1
|
Lin S, Yang Y, Zhou Z, Li W, Wang X, Liu Y, Bi Y, Mao J. Regulation mechanism of microRNAs in cardiac cells-derived exosomes in cell crosstalk. Front Pharmacol 2024; 15:1399850. [PMID: 39228519 PMCID: PMC11368792 DOI: 10.3389/fphar.2024.1399850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 07/22/2024] [Indexed: 09/05/2024] Open
Abstract
The heart is a multicellular system, and the intercellular crosstalk mechanism is very important for the growth and development of the heart and even the organs, tissues, and cells at a distance. As a kind of extracellular vesicle, exosomes are released by different types of cells and can carry specific genetic material, endosomal proteins, cytokines, etc., which are the main material basis for mediating cell crosstalk mechanism. Among them, microRNA carried by cardiac cells-derived exosomes have highly conserved sequences and play a key role in regulating the function of organs, tissues, and cells related to cardiovascular diseases and their complications and comorbidities, which have attracted extensive attention in the medical community in recent years. Following up on the latest research progress at home and abroad, this review systematically summarized the regulatory role of microRNA from cardiac cells-derived exosomes in various cell crosstalk, including not only cardiac cells (including cardiomyocytes, fibroblasts, myofibroblast, cardiac progenitor cells, cardiac microvascular endothelial cells, cardiosphere-derived cells, etc.) but also tumor cells, bone marrow progenitor cells, and other tissue cells, in order to provide a reference for the prevention and treatment of cardiovascular diseases and their complications and comorbidities.
Collapse
Affiliation(s)
- Shanshan Lin
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yuanjian Yang
- Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Zhou Zhou
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Wen Li
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xianliang Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Yu Liu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Yingfei Bi
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Jingyuan Mao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| |
Collapse
|
2
|
Zhao L, Qian X, Ren Z, Wang A. miR-31-5p suppresses myocardial hypertrophy by targeting Nfatc2ip. J Cell Mol Med 2024; 28:e18413. [PMID: 38894694 PMCID: PMC11187844 DOI: 10.1111/jcmm.18413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 04/20/2024] [Accepted: 05/02/2024] [Indexed: 06/21/2024] Open
Abstract
Cardiac hypertrophy, worldwide known as an adaptive functional compensatory state of myocardial stress, is mainly believed to proceed to severe heart diseases, even to sudden death. Emerging studies have explored the microRNA alteration during hypertrophy. However, the mechanisms of microRNAs involved in cardiac hypertrophy are still uncertain. We studied young rats to establish abdominal aorta coarctation (AAC) for 4 weeks. With the significant downregulated cardiac function and upregulated hypertrophic biomarkers, AAC-induced rats showed enlarged myocardiocytes and alterations in microRNAs, especially downregulated miR-31-5p. miR-31-5p targets the 3'UTR of Nfatc2ip and inhibits myocardial hypertrophy in vitro and in vivo. Furthermore, we verified that Nfatc2ip is necessary and sufficient for cardiac hypertrophy in neonatal rat cardiomyocytes. Moreover, we found miR-31-5p inhibited the colocalization of Nfatc2ip and hypertrophic gene β-Mhc. Luciferase assay and ChiP-qPCR test demonstrated that Nfatc2ip binded to the core-promoter of β-Mhc and enhanced its transcriptional activity. Above all, our study found a new pathway, mir-31-5p/Nfatc2ip/β-Mhc, which is involved in cardiac hypertrophy, suggesting a potential target for intervention of cardiac hypertrophy.
Collapse
Affiliation(s)
- Lamei Zhao
- Department of Cardiology1st Affiliated Hospital of Anhui Medical UniversityHefeiAnhuiChina
| | - Xiaotao Qian
- Department of Oncology, Hefei Cancer HospitalChinese Academy of SciencesHefeiAnhuiChina
| | - Zhenxing Ren
- Department of Anatomy, The Research Center of Basic Integrative MedicineGuangzhou University of Traditional Chinese MedicineGuangzhouGuangdongChina
| | - Ailing Wang
- Department of Cardiology1st Affiliated Hospital of Anhui Medical UniversityHefeiAnhuiChina
| |
Collapse
|
3
|
Kovácsházi C, Hambalkó S, Sayour NV, Gergely TG, Brenner GB, Pelyhe C, Kapui D, Weber BY, Hültenschmidt AL, Pállinger É, Buzás EI, Zolcsák Á, Kiss B, Bozó T, Csányi C, Kósa N, Kellermayer M, Farkas R, Karvaly GB, Wynne K, Matallanas D, Ferdinandy P, Giricz Z. Effect of hypercholesterolemia on circulating and cardiomyocyte-derived extracellular vesicles. Sci Rep 2024; 14:12016. [PMID: 38797778 PMCID: PMC11128454 DOI: 10.1038/s41598-024-62689-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 05/20/2024] [Indexed: 05/29/2024] Open
Abstract
Hypercholesterolemia (HC) induces, propagates and exacerbates cardiovascular diseases via various mechanisms that are yet not properly understood. Extracellular vesicles (EVs) are involved in the pathomechanism of these diseases. To understand how circulating or cardiac-derived EVs could affect myocardial functions, we analyzed the metabolomic profile of circulating EVs, and we performed an in-depth analysis of cardiomyocyte (CM)-derived EVs in HC. Circulating EVs were isolated with Vezics technology from male Wistar rats fed with high-cholesterol or control chow. AC16 human CMs were treated with Remembrane HC supplement and EVs were isolated from cell culture supernatant. The biophysical properties and the protein composition of CM EVs were analyzed. THP1-ASC-GFP cells were treated with CM EVs, and monocyte activation was measured. HC diet reduced the amount of certain phosphatidylcholines in circulating EVs, independently of their plasma level. HC treatment significantly increased EV secretion of CMs and greatly modified CM EV proteome, enriching several proteins involved in tissue remodeling. Regardless of the treatment, CM EVs did not induce the activation of THP1 monocytes. In conclusion, HC strongly affects the metabolome of circulating EVs and dysregulates CM EVs, which might contribute to HC-induced cardiac derangements.
Collapse
Affiliation(s)
- Csenger Kovácsházi
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Szabolcs Hambalkó
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Nabil V Sayour
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Tamás G Gergely
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Gábor B Brenner
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Csilla Pelyhe
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Dóra Kapui
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Bennet Y Weber
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | | | - Éva Pállinger
- Institute of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
| | - Edit I Buzás
- Institute of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
- ELKH-SE Translational Extracellular Vesicle Research Group, Budapest, Hungary
- HCEMM-SU Extracellular Vesicle Research Group, Budapest, Hungary
| | - Ádám Zolcsák
- Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
| | - Bálint Kiss
- Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
- HUNREN-SE Biophysical Virology Research Group, Budapest, Hungary
| | - Tamás Bozó
- Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
| | - Csilla Csányi
- Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
| | - Nikolett Kósa
- Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
| | - Miklós Kellermayer
- Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
- HUNREN-SE Biophysical Virology Research Group, Budapest, Hungary
| | - Róbert Farkas
- Department of Laboratory Medicine, Laboratory of Mass Spectrometry and Separation Technology, Semmelweis University, Budapest, Hungary
| | - Gellért B Karvaly
- Department of Laboratory Medicine, Laboratory of Mass Spectrometry and Separation Technology, Semmelweis University, Budapest, Hungary
| | - Kieran Wynne
- Systems Biology Ireland and School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
- UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
| | - David Matallanas
- Systems Biology Ireland and School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
| | - Zoltán Giricz
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary.
- Pharmahungary Group, Szeged, Hungary.
| |
Collapse
|
4
|
Zhang L, Xie F, Zhang F, Lu B. The potential roles of exosomes in pathological cardiomyocyte hypertrophy mechanisms and therapy: A review. Medicine (Baltimore) 2024; 103:e37994. [PMID: 38669371 PMCID: PMC11049793 DOI: 10.1097/md.0000000000037994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Accepted: 03/29/2024] [Indexed: 04/28/2024] Open
Abstract
Pathological cardiac hypertrophy, characterized by the enlargement of cardiac muscle cells, leads to serious cardiac conditions and stands as a major global health issue. Exosomes, comprising small lipid bilayer vesicles, are produced by various cell types and found in numerous bodily fluids. They play a pivotal role in intercellular communication by transferring bioactive cargos to recipient cells or activating signaling pathways in target cells. Exosomes from cardiomyocytes, endothelial cells, fibroblasts, and stem cells are key in regulating processes like cardiac hypertrophy, cardiomyocyte survival, apoptosis, fibrosis, and angiogenesis within the context of cardiovascular diseases. This review delves into exosomes' roles in pathological cardiac hypertrophy, first elucidating their impact on cell communication and signaling pathways. It then advances to discuss how exosomes affect key hypertrophic processes, including metabolism, fibrosis, oxidative stress, and angiogenesis. The review culminates by evaluating the potential of exosomes as biomarkers and their significance in targeted therapeutic strategies, thus emphasizing their critical role in the pathophysiology and management of cardiac hypertrophy.
Collapse
Affiliation(s)
- Lijun Zhang
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Fang Xie
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Fengmei Zhang
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Beiyao Lu
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
5
|
Dai S, Xu M, Pang Q, Sun J, Lin X, Chu X, Guo C, Xu J. Hypoxia macrophage-derived exosomal miR-26b-5p targeting PTEN promotes the development of keloids. BURNS & TRAUMA 2024; 12:tkad036. [PMID: 38434721 PMCID: PMC10905499 DOI: 10.1093/burnst/tkad036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Revised: 05/11/2023] [Accepted: 06/21/2023] [Indexed: 03/05/2024]
Abstract
Background Hypoxia is the typical characteristic of keloids. The development of keloids is closely related to the abnormal phenotypic transition of macrophages. However, the role of exosomal microRNAs (miRNAs) derived from hypoxic macrophages in keloids remains unclear. This study aimed to explore the role of hypoxic macrophage-derived exosomes (HMDE) in the occurrence and development of keloids and identify the critical miRNA. Methods The expression of CD206+ M2 macrophage in keloids and normal skin tissues was examined through immunofluorescence. The polarization of macrophages under a hypoxia environment was detected through flow cytometry. The internalization of macrophage-derived exosomes in human keloid fibroblasts (HKFs) was detected using a confocal microscope. miRNA sequencing was used to explore the differentially expressed miRNAs in exosomes derived from the normoxic and hypoxic macrophage. Subsequently, the dual-luciferase reporter assay verified that phosphatase and tension homolog (PTEN) was miR-26b-5p's target. The biological function of macrophage-derived exosomes, miR-26b-5p and PTEN were detected using the CCK-8, wound-healing and Transwell assays. Western blot assay was used to confirm the miR-26b-5p's underlying mechanisms and PTEN-PI3K/AKT pathway. Results We demonstrated that M2-type macrophages were enriched in keloids and that hypoxia treatment could polarize macrophages toward M2-type. Compared with normoxic macrophages-derived exosomes (NMDE), HMDE promote the proliferation, migration and invasion of HKFs. A total of 38 differential miRNAs (18 upregulated and 20 downregulated) were found between the NMDE and HMDE. miR-26b-5p was enriched in HMDE, which could be transmitted to HKFs. According to the results of the functional assay, exosomal miR-26b-5p produced by macrophages facilitated HKFs' migration, invasion and proliferation via the PTEN-PI3K/AKT pathway. Conclusions The highly expressed miR-26b-5p in HMDE promotes the development of keloids via the PTEN-PI3K/AKT pathway.
Collapse
Affiliation(s)
- Siya Dai
- Department of Plastic Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Shangcheng District, Hangzhou, China
| | - Mingyuan Xu
- Department of Plastic Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Shangcheng District, Hangzhou, China
| | - Qianqian Pang
- Department of Plastic Surgery, Ningbo Second Hospital, 41 Xibei Street, Ningbo, China
| | - Jiaqi Sun
- Department of Plastic Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Shangcheng District, Hangzhou, China
| | - Xiaohu Lin
- Department of Plastic and Reconstructive Surgery, Zhejiang Provincial People's Hospital, 158 Shangtang Road, Gongshu District, Hangzhou, China
| | - Xi Chu
- Department of Plastic Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Shangcheng District, Hangzhou, China
| | - Chunyi Guo
- Department of Plastic Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Shangcheng District, Hangzhou, China
| | - Jinghong Xu
- Department of Plastic Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Shangcheng District, Hangzhou, China
| |
Collapse
|
6
|
Zheng L, Ling W, Zhu D, Li Z, Li Y, Zhou H, Kong L. Roquin-1 resolves sepsis-associated acute liver injury by regulating inflammatory profiles via miRNA cargo in extracellular vesicles. iScience 2023; 26:107295. [PMID: 37554446 PMCID: PMC10405074 DOI: 10.1016/j.isci.2023.107295] [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: 05/29/2022] [Revised: 01/05/2023] [Accepted: 07/03/2023] [Indexed: 08/10/2023] Open
Abstract
Sepsis-associated acute liver injury (SALI) is an independent risk for sepsis-induced death orchestrated by innate and adaptive immune responses. Here, we found that Roquin-1 was decreased during SALI and expressed mainly in monocyte-derived macrophages. Meanwhile, Roquin-1 was correlated with the inflammatory profiles in humans and mice. Mechanically, Roquin-1 in macrophages promoted Ago2-K258-ubiquitination and inhibited Ago2-S387/S828-phosphorylation. Ago2-S387-phosphorylation inhibited Ago2-miRNA's complex location in multivesicular bodies and sorting in macrophages-derived extracellular vesicles (MDEVs), while Ago2-S828-phosphorylation modulated the binding between Ago2 and miRNAs by special miRNAs-motifs. Then, the anti-inflammatory miRNAs in MDEVs decreased TSC22D2 expression directly, upregulated Tregs-differentiation via TSC22D2-STAT3 signaling, and inhibited M1-macrophage-polarization by TSC22D2-AMPKα-mTOR pathway. Furthermore, WT MDEVs in mice alleviated SALI by increasing Tregs ratio and decreasing M1-macrophage frequency synchronously. Our study showed that Roquin-1 in macrophages increased Tregs-differentiation and decreased M1-macrophage-polarization simultaneously via miRNA in MDEVs, suggesting Roquin-1 can be used as a potential tool for SALI treatment and MDEVs engineering.
Collapse
Affiliation(s)
- Lei Zheng
- Hepatobiliary Center/Liver Transplantation Center, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, P.R. China
- Department of General Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao-tong University School of Medicine, 639 Zhizaoju Road, Shanghai 200011, P.R. China
| | - Wei Ling
- Hepatobiliary Center/Liver Transplantation Center, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, P.R. China
| | - Deming Zhu
- Hepatobiliary Center/Liver Transplantation Center, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, P.R. China
| | - Zhi Li
- Hepatobiliary Center/Liver Transplantation Center, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, P.R. China
| | - Yousheng Li
- Department of General Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao-tong University School of Medicine, 639 Zhizaoju Road, Shanghai 200011, P.R. China
| | - Haoming Zhou
- Hepatobiliary Center/Liver Transplantation Center, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, P.R. China
| | - Lianbao Kong
- Hepatobiliary Center/Liver Transplantation Center, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, P.R. China
| |
Collapse
|
7
|
Wang H, Ye X, Spanos M, Wang H, Yang Z, Li G, Xiao J, Zhou L. Exosomal Non-Coding RNA Mediates Macrophage Polarization: Roles in Cardiovascular Diseases. BIOLOGY 2023; 12:745. [PMID: 37237557 PMCID: PMC10215119 DOI: 10.3390/biology12050745] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 04/26/2023] [Accepted: 05/11/2023] [Indexed: 05/28/2023]
Abstract
Extracellular vesicles (EVs) or exosomes are nanosized extracellular particles that contain proteins, DNA, non-coding RNA (ncRNA) and other molecules, which are widely present in biofluids throughout the body. As a key mediator of intercellular communication, EVs transfer their cargoes to target cells and activate signaling transduction. Increasing evidence shows that ncRNA is involved in a variety of pathological and physiological processes through various pathways, particularly the inflammatory response. Macrophage, one of the body's "gatekeepers", plays a crucial role in inflammatory reactions. Generally, macrophages can be classified as pro-inflammatory type (M1) or anti-inflammatory type (M2) upon their phenotypes, a phenomenon termed macrophage polarization. Increasing evidence indicates that the polarization of macrophages plays important roles in the progression of cardiovascular diseases (CVD). However, the role of exosomal ncRNA in regulating macrophage polarization and the role of polarized macrophages as an important source of EV in CVD remains to be elucidated. In this review, we summarize the role and molecular mechanisms of exosomal-ncRNA in regulating macrophage polarization during CVD development, focusing on their cellular origins, functional cargo, and their detailed effects on macrophage polarization. We also discuss the role of polarized macrophages and their derived EV in CVD as well as the therapeutic prospects of exosomal ncRNA in the treatment of CVD.
Collapse
Affiliation(s)
- Hongyun Wang
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, 333 Nan Chen Road, Shanghai 200444, China
- Institute of Geriatrics, Affiliated Nantong Hospital of Shanghai University (The Sixth People’s Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China
| | - Xuan Ye
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, 333 Nan Chen Road, Shanghai 200444, China
- Department of Cardiology, First Affiliated Hospital of Nanjing Medical University, Nanjing 210008, China
| | - Michail Spanos
- Division of Cardiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Huanxin Wang
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, 333 Nan Chen Road, Shanghai 200444, China
| | - Zijiang Yang
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, 333 Nan Chen Road, Shanghai 200444, China
| | - Guoping Li
- Division of Cardiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Junjie Xiao
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, 333 Nan Chen Road, Shanghai 200444, China
- Institute of Geriatrics, Affiliated Nantong Hospital of Shanghai University (The Sixth People’s Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China
| | - Lei Zhou
- Department of Cardiology, First Affiliated Hospital of Nanjing Medical University, Nanjing 210008, China
| |
Collapse
|
8
|
Jiapaer Z, Li C, Yang X, Sun L, Chatterjee E, Zhang L, Lei J, Li G. Extracellular Non-Coding RNAs in Cardiovascular Diseases. Pharmaceutics 2023; 15:pharmaceutics15010155. [PMID: 36678784 PMCID: PMC9865796 DOI: 10.3390/pharmaceutics15010155] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 12/22/2022] [Accepted: 12/27/2022] [Indexed: 01/04/2023] Open
Abstract
Cardiovascular diseases (CVDs) remain the world's leading cause of death despite the best available healthcare and therapy. Emerging as a key mediator of intercellular and inter-organ communication in CVD pathogenesis, extracellular vesicles (EVs) are a heterogeneous group of membrane-enclosed nano-sized vesicles released by virtually all cells, of which their RNA cargo, especially non-coding RNAs (ncRNA), has been increasingly recognized as a promising diagnostic and therapeutic target. Recent evidence shows that ncRNAs, such as small ncRNAs, circular RNAs, and long ncRNAs, can be selectively sorted into EVs or other non-vesicular carriers and modulate various biological processes in recipient cells. In this review, we summarize recent advances in the literature regarding the origin, extracellular carrier, and functional mechanisms of extracellular ncRNAs with a focus on small ncRNAs, circular RNAs, and long ncRNAs. The pathophysiological roles of extracellular ncRNAs in various CVDs, including atherosclerosis, ischemic heart diseases, hypertension, cardiac hypertrophy, and heart failure, are extensively discussed. We also provide an update on recent developments and challenges in using extracellular ncRNAs as biomarkers or therapeutical targets in these CVDs.
Collapse
Affiliation(s)
- Zeyidan Jiapaer
- College of Life Science & Technology, Xinjiang University, Urumqi 830046, China
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, Urumqi 830046, China
| | - Chengyu Li
- College of Life Science & Technology, Xinjiang University, Urumqi 830046, China
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, Urumqi 830046, China
| | - Xinyu Yang
- Fangshan Hospital Beijing University of Chinese Medicine, Beijing 102400, China
| | - Lingfei Sun
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Emeli Chatterjee
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Lingying Zhang
- College of Life Science & Technology, Xinjiang University, Urumqi 830046, China
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, Urumqi 830046, China
| | - Ji Lei
- Center for Transplantation Science, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
- Correspondence: (J.L.); (G.L.)
| | - Guoping Li
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
- Correspondence: (J.L.); (G.L.)
| |
Collapse
|
9
|
Gao R, Li X. Extracellular Vesicles and Pathological Cardiac Hypertrophy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1418:17-31. [PMID: 37603270 DOI: 10.1007/978-981-99-1443-2_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/22/2023]
Abstract
Pathological cardiac hypertrophy is a well-recognized risk factor for cardiovascular diseases (CVDs). Although lots of efforts have been made to illustrate the underlying molecular mechanisms, many issues remain undiscovered. Recently, intercellular communication by delivering small molecules between different cell types in the progression of cardiac hypertrophy has been reported, including bioactive nucleic acids or proteins. These extracellular vesicles (EVs) may act in an autocrine or paracrine manner between cardiomyocytes and noncardiomyocytes to provoke or inhibit cardiac remodeling and hypertrophy. Besides, EVs can be used as novel diagnostic or prognostic biomarkers in cardiac hypertrophy and also may serve as potential therapeutic targets due to its biocompatible nature and low immunogenicity. In this chapter, we will first summarize the current knowledge about EVs from different cells in pathological cardiac hypertrophy. Then, we will focus on the value of EVs as therapeutic agents and biomarkers for pathological myocardial hypertrophy.
Collapse
Affiliation(s)
- Rongrong Gao
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xinli Li
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
10
|
Okamura A, Yoshioka Y, Saito Y, Ochiya T. Can Extracellular Vesicles as Drug Delivery Systems Be a Game Changer in Cardiac Disease? Pharm Res 2022; 40:889-908. [PMID: 36577860 PMCID: PMC10126064 DOI: 10.1007/s11095-022-03463-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 12/13/2022] [Indexed: 12/29/2022]
Abstract
Cardiac diseases such as myocardial infarction and heart failure have been the leading cause of death worldwide for more than 20 years, and new treatments continue to be investigated. Heart transplantation, a curative treatment for severe cardiac dysfunction, is available to only a small number of patients due to the rarity of donors and high costs. Cardiac regenerative medicine using embryonic stem cells and induced pluripotent stem cells is expected to be a new alternative to heart transplantation, but it has problems such as induction of immune response, tumor formation, and low survival rate of transplanted cells. On the other hand, there has been a focus on cell-free therapy using extracellular vesicles (EVs) due to their high biocompatibility and target specificity. Exosomes, one type of EV, play a role in the molecular transport system in vivo and can be considered a drug delivery system (DDS) innate to all living things. Exosomes contain nucleic acids and proteins, which are transported from secretory cells to recipient cells. Molecules in exosomes are encapsulated in a lipid bilayer, which allows them to exist stably in body fluids without being affected by nuclease degradation enzymes. Therefore, the therapeutic use of exosomes as DDSs has been widely explored and is being used in clinical trials and other clinical settings. This review summarizes the current topics of EVs as DDSs in cardiac disease.
Collapse
Affiliation(s)
- Akihiko Okamura
- Department of Molecular and Cellular Medicine, Institute of Medical Science, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-Ku, Tokyo, 160-0023, Japan.,Department of Cardiovascular Medicine, Nara Medical University, 840 Shijo-Cho, Kashihara, Nara, 634-8522, Japan
| | - Yusuke Yoshioka
- Department of Molecular and Cellular Medicine, Institute of Medical Science, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-Ku, Tokyo, 160-0023, Japan
| | - Yoshihiko Saito
- Department of Cardiovascular Medicine, Nara Medical University, 840 Shijo-Cho, Kashihara, Nara, 634-8522, Japan
| | - Takahiro Ochiya
- Department of Molecular and Cellular Medicine, Institute of Medical Science, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-Ku, Tokyo, 160-0023, Japan.
| |
Collapse
|
11
|
Sun Z, Liu X, Lu M, Zhang X, Sun J. Serum-derived exosomes induce proinflammatory cytokines production in Cynoglossus semilaevis via miR-133-3p. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2022; 136:104497. [PMID: 35921922 DOI: 10.1016/j.dci.2022.104497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 07/18/2022] [Accepted: 07/18/2022] [Indexed: 06/15/2023]
Abstract
Exosomes are small membrane-enclosed vesicles secreted by various types of cells. In mammals, a wide range of physiological and pathological functions have been confirmed and attributed to EVs carrying a variety of molecular cargoes, including miRNAs. However, studies on the biological functions and related molecular mechanisms of serum exosomes isolated from teleost fish are limited. Indeed, the molecular mechanisms underlying the effects of serum exosomes on immune responses and inflammatory processes are unknown. Chinese tongue sole (Cynoglossus semilaevis) is an economically important species used widely in industrial aquaculture. Vibrio harveyi, a common bacterial pathogen that infects C. semilaevis and some other fish, causes excessive inflammatory reactions, which are characterized by skin ulceration. Here, we isolated serum-derived exosomes from C. semilaevis and investigated their effects on inflammatory processes following V. harveyi infection. We found that compared with uninfected fish, exosome abundance in infected fish blood increased with bacterial infection time, while expression of TNF-α increased, and that of IL-10 decreased, significantly. Moreover, artificial infection studies demonstrated that injection of serum exosomes isolated from infected fish increased expression of TNF-α, IL-6, and IL-8, which is consistent with the increase in proinflammatory cytokines induced by V. harveyi infection. To further investigate the mechanisms by which exosomes increase proinflammatory cytokine production, we performed miRNA expression profiling and found that 26 differentially expressed miRNAs were associated with bacterial infection and immune responses; of these, miR-133-3p was considerably more abundant in serum exosomes from infected fish. Bioinformatics analysis suggested that miR-133-3p inhibits NF-κB signaling pathways by targeting PP2A and affecting cytokine release. We also found that miR-133-3p increased expression of TNF-α, IL-6, and IL-8 in fish blood and kidney, whereas an miR-133-3p inhibitor showed the opposite results. Thus, the data suggest that serum exosomes participate in innate immunity in teleost fish by promoting inflammatory responses to bacterial infection. Exosome-mediated transfer of miR-133-3p increases expression of proinflammatory cytokines in C. semilaevis, resulting in excessive inflammatory responses during V. harveyi infection. These data may lead to development of methods and strategies that control skin ulceration in Chinese tongue sole.
Collapse
Affiliation(s)
- Zhanpeng Sun
- Tianjin Key Laboratory of Animal and Plant Resistance, College of Life Sciences, Tianjin Normal University, Tianjin, China; Faculty of Education, Tianjin Normal University, Tianjin, China.
| | - Xiaozhu Liu
- Tianjin Key Laboratory of Animal and Plant Resistance, College of Life Sciences, Tianjin Normal University, Tianjin, China.
| | - Meiyi Lu
- Tianjin Key Laboratory of Animal and Plant Resistance, College of Life Sciences, Tianjin Normal University, Tianjin, China.
| | - Xiao Zhang
- Tianjin Key Laboratory of Animal and Plant Resistance, College of Life Sciences, Tianjin Normal University, Tianjin, China.
| | - Jinsheng Sun
- Tianjin Key Laboratory of Animal and Plant Resistance, College of Life Sciences, Tianjin Normal University, Tianjin, China.
| |
Collapse
|
12
|
Wang Y, Zhu J, Chen J, Xu R, Groth T, Wan H, Zhou G. The Signaling Pathways Induced by Exosomes in Promoting Diabetic Wound Healing: A Mini-Review. Curr Issues Mol Biol 2022; 44:4960-4976. [PMID: 36286052 PMCID: PMC9600352 DOI: 10.3390/cimb44100337] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 10/08/2022] [Accepted: 10/13/2022] [Indexed: 11/16/2022] Open
Abstract
Impaired healing of diabetic wounds harms patients' quality of life and even leads to disability and death, which is an urgent issue to be solved clinically. Despite the great progress that has been achieved, it remains a worldwide challenge to develop effective therapeutic treatments for diabetic wounds. Recently, exosomes have attracted special attention because they can be involved in immune response, antigen presentation, cell migration, cell differentiation, tumor invasion and other processes. Meanwhile, exosomes have been proven to hold great potential in the treatment of diabetic wounds. Mechanistic studies of exosomes based on signaling pathways could not only help to uncover the mechanisms by which exosomes promote diabetic wound healing but could also provide a theoretical basis for the clinical application of exosomes. Herein, our mini-review aims to summarize the progress of research on the use of various exosomes derived from different cell types to promote diabetic wound healing, with a focus on the classical signaling pathways, including PI3K/Akt, Wnt, NF-κB, MAPK, Notch, Nrf2, HIF-1α/VEGF and TGF-β/Smad. The results show that exosomes could regulate these signaling pathways to down-regulate inflammation, reduce oxidative stress, increase angiogenesis, promote fibroblast proliferation, induce re-epithelization and inhibit scar formation, making exosomes attractive candidates for the treatment of diabetic wounds.
Collapse
Affiliation(s)
- Yanying Wang
- The Second Clinical Medical College, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou 310053, China
| | - Jiayan Zhu
- College of Life Science, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou 310053, China
| | - Jing Chen
- College of Life Science, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou 310053, China
| | - Ruojiao Xu
- College of Life Science, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou 310053, China
| | - Thomas Groth
- Department Biomedical Materials, Institute of Pharmacy, Martin Luther University Halle-Wittenberg, D-06099 Halle (Saale), Germany
| | - Haitong Wan
- College of Life Science, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou 310053, China
- Correspondence: (H.W.); (G.Z.)
| | - Guoying Zhou
- The Second Clinical Medical College, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou 310053, China
- College of Life Science, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou 310053, China
- Correspondence: (H.W.); (G.Z.)
| |
Collapse
|
13
|
Fang J, Zhang Y, Chen D, Zheng Y, Jiang J. Exosomes and Exosomal Cargos: A Promising World for Ventricular Remodeling Following Myocardial Infarction. Int J Nanomedicine 2022; 17:4699-4719. [PMID: 36217495 PMCID: PMC9547598 DOI: 10.2147/ijn.s377479] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Accepted: 09/21/2022] [Indexed: 11/23/2022] Open
Abstract
Exosomes are a pluripotent group of extracellular nanovesicles secreted by all cells that mediate intercellular communications. The effective information within exosomes is primarily reflected in exosomal cargos, including proteins, lipids, DNAs, and non-coding RNAs (ncRNAs), the most intensively studied molecules. Cardiac resident cells (cardiomyocytes, fibroblasts, and endothelial cells) and foreign cells (infiltrated immune cells, cardiac progenitor cells, cardiosphere-derived cells, and mesenchymal stem cells) are involved in the progress of ventricular remodeling (VR) following myocardial infarction (MI) via transferring exosomes into target cells. Here, we summarize the pathological mechanisms of VR following MI, including cardiac myocyte hypertrophy, cardiac fibrosis, inflammation, pyroptosis, apoptosis, autophagy, angiogenesis, and metabolic disorders, and the roles of exosomal cargos in these processes, with a focus on proteins and ncRNAs. Continued research in this field reveals a novel diagnostic and therapeutic strategy for VR.
Collapse
Affiliation(s)
- Jiacheng Fang
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, People’s Republic of China
| | - Yuxuan Zhang
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, People’s Republic of China
| | - Delong Chen
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, People’s Republic of China
| | - Yiyue Zheng
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, People’s Republic of China
| | - Jun Jiang
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, People’s Republic of China,Correspondence: Jun Jiang, Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, No. 88 Jiefang Road, Hangzhou, Zhejiang, 310009, People’s Republic of China, Tel/Fax +86 135 8870 6891, Email
| |
Collapse
|
14
|
Extracellular Vesicles, Inflammation, and Cardiovascular Disease. Cells 2022; 11:cells11142229. [PMID: 35883672 PMCID: PMC9320258 DOI: 10.3390/cells11142229] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 07/11/2022] [Accepted: 07/12/2022] [Indexed: 12/15/2022] Open
Abstract
Cardiovascular disease is a leading cause of death worldwide. The underlying mechanisms of most cardiovascular disorders involve innate and adaptive immune responses, and extracellular vesicles are implicated in both. In this review, we describe the mechanistic role of extracellular vesicles at the intersection of inflammatory processes and cardiovascular disease. Our discussion focuses on atherosclerosis, myocardial ischemia and ischemic heart disease, heart failure, aortic aneurysms, and valvular pathology.
Collapse
|
15
|
Lee S, Ko JH, Kim SN. The Extracellular MicroRNAs on Inflammation: A Literature Review of Rodent Studies. Biomedicines 2022; 10:1601. [PMID: 35884901 PMCID: PMC9312877 DOI: 10.3390/biomedicines10071601] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/03/2022] [Accepted: 07/03/2022] [Indexed: 11/17/2022] Open
Abstract
Inflammation is an indispensable biological process stimulated by infection and injuries. Inflammatory mechanisms related to extracellular vesicles (EVs), which are small membrane structures carrying various molecules, were summarized in this review. Emerging evidence from animal studies has highlighted the role of EVs in modulating inflammatory responses, by transporting various molecules involved in host defense. In this review, we have discussed the role of EV miRNAs in inflammation. Rodent studies associated with extracellular miRNAs in inflammatory diseases, published from 2012 to 2022, were explored from PUBMED, EMBASE, and MEDLINE. A total of 95 studies were reviewed. In summary, EV-associated miRNAs play a key role in various diseases, including organ injury, immune dysfunction, neurological disease, metabolic syndrome, vesicular disease, arthritis, cancer, and other inflammatory diseases. Diverse EV-associated miRNAs regulate inflammasome activation and pro- and anti-inflammatory cytokine levels by targeting genes.
Collapse
Affiliation(s)
- Seri Lee
- College of Korean Medicine, Dongguk University, Goyang 10326, Korea; (S.L.); (J.H.K.)
- Graduate School, Dongguk University, Seoul 04620, Korea
| | - Jade Heejae Ko
- College of Korean Medicine, Dongguk University, Goyang 10326, Korea; (S.L.); (J.H.K.)
| | - Seung-Nam Kim
- College of Korean Medicine, Dongguk University, Goyang 10326, Korea; (S.L.); (J.H.K.)
| |
Collapse
|
16
|
Jia G, Liang C, Li W, Dai H. MiR-410-3p facilitates Angiotensin II-induced cardiac hypertrophy by targeting Smad7. Bioengineered 2021; 13:119-127. [PMID: 34951337 PMCID: PMC8805929 DOI: 10.1080/21655979.2021.2009968] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
MicroRNAs (miRNAs) have emerged as important regulators in the development of cardiovascular diseases. miR-410-3p was shown to play a protective or detrimental role in the progression in cardiovascular events. However, the exact role and the underlying mechanism of miR-410-3p in cardiac hypertrophy have not been documented. The current work was aimed to determine the role and underlying mechanism of miR-410-3p on Angiotensin II (Ang II) induced cardiac hypertrophy. FITC-phalloidin staining was used for determination of cardiomyocyte surface area. Quantitative reverse transcription polymerase chain reaction (qRT-PCR) was performed to identify mRNA expression level of hypertrophic markers. Smad7 protein expression level was analyzed using Western blot. Dual-luciferase reporter assay was used to examine the regulatory function of miR-410-3p on Smad7. MiR-410-3p was found significantly up-regulated in Ang II–induced cardiac hypertrophy. MiR-410-3p inhibitor remarkably alleviated cardiomyocyte hypertrophic changes. Dual-luciferase reporter assay result indicated that miR-410-3p directly targeted Smad7 and miR-410-3p inhibitor effectively prevented Ang II triggered down-regulation of Smad7. Moreover, Smad7 overexpression significantly reversed the pro-hypertrophic effect of miR-410-3p. In summary, our findings revealed that miR-410-3p mediated Ang II–induced cardiac hypertrophy via targeting inhibition of Smad7.
Collapse
Affiliation(s)
- Guizhi Jia
- Department of Physiology, Jinzhou Medical University, Jinzhou, Liaoning, People's Republic of China
| | - Chunguang Liang
- School of Nursing, Jinzhou Medical University, Jinzhou, Liaoning, People's Republic of China
| | - Wenhui Li
- Experimental Teaching Center of Basic Medicine, Jinzhou Medical University, Jinzhou, Liaoning, People's Republic of China
| | - Hongliang Dai
- School of Nursing, Jinzhou Medical University, Jinzhou, Liaoning, People's Republic of China
| |
Collapse
|
17
|
Boucher AC, Caldwell KJ, Crispino JD, Flerlage JE. Clinical and biological aspects of myeloid leukemia in Down syndrome. Leukemia 2021; 35:3352-3360. [PMID: 34518645 PMCID: PMC8639661 DOI: 10.1038/s41375-021-01414-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 08/30/2021] [Accepted: 09/01/2021] [Indexed: 02/08/2023]
Abstract
Children with Down syndrome are at an elevated risk of leukemia, especially myeloid leukemia (ML-DS). This malignancy is frequently preceded by transient abnormal myelopoiesis (TAM), which is self-limited expansion of fetal liver-derived megakaryocyte progenitors. An array of international studies has led to consensus in treating ML-DS with reduced-intensity chemotherapy, leading to excellent outcomes. In addition, studies performed in the past 20 years have revealed many of the genetic and epigenetic features of the tumors, including GATA1 mutations that are arguably associated with all cases of both TAM and ML-DS. Despite these advances in understanding the clinical and biological aspects of ML-DS, little is known about the mechanisms of relapse. Upon relapse, patients face a poor outcome, and there is no consensus on treatment. Future studies need to be focused on this challenging aspect of leukemia in children with DS.
Collapse
Affiliation(s)
- Austin C Boucher
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Kenneth J Caldwell
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - John D Crispino
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA.
| | - Jamie E Flerlage
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA.
| |
Collapse
|
18
|
Roles of Exosomes in Cardiac Fibroblast Activation and Fibrosis. Cells 2021; 10:cells10112933. [PMID: 34831158 PMCID: PMC8616203 DOI: 10.3390/cells10112933] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 10/22/2021] [Accepted: 10/26/2021] [Indexed: 12/23/2022] Open
Abstract
Alterations in the accumulation and composition of the extracellular matrix are part of the normal tissue repair process. During fibrosis, this process becomes dysregulated and excessive extracellular matrix alters the biomechanical properties and function of tissues involved. Historically fibrosis was thought to be progressive and irreversible; however, studies suggest that fibrosis is a dynamic process whose progression can be stopped and even reversed. This realization has led to an enhanced pursuit of therapeutic agents targeting fibrosis and extracellular matrix-producing cells. In many organs, fibroblasts are the primary cells that produce the extracellular matrix. In response to diverse mechanical and biochemical stimuli, these cells are activated or transdifferentiate into specialized cells termed myofibroblasts that have an enhanced capacity to produce extracellular matrix. It is clear that interactions between diverse cells of the heart are able to modulate fibroblast activation and fibrosis. Exosomes are a form of extracellular vesicle that play an important role in intercellular communication via the cargo that they deliver to target cells. While relatively recently discovered, exosomes have been demonstrated to play important positive and negative roles in the regulation of fibroblast activation and tissue fibrosis. These roles as well as efforts to engineer exosomes as therapeutic tools will be discussed.
Collapse
|
19
|
Mosquera-Heredia MI, Morales LC, Vidal OM, Barceló E, Silvera-Redondo C, Vélez JI, Garavito-Galofre P. Exosomes: Potential Disease Biomarkers and New Therapeutic Targets. Biomedicines 2021; 9:1061. [PMID: 34440265 PMCID: PMC8393483 DOI: 10.3390/biomedicines9081061] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 08/10/2021] [Accepted: 08/11/2021] [Indexed: 02/07/2023] Open
Abstract
Exosomes are extracellular vesicles released by cells, both constitutively and after cell activation, and are present in different types of biological fluid. Exosomes are involved in the pathogenesis of diseases, such as cancer, neurodegenerative diseases, pregnancy disorders and cardiovascular diseases, and have emerged as potential non-invasive biomarkers for the detection, prognosis and therapeutics of a myriad of diseases. In this review, we describe recent advances related to the regulatory mechanisms of exosome biogenesis, release and molecular composition, as well as their role in health and disease, and their potential use as disease biomarkers and therapeutic targets. In addition, the advantages and disadvantages of their main isolation methods, characterization and cargo analysis, as well as the experimental methods used for exosome-mediated drug delivery, are discussed. Finally, we present potential perspectives for the use of exosomes in future clinical practice.
Collapse
Affiliation(s)
- Maria I. Mosquera-Heredia
- Department of Medicine, Universidad del Norte, Barranquilla 081007, Colombia; (L.C.M.); (O.M.V.); (C.S.-R.)
| | - Luis C. Morales
- Department of Medicine, Universidad del Norte, Barranquilla 081007, Colombia; (L.C.M.); (O.M.V.); (C.S.-R.)
| | - Oscar M. Vidal
- Department of Medicine, Universidad del Norte, Barranquilla 081007, Colombia; (L.C.M.); (O.M.V.); (C.S.-R.)
| | - Ernesto Barceló
- Instituto Colombiano de Neuropedagogía, Barranquilla 080020, Colombia;
| | - Carlos Silvera-Redondo
- Department of Medicine, Universidad del Norte, Barranquilla 081007, Colombia; (L.C.M.); (O.M.V.); (C.S.-R.)
| | - Jorge I. Vélez
- Department of Industrial Engineering, Universidad del Norte, Barranquilla 081007, Colombia;
| | - Pilar Garavito-Galofre
- Department of Medicine, Universidad del Norte, Barranquilla 081007, Colombia; (L.C.M.); (O.M.V.); (C.S.-R.)
| |
Collapse
|
20
|
The Impact of microRNAs in Renin-Angiotensin-System-Induced Cardiac Remodelling. Int J Mol Sci 2021; 22:ijms22094762. [PMID: 33946230 PMCID: PMC8124994 DOI: 10.3390/ijms22094762] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 04/27/2021] [Accepted: 04/27/2021] [Indexed: 02/06/2023] Open
Abstract
Current knowledge on the renin-angiotensin system (RAS) indicates its central role in the pathogenesis of cardiovascular remodelling via both hemodynamic alterations and direct growth and the proliferation effects of angiotensin II or aldosterone resulting in the hypertrophy of cardiomyocytes, the proliferation of fibroblasts, and inflammatory immune cell activation. The noncoding regulatory microRNAs has recently emerged as a completely novel approach to the study of the RAS. A growing number of microRNAs serve as mediators and/or regulators of RAS-induced cardiac remodelling by directly targeting RAS enzymes, receptors, signalling molecules, or inhibitors of signalling pathways. Specifically, microRNAs that directly modulate pro-hypertrophic, pro-fibrotic and pro-inflammatory signalling initiated by angiotensin II receptor type 1 (AT1R) stimulation are of particular relevance in mediating the cardiovascular effects of the RAS. The aim of this review is to summarize the current knowledge in the field that is still in the early stage of preclinical investigation with occasionally conflicting reports. Understanding the big picture of microRNAs not only aids in the improved understanding of cardiac response to injury but also leads to better therapeutic strategies utilizing microRNAs as biomarkers, therapeutic agents and pharmacological targets.
Collapse
|
21
|
Adipocyte, Immune Cells, and miRNA Crosstalk: A Novel Regulator of Metabolic Dysfunction and Obesity. Cells 2021; 10:cells10051004. [PMID: 33923175 PMCID: PMC8147115 DOI: 10.3390/cells10051004] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 04/20/2021] [Accepted: 04/23/2021] [Indexed: 02/06/2023] Open
Abstract
Obesity is characterized as a complex and multifactorial excess accretion of adipose tissue (AT) accompanied with alterations in the immune response that affects virtually all age and socioeconomic groups around the globe. The abnormal accumulation of AT leads to several metabolic diseases, including nonalcoholic fatty liver disorder (NAFLD), low-grade inflammation, type 2 diabetes mellitus (T2DM), cardiovascular disorders (CVDs), and cancer. AT is an endocrine organ composed of adipocytes and immune cells, including B-Cells, T-cells and macrophages. These immune cells secrete various cytokines and chemokines and crosstalk with adipokines to maintain metabolic homeostasis and low-grade chronic inflammation. A novel form of adipokines, microRNA (miRs), is expressed in many developing peripheral tissues, including ATs, T-cells, and macrophages, and modulates the immune response. miRs are essential for insulin resistance, maintaining the tumor microenvironment, and obesity-associated inflammation (OAI). The abnormal regulation of AT, T-cells, and macrophage miRs may change the function of different organs including the pancreas, heart, liver, and skeletal muscle. Since obesity and inflammation are closely associated, the dysregulated expression of miRs in inflammatory adipocytes, T-cells, and macrophages suggest the importance of miRs in OAI. Therefore, in this review article, we have elaborated the role of miRs as epigenetic regulators affecting adipocyte differentiation, immune response, AT browning, adipogenesis, lipid metabolism, insulin resistance (IR), glucose homeostasis, obesity, and metabolic disorders. Further, we will discuss a set of altered miRs as novel biomarkers for metabolic disease progression and therapeutic targets for obesity.
Collapse
|