1
|
Zhang S, Zhao X, Lv Y, Niu J, Wei X, Luo Z, Wang X, Chen XL. Exosomes of different cellular origins: prospects and challenges in the treatment of acute lung injury after burns. J Mater Chem B 2024. [PMID: 39704476 DOI: 10.1039/d4tb02351j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2024]
Abstract
Acute lung injury (ALI) is a critical clinical disease caused by direct factors (inhalation injury, gastroesophageal reflux, etc.) or indirect factors (including infection, sepsis, burn, shock, trauma, acute pancreatitis, fat embolism, drug overdose, etc.). ALI is characterized mainly by diffuse interstitial and alveolar edema caused by an uncontrolled inflammatory response and damage to the alveoli-capillary barrier and has very high morbidity and mortality rates. Currently, there is no effective treatment strategy other than mechanical ventilation, fluid management or other supportive treatments. Exosomes are nanovesicle-like vesicles with double-membrane structures detached from the cell membrane or secreted by cells. These vesicles can be used as drug carriers because of their unique biological properties, such as anti-inflammatory, anti-apoptotic, pro-cell growth and immunomodulatory functions, and have been applied in the treatment of ALI in recent years. In this study, the mechanism and pathophysiological characteristics of ALI were first systematically described. The different cellular sources and characteristics of exosomes are summarized, and their functions and value as drug carriers in the treatment of ALI are discussed, as are the challenges that may be faced in the treatment of ALI with exosomes.
Collapse
Affiliation(s)
- Shuo Zhang
- Department of Burns, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China.
| | - Xinyu Zhao
- Department of Burns, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China.
| | - Yang Lv
- Plastic Surgery Department, The Second Affiliated Hospital of Anhui Medical University, 230061, P. R. China
| | - Jianguo Niu
- School of Biomedical Engineering, Anhui Medical University, Hefei 230022, China.
| | - Xiaolong Wei
- School of Biomedical Engineering, Anhui Medical University, Hefei 230022, China.
| | - Zhiwen Luo
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai 200040, P. R. China.
| | - Xianwen Wang
- School of Biomedical Engineering, Anhui Medical University, Hefei 230022, China.
| | - Xu-Lin Chen
- Department of Burns, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China.
| |
Collapse
|
2
|
Zolfaghari Dehkharghani M, Mousavi S, Kianifard N, Fazlzadeh A, Parsa H, Tavakoli Pirzaman A, Fazlollahpour-Naghibi A. Importance of long non-coding RNAs in the pathogenesis, diagnosis, and treatment of myocardial infarction. IJC HEART & VASCULATURE 2024; 55:101529. [PMID: 39498345 PMCID: PMC11532444 DOI: 10.1016/j.ijcha.2024.101529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 10/02/2024] [Accepted: 10/07/2024] [Indexed: 11/07/2024]
Abstract
Myocardial infarction (MI), a major global cause of mortality and morbidity, continues to pose a significant burden on public health. Despite advances in understanding its pathogenesis, there remains a need to elucidate the intricate molecular mechanisms underlying MI progression. Long non-coding RNAs (lncRNAs) have emerged as key regulators in diverse biological processes, yet their specific roles in MI pathophysiology remain elusive. Conducting a thorough review of literature using PubMed and Google Scholar databases, we investigated the involvement of lncRNAs in MI, focusing on their regulatory functions and downstream signaling pathways. Our analysis revealed extensive dysregulation of lncRNAs in MI, impacting various biological processes through diverse mechanisms. Notably, lncRNAs act as crucial modulators of gene expression and signaling cascades, functioning as decoys, regulators, and scaffolds. Furthermore, studies identified the multifaceted roles of lncRNAs in modulating inflammation, apoptosis, autophagy, necrosis, fibrosis, remodeling, and ischemia-reperfusion injury during MI progression. Recent research highlights the pivotal contribution of lncRNAs to MI pathogenesis, offering novel insights into potential therapeutic interventions. Moreover, the identification of circulating lncRNA signatures holds promise for the development of non-invasive diagnostic biomarkers. In summary, findings underscore the significance of lncRNAs in MI pathophysiology, emphasizing their potential as therapeutic targets and diagnostic tools for improved patient management and outcomes.
Collapse
Affiliation(s)
| | - Safa Mousavi
- School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nazanin Kianifard
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amin Fazlzadeh
- School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamid Parsa
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | | |
Collapse
|
3
|
Dabrowska S, Turano E, Scambi I, Virla F, Nodari A, Pezzini F, Galiè M, Bonetti B, Mariotti R. A Cellular Model of Amyotrophic Lateral Sclerosis to Study the Therapeutic Effects of Extracellular Vesicles from Adipose Mesenchymal Stem Cells on Microglial Activation. Int J Mol Sci 2024; 25:5707. [PMID: 38891895 PMCID: PMC11171908 DOI: 10.3390/ijms25115707] [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: 03/19/2024] [Revised: 05/16/2024] [Accepted: 05/17/2024] [Indexed: 06/21/2024] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease characterized by the progressive degeneration of upper and lower motor neurons (MNs) in the brain and spinal cord, leading to progressive paralysis and death. Increasing evidence indicates that neuroinflammation plays an important role in ALS's pathogenesis and disease progression. Neuroinflammatory responses, primarily driven by activated microglia and astrocytes, and followed by infiltrating peripheral immune cells, contribute to exacerbate/accelerate MN death. In particular, the role of the microglia in ALS remains unclear, partly due to the lack of experimental models that can fully recapitulate the complexity of ALS's pathology. In this study, we developed and characterized a microglial cell line, SIM-A9-expressing human mutant protein Cu+/Zn+ superoxide dismutase_1 (SIM-A9hSOD1(G93A)), as a suitable model in vitro mimicking the microglia activity in ALS. The expression of hSOD1(G93A) in SIM-A9 cells induced a change in their metabolic activity, causing polarization into a pro-inflammatory phenotype and enhancing reactive oxygen species production, which is known to activate cell death processes and apoptosis. Afterward, we used our microglial model as an experimental set-up to investigate the therapeutic action of extracellular vesicles isolated from adipose mesenchymal stem cells (ASC-EVs). ASC-EVs represent a promising therapeutic treatment for ALS due to their neuroprotective and immunomodulatory properties. Here, we demonstrated that treatment with ASC-EVs is able to modulate activated ALS microglia, reducing their metabolic activity and polarizing their phenotype toward an anti-inflammatory one through a mechanism of reduction of reactive oxygen species.
Collapse
Affiliation(s)
- Sylwia Dabrowska
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Strada Le Grazie 8, 37134 Verona, Italy; (S.D.); (E.T.); (I.S.); (F.V.); (A.N.); (M.G.)
- NeuroRepair Department, Mossakowski Medical Research Institute, Polish Academy of Sciences, Pawinskiego Street 5, 02-106 Warsaw, Poland
| | - Ermanna Turano
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Strada Le Grazie 8, 37134 Verona, Italy; (S.D.); (E.T.); (I.S.); (F.V.); (A.N.); (M.G.)
| | - Ilaria Scambi
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Strada Le Grazie 8, 37134 Verona, Italy; (S.D.); (E.T.); (I.S.); (F.V.); (A.N.); (M.G.)
| | - Federica Virla
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Strada Le Grazie 8, 37134 Verona, Italy; (S.D.); (E.T.); (I.S.); (F.V.); (A.N.); (M.G.)
| | - Alice Nodari
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Strada Le Grazie 8, 37134 Verona, Italy; (S.D.); (E.T.); (I.S.); (F.V.); (A.N.); (M.G.)
| | - Francesco Pezzini
- Department of Surgery, Dentistry, Paediatrics and Gynaecology (Child Neurology and Psychiatry), University of Verona, 37134 Verona, Italy;
| | - Mirco Galiè
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Strada Le Grazie 8, 37134 Verona, Italy; (S.D.); (E.T.); (I.S.); (F.V.); (A.N.); (M.G.)
| | - Bruno Bonetti
- Neurology Unit, Azienda Ospedaliera Universitaria Integrata, 37126 Verona, Italy;
| | - Raffaella Mariotti
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Strada Le Grazie 8, 37134 Verona, Italy; (S.D.); (E.T.); (I.S.); (F.V.); (A.N.); (M.G.)
| |
Collapse
|
4
|
Caño-Carrillo S, Castillo-Casas JM, Franco D, Lozano-Velasco E. Unraveling the Signaling Dynamics of Small Extracellular Vesicles in Cardiac Diseases. Cells 2024; 13:265. [PMID: 38334657 PMCID: PMC10854837 DOI: 10.3390/cells13030265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 01/28/2024] [Accepted: 01/30/2024] [Indexed: 02/10/2024] Open
Abstract
Effective intercellular communication is essential for cellular and tissue balance maintenance and response to challenges. Cellular communication methods involve direct cell contact or the release of biological molecules to cover short and long distances. However, a recent discovery in this communication network is the involvement of extracellular vesicles that host biological contents such as proteins, nucleic acids, and lipids, influencing neighboring cells. These extracellular vesicles are found in body fluids; thus, they are considered as potential disease biomarkers. Cardiovascular diseases are significant contributors to global morbidity and mortality, encompassing conditions such as ischemic heart disease, cardiomyopathies, electrical heart diseases, and heart failure. Recent studies reveal the release of extracellular vesicles by cardiovascular cells, influencing normal cardiac function and structure. However, under pathological conditions, extracellular vesicles composition changes, contributing to the development of cardiovascular diseases. Investigating the loading of molecular cargo in these extracellular vesicles is essential for understanding their role in disease development. This review consolidates the latest insights into the role of extracellular vesicles in diagnosis and prognosis of cardiovascular diseases, exploring the potential applications of extracellular vesicles in personalized therapies, shedding light on the evolving landscape of cardiovascular medicine.
Collapse
Affiliation(s)
| | | | | | - Estefanía Lozano-Velasco
- Cardiovascular Development Group, Department of Experimental Biology, University of Jaén, 23071 Jaén, Spain; (S.C.-C.); (J.M.C.-C.); (D.F.)
| |
Collapse
|
5
|
Yin X, Lin L, Fang F, Zhang B, Shen C. Mechanisms and Optimization Strategies of Paracrine Exosomes from Mesenchymal Stem Cells in Ischemic Heart Disease. Stem Cells Int 2023; 2023:6500831. [PMID: 38034060 PMCID: PMC10686715 DOI: 10.1155/2023/6500831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 10/11/2023] [Accepted: 10/25/2023] [Indexed: 12/02/2023] Open
Abstract
The morbidity and mortality of myocardial infarction (MI) are increasing worldwide. Mesenchymal stem cells (MSCs) are multipotent stem cells with self-renewal and differentiation capabilities that are essential in tissue healing and regenerative medicine. However, the low implantation and survival rates of transplanted cells hinder the widespread clinical use of stem cells. Exosomes are naturally occurring nanovesicles that are secreted by cells and promote the repair of cardiac function by transporting noncoding RNA and protein. In recent years, MSC-derived exosomes have been promising cell-free treatment tools for improving cardiac function and reversing cardiac remodeling. This review describes the biological properties and therapeutic potential of exosomes and summarizes some engineering approaches for exosomes optimization to enhance the targeting and therapeutic efficacy of exosomes in MI.
Collapse
Affiliation(s)
- Xiaorong Yin
- Department of Clinical Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, China
| | - Lizhi Lin
- Department of Clinical Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, China
| | - Fang Fang
- Department of Cardiology, Jining Key Laboratory for Diagnosis and Treatment of Cardiovascular Diseases, Affiliated Hospital of Jining Medical University, Jining, Shandong, China
| | - Bin Zhang
- Department of Laboratory Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, China
| | - Cheng Shen
- Department of Cardiology, Jining Key Laboratory for Diagnosis and Treatment of Cardiovascular Diseases, Affiliated Hospital of Jining Medical University, Jining, Shandong, China
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| |
Collapse
|
6
|
Fan M, Liang T, Xie F, Ma P, Li J. Exosomal circ_HIPK3 reduces apoptosis in H2O2-induced AC16 cardiomyocytes through miR-33a-5p/IRS1 axis. Transpl Immunol 2023; 80:101862. [PMID: 37230396 DOI: 10.1016/j.trim.2023.101862] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 04/06/2023] [Accepted: 05/20/2023] [Indexed: 05/27/2023]
Abstract
BACKGROUND Exosomal circular RNAs (circRNAs) has been revealed to participate in the processes of cellular angiogenesis, growth and metastasis. Herein, the goal of this work was to investigate the role of exosomal circ_HIPK3 in cardiomyocyte apoptosis. METHODS Exosomes were isolated using ultracentrifugation method and observed by transmission electron microscopy (TEM). Western blot was used to detect exosomes markers. The experimental group AC16 cells were exposed to hydrogen peroxide (H2O2). Levels of genes and proteins was detected by qRT-PCR and Western blot. EdU assay, CCK8 assay, flow cytometry, and Western blot were utilized to detect the function of exosomal circ_HIPK3 in proliferation, and apoptosis. The target relationship between miR-33a-5p and circ_HIPK3 or IRS1 (insulin receptor substrate 1). RESULTS Circ_HIPK3 was packaged into exosomes and derived from AC16 cells. The expression of circ_HIPK3 was decreased by H2O2 treatment in AC16 cells, which also led to the decrease of circ_HIPK3 in exosomes. Functional analysis showed exosomal circ_HIPK3 promoted AC16 cell proliferation and reduced cell apoptosis under H2O2 treatment. Mechanistically, circ_HIPK3 acted as a sponge of miR-33a-5p to up-regulate the expression of its target IRS1. Functionally, forced expression of miR-33a-5p reversed the reduction of exosomal circ_HIPK3 in apoptosis of H2O2-stimulated AC16 cells. Moreover, miR-33a-5p inhibition contributed to the proliferation of H2O2-stimulated AC16 cells, which was abolished by IRS1 silencing. CONCLUSION Exosomal circ_HIPK3 reduced H2O2-induced AC16 cardiomyocyte apoptosis through miR-33a-5p/IRS1 axis, suggesting a novel insight into the pathology of myocardial infarction.
Collapse
Affiliation(s)
- Ming Fan
- Department of Cardiovascular Surgery, 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Ting Liang
- Department of Ophthalmology, 4th Affiliated Hospital of Harbin Medical University, Harbin City, Heilongjiang Province, China
| | - Fei Xie
- Department of Cardiovascular Surgery, 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Pingwei Ma
- Department of Cardiovascular Surgery, 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Junquan Li
- Department of Cardiovascular Surgery, 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China.
| |
Collapse
|
7
|
Liang W, Sagar S, Ravindran R, Najor RH, Quiles JM, Chi L, Diao RY, Woodall BP, Leon LJ, Zumaya E, Duran J, Cauvi DM, De Maio A, Adler ED, Gustafsson ÅB. Mitochondria are secreted in extracellular vesicles when lysosomal function is impaired. Nat Commun 2023; 14:5031. [PMID: 37596294 PMCID: PMC10439183 DOI: 10.1038/s41467-023-40680-5] [Citation(s) in RCA: 56] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 08/04/2023] [Indexed: 08/20/2023] Open
Abstract
Mitochondrial quality control is critical for cardiac homeostasis as these organelles are responsible for generating most of the energy needed to sustain contraction. Dysfunctional mitochondria are normally degraded via intracellular degradation pathways that converge on the lysosome. Here, we identified an alternative mechanism to eliminate mitochondria when lysosomal function is compromised. We show that lysosomal inhibition leads to increased secretion of mitochondria in large extracellular vesicles (EVs). The EVs are produced in multivesicular bodies, and their release is independent of autophagy. Deletion of the small GTPase Rab7 in cells or adult mouse heart leads to increased secretion of EVs containing ubiquitinated cargos, including intact mitochondria. The secreted EVs are captured by macrophages without activating inflammation. Hearts from aged mice or Danon disease patients have increased levels of secreted EVs containing mitochondria indicating activation of vesicular release during cardiac pathophysiology. Overall, these findings establish that mitochondria are eliminated in large EVs through the endosomal pathway when lysosomal degradation is inhibited.
Collapse
Affiliation(s)
- Wenjing Liang
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Shakti Sagar
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Rishith Ravindran
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Rita H Najor
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Justin M Quiles
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Liguo Chi
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Rachel Y Diao
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Benjamin P Woodall
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Leonardo J Leon
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Erika Zumaya
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Jason Duran
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - David M Cauvi
- Department of Surgery, University of California San Diego, La Jolla, CA, USA
| | - Antonio De Maio
- Department of Surgery, University of California San Diego, La Jolla, CA, USA
| | - Eric D Adler
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Åsa B Gustafsson
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA.
- Department of Pharmacology, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
8
|
Ong-Meang V, Blanzat M, Savchenko L, Perquis L, Guardia M, Pizzinat N, Poinsot V. Extracellular Vesicles Produced by the Cardiac Microenvironment Carry Functional Enzymes to Produce Lipid Mediators In Situ. Int J Mol Sci 2023; 24:ijms24065866. [PMID: 36982939 PMCID: PMC10056942 DOI: 10.3390/ijms24065866] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/27/2023] [Accepted: 03/15/2023] [Indexed: 03/22/2023] Open
Abstract
The impact of the polyunsaturated fatty acids (PUFAs) at physiological concentrations on the composition of eicosanoids transported within the extracellular vesicles (EVs) of rat bone marrow mesenchymal stem cells and cardiomyoblasts was reported by our group in 2020. The aim of this article was to extend this observation to cells from the cardiac microenvironment involved in the processes of inflammation, namely mouse J774 macrophages and rat heart mesenchymal stem cells cMSCs. Moreover, to enhance our capacity to understand the paracrine exchange between these orchestrators of cardiac inflammation, we investigated some machinery involved in the eicosanoid’s synthesis transported by the EVs produced by these cells (including the two formerly described cells: bone marrow mesenchymal stem cells BM-MSC and cardiomyoblasts H9c2). We analyzed the oxylipin and the enzymatic content of the EVs collected from cell cultures supplemented (or not) with PUFAs. We prove that large eicosanoid profiles are exported in the EVs by the cardiac microenvironment cells, but also that these EVs carry some critical and functional biosynthetic enzymes, allowing them to synthesize inflammation bioactive compounds by sensing their environment. Moreover, we demonstrate that these are functional. This observation reinforces the hypothesis that EVs are key factors in paracrine signaling, even in the absence of the parent cell. We also reveal a macrophage-specific behavior, as we observed a radical change in the lipid mediator profile when small EVs derived from J774 cells were exposed to PUFAs. To summarize, we prove that the EVs, due to the carried functional enzymes, can alone produce bioactive compounds, in the absence of the parent cell, by sensing their environment. This makes them potential circulating monitoring entities.
Collapse
Affiliation(s)
- Varravaddheay Ong-Meang
- Inserm, CNRS, Institut des Maladies Métaboliques et Cardiovasculaires U1964, Université Toulouse III—Paul Sabatier, BP 84225, CEDEX 4, F-31432 Toulouse, France
| | - Muriel Blanzat
- CNRS, Laboratoire IMRCP UMR 5623, Université Toulouse III—Paul Sabatier, CEDEX 9, F-31062 Toulouse, France
| | - Lesia Savchenko
- Inserm, CNRS, Institut des Maladies Métaboliques et Cardiovasculaires U1964, Université Toulouse III—Paul Sabatier, BP 84225, CEDEX 4, F-31432 Toulouse, France
| | - Lucie Perquis
- CNRS, Laboratoire IMRCP UMR 5623, Université Toulouse III—Paul Sabatier, CEDEX 9, F-31062 Toulouse, France
| | - Mégane Guardia
- Inserm, CNRS, Institut des Maladies Métaboliques et Cardiovasculaires U1964, Université Toulouse III—Paul Sabatier, BP 84225, CEDEX 4, F-31432 Toulouse, France
| | - Nathalie Pizzinat
- Inserm, CNRS, Institut des Maladies Métaboliques et Cardiovasculaires U1964, Université Toulouse III—Paul Sabatier, BP 84225, CEDEX 4, F-31432 Toulouse, France
| | - Verena Poinsot
- Inserm, CNRS, Institut des Maladies Métaboliques et Cardiovasculaires U1964, Université Toulouse III—Paul Sabatier, BP 84225, CEDEX 4, F-31432 Toulouse, France
- Correspondence:
| |
Collapse
|
9
|
Liang W, Diao RY, Quiles JM, Najor RH, Chi L, Woodall BP, Leon LJ, Duran J, Cauvi DM, De Maio A, Adler ED, Gustafsson ÃSB. The Small GTPase Rab7 Regulates Release of Mitochondria in Extracellular Vesicles in Response to Lysosomal Dysfunction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.11.528148. [PMID: 36824711 PMCID: PMC9949095 DOI: 10.1101/2023.02.11.528148] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
Mitochondrial quality control is critical for cardiac homeostasis as these organelles are responsible for generating most of the energy needed to sustain contraction. Dysfunctional mitochondria are normally degraded via intracellular degradation pathways that converge on the lysosome. Here, we identified an alternative mechanism to eliminate mitochondria when lysosomal function is compromised. We show that lysosomal inhibition leads to increased secretion of mitochondria in large extracellular vesicles (EVs). The EVs are produced in multivesicular bodies, and their release is independent of autophagy. Deletion of the small GTPase Rab7 in cells or adult mouse heart leads to increased secretion of EVs containing ubiquitinated cargos, including intact mitochondria. The secreted EVs are captured by macrophages without activating inflammation. Hearts from aged mice or Danon disease patients have increased levels of secreted EVs containing mitochondria indicating activation of vesicular release during cardiac pathophysiology. Overall, these findings establish that mitochondria are eliminated in large EVs through the endosomal pathway when lysosomal degradation is inhibited.
Collapse
|
10
|
Cheng P, Wang X, Liu Q, Yang T, Qu H, Zhou H. Extracellular vesicles mediate biological information delivery: A double-edged sword in cardiac remodeling after myocardial infarction. Front Pharmacol 2023; 14:1067992. [PMID: 36909157 PMCID: PMC9992194 DOI: 10.3389/fphar.2023.1067992] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 02/13/2023] [Indexed: 02/24/2023] Open
Abstract
Acute myocardial infarction (AMI) is a severe ischemic disease with high morbidity and mortality worldwide. Maladaptive cardiac remodeling is a series of abnormalities in cardiac structure and function that occurs following myocardial infarction (MI). The pathophysiology of this process can be separated into two distinct phases: the initial inflammatory response, and the subsequent longer-term scar revision that includes the regression of inflammation, neovascularization, and fibrotic scar formation. Extracellular vesicles are nano-sized lipid bilayer vesicles released into the extracellular environment by eukaryotic cells, containing bioinformatic transmitters which are essential mediators of intercellular communication. EVs of different cellular origins play an essential role in cardiac remodeling after myocardial infarction. In this review, we first introduce the pathophysiology of post-infarction cardiac remodeling, as well as the biogenesis, classification, delivery, and functions of EVs. Then, we explore the dual role of these small molecule transmitters delivered by EVs in post-infarction cardiac remodeling, including the double-edged sword of pro-and anti-inflammation, and pro-and anti-fibrosis, which is significant for post-infarction cardiac repair. Finally, we discuss the pharmacological and engineered targeting of EVs for promoting heart repair after MI, thus revealing the potential value of targeted modulation of EVs and its use as a drug delivery vehicle in the therapeutic process of post-infarction cardiac remodeling.
Collapse
Affiliation(s)
- Peipei Cheng
- Institute of Cardiovascular Disease of Integrated Traditional Chinese and Western Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Branch of National Clinical Research Center for Chinese Medicine Cardiology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xinting Wang
- Institute of Cardiovascular Disease of Integrated Traditional Chinese and Western Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Branch of National Clinical Research Center for Chinese Medicine Cardiology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qian Liu
- Institute of Cardiovascular Disease of Integrated Traditional Chinese and Western Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Branch of National Clinical Research Center for Chinese Medicine Cardiology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Tao Yang
- Institute of Cardiovascular Disease of Integrated Traditional Chinese and Western Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Branch of National Clinical Research Center for Chinese Medicine Cardiology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Huiyan Qu
- Branch of National Clinical Research Center for Chinese Medicine Cardiology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Department of Cardiovascular Disease, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hua Zhou
- Institute of Cardiovascular Disease of Integrated Traditional Chinese and Western Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Branch of National Clinical Research Center for Chinese Medicine Cardiology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Department of Cardiovascular Disease, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
11
|
Ghovvati M, Kharaziha M, Ardehali R, Annabi N. Recent Advances in Designing Electroconductive Biomaterials for Cardiac Tissue Engineering. Adv Healthc Mater 2022; 11:e2200055. [PMID: 35368150 PMCID: PMC9262872 DOI: 10.1002/adhm.202200055] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 03/12/2022] [Indexed: 12/19/2022]
Abstract
Implantable cardiac patches and injectable hydrogels are among the most promising therapies for cardiac tissue regeneration following myocardial infarction. Incorporating electrical conductivity into these patches and hydrogels is found to be an efficient method to improve cardiac tissue function. Conductive nanomaterials such as carbon nanotube, graphene oxide, gold nanorod, as well as conductive polymers such as polyaniline, polypyrrole, and poly(3,4-ethylenedioxythiophene):polystyrene sulfonate are appealing because they possess the electroconductive properties of semiconductors with ease of processing and have potential to restore electrical signaling propagation through the infarct area. Numerous studies have utilized these materials for regeneration of biological tissues that possess electrical activities, such as cardiac tissue. In this review, recent studies on the use of electroconductive materials for cardiac tissue engineering and their fabrication methods are summarized. Moreover, recent advances in developing electroconductive materials for delivering therapeutic agents as one of emerging approaches for treating heart diseases and regenerating damaged cardiac tissues are highlighted.
Collapse
Affiliation(s)
- Mahsa Ghovvati
- Department of Chemical and Biomolecular Engineering, University of California - Los Angeles, Los Angeles, CA 90095, USA
| | - Mahshid Kharaziha
- Biomaterials Research Group, Department of Materials Engineering, Isfahan University of Technology, Isfahan 84156-83111, Iran
| | - Reza Ardehali
- Division of Cardiology, Department of Internal Medicine, David Geffen School of Medicine, University of California – Los Angeles, Los Angeles, CA 90095, USA
| | - Nasim Annabi
- Department of Chemical and Biomolecular Engineering, University of California - Los Angeles, Los Angeles, CA 90095, USA
- Department of Bioengineering, University of California - Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
12
|
Lan Z, Wang T, Zhang L, Jiang Z, Zou X. CircSLC8A1 Exacerbates Hypoxia-Induced Myocardial Injury via Interacting with MiR-214-5p to Upregulate TEAD1 Expression. Int Heart J 2022; 63:591-601. [PMID: 35650159 DOI: 10.1536/ihj.21-547] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Circular RNAs (circRNAs) act as important regulators in myocardial infarction (MI). This study aimed to explore the regulatory mechanism of circRNA solute carrier family 8 member A1 antisense RNA 1 (circSLC8A1) in hypoxia-induced myocardial injury.Exosomes were isolated by ultracentrifugation and identified by microscopic observation or protein detection. Protein levels were examined by Western blot. CircSLC8A1, microRNA-214-5p (miR-214-5p), and TEA domain transcription factor 1 (TEAD1) levels were determined via quantitative real-time polymerase chain reaction (qRT-PCR). Cell viability and apoptosis were analyzed by 3-(4,5-dimethylthiazol-2-y1)-2,5-diphenyl tetrazolium bromide (MTT) and flow cytometry, respectively. Inflammatory cytokines were measured using enzyme-linked immunosorbent assay (ELISA). Oxidative stress was assessed by reactive oxygen species (ROS) production, malondialdehyde (MDA) level, and superoxide dismutase (SOD) activity through the corresponding detection kits. Target analysis was performed by dual-luciferase reporter assay, RNA immunoprecipitation (RIP) assay, and pull-down assay.Exosomes released circSLC8A1 from hypoxic cardiomyocytes. Exosomal circSLC8A1 exacerbated hypoxia-induced repression of cell viability but promotion of cell apoptosis, inflammation, and oxidative stress. Knockdown of circSLC8A1 ameliorated hypoxia-mediated cell injury. CircSLC8A1 directly targeted miR-214-5p and miR-214-5p downregulation reverted the effects of si-circSLC8A1 on hypoxia-treated cardiomyocytes. TEAD1 was a target of miR-214-5p and circSLC8A1 upregulated TEAD1 level via targeting miR-214-5p. In addition, miR-214-5p inhibited hypoxia-caused cell injury by downregulating the expression of TEAD1.These results suggested that circSLC8A1 aggravated cell damages in hypoxia-treated cardiomyocytes by the regulation of TEAD1 via sponging miR-214-5p.
Collapse
Affiliation(s)
- Zhong Lan
- Department of Internal Medicine-Cardiovascular, The Fifth Affiliated Hospital of Southern Medical University
| | - Tao Wang
- Department of Cardiac Surgery, The Fifth Affiliated Hospital of Southern Medical University
| | - Lihong Zhang
- Department of Internal Medicine-Cardiovascular, The Fifth Affiliated Hospital of Southern Medical University
| | - Zhizhong Jiang
- Department of Internal Medicine-Cardiovascular, The Fifth Affiliated Hospital of Southern Medical University
| | - Xiaoming Zou
- Department of Cardiac Surgery, The Fifth Affiliated Hospital of Southern Medical University
| |
Collapse
|
13
|
Han YC, Xie HZ, Lu B, Xiang RL, Li JY, Qian H, Zhang SY. Effect of berberine on global modulation of lncRNAs and mRNAs expression profiles in patients with stable coronary heart disease. BMC Genomics 2022; 23:400. [PMID: 35619068 PMCID: PMC9134690 DOI: 10.1186/s12864-022-08641-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 05/16/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Berberine (BBR) is an isoquinoline alkaloid found in the Berberis species. It was found to have protected effects in cardiovascular diseases. Here, we investigated the effect the regulatory function of long noncoding RNAs (lncRNAs) during the treatment of stable coronary heart disease (CHD) using BBR. We performed microarray analyses to identify differentially expressed (DE) lncRNAs and mRNAs between whole blood samples from 5 patients with stable CHD taking BBR and 5 no BBR volunteers. DE lncRNAs and mRNAs were validated by quantitative real-time PCR. RESULTS A total of 1703 DE lncRNAs and 912 DE mRNAs were identified. Kyoto Encyclopedia of Genes and Genomes pathway analysis indicated DE mRNAs might be associated with mammalian target of rapamycin and mitogen-activated protein kinase pathway. These pathways may be involved in the healing process after CHD. To study the relationship between mRNAs encoding transcription factors (DNA damage inducible transcript 3, sal-like protein 4 and estrogen receptor alpha gene) and CHD related de mRNAs, we performed protein and protein interaction analysis on their corresponding proteins. AKT and apoptosis pathway were significant enriched in protein and protein interaction network. BBR may affect downstream apoptosis pathways through DNA damage inducible transcript 3, sal-like protein 4 and estrogen receptor alpha gene. Growth arrest-specific transcript 5 might regulate CHD-related mRNAs through competing endogenous RNA mechanism and may be the downstream target gene regulated by BBR. Verified by the quantitative real-time PCR, we identified 8 DE lncRNAs that may relate to CHD. We performed coding and non-coding co-expression and competing endogenous RNA mechanism analysis of these 8 DE lncRNAs and CHD-related DE mRNA, and predicted their subcellular localization and N6-methyladenosine modification sites. CONCLUSION Our research found that BBR may affect mammalian target of rapamycin, mitogen-activated protein kinase, apoptosis pathway and growth arrest-specific transcript 5 in the process of CHD. These pathways may be involved in the healing process after CHD. Our research might provide novel insights for functional research of BBR.
Collapse
Affiliation(s)
- Ye-Chen Han
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Shuaifuyuan, Dongcheng District, Beijing, 100730, China
| | - Hong-Zhi Xie
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Shuaifuyuan, Dongcheng District, Beijing, 100730, China
| | - Bo Lu
- Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Ruo-Lan Xiang
- Department of Physiology and Pathophysiology, Peking University School of Basic Medical Sciences, Beijing, 100191, China
| | - Jing-Yi Li
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Shuaifuyuan, Dongcheng District, Beijing, 100730, China
| | - Hao Qian
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Shuaifuyuan, Dongcheng District, Beijing, 100730, China
| | - Shu-Yang Zhang
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Shuaifuyuan, Dongcheng District, Beijing, 100730, China.
| |
Collapse
|
14
|
The Mechanisms Underlying the Beneficial Effects of Stem Cell-Derived Exosomes in Repairing Ischemic Tissue Injury. J Cardiovasc Transl Res 2022; 15:524-534. [PMID: 35484464 DOI: 10.1007/s12265-022-10263-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 04/20/2022] [Indexed: 12/12/2022]
Abstract
Ischemic diseases are life-threatening, and the incidence increases as people's lifestyles change. Medications and surgical intervention offer limited benefit, and stem cell therapy has emerged as a potential approach for treating ischemic diseases. The exosomes secreted by stem cells have attracted more attention because they do not trigger the immune response and can be used as drug carriers. The non-coding RNA (ncRNA) carried by exosomes plays a key role in mediating exosome's beneficial effect, which can be further enhanced when combined with nanomaterials to improve its retention time. Here, we review the downstream target molecules and signal pathways of ncRNA and summarize recent advances of some nanomaterials used to encapsulate exosomes and promote ischemic tissue repair. We highlight the imprinting of exosomes from parent cells and discuss how the inflammasome pathway may be targeted for the development of novel therapy for ischemic diseases.
Collapse
|
15
|
Zheng YL, Wang WD, Cai PY, Zheng F, Zhou YF, Li MM, Du JR, Lin S, Lin HL. Stem cell-derived exosomes in the treatment of acute myocardial infarction in preclinical animal models: a meta-analysis of randomized controlled trials. Stem Cell Res Ther 2022; 13:151. [PMID: 35395872 PMCID: PMC8994329 DOI: 10.1186/s13287-022-02833-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 03/28/2022] [Indexed: 11/10/2022] Open
Abstract
Background Exosomes (EXOs) derived from stem cells have become a potential new treatment for acute myocardial infarction (AMI). However, their impact is still not fully understood. Therefore, we performed this meta-analysis to systematically review the efficacy of EXOs on AMI in preclinical animal models. Methods We searched PubMed, EMBASE, and the Web of Science from September 1, 1980 to September 1, 2021, to retrieve the studies reporting the therapeutic effects of EXOs on AMI animal models. Secondary endpoints include the fractional shortening (FS), infarct size (IS), fibrosis area (FA), the TNF-α, IL-6 and IL-10 levels, the apoptosis rate and the number of autophagic vesicles. Two authors independently screened the articles based on inclusion and exclusion criteria. All statistical analyses were conducted using Stata14.0. Results Ten studies satisfied the inclusion criteria. Pooled analyses demonstrated that the levels of LVEF (WMD = 3.67%; 95% CI 2.28–5.07%; P = 0.000), FS (WMD = 3.69%; 95% CI 2.06–5.33%; P = 0.000), IS (WMD = −4.52%, 95% CI − 7.14 to − 1.9%; P = 0.001), and FA (WMD = −7.04%, 95% CI − 8.74 to − 5.34%; P = 0.000), TNF-α (WMD = −3.09, 95% CI − 5.47 to − 0.72; P = 0.011), TL-6 (WMD = −6.34, 95% CI − 11.2 to − 1.49; P < 0.01), TL-10 (WMD = 6.37, 95% CI 1.53–11.21; P = 0.01), the apoptosis rate (WMD = −8.23, 95% CI − 15.29 to − 1.17; P = 0.000), and the number of autophagic vesicles (WMD = −4.52, 95% CI − 7.43 to − 1.62; P = 0.000). Subgroup analysis showed that the EXOs were derived from HMSCs. Subgroup analysis showed that the EXOs derived from HMSCs, and that exosome therapy immediately after myocardial infarction can better improve the LVEF. Conclusions: EXOs therapy has the potential to improve cardiac function, fibrogenesis, and inflammatory response, as well as reducing cell apoptosis and autophagy in preclinical AMI animal models. This can inform future human clinical trials of EXOs. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-022-02833-z.
Collapse
Affiliation(s)
- Yan-Li Zheng
- Department of Cardiology, The Second Affiliated Hospital of Fujian Medical University, No. 34 North Zhongshan Road, Quanzhou, 362000, Fujian Province, China
| | - Wan-da Wang
- Department of Cardiology, The Second Affiliated Hospital of Fujian Medical University, No. 34 North Zhongshan Road, Quanzhou, 362000, Fujian Province, China
| | - Ping-Yu Cai
- Department of Cardiology, The Second Affiliated Hospital of Fujian Medical University, No. 34 North Zhongshan Road, Quanzhou, 362000, Fujian Province, China
| | - Feng Zheng
- Department of Neurosurgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China
| | - Yi-Fan Zhou
- Department of Cardiology, The Second Affiliated Hospital of Fujian Medical University, No. 34 North Zhongshan Road, Quanzhou, 362000, Fujian Province, China
| | - Mei-Mei Li
- Department of Cardiology, The Second Affiliated Hospital of Fujian Medical University, No. 34 North Zhongshan Road, Quanzhou, 362000, Fujian Province, China
| | - Jing-Ru Du
- Department of Cardiology, The Second Affiliated Hospital of Fujian Medical University, No. 34 North Zhongshan Road, Quanzhou, 362000, Fujian Province, China
| | - Shu Lin
- Department of Cardiology, The Second Affiliated Hospital of Fujian Medical University, No. 34 North Zhongshan Road, Quanzhou, 362000, Fujian Province, China. .,Centre of Neurological and Metabolic Research, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China. .,Diabetes and Metabolism Division, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Sydney, NSW, 2010, Australia.
| | - Hui-Li Lin
- Department of Cardiology, The Second Affiliated Hospital of Fujian Medical University, No. 34 North Zhongshan Road, Quanzhou, 362000, Fujian Province, China.
| |
Collapse
|
16
|
Recent Advances in the Application of Mesenchymal Stem Cell-Derived Exosomes for Cardiovascular and Neurodegenerative Disease Therapies. Pharmaceutics 2022; 14:pharmaceutics14030618. [PMID: 35335993 PMCID: PMC8949563 DOI: 10.3390/pharmaceutics14030618] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 03/02/2022] [Accepted: 03/09/2022] [Indexed: 12/17/2022] Open
Abstract
Exosomes are naturally occurring nanoscale vesicles that are released and received by almost all cells in the body. Exosomes can be transferred between cells and contain various molecular constitutes closely related to their origin and function, including proteins, lipids, and RNAs. The importance of exosomes in cellular communication makes them important vectors for delivering a variety of drugs throughout the body. Exosomes are ubiquitous in the circulatory system and can reach the site of injury or disease through a variety of biological barriers. Due to its unique structure and rich inclusions, it can be used for the diagnosis and treatment of diseases. Mesenchymal stem-cell-derived exosomes (MSCs-Exo) inherit the physiological functions of MSCs, including repairing and regenerating tissues, suppressing inflammatory responses, and regulating the body’s immunity; therefore, MSCs-Exo can be used as a natural drug delivery carrier with therapeutic effects, and has been increasingly used in the treatment of cardiovascular diseases and neurodegenerative diseases. Here, we summarize the research progress of MSCs-Exo as drug delivery vectors and their application for various drug deliveries, providing ideas and references for the study of MSCs-Exo in recent years.
Collapse
|
17
|
Yang L, Wang T, Zhang X, Zhang H, Yan N, Zhang G, Yan R, Li Y, Yu J, He J, Jia S, Wang H. Exosomes derived from human placental mesenchymal stem cells ameliorate myocardial infarction via anti-inflammation and restoring gut dysbiosis. BMC Cardiovasc Disord 2022; 22:61. [PMID: 35172728 PMCID: PMC8851843 DOI: 10.1186/s12872-022-02508-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 02/09/2022] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Myocardial infarction (MI) represents a severe cardiovascular disease with limited therapeutic agents. This study was aimed to elucidate the role of the exosomes derived from human placental mesenchymal stem cells (PMSCs-Exos) in MI. METHODS PMSCs were isolated and cultured in vitro, with identification by both transmission electron microscopy (TEM) and nanoparticle tracking analysis (NTA). To further investigate the effects of PMSC-Exos on MI, C57BL/6 mice were randomly divided into Sham group, MI group, and PMSC-Exos group. After 4 weeks of the intervention, cardiac function was assessed by cardiac echocardiography, electrocardiogram and masson trichrome staining; lipid indicators were determined by automatic biochemical instrument; inflammatory cytokines were measured by cytometric bead array (CBA); gut microbiota, microbial metabolites short chain fatty acids (SCFAs) as well as lipopolysaccharide (LPS) were separately investigated by 16S rRNA high throughput sequencing, gas chromatography mass spectrometry (GC-MS) and tachypleus amebocyte lysate kit; transcriptome analysis was used to test the transcriptional components (mRNA\miRNA\cirRNA\lncRNA) of PMSC-Exos. RESULTS We found that human PMSC-Exos were obtained and identified with high purity and uniformity. MI model was successfully established. Compared to MI group, PMSC-Exos treatment ameliorated myocardial fibrosis and left ventricular (LV) remodeling (P < 0.05). Moreover, PMSC-Exos treatment obviously decreased MI molecular markers (AST/BNP/MYO/Tn-I/TC), pro-inflammatory indicators (IL-1β, IL-6, TNF-α, MCP-1), as well as increased HDL in comparison with MI group (all P < 0.05). Intriguingly, PMSC-Exos intervention notably modulated gut microbial community via increasing the relative abundances of Bacteroidetes, Proteobacteria, Verrucomicrobia, Actinobacteria, Akkermansia, Bacteroides, Bifidobacterium, Thauera and Ruminiclostridium, as well as decreasing Firmicutes (all P < 0.05), compared with MI group. Furthermore, PMSC-Exos supplementation increased gut microbiota metabolites SCFAs (butyric acid, isobutyric acid and valeric acid) and decreased LPS in comparison with MI group (all P < 0.05). Correlation analysis indicated close correlations among gut microbiota, microbial SCFAs and inflammation in MI. CONCLUSIONS Our study highlighted that PMSC-Exos intervention alleviated MI via modulating gut microbiota and suppressing inflammation.
Collapse
Affiliation(s)
- Libo Yang
- Clinical Medical College, Ningxia Medical University, Yinchuan, 750004 Ningxia China
- Heart Centre and Department of Cardiovascular Diseases, General Hospital of Ningxia Medical University, Yinchuan, 750004 Ningxia China
- Ningxia Key Laboratory of Vascular Injury and Repair Research, Ningxia Medical University, Yinchuan, 750004 China
| | - Ting Wang
- Department of Pathogenic Biology and Medical Immunology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004 Ningxia China
| | - Xiaoxia Zhang
- College of Traditional Chinese Medicine, Ningxia Medical University, Yinchuan, 750004 Ningxia China
| | - Hua Zhang
- Heart Centre and Department of Cardiovascular Diseases, General Hospital of Ningxia Medical University, Yinchuan, 750004 Ningxia China
| | - Ning Yan
- Clinical Medical College, Ningxia Medical University, Yinchuan, 750004 Ningxia China
- Heart Centre and Department of Cardiovascular Diseases, General Hospital of Ningxia Medical University, Yinchuan, 750004 Ningxia China
- Ningxia Key Laboratory of Vascular Injury and Repair Research, Ningxia Medical University, Yinchuan, 750004 China
| | - Guoshan Zhang
- Heart Centre and Department of Cardiovascular Diseases, General Hospital of Ningxia Medical University, Yinchuan, 750004 Ningxia China
| | - Ru Yan
- Heart Centre and Department of Cardiovascular Diseases, General Hospital of Ningxia Medical University, Yinchuan, 750004 Ningxia China
- Ningxia Key Laboratory of Vascular Injury and Repair Research, Ningxia Medical University, Yinchuan, 750004 China
| | - Yiwei Li
- Department of Pathogenic Biology and Medical Immunology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004 Ningxia China
| | - Jingjing Yu
- Clinical Medical College, Ningxia Medical University, Yinchuan, 750004 Ningxia China
- Department of Beijing National Biochip Research Center Sub-Center in Ningxia, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Jun He
- Heart Centre and Department of Cardiovascular Diseases, General Hospital of Ningxia Medical University, Yinchuan, 750004 Ningxia China
- Ningxia Key Laboratory of Vascular Injury and Repair Research, Ningxia Medical University, Yinchuan, 750004 China
| | - Shaobin Jia
- Clinical Medical College, Ningxia Medical University, Yinchuan, 750004 Ningxia China
- Heart Centre and Department of Cardiovascular Diseases, General Hospital of Ningxia Medical University, Yinchuan, 750004 Ningxia China
- Ningxia Key Laboratory of Vascular Injury and Repair Research, Ningxia Medical University, Yinchuan, 750004 China
| | - Hao Wang
- Department of Pathogenic Biology and Medical Immunology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004 Ningxia China
| |
Collapse
|
18
|
Low-density lipoprotein receptor and apolipoprotein A 5, myocardial infarction biomarkers in plasma-derived exosomes. J Cardiol 2022; 79:605-610. [DOI: 10.1016/j.jjcc.2021.10.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 09/16/2021] [Accepted: 09/21/2021] [Indexed: 11/20/2022]
|
19
|
Zhou Q, Deng J, Yao J, Song J, Meng D, Zhu Y, Xu M, Liang Y, Xu J, Sluijter JP, Xiao J. Exercise downregulates HIPK2 and HIPK2 inhibition protects against myocardial infarction. EBioMedicine 2021; 74:103713. [PMID: 34837851 PMCID: PMC8626841 DOI: 10.1016/j.ebiom.2021.103713] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 10/11/2021] [Accepted: 11/09/2021] [Indexed: 12/13/2022] Open
Abstract
Background Exercise can protect myocardial infarction (MI) and downregulate cardiac Homeodomain-Interacting Protein Kinase 2 (HIPK2). However, the role of HIPK2 in MI is unclear. Methods HIPK2–/– mice and miR-222–/– rats, HIPK2 inhibitor (PKI1H) and adeno-associated virus serotype 9 (AAV9) carrying miR-222 were applied in the study. Animals were subjected to running, swimming, acute MI or post-MI remodeling. HIPK2 inhibition and P53 activator were used in neonatal rat cardiomyocytes (NRCMs) and human embryonic stem cell-derived cardiomyocytes (hESC-CMs) subjected to oxygen glucose deprivation/reperfusion (OGD/R). Serum miR-222 levels were analyzed in healthy people and MI patients that were survival or readmitted to the hospital and/or died. Findings Cardiac HIPK2 protein levels were reduced by exercise while increased in MI. In vitro, HIPK2 suppression by lentiviral vectors or inhibitor prevented apoptosis induced by OGD/R in NRCMs and hESC-CMs. HIPK2 inhibitor-treated mice and HIPK2–/– mice reduced infarct size after acute MI, and preserved cardiac function in MI remodeling. Mechanistically, protective effect against apoptosis by HIPK2 suppression was reversed by P53 activators. Furthermore, increasing levels of miR-222, targeting HIPK2, protected post-MI cardiac dysfunction, whereas cardiac dysfunction post-MI was aggravated in miR-222–/– rats. Moreover, serum miR-222 levels were significantly reduced in MI patients, as well as in MI patients that were readmitted to the hospital and/or died compared to those not. Interpretation Exercise-induced HIPK2 suppression attenuates cardiomyocytes apoptosis and protects MI by decreasing P-P53. Inhibition of HIPK2 represents a potential novel therapeutic intervention for MI. Funding This work was supported by the grants from National Key Research and Development Project (2018YFE0113500 to JJ Xiao), National Natural Science Foundation of China (82020108002, 81722008, and 81911540486 to JJ Xiao, 81400647 to MJ Xu, 81800265 to YJ Liang), Innovation Program of Shanghai Municipal Education Commission (2017-01-07-00-09-E00042 to JJ Xiao), the grant from Science and Technology Commission of Shanghai Municipality (18410722200 and 17010500100 to JJ Xiao), the “Dawn” Program of Shanghai Education Commission (19SG34 to JJ Xiao), Shanghai Sailing Program (21YF1413200 to QL Zhou). JS is supported by Horizon2020 ERC-2016-COG EVICARE (725229).
Collapse
Affiliation(s)
- Qiulian Zhou
- Shanghai Engineering Research Center of Organ Repair, Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China; Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, Shanghai, 200444, China
| | - Jiali Deng
- Shanghai Engineering Research Center of Organ Repair, Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China
| | - Jianhua Yao
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Jiaxin Song
- Shanghai Engineering Research Center of Organ Repair, Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China
| | - Danni Meng
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, Shanghai, 200444, China
| | - Yujiao Zhu
- Shanghai Engineering Research Center of Organ Repair, Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China
| | - Minjun Xu
- Shanghai Engineering Research Center of Organ Repair, Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China
| | - Yajun Liang
- Shanghai Engineering Research Center of Organ Repair, Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China
| | - Jiahong Xu
- Department of Cardiology, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Joost Pg Sluijter
- Department of Cardiology, Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht, 3508GA, the Netherlands; UMC Utrecht Regenerative Medicine Center, University Medical Center, Utrecht University, Utrecht, 3508GA, the Netherlands
| | - Junjie Xiao
- Shanghai Engineering Research Center of Organ Repair, Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China; Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, Shanghai, 200444, China.
| |
Collapse
|
20
|
M1 Bone Marrow-Derived Macrophage-Derived Extracellular Vesicles Inhibit Angiogenesis and Myocardial Regeneration Following Myocardial Infarction via the MALAT1/MicroRNA-25-3p/CDC42 Axis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:9959746. [PMID: 34745428 PMCID: PMC8570847 DOI: 10.1155/2021/9959746] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 09/08/2021] [Accepted: 10/04/2021] [Indexed: 11/18/2022]
Abstract
Myocardial infarction (MI) is a severe cardiovascular disease. Some M1 macrophage-derived extracellular vesicles (EVs) are involved in the inhibition of angiogenesis and acceleration dysfunction during MI. However, the potential mechanism of M1 phenotype bone marrow-derived macrophages- (BMMs-) EVs (M1-BMMs-EVs) in MI is largely unknown. This study sought to investigate whether M1-BMMs-EVs increased CDC42 expression and activated the MEK/ERK pathway by carrying lncRNA MALAT1 and competitively binding to miR-25-3p, thus inhibiting angiogenesis and myocardial regeneration after MI. After EV treatment, the cardiac function, infarct size, fibrosis, angiogenesis, and myocardial regeneration of MI mice and the viability, proliferation and angiogenesis of oxygen-glucose deprivation- (OGD-) treated myocardial microvascular endothelial cells (MMECs) were assessed. MALAT1 expression in MI mice, cells, and EVs was detected. MALAT1 downstream microRNAs (miRs), genes, and pathways were predicted and verified. MALAT1 and miR-25-3p were intervened to evaluate EV effects on OGD-treated cells. In MI mice, EV treatment aggravated MI and inhibited angiogenesis and myocardial regeneration. In OGD-treated cells, EV treatment suppressed cell viability, proliferation, and angiogenesis. MALAT1 was highly expressed in MI mice, OGD-treated MMECs, M1-BMMs, and EVs. Silencing MALAT1 weakened the inhibition of EV treatment on OGD-treated cells. MALAT1 sponged miR-25-3p to upregulate CDC42. miR-25-3p overexpression promoted OGD-treated cell viability, proliferation, and angiogenesis. The MEK/ERK pathway was activated after EV treatment. Collectively, M1-BMMs-EVs inhibited angiogenesis and myocardial regeneration following MI via the MALAT1/miR-25-3p/CDC42 axis and the MEK/ERK pathway activation.
Collapse
|
21
|
Wang J, Xu T, Xu M. Roles and Mechanisms of TGR5 in the Modulation of CD4 + T Cell Functions in Myocardial Infarction. J Cardiovasc Transl Res 2021; 15:350-359. [PMID: 34402028 DOI: 10.1007/s12265-021-10164-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 08/02/2021] [Indexed: 10/20/2022]
Abstract
Bile acid receptor TGR5 has been proved to play protective roles in the process of myocardial infarction (MI). Recently, we found spleen weight of Tgr5+/+ mice was increased at 7-day post-MI but not in Tgr5-/- mice. Since the spleen is one of the main resources of immune and inflammatory cells post-MI, we conducted flow cytometry analysis of multiple immune cells in the heart post-MI. It showed the recruitment of CD4+ T cells and CD8+ T cells was continuously more in the heart of Tgr5-/- mice post-MI until 7 days after MI. Furthermore, CD4-specific TGR5 depletion mice exhibited aggravated ischemic injury. The mRNA expressions of the markers of Th1 and Treg were upregulated in the heart of Tgr5-/- mice at 7-day post-MI. These results suggested TGR5 modulates CD4+ T cell functions and subsets distribution in the heart, and plays protective roles in myocardial infarction.
Collapse
Affiliation(s)
- Jiaxing Wang
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, 100191, China.,NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptide, Peking University Third Hospital, Beijing, 100191, China.,Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Third Hospital, Beijing, 100191, China.,Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University Third Hospital, Beijing, 100191, China
| | - Tan Xu
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, 100191, China.,NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptide, Peking University Third Hospital, Beijing, 100191, China.,Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Third Hospital, Beijing, 100191, China.,Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University Third Hospital, Beijing, 100191, China
| | - Ming Xu
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, 100191, China. .,NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptide, Peking University Third Hospital, Beijing, 100191, China. .,Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Third Hospital, Beijing, 100191, China. .,Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University Third Hospital, Beijing, 100191, China. .,State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China.
| |
Collapse
|
22
|
Viola M, de Jager SCA, Sluijter JPG. Targeting Inflammation after Myocardial Infarction: A Therapeutic Opportunity for Extracellular Vesicles? Int J Mol Sci 2021; 22:ijms22157831. [PMID: 34360595 PMCID: PMC8346058 DOI: 10.3390/ijms22157831] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 07/15/2021] [Accepted: 07/19/2021] [Indexed: 12/12/2022] Open
Abstract
After myocardial infarction (MI), a strong inflammatory response takes place in the heart to remove the dead tissue resulting from ischemic injury. A growing body of evidence suggests that timely resolution of this inflammatory process may aid in the prevention of adverse cardiac remodeling and heart failure post-MI. The present challenge is to find a way to stimulate this process without interfering with the reparative role of the immune system. Extracellular vesicles (EVs) are natural membrane particles that are released by cells and carry different macromolecules, including proteins and non-coding RNAs. In recent years, EVs derived from various stem and progenitor cells have been demonstrated to possess regenerative properties. They can provide cardioprotection via several mechanisms of action, including immunomodulation. In this review, we summarize the role of the innate immune system in post-MI healing. We then discuss the mechanisms by which EVs modulate cardiac inflammation in preclinical models of myocardial injury through regulation of monocyte influx and macrophage function. Finally, we provide suggestions for further optimization of EV-based therapy to improve its potential for the treatment of MI.
Collapse
Affiliation(s)
- Margarida Viola
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands;
- UMC Utrecht Regenerative Medicine Center, Circulatory Health Laboratory, University Utrecht, 3584 CS Utrecht, The Netherlands
| | - Saskia C. A. de Jager
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands;
- UMC Utrecht Regenerative Medicine Center, Circulatory Health Laboratory, University Utrecht, 3584 CS Utrecht, The Netherlands
- Correspondence: (S.C.A.d.J.); (J.P.G.S.)
| | - Joost P. G. Sluijter
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands;
- UMC Utrecht Regenerative Medicine Center, Circulatory Health Laboratory, University Utrecht, 3584 CS Utrecht, The Netherlands
- Correspondence: (S.C.A.d.J.); (J.P.G.S.)
| |
Collapse
|
23
|
Wu Y, Wu M, Yang J, Li Y, Peng W, Wu M, Yu C, Fang M. Silencing CircHIPK3 Sponges miR-93-5p to Inhibit the Activation of Rac1/PI3K/AKT Pathway and Improves Myocardial Infarction-Induced Cardiac Dysfunction. Front Cardiovasc Med 2021; 8:645378. [PMID: 33996942 PMCID: PMC8119651 DOI: 10.3389/fcvm.2021.645378] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 03/12/2021] [Indexed: 01/24/2023] Open
Abstract
The ceRNA network involving circular RNAs (circRNAs) is essential in the cardiovascular system. We investigated the underlying ceRNA network involving circHIPK3 in myocardial infarction (MI). After an MI model was established, cardiac function was verified, and myocardial tissue damage in mice with MI was evaluated. A hypoxia model of cardiomyocytes was used to simulate MI in vivo, and the expression of and targeting relationships among circHIPK3, miR-93-5p, and Rac1 were verified. The apoptosis of cardiomyocyte was identified. Gain- and loss-of-functions were performed to verify the ceRNA mechanism. The MI-modeled mice showed cardiac dysfunction and enlarged infarct size. CircHIPK3 was highly expressed in mouse and cell models of MI. Silencing circHIPK3 reduced infarct size, myocardial collagen deposition, and myocardial apoptosis rate and improved cardiac function. CircHIPK3 sponged miR-93-5p, and miR-93-5p targeted Rac1. Overexpression of miR-93-5p inhibited MI-induced cardiomyocyte injury and eliminated the harmful effect of circHIPK3. CircHIPK3 acted as ceRNA to absorb miR-93-5p, thus promoting the activation of the Rac1/PI3K/AKT pathway. We highlighted that silencing circHIPK3 can upregulate miR-93-5p and then inhibit the activation of Rac1/PI3K/Akt pathway, which can improve MI-induced cardiac dysfunction.
Collapse
Affiliation(s)
- Yijin Wu
- Department of Intensive Care Unit of Cardiac Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangdong Cardiovascular Institute, Guangzhou, China
| | - Min Wu
- Department of Cardiac Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangdong Cardiovascular Institute, Guangzhou, China
| | - Jue Yang
- Department of Cardiac Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangdong Cardiovascular Institute, Guangzhou, China
| | - Ying Li
- Department of Cardiac Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangdong Cardiovascular Institute, Guangzhou, China
| | - Wenying Peng
- Department of Intensive Care Unit of Cardiac Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangdong Cardiovascular Institute, Guangzhou, China
| | - Meifen Wu
- Department of Intensive Care Unit of Cardiac Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangdong Cardiovascular Institute, Guangzhou, China
| | - Changjiang Yu
- Department of Cardiac Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangdong Cardiovascular Institute, Guangzhou, China,Changjiang Yu
| | - Miaoxian Fang
- Department of Intensive Care Unit of Cardiac Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangdong Cardiovascular Institute, Guangzhou, China,*Correspondence: Miaoxian Fang
| |
Collapse
|
24
|
Adipose-Derived Mesenchymal Stem Cells-Derived Exosomes Carry MicroRNA-671 to Alleviate Myocardial Infarction Through Inactivating the TGFBR2/Smad2 Axis. Inflammation 2021; 44:1815-1830. [PMID: 33881681 PMCID: PMC8460592 DOI: 10.1007/s10753-021-01460-9] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/11/2021] [Accepted: 03/29/2021] [Indexed: 12/14/2022]
Abstract
Mesenchymal stem cells (MSCs) and their derived extracellular vesicles have been reported as promising tools for the management of heart disease. The aim of this study was to explore the function of adipose-derived MSCs (adMSCs)-derived exosomes (Exo) in the progression of myocardial infarction (MI) and the molecules involved. Mouse cardiomyocytes were treated with oxygen-glucose deprivation (OGD) to mimic an MI condition in vitro. The adMSCs-derived Exo were identified and administrated into the OGD-treated cardiomyocytes, and then the viability and apoptosis of cells, and the secretion of fibrosis- and inflammation-related cytokines in cells were determined. Differentially expressed microRNAs (miRNAs) in cells after Exo treatment were screened using a microarray analysis. The downstream molecules regulated by miR-671 were explored through bioinformatic analysis. Involvements of miR-671 and transforming growth factor beta receptor 2 (TGFBR2) in the exosome-mediated events were confirmed by rescue experiments. A murine model with MI was induced and treated with Exo for functional experiments in vivo. Compared to phosphate-buffered saline treatment, the Exo treatment significantly enhanced viability while reduced apoptosis of cardiomyocytes, and in reduced myocardial fibrosis and inflammation both in vitro and in vivo. miR-671 was significantly upregulated in cells after Exo treatment. Downregulation of miR-671 blocked the protective functions of Exo. miR-671 targeted TGFBR2 and suppressed phosphorylation of Smad2. Artificial downregulation of TGFBR2 enhanced viability of the OGD-treated cardiomyocytes. This study suggested that adMSC-derived exosomal miR-671 directly targets TGFBR2 and reduces Smad2 phosphorylation to alleviate MI-like symptoms both in vivo and in vitro.
Collapse
|
25
|
Wu Z, Cheng S, Wang S, Li W, Liu J. RETRACTED:BMSCs-derived exosomal microRNA-150-5p attenuates myocardial infarction in mice. Int Immunopharmacol 2021; 93:107389. [PMID: 33582480 DOI: 10.1016/j.intimp.2021.107389] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 01/05/2021] [Accepted: 01/08/2021] [Indexed: 12/31/2022]
Abstract
This article has been retracted: please see Elsevier Policy on Article Withdrawal (http://www.elsevier.com/locate/withdrawalpolicy). This article has been retracted at the request of the Editor-in-Chief. Concern was raised about the reliability of the Western blot results in Figs. 2B and 7B, which appear to have the same eyebrow shaped phenotype as many other publications tabulated here (https://docs.google.com/spreadsheets/d/149EjFXVxpwkBXYJOnOHb6RhAqT4a2llhj9LM60MBffM/edit#gid=0 [docs.google.com]). Concerns were also raised over the provenance of the flow cytometry plots in Fig. 1F. The journal requested the corresponding author comment on these concerns and provide the raw data. However, the authors were not responsive to the request for comment. Since original data could not be provided, the overall validity of the results could not be confirmed. Therefore, the Editor-in-Chief decided to retract the article.
Collapse
Affiliation(s)
- Zheng Wu
- Department of 28 Division of Cardiovascular, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing 100029, China
| | - Shujuan Cheng
- Department of 28 Division of Cardiovascular, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing 100029, China
| | - Shaoping Wang
- Department of 28 Division of Cardiovascular, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing 100029, China
| | - Wenzheng Li
- Department of 28 Division of Cardiovascular, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing 100029, China
| | - Jinghua Liu
- Department of 28 Division of Cardiovascular, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing 100029, China.
| |
Collapse
|
26
|
Abstract
Abstract
Purpose of Review
The regenerative capacity of the heart is insufficient to compensate for the pathological loss of cardiomyocytes during a large injury, such as a myocardial infarction. Therapeutic options for patients after cardiac infarction are limited: treatment with drugs that only treat the symptoms or extraordinary measures, such as heart transplantation. Cell therapies offer a promising strategy for cardiac regeneration. In this brief review, the major issues in these areas are discussed, and possible directions for future research are indicated.
Recent Findings
Cardiac regeneration can be obtained by at least two strategies: the first is direct to generate an ex vivo functional myocardial tissue that replaces damaged tissue; the second approach aims to stimulate endogenous mechanisms of cardiac repair. However, current cell therapies are still hampered by poor translation into actual clinical applications.
Summary
In this scenario, recent advancements in cell biology and biomaterial-based technologies can play a key role to design effective therapeutic approaches.
Collapse
|
27
|
Huang H, Xu Z, Qi Y, Zhang W, Zhang C, Jiang M, Deng S, Wang H. Exosomes from SIRT1-Overexpressing ADSCs Restore Cardiac Function by Improving Angiogenic Function of EPCs. MOLECULAR THERAPY. NUCLEIC ACIDS 2020; 21:737-750. [PMID: 32771925 PMCID: PMC7412761 DOI: 10.1016/j.omtn.2020.07.007] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 06/19/2020] [Accepted: 07/06/2020] [Indexed: 12/15/2022]
Abstract
Acute myocardial infarction (AMI) is one of the leading causes of mortality in cardiovascular diseases. The aim of this study was to investigate whether exosomes from Sirtuin 1 (SIRT1)-overexpressing adipose-derived stem cells (ADSCs) had a protective effect on AMI. The expression of C-X-C chemokine receptor type 7 (CXCR7) was significantly downregulated in peripheral blood endothelial progenitor cells (EPCs) from AMI patients (AMI-EPCs) compared with that in healthy donors, which coincided with impaired tube formation. The exosomes from SIRT1 overexpression in ADSCs (ADSCs-SIRT1-Exos) increased the expression of C-X-C motif chemokine 12 (CXCL12) and nuclear factor E2 related factor 2 (Nrf2) in AMI-EPCs, which promoted migration and tube formation of AMI-EPCs, and overexpression of CXCR7 helped AMI-EPCs to restore the function of cell migration and tube formation. Moreover, CXCR7 was downregulated in the myocardium of AMI mice, and knockout of CXCR7 exacerbated AMI-induced impairment of cardiovascular function. Injection of ADSCs-SIRT1-Exos increased the survival and promoted the recovery of myocardial function with reduced infarct size and post-AMI left ventricular remodeling, induced vasculogenesis, and decreased AMI-induced myocardial inflammation. These findings showed that ADSCs-SIRT1-Exos may recruit EPCs to the repair area and that this recruitment may be mediated by Nrf2/CXCL12/CXCR7 signaling.
Collapse
Affiliation(s)
- Hui Huang
- Department of Cardiology, Shanghai Pudong New Area Gongli Hospital, Shanghai 200135, P.R. China
| | - Zhenxing Xu
- Department of Cardiology, Shanghai Pudong New Area Gongli Hospital, Shanghai 200135, P.R. China
| | - Yuan Qi
- Department of Cardiology, Shanghai Pudong New Area Gongli Hospital, Shanghai 200135, P.R. China
| | - Wei Zhang
- Department of Cardiology, Shanghai Pudong New Area Gongli Hospital, Shanghai 200135, P.R. China
| | - Chenjun Zhang
- Department of Cardiology, Shanghai Pudong New Area Gongli Hospital, Shanghai 200135, P.R. China
| | - Mei Jiang
- Department of Neurology, Shanghai Pudong New Area Gongli Hospital, Shanghai 200135, P.R. China
| | - Shengqiong Deng
- Department of Clinical Laboratory, Shanghai Pudong New Area Gongli Hospital, Shanghai 200135, P.R. China
| | - Hairong Wang
- Department of Cardiology, Shanghai Pudong New Area Gongli Hospital, Shanghai 200135, P.R. China.
| |
Collapse
|
28
|
Mesenchymal and Induced Pluripotent Stem Cells-Derived Extracellular Vesicles: The New Frontier for Regenerative Medicine? Cells 2020; 9:cells9051163. [PMID: 32397132 PMCID: PMC7290733 DOI: 10.3390/cells9051163] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 05/02/2020] [Accepted: 05/04/2020] [Indexed: 12/13/2022] Open
Abstract
Regenerative medicine aims to repair damaged, tissues or organs for the treatment of various diseases, which have been poorly managed with conventional drugs and medical procedures. To date, multimodal regenerative methods include transplant of healthy organs, tissues, or cells, body stimulation to activate a self-healing response in damaged tissues, as well as the combined use of cells and bio-degradable scaffold to obtain functional tissues. Certainly, stem cells are promising tools in regenerative medicine due to their ability to induce de novo tissue formation and/or promote organ repair and regeneration. Currently, several studies have shown that the beneficial stem cell effects, especially for mesenchymal stem cells (MSCs) and induced pluripotent stem cells (iPSCs) in damaged tissue restore are not dependent on their engraftment and differentiation on the injury site, but rather to their paracrine activity. It is now well known that paracrine action of stem cells is due to their ability to release extracellular vesicles (EVs). EVs play a fundamental role in cell-to-cell communication and are directly involved in tissue regeneration. In the present review, we tried to summarize the molecular mechanisms through which MSCs and iPSCs-derived EVs carry out their therapeutic action and their possible application for the treatment of several diseases.
Collapse
|
29
|
He N, Zhang Y, Zhang S, Wang D, Ye H. Exosomes: Cell-Free Therapy for Cardiovascular Diseases. J Cardiovasc Transl Res 2020; 13:713-721. [PMID: 32333198 DOI: 10.1007/s12265-020-09966-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 02/04/2020] [Indexed: 12/20/2022]
Abstract
Cardiovascular diseases (CVDs) are an important cause of death and disease worldwide. Because injured cardiac tissue cannot be repaired itself, it is urgent to develop other alternate therapies. Stem cells can be differentiated into cardiomyocytes, endothelial cells, and vascular smooth muscle cells for the treatment of CVDs. Therefore, cell therapy has recently been considered a viable treatment option that can significantly improve cardiac function. Nonetheless, implanted stem cells rarely survive in the recipient heart, suggesting that the benefits of stem cell therapy may involve other mechanisms. Exosomes derived from stem cells have a myocardial protection function after myocardial injury, and may be a promising and effective therapy for CVDs. Here, we discuss the application and mechanism of exosomes derived from stem cells in the diagnosis and treatment of CVDs and provide evidence for the application of exosomes in CVDs. Graphical Abstract.
Collapse
Affiliation(s)
- Nana He
- Department of Cardiology, HwaMei Hospital (previously named Ningbo No. 2 Hospital), University of Chinese Academy of Sciences, 41 Xibei Street, Ningbo, 315010, Zhejiang, China
- Department of Experimental Medical Science, HwaMei Hospital, University of Chinese Academy of Sciences, Ningbo, China
- Key Laboratory of Diagnosis and Treatment of Digestive System Tumors of Zhejiang Province, Ningbo, China
| | - Yuelin Zhang
- Department of Medicine, University of Ningbo, Ningbo, China
| | - Shun Zhang
- Department of Experimental Medical Science, HwaMei Hospital, University of Chinese Academy of Sciences, Ningbo, China
- Key Laboratory of Diagnosis and Treatment of Digestive System Tumors of Zhejiang Province, Ningbo, China
| | - Dongjuan Wang
- Department of Cardiology, HwaMei Hospital (previously named Ningbo No. 2 Hospital), University of Chinese Academy of Sciences, 41 Xibei Street, Ningbo, 315010, Zhejiang, China
| | - Honghua Ye
- Department of Cardiology, HwaMei Hospital (previously named Ningbo No. 2 Hospital), University of Chinese Academy of Sciences, 41 Xibei Street, Ningbo, 315010, Zhejiang, China.
| |
Collapse
|
30
|
Effects of Colchicine on Atherosclerotic Plaque Stabilization: a Multimodality Imaging Study in an Animal Model. J Cardiovasc Transl Res 2020; 14:150-160. [PMID: 32140929 DOI: 10.1007/s12265-020-09974-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Accepted: 02/14/2020] [Indexed: 02/07/2023]
Abstract
Colchicine demonstrated clinical benefits in the treatment of stable coronary artery disease. Our aim was to evaluate the effects of colchicine on atherosclerotic plaque stabilization. Atherosclerosis was induced in the abdominal aorta of 20 rabbits with high-cholesterol diet and balloon endothelial denudation. Rabbits were randomized to receive either colchicine or placebo. All animals underwent MRI, 18F-FDG PET/CT, optical coherence tomography (OCT), and histology. Similar progression of atherosclerotic burden was observed in the two groups as relative increase of normalized wall index (NWI). Maximum 18F-FDG standardized uptake value (meanSUVmax) decreased after colchicine treatment, while it increased in the placebo group with a trend toward significance. Animals with higher levels of cholesterol showed significant differences in favor to colchicine group, both as NWI at the end of the protocol and as relative increase in meanSUVmax. Colchicine may stabilize atherosclerotic plaque by reducing inflammatory activity and plaque burden, without altering macrophage infiltration or plaque typology.
Collapse
|
31
|
Wang J, Liu S, Li G, Xiao J. Exercise Regulates the Immune System. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1228:395-408. [PMID: 32342473 DOI: 10.1007/978-981-15-1792-1_27] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The profound effect of exercise on the normal functioning of the immune system has been well-known. Exercise and immune regulation are interrelated and affect each other. Exercise changes immune regulation by affecting leucocytes, red blood cells, and cytokines, etc. Regular exercise could reduce the risk of chronic metabolic and cardiorespiratory diseases, partially by the anti-inflammatory effects of exercise. However, these effects are also likely to be responsible for the suppressed immunity that make our bodies more susceptible to infections. Here we summarize the known mechanisms by which exercise-both acute and chronic-exerts its immune regulation effects.
Collapse
Affiliation(s)
- Jing Wang
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, Shanghai, China
| | - Shuqin Liu
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, Shanghai, China
| | - Guoping Li
- Cardiovascular Division of the Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Junjie Xiao
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, Shanghai, China.
| |
Collapse
|
32
|
Ma Z, Wang Y, Li H. Applications of extracellular vesicles in tissue regeneration. BIOMICROFLUIDICS 2020; 14:011501. [PMID: 32002105 PMCID: PMC6984977 DOI: 10.1063/1.5127077] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 01/15/2020] [Indexed: 05/05/2023]
Abstract
Extracellular vesicles (EVs) can be classified into several types based on their different biosyntheses or release pathways, including exosomes, microvesicles, apoptotic bodies, and large oncosomes. As they contain DNAs, RNAs, proteins, and other bioactive signals, EVs have been utilized in the diagnosis field for a long time. Considering the fact that stem cells have been widely used for tissue regeneration and EVs possess similar biological properties to their source cells, tissue regeneration abilities of EVs have recently attracted much attention in the regenerative medicine field. In this paper, recent advances and challenges of EVs applied in the repair and regeneration of damaged tissues, such as skin, heart, liver, kidney, bone, and central nervous system, have been summarized. Specifically, critical bioactive molecules, which are encapsulated within EVs and play significant roles in the tissue regeneration, have been highlighted. Finally, the prospects and future development directions of the application of EVs in the field of tissue regeneration have been discussed.
Collapse
Affiliation(s)
| | | | - Haiyan Li
- Author to whom correspondence should be addressed:. Tel.: +86 18717902901
| |
Collapse
|
33
|
Carotenuto F, Teodori L, Maccari AM, Delbono L, Orlando G, Di Nardo P. Turning regenerative technologies into treatment to repair myocardial injuries. J Cell Mol Med 2019; 24:2704-2716. [PMID: 31568640 PMCID: PMC7077550 DOI: 10.1111/jcmm.14630] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 06/28/2019] [Accepted: 07/23/2019] [Indexed: 02/06/2023] Open
Abstract
Regenerative therapies including stem cell treatments hold promise to allow curing patients affected by severe cardiac muscle diseases. However, the clinical efficacy of stem cell therapy remains elusive, so far. The two key roadblocks that still need to be overcome are the poor cell engraftment into the injured myocardium and the limited knowledge of the ideal mixture of bioactive factors to be locally delivered for restoring heart function. Thus, therapeutic strategies for cardiac repair are directed to increase the retention and functional integration of transplanted cells in the damaged myocardium or to enhance the endogenous repair mechanisms through cell‐free therapies. In this context, biomaterial‐based technologies and tissue engineering approaches have the potential to dramatically impact cardiac translational medicine. This review intends to offer some consideration on the cell‐based and cell‐free cardiac therapies, their limitations and the possible future developments.
Collapse
Affiliation(s)
- Felicia Carotenuto
- Centro Interdipartimentale di Medicina Rigenerativa, Università di Roma Tor Vergata, Rome, Italy.,Dipartimento di Scienze Cliniche e Medicina Traslazionale, Università di Roma Tor Vergata, Rome, Italy.,Diagnostics and Metrology (FSN-TECFIS-DIM), ENEA, C.R. Frascati, Rome, Italy
| | - Laura Teodori
- Diagnostics and Metrology (FSN-TECFIS-DIM), ENEA, C.R. Frascati, Rome, Italy
| | - Anna Maria Maccari
- Centro Interdipartimentale di Medicina Rigenerativa, Università di Roma Tor Vergata, Rome, Italy.,Dipartimento di Scienze Cliniche e Medicina Traslazionale, Università di Roma Tor Vergata, Rome, Italy
| | - Luciano Delbono
- Wake Forest University School of Medicine, Winston Salem, NC, USA
| | - Giuseppe Orlando
- Wake Forest University School of Medicine, Winston Salem, NC, USA.,Department of Surgery, Wake Forest University School of Medicine, Winston Salem, NC, USA
| | - Paolo Di Nardo
- Centro Interdipartimentale di Medicina Rigenerativa, Università di Roma Tor Vergata, Rome, Italy.,Dipartimento di Scienze Cliniche e Medicina Traslazionale, Università di Roma Tor Vergata, Rome, Italy.,I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| |
Collapse
|
34
|
Exosomes in Cardiovascular Diseases and Treatment: Experimental and Clinical Aspects. J Cardiovasc Transl Res 2019; 12:1-2. [DOI: 10.1007/s12265-018-9860-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 12/21/2018] [Indexed: 12/26/2022]
|