao Paulo, SP, 05403-000, Brazil.
| |
Collapse
|
49
|
Dixon DL, Lawrence MD, Bihari S, De Pasquale CG, Griggs KM, Bersten AD. Systemic Markers of Monocyte Activation in Acute Pulmonary Oedema. Heart Lung Circ 2020; 30:404-413. [PMID: 32713768 DOI: 10.1016/j.hlc.2020.06.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 05/21/2020] [Accepted: 06/15/2020] [Indexed: 11/24/2022]
Abstract
BACKGROUND Hydrostatic lung injury followed by pulmonary remodelling variably complicates cardiogenic acute pulmonary oedema (APO). Pulmonary remodelling may be regulated by the balance between distinct phenotypes of pulmonary macrophages; activated/inflammatory (M1), and reparative/anti-inflammatory (M2), derived from circulating monocyte populations. The aim of this study was to identify biomarkers in peripheral blood that are consistent with hydrostatic lung injury and pulmonary remodelling in APO and which follow the variable clinical course. METHODS To examine peripheral markers of lung inflammation, resolution and remodelling, 18 patients, admitted to the intensive care unit (ICU) with a clinical diagnosis of APO, were enrolled. Admission, 12- and 24-hour post-admission bloods were assayed for cytokines by ELISA (R&D Systems, Minneapolis, MN, USA) and leukocyte surface markers by flow cytometry. RESULTS Admission PaO2 to FiO2 ratio was positively correlated with Mon 2 (intermediate) monocyte prevalence, through increasing ratio of CD16+ monocytes to CD11b+ and CD40+ monocytes, and negatively correlated with Mon 1 (classical) monocyte prevalence, through decreasing ratio of CD16+ monocytes to CD62L+. Secondary cohort analysis compared 10 APO patients with established chronic heart failure (CHF) to eight without CHF. An increase in monocyte chemotactic peptide (MCP)-1, monocyte prevalence, and CD16-CD62L+ monocytes with CHF, all characteristic of monocyte activation to a Mon 1 phenotype, were found in the CHF APO patients. CONCLUSIONS Increased systemic monocyte prevalence and expression of cell surface markers suggest a Mon 1 profile in CHF patients during episodes of APO. Future studies should define the role of systemic monocyte prevalence and activation in decompensated CHF.
Collapse
Affiliation(s)
- Dani-Louise Dixon
- Intensive and Critical Care Unit, Flinders Medical Centre, Adelaide, SA, Australia; Department of Critical Care Medicine, Flinders University, Adelaide, SA, Australia.
| | - Mark D Lawrence
- Department of Critical Care Medicine, Flinders University, Adelaide, SA, Australia
| | - Shailesh Bihari
- Intensive and Critical Care Unit, Flinders Medical Centre, Adelaide, SA, Australia; Department of Critical Care Medicine, Flinders University, Adelaide, SA, Australia
| | - Carmine G De Pasquale
- Cardiac Services, Flinders Medical Centre, Adelaide, SA, Australia; Department of Medicine, Flinders University, Adelaide, SA, Australia
| | - Kim M Griggs
- Department of Critical Care Medicine, Flinders University, Adelaide, SA, Australia
| | - Andrew D Bersten
- Intensive and Critical Care Unit, Flinders Medical Centre, Adelaide, SA, Australia; Department of Critical Care Medicine, Flinders University, Adelaide, SA, Australia
| |
Collapse
|
50
|
Hitscherich PG, Xie LH, Del Re D, Lee EJ. The effects of macrophages on cardiomyocyte calcium-handling function using in vitro culture models. Physiol Rep 2020; 7:e14137. [PMID: 31301118 PMCID: PMC6640591 DOI: 10.14814/phy2.14137] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 04/18/2019] [Accepted: 04/23/2019] [Indexed: 12/11/2022] Open
Abstract
Following myocardial infarction (MI), myocardial inflammation plays a crucial role in the pathogenesis of MI injury and macrophages are among the key cells activated during the initial phases of the host response regulating the healing process. While macrophages have emerged as attractive effectors in tissue injury and repair, the contribution of macrophages on cardiac cell function and survival is not fully understood due to complexity of the in vivo inflammatory microenvironment. Understanding the key cells involved and how they communicate with one another is of paramount importance for the development of effective clinical treatments. Here, novel in vitro myocardial inflammation models were developed to examine how both direct and indirect interactions with polarized macrophage subsets present in the post-MI microenvironment affect cardiomyocyte function. The indirect model using conditioned medium from polarized macrophage subsets allowed examination of the effects of macrophage-derived factors on stem cell-derived cardiomyocyte function for up to 3 days. The results from the indirect model demonstrated that pro-inflammatory macrophage-derived factors led to a significant downregulation of cardiac troponin T (cTnT) and sarcoplasmic/endoplasmic reticulum calcium ATPase (Serca2) gene expression. It also demonstrated that inhibition of macrophage-secreted matricellular protein, osteopontin (OPN), led to a significant decrease in cardiomyocyte store-operated calcium entry (SOCE). In the direct model, stem cell-derived cardiomyocytes were co-cultured with polarized macrophage subsets for up to 3 days. It was demonstrated that anti-inflammatory macrophages significantly increased cardiomyocyte Ca2+ fractional release while macrophages independent of their subtypes led to significant downregulation of SOCE response in cardiomyocytes. This study describes simplified and controlled in vitro myocardial inflammation models, which allow examination of potential beneficial and deleterious effects of macrophages on cardiomyocytes and vise versa. This can lead to our improved understanding of the inflammatory microenvironment post-MI, otherwise difficult to directly investigate in vivo or by using currently available in vitro models.
Collapse
Affiliation(s)
- Pamela G Hitscherich
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, New Jersey
| | - Lai-Hua Xie
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, New Jersey
| | - Dominic Del Re
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, New Jersey
| | - Eun Jung Lee
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, New Jersey
| |
Collapse
|