1
|
Banikarimi SP, Mellati A, Abasi M, Soleimani M, Ghiass MA, Ahmadi Tafti SH, Boroumand S, Hasanzadeh E. Cardiac tissue regeneration by microfluidic generated cardiac cell-laden calcium alginate microgels and mesenchymal stem cell extracted exosomes on myocardial infarction model. Int J Biol Macromol 2024; 292:139247. [PMID: 39733869 DOI: 10.1016/j.ijbiomac.2024.139247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 12/19/2024] [Accepted: 12/25/2024] [Indexed: 12/31/2024]
Abstract
Regenerative medicine is one of the effective approaches for myocardial infarcted (MI) tissue due to the low capacity of heart for regeneration. However, cell therapy with local administration has shown poor cell retention in the targeted area and limited engraftment capacity at the intended location, resulting in inadequate tissue regeneration. The present study involves mesenchymal stem cell-derived exosomes and encapsulated cells in small and injectable calcium alginate microgels by a specialized microfluidic device to decrease inflammation and increase cell retention in the infarcted tissue. The results have shown that our microfluidic system can produce monodisperse cardiac cell-laden alginate microgels within the size range of <100 μm that are easily injectable. Our in vivo findings on the MI rat model demonstrated that the combination of cardiac cell-laden calcium alginate microgels with mesenchymal stem cells derived exosomes resulted in a higher increase in echocardiography, heart-specific gene expressions, and cardiac markers results compared to the other groups. However, the administration of exosomes or cardiac cells separately has shown a small amount of regeneration. Encapsulating cardiac cells of specific sizes along with exosomes produced from mesenchymal stem cells can be potentially applied as an effective method for regenerating the myocardium following infarction.
Collapse
Affiliation(s)
- Seyedeh Parnian Banikarimi
- Department of Tissue Engineering & Regenerative Medicine, School of Advanced Technologies in Medicine, Mazandaran University of Medical Sciences, Sari, Iran; Student Research Committee, School of Advanced Technologies in Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Amir Mellati
- Department of Tissue Engineering & Regenerative Medicine, School of Advanced Technologies in Medicine, Mazandaran University of Medical Sciences, Sari, Iran; Molecular and Cell Biology Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mozhgan Abasi
- Department of Tissue Engineering & Regenerative Medicine, School of Advanced Technologies in Medicine, Mazandaran University of Medical Sciences, Sari, Iran; Immunogenetics Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Masoud Soleimani
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Adel Ghiass
- Tissue Engineering Department, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Seyed Hossein Ahmadi Tafti
- Research Center for Advanced Technologies in Cardiovascular Medicine, Cardiovascular Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Safieh Boroumand
- Research Center for Advanced Technologies in Cardiovascular Medicine, Cardiovascular Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Elham Hasanzadeh
- Department of Tissue Engineering & Regenerative Medicine, School of Advanced Technologies in Medicine, Mazandaran University of Medical Sciences, Sari, Iran; Molecular and Cell Biology Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran; Immunogenetics Research Center, Mazandaran University of Medical Sciences, Sari, Iran.
| |
Collapse
|
2
|
Samiminemati A, Shahzaib M, Moriello C, Alessio N, Aprile D, Squillaro T, Di Bernardo G, Galderisi U. Mesenchymal Stromal Cell (MSC) Isolation and Induction of Acute and Replicative Senescence. Methods Mol Biol 2024. [PMID: 39714585 DOI: 10.1007/7651_2024_580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Mesenchymal stromal cells (MSCs) are a heterogeneous population of non-hematopoietic adult stem cells derived from the embryonic mesoderm. They possess self-renewal and multipotent differentiation capabilities, allowing them to give rise to mesodermal cell types, such as osteoblasts, chondroblasts, and adipocytes, as well as non-mesodermal cells, including neuron-like cells and endothelial cells. MSCs play a vital role in maintaining homeostasis across various tissues by facilitating tissue repair, immune regulation, and inflammatory response balance. Initially identified in bone marrow, MSCs have since been found in multiple tissues, including muscle, adipose tissue, and dental pulp, and are characterized by specific surface markers and differentiation abilities.Aging induces cellular senescence, an irreversible growth arrest linked to various stressors, which has significant implications for regenerative medicine. While initially viewed as a protective mechanism against tumorigenesis, the accumulation of senescent cells, particularly in MSCs, leads to age-related diseases through the senescence-associated secretory phenotype (SASP). The onset of senescence in MSCs diminishes their therapeutic potential and contributes to homeostatic imbalance. Key drivers of MSC senescence include genetic damage, noncoding RNA, and mitochondrial dysfunction, among others.This study outlines the principal methodologies for the isolation and characterization of MSCs, alongside techniques to induce acute senescence via hydrogen peroxide or irradiation, as well as replicative senescence, to investigate senescence-related changes in vitro. Understanding the mechanisms of MSC senescence will provide critical insights into the molecular pathways of aging and pave way for advancements in cellular therapies targeting age-related diseases.
Collapse
Affiliation(s)
- Afshin Samiminemati
- Department of Experimental Medicine, Biotechnology, and Molecular Biology Section, Luigi Vanvitelli Campania University, Naples, Italy
| | - Mohd Shahzaib
- Department of Experimental Medicine, Biotechnology, and Molecular Biology Section, Luigi Vanvitelli Campania University, Naples, Italy
| | - Claudia Moriello
- Department of Experimental Medicine, Biotechnology, and Molecular Biology Section, Luigi Vanvitelli Campania University, Naples, Italy
| | - Nicola Alessio
- Department of Experimental Medicine, Biotechnology, and Molecular Biology Section, Luigi Vanvitelli Campania University, Naples, Italy
| | - Domenico Aprile
- Department of Experimental Medicine, Biotechnology, and Molecular Biology Section, Luigi Vanvitelli Campania University, Naples, Italy
| | - Tiziana Squillaro
- Department of Life Sciences, Health and Health Professions, Link Campus University, Rome, Italy
| | - Giovanni Di Bernardo
- Department of Experimental Medicine, Biotechnology, and Molecular Biology Section, Luigi Vanvitelli Campania University, Naples, Italy
- Sbarro Health Research Organization, Temple University, Philadelphia, PA, USA
| | - Umberto Galderisi
- Department of Experimental Medicine, Biotechnology, and Molecular Biology Section, Luigi Vanvitelli Campania University, Naples, Italy.
- Sbarro Health Research Organization, Temple University, Philadelphia, PA, USA.
- Genome and Stem Cell Center (GENKÖK), Erciyes University, Kayseri, Turkey.
| |
Collapse
|
3
|
Razavi ZS, Farokhi S, Mahmoudvand G, Karimi-Rouzbahani A, Farasati-Far B, Tahmasebi-Ghorabi S, Pazoki-Toroudi H, Saadat-Fakhr M, Afkhami H. Stem cells and bio scaffolds for the treatment of cardiovascular diseases: new insights. Front Cell Dev Biol 2024; 12:1472103. [PMID: 39726717 PMCID: PMC11669526 DOI: 10.3389/fcell.2024.1472103] [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: 07/28/2024] [Accepted: 10/01/2024] [Indexed: 12/28/2024] Open
Abstract
Mortality and morbidity from cardiovascular diseases are common worldwide. In order to improve survival and quality of life for this patient population, extensive efforts are being made to establish effective therapeutic modalities. New treatment options are needed, it seems. In addition to treating cardiovascular diseases, cell therapy is one of the most promising medical platforms. One of the most effective therapeutic approaches in this area is stem cell therapy. In stem cell biology, multipotent stem cells and pluripotent stem cells are divided into two types. There is evidence that stem cell therapy could be used as a therapeutic approach for cardiovascular diseases based on multiple lines of evidence. The effectiveness of stem cell therapies in humans has been studied in several clinical trials. In spite of the challenges associated with stem cell therapy, it appears that resolving them may lead to stem cells being used in cardiovascular disease patients. This may be an effective therapeutic approach. By mounting these stem cells on biological scaffolds, their effect can be enhanced.
Collapse
Affiliation(s)
- Zahra Sadat Razavi
- Physiology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Simin Farokhi
- Student Research Committee, USERN Office, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Golnaz Mahmoudvand
- Student Research Committee, USERN Office, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Arian Karimi-Rouzbahani
- Student Research Committee, USERN Office, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Bahareh Farasati-Far
- Department of Chemistry, Iran University of Science and Technology, Tehran, Iran
| | - Samaneh Tahmasebi-Ghorabi
- Master of Health Education, Research Expert, Clinical Research Development Unit, Emam Khomeini Hospital, Ilam University of Medical Sciences, Ilam, Iran
| | | | - Masoud Saadat-Fakhr
- Faculty of Medicine, Tehran Medical Sciences Branch, Islamic Azad University, Tehran, Iran
| | - Hamed Afkhami
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran
- Department of Medical Microbiology, Faculty of Medicine, Shahed University, Tehran, Iran
| |
Collapse
|
4
|
Wang S, Wang Z, Shen Z, Zhang M, Jin D, Zheng K, Liu X, Chai M, Wang Z, Chi A, Ostrovidov S, Wu H, Shao D, Liu G, Wu K, Leong KW, Shi X. Magnetic soft microrobots for erectile dysfunction therapy. Proc Natl Acad Sci U S A 2024; 121:e2407809121. [PMID: 39556757 PMCID: PMC11626158 DOI: 10.1073/pnas.2407809121] [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: 04/24/2024] [Accepted: 10/08/2024] [Indexed: 11/20/2024] Open
Abstract
Erectile dysfunction (ED) is a major threat to male fertility and quality of life, and mesenchymal stromal cells (MSCs) are a promising therapeutic option. However, therapeutic outcomes are compromised by low MSC retention and survival rates in corpus cavernosum tissue. Here, we developed an innovative magnetic soft microrobot comprising an ultrasoft hydrogel microsphere embedded with a magnetic nanoparticle chain for MSC delivery. This design also features phenylboronic acid groups for scavenging reactive oxygen species (ROS). With a Young's modulus of less than 1 kPa, the ultrasoft microrobot adapts its shape within narrow blood vessels, ensuring a uniform distribution of MSCs within the corpus cavernosum. Our findings showed that compared with traditional MSC injections, the MSC delivery microrobot (MSC-Rob) significantly enhanced MSC retention and survival. In both rat and beagle ED models, MSC-Rob treatment accelerated the repair of corpus cavernosum tissue and restored erectile function. Single-cell RNA sequencing (scRNA-seq) revealed that MSC-Rob treatment facilitates nerve and blood vessel regeneration in the corpus cavernosum by increasing the presence of regenerative macrophages. Overall, our MSC-Rob not only advances the clinical application of MSCs for ED therapy but also broadens the scope of microrobots for other cell therapies.
Collapse
Affiliation(s)
- Shuting Wang
- National Engineering Research Centre for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou510006, P. R. China
- School of Materials Science and Engineering, South China University of Technology, Guangzhou510640, P. R. China
- Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou510006, P. R. China
| | - Zhenqing Wang
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan250021, P. R. China
| | - Zongshan Shen
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou510055, P. R. China
| | - Min Zhang
- Department of Andrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou510080, P. R. China
| | - Dongdong Jin
- School of Materials Science and Engineering, Harbin Institute of Technology, Shenzhen518071, P. R. China
| | - Ke Zheng
- School of Materials Science and Engineering, Dongguan University of Technology, Dongguan523808, P. R. China
| | - Xuemin Liu
- National Engineering Research Centre for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou510006, P. R. China
| | - Muyuan Chai
- National Engineering Research Centre for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou510006, P. R. China
| | - Zhenxing Wang
- National Engineering Research Centre for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou510006, P. R. China
| | - Ani Chi
- National Engineering Research Centre for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou510006, P. R. China
| | - Serge Ostrovidov
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Chiyoda-ku113-8510, Tokyo, Japan
| | - Hongkai Wu
- Department of Chemistry, The Hong Kong University of Science and Technology, Hong Kong999077, P. R. China
| | - Dan Shao
- National Engineering Research Centre for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou510006, P. R. China
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou511442, P. R. China
- Department of Biomedical Engineering Columbia University, New York, NY10032
| | - Guihua Liu
- Reproductive Centre The Sixth Affiliated Hospital Sun Yat-sen University, Guangzhou510655, P. R. China
| | - Kai Wu
- National Engineering Research Centre for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou510006, P. R. China
| | - Kam W. Leong
- National Engineering Research Centre for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou510006, P. R. China
| | - Xuetao Shi
- National Engineering Research Centre for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou510006, P. R. China
- School of Materials Science and Engineering, South China University of Technology, Guangzhou510640, P. R. China
- Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou510006, P. R. China
| |
Collapse
|
5
|
Kamar SA, Naiem Hamdy K, El-Nefiawy NE, Mohammed H, Fetouh MA. Exploring Mesenchymal Stem Cells versus Minoxidil for Androgenic Alopecia Treatment: A Detailed Animal-Based Histological and Morphometric Study. Cells Tissues Organs 2024:1-17. [PMID: 39527940 DOI: 10.1159/000542547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 11/08/2024] [Indexed: 11/16/2024] Open
Abstract
INTRODUCTION Androgenic alopecia (AGA), a hair loss condition caused by dihydrotestosterone binding to hair follicle receptors, negatively impacts quality of life for both men and women. Current treatments like minoxidil and finasteride have limitations, highlighting the need for alternative therapies, such as human umbilical cord blood-derived mesenchymal stem cells (HUCB-MSCs). METHODS In this study, forty-eight adult male Wistar albino rats (3 months old) were used. The control group (Group I) received no treatment, while the other rats underwent AGA induction via daily subcutaneous testosterone injections (100 mg/kg). These rats developed alopecia and were divided into three groups: AGA (Group II), AGA plus daily minoxidil spray (Group III), and AGA plus a single intradermal injection of HUCB-MSCs (1 mL containing 1 × 105 cells, Group IV). After 4 weeks, the rats were sacrificed, and skin specimens were prepared for histological analysis using H&E, Masson's trichrome, and immunohistochemical staining for CK 19, vascular endothelial growth factor (VEGF), and TUNEL antibodies. RESULTS It was shown that HUCB-MSC treatment reversed structural damage to hair and follicles, normalizing conditions within 1-week post-injection. The treatment enhanced the anagen phase, suppressed telogen and catagen phases, reduced apoptosis, and increased VEGF and CK 19 immune reactions. Observational follow-up for Groups III and IV revealed that while the minoxidil group experienced significant hair loss after 37 days, the stem cell group exhibited dense and long hair covering the treated area. CONCLUSION HUCB-MSC therapy demonstrated superior efficacy over minoxidil with no observed side effects, indicating its potential as a promising alternative for AGA treatment.
Collapse
Affiliation(s)
- Sherif A Kamar
- Department Anatomy and Embryology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
- Department of Basic Medical Sciences, Faculty of Dentistry, Al-Ahliyya Amman University, Amman, Jordan
| | - Khaled Naiem Hamdy
- Department Anatomy and Embryology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | | | - Heba Mohammed
- Department Anatomy and Embryology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Marwa A Fetouh
- Department Anatomy and Embryology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| |
Collapse
|
6
|
Seibt LE, Antonio EL, AzevedoTeixeira IL, de Oliveira HA, Dias ARL, Neves Dos Santos LF, Serra AJ. Mesenchymal Stem Cells Increase Resistance Against Ventricular Arrhythmias Provoked in Rats with Myocardial Infarction. Stem Cell Rev Rep 2024; 20:2293-2302. [PMID: 39172208 DOI: 10.1007/s12015-024-10773-9] [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] [Accepted: 08/06/2024] [Indexed: 08/23/2024]
Abstract
This study evaluated the role of the mesenchymal stem cells derived from adipose tissue (MSCs) in provoked ventricular arrhythmias (VAs) in animals with myocardial infarction (MI). The experimental groups were: sham, subjected to sham surgery and intramyocardial saline injection; MIV, infarcted rats subjected to intramyocardial saline injection; MI + MSCs, infarcted rats subjected to intramyocardial MSCs injection. Injections were performed two days after infarction and the arrhythmogenic inducibility experiment was performed the next day. Only 35% of the MI + MSCs group developed VAs, while the one in the MIV group was 65%. The proportion of nonsustained ventricular tachycardia, sustained tachycardia, and ventricular fibrillation was similar between the infarcted groups, but MSCs animals had shorter duration of nonsustained ventricular tachycardia. However, MSCs increased connexin 43 content in the remote area, even above the levels found in the sham group. MSCs prevented the increase of IL-1β in the different areas of the myocardium. There was higher carbonylation and content of 4-hydroxynonenal (4-HNE, a marker of lipoperoxidation) in the myocardium of infarcted rats, but MSCs attenuated the increase of 4-HNE in the infarcted area. In conclusion, MSCs have a protective effect against the development of arrhythmias, but do not imply a significant benefit for animals that have developed VAs. It is possible to think that the cardioprotection of MSCs involves anti-inflammatory/oxidative actions and improvement in the formation of communicating junctions.Graphical abstract.
Collapse
Affiliation(s)
- Larissa Emília Seibt
- Laboratory of Physiology and Cardiac Pathophysiology, Department of Medicine, Federal University of São Paulo, São Paulo, SP, Brazil
| | - Ednei Luiz Antonio
- Laboratory of Physiology and Cardiac Pathophysiology, Department of Medicine, Federal University of São Paulo, São Paulo, SP, Brazil
| | - Ighor Luiz AzevedoTeixeira
- Laboratory of Physiology and Cardiac Pathophysiology, Department of Medicine, Federal University of São Paulo, São Paulo, SP, Brazil
| | - Helenita Antonia de Oliveira
- Laboratory of Physiology and Cardiac Pathophysiology, Department of Medicine, Federal University of São Paulo, São Paulo, SP, Brazil
| | - André Rodrigues Lourenço Dias
- Laboratory of Physiology and Cardiac Pathophysiology, Department of Medicine, Federal University of São Paulo, São Paulo, SP, Brazil
| | | | - Andrey Jorge Serra
- Laboratory of Physiology and Cardiac Pathophysiology, Department of Medicine, Federal University of São Paulo, São Paulo, SP, Brazil.
| |
Collapse
|
7
|
Tian G, Yin H, Zheng J, Yu R, Ding Z, Yan Z, Tang Y, Wu J, Ning C, Yuan X, Liao C, Sui X, Zhao Z, Liu S, Guo W, Guo Q. Promotion of osteochondral repair through immune microenvironment regulation and activation of endogenous chondrogenesis via the release of apoptotic vesicles from donor MSCs. Bioact Mater 2024; 41:455-470. [PMID: 39188379 PMCID: PMC11347043 DOI: 10.1016/j.bioactmat.2024.07.034] [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: 04/24/2024] [Revised: 07/28/2024] [Accepted: 07/30/2024] [Indexed: 08/28/2024] Open
Abstract
Utilizing transplanted human umbilical cord mesenchymal stem cells (HUMSCs) for cartilage defects yielded advanced tissue regeneration, but the underlying mechanism remain elucidated. Early after HUMSCs delivery to the defects, we observed substantial apoptosis. The released apoptotic vesicles (apoVs) of HUMSCs promoted cartilage regeneration by alleviating the chondro-immune microenvironment. ApoVs triggered M2 polarization in macrophages while simultaneously facilitating the chondrogenic differentiation of endogenous MSCs. Mechanistically, in macrophages, miR-100-5p delivered by apoVs activated the MAPK/ERK signaling pathway to promote M2 polarization. In MSCs, let-7i-5p delivered by apoVs promoted chondrogenic differentiation by targeting the eEF2K/p38 MAPK axis. Consequently, a cell-free cartilage regeneration strategy using apoVs combined with a decellularized cartilage extracellular matrix (DCM) scaffold effectively promoted the regeneration of osteochondral defects. Overall, new mechanisms of cartilage regeneration by transplanted MSCs were unconcealed in this study. Moreover, we provided a novel experimental basis for cell-free tissue engineering-based cartilage regeneration utilizing apoVs.
Collapse
Affiliation(s)
- Guangzhao Tian
- School of Medicine, Nankai University, Tianjin, 300071, China
- Institute of Orthopedies, Department of Orthopedics, The Fourth Medical Center of PLA General Hospital, National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, 51 Fucheng Road, Haidian District, Beijing, 100142, China
| | - Han Yin
- Institute of Orthopedies, Department of Orthopedics, The Fourth Medical Center of PLA General Hospital, National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, 51 Fucheng Road, Haidian District, Beijing, 100142, China
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jinxuan Zheng
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510055, China
| | - Rongcheng Yu
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510055, China
| | - Zhengang Ding
- Institute of Orthopedies, Department of Orthopedics, The Fourth Medical Center of PLA General Hospital, National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, 51 Fucheng Road, Haidian District, Beijing, 100142, China
| | - Zineng Yan
- Institute of Orthopedies, Department of Orthopedics, The Fourth Medical Center of PLA General Hospital, National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, 51 Fucheng Road, Haidian District, Beijing, 100142, China
| | - Yiqi Tang
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510055, China
| | - Jiang Wu
- Institute of Orthopedies, Department of Orthopedics, The Fourth Medical Center of PLA General Hospital, National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, 51 Fucheng Road, Haidian District, Beijing, 100142, China
| | - Chao Ning
- Institute of Orthopedies, Department of Orthopedics, The Fourth Medical Center of PLA General Hospital, National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, 51 Fucheng Road, Haidian District, Beijing, 100142, China
| | - Xun Yuan
- Institute of Orthopedies, Department of Orthopedics, The Fourth Medical Center of PLA General Hospital, National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, 51 Fucheng Road, Haidian District, Beijing, 100142, China
| | - Chenxi Liao
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510055, China
| | - Xiang Sui
- Institute of Orthopedies, Department of Orthopedics, The Fourth Medical Center of PLA General Hospital, National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, 51 Fucheng Road, Haidian District, Beijing, 100142, China
| | - Zhe Zhao
- Institute of Orthopedies, Department of Orthopedics, The Fourth Medical Center of PLA General Hospital, National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, 51 Fucheng Road, Haidian District, Beijing, 100142, China
| | - Shuyun Liu
- Institute of Orthopedies, Department of Orthopedics, The Fourth Medical Center of PLA General Hospital, National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, 51 Fucheng Road, Haidian District, Beijing, 100142, China
| | - Weimin Guo
- Department of Orthopaedic Surgery, Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, First Affiliated Hospital Sun Yat-Sen University, Guangzhou, 510080, China
| | - Quanyi Guo
- School of Medicine, Nankai University, Tianjin, 300071, China
- Institute of Orthopedies, Department of Orthopedics, The Fourth Medical Center of PLA General Hospital, National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, 51 Fucheng Road, Haidian District, Beijing, 100142, China
| |
Collapse
|
8
|
Xiao Z, Li S, Wu X, Chen X, Yan D, He J. GATA-4 overexpressing BMSC-derived exosomes suppress H/R-induced cardiomyocyte ferroptosis. iScience 2024; 27:110784. [PMID: 39391723 PMCID: PMC11466636 DOI: 10.1016/j.isci.2024.110784] [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: 01/30/2024] [Revised: 07/01/2024] [Accepted: 08/19/2024] [Indexed: 10/12/2024] Open
Abstract
Bone marrow mesenchymal stem cell (BMSC)-derived exosomes overexpressing GATA-4 (Exosoe-GATA-4) can protect cardiac function. Mitochondrial permeability transition pore (mPTP) has a crucial role in ferroptosis. This study aimed to assess the mechanism of Exosoe-GATA-4 in myocardial ischemia/reperfusion (I/R) injury. Exos were successfully excreted, and 185 differential expression miRNAs were obtained using bioinformatics. The Exosoe-GATA-4 effectively suppressed hypoxia/reoxygenation (H/R)-induced cardiomyocytes' ferroptosis, while the effects were reversed by miR-330-3p inhibitor. miR-330-3p targeted negative regulated BAP1. The effects of miR-330-3p inhibitor were reversed by knock-down BAP1. Also, BAP1 reversed the effects of Exosoe-GATA-4 on H/R-induced cardiomyocytes' ferroptosis by downregulating SLC7A11. Mechanistically, BAP1 interacted with IP3R and increased cardiomyocytes' Ca2+ level, causing mPTP opening and mitochondrial dysfunction, promoting H/R-induced cardiomyocytes' ferroptosis. Moreover, hydrogen sulfide (H2S) content was increased and regulated the keap1/Nrf2 signaling pathway by Exosoe-GATA-4 treated. Exosoe-GATA-4 effectively suppresses H/R-induced cardiomyocytes' ferroptosis by upregulating miR-330-3p, which regulates the BAP1/SLC7A11/IP3R axis and inhibits mPTP opening.
Collapse
Affiliation(s)
- Zhiyuan Xiao
- Department of Medical Intensive Care Unit, the First People′s Hospital of Yunnan Province, No.157 Jinbi Road, Kunming, Yunnan 650032, China
| | - Si Li
- The Affiliated Hospital of Kunming University of Science and Technology, Kunming 650500, P.R. China
| | - Xinxin Wu
- Yunnan University of Traditional Chinese Medicine, No.1076 Yuhua Road, Kunming, Yunnan 650500, China
| | - Xinhao Chen
- The Affiliated Hospital of Kunming University of Science and Technology, Kunming 650500, P.R. China
| | - Dan Yan
- Department of Medical Intensive Care Unit, the First People′s Hospital of Yunnan Province, No.157 Jinbi Road, Kunming, Yunnan 650032, China
| | - Jigang He
- Department of Cardiovascular Surgery, the First People′s Hospital of Yunnan Province, No.157 Jinbi Road, Kunming, Yunnan 650032, China
| |
Collapse
|
9
|
Xu C, Xie Y, Wang B. Genetically modified mesenchymal stromal cells: a cell-based therapy offering more efficient repair after myocardial infarction. Stem Cell Res Ther 2024; 15:323. [PMID: 39334266 PMCID: PMC11438184 DOI: 10.1186/s13287-024-03942-7] [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: 06/27/2024] [Accepted: 09/16/2024] [Indexed: 09/30/2024] Open
Abstract
Myocardial infarction (MI) is a serious complication of coronary artery disease. This condition is common worldwide and has a profound impact on patients' lives and quality of life. Despite significant advances in the treatment of heart disease in modern medicine, the efficient treatment of MI still faces a number of challenges. Problems such as scar formation and loss of myocardial function after a heart attack still limit patients' recovery. Therefore, the search for a new therapeutic tool that can promote repair and regeneration of myocardial tissue has become crucial. In this context, mesenchymal stromal cells (MSCs) have attracted much attention as a potential therapeutic tool. MSCs are a class of adult stem cells with multidirectional differentiation potential, derived from bone marrow, fat, placenta and other tissues, and possessing properties such as self-renewal and immunomodulation. The application of MSCs may provide a new direction for the treatment of MI. These stem cells have the potential to differentiate into cardiomyocytes and vascular endothelial cells in damaged tissue and to repair and protect myocardial tissue through anti-inflammatory, anti-fibrotic and pro-neovascularization mechanisms. However, the clinical results of MSCs transplantation for the treatment of MI are less satisfactory due to the limitations of the native function of MSCs. Genetic modification has overcome problems such as the low survival rate of transplanted MSCs in vivo and enhanced their functions of promoting neovascularization and differentiation into cardiomyocytes, paving the way for them to become an effective tool for repair therapy after MI. In previous studies, MSCs have shown some therapeutic potential in experimental animals and preliminary clinical trials. This review aims to provide readers with a comprehensive and in-depth understanding to promote the wider application of engineering MSCs in the field of MI therapy, offering new hope for recovery and improved survival of cardiac patients.
Collapse
Affiliation(s)
- Congwang Xu
- Clinical Stem Cell Center, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese, Medicine321 Zhongshan Road, Nanjing, 210008, People's Republic of China
| | - Yuanyuan Xie
- Clinical Stem Cell Center, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210000, People's Republic of China
| | - Bin Wang
- Clinical Stem Cell Center, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese, Medicine321 Zhongshan Road, Nanjing, 210008, People's Republic of China.
- Clinical Stem Cell Center, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210000, People's Republic of China.
| |
Collapse
|
10
|
Farag A, Koung Ngeun S, Kaneda M, Aboubakr M, Tanaka R. Optimizing Cardiomyocyte Differentiation: Comparative Analysis of Bone Marrow and Adipose-Derived Mesenchymal Stem Cells in Rats Using 5-Azacytidine and Low-Dose FGF and IGF Treatment. Biomedicines 2024; 12:1923. [PMID: 39200387 PMCID: PMC11352160 DOI: 10.3390/biomedicines12081923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 08/09/2024] [Accepted: 08/19/2024] [Indexed: 09/02/2024] Open
Abstract
Mesenchymal stem cells (MSCs) exhibit multipotency, self-renewal, and immune-modulatory properties, making them promising in regenerative medicine, particularly in cardiovascular treatments. However, optimizing the MSC source and induction method of cardiac differentiation is challenging. This study compares the cardiomyogenic potential of bone marrow (BM)-MSCs and adipose-derived (AD)-MSCs using 5-Azacytidine (5-Aza) alone or combined with low doses of Fibroblast Growth Factor (FGF) and Insulin-like Growth Factor (IGF). BM-MSCs and AD-MSCs were differentiated using two protocols: 10 μmol 5-Aza alone and 10 μmol 5-Aza with 1 ng/mL FGF and 10 ng/mL IGF. Morphological, transcriptional, and translational analyses, along with cell viability assessments, were performed. Both the MSC types exhibited similar morphological changes; however, AD-MSCs achieved 70-80% confluence faster than BM-MSCs. Surface marker profiling confirmed CD29 and CD90 positivity and CD45 negativity. The differentiation protocols led to cell flattening and myotube formation, with earlier differentiation in AD-MSCs. The combined protocol reduced cell mortality in BM-MSCs and enhanced the expression of cardiac markers (MEF2c, Troponin I, GSK-3β), particularly in BM-MSCs. Immunofluorescence confirmed cardiac-specific protein expression in all the treated groups. Both MSC types exhibited the expression of cardiac-specific markers indicative of cardiomyogenic differentiation, with the combined treatment showing superior efficiency for BM-MSCs.
Collapse
Affiliation(s)
- Ahmed Farag
- Veterinary Teaching Hospital, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Tokyo 183-8509, Japan
- Department of Surgery, Anesthesiology, and Radiology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44519, Egypt
| | - Sai Koung Ngeun
- Laboratory of Veterinary Diagnostic Imaging, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Tokyo 183-8509, Japan;
| | - Masahiro Kaneda
- Laboratory of Veterinary Anatomy, Division of Animal Life Science, Tokyo University of Agriculture and Technology, Tokyo 183-8509, Japan;
| | - Mohamed Aboubakr
- Department of Pharmacology, Faculty of Veterinary Medicine, Benha University, Toukh 13736, Egypt;
| | - Ryou Tanaka
- Veterinary Teaching Hospital, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Tokyo 183-8509, Japan
| |
Collapse
|
11
|
Xu J, Zhao J, Chen H, Tan X, Zhang W, Xia Z, Yao D, Lei Y, Xu B, Wei Z, Hu J. Mesenchymal stromal cell-derived exosomes protect against abdominal aortic aneurysm formation through CD74 modulation of macrophage polarization in mice. Stem Cell Res Ther 2024; 15:242. [PMID: 39098899 PMCID: PMC11299418 DOI: 10.1186/s13287-024-03808-y] [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: 04/13/2024] [Accepted: 06/18/2024] [Indexed: 08/06/2024] Open
Abstract
BACKGROUND Mesenchymal stromal cell (MSC)-derived exosomes (MSC-Exo) have been recognized for their significant role in regulating macrophage polarization, a process crucial to the pathogenesis of abdominal aortic aneurysm (AAA). However, the therapeutic effects of MSC-Exo on AAA remain largely unexplored. Therefore, this study aimed to investigate the functional and mechanistic aspects of MSC-Exo in the progression of AAA. METHODS The MSC-derived exosomes were characterized using Transmission Electron Microscopy, Nanoparticle Tracking Analysis, and Western blotting. An experimental mouse model of AAA was established through the administration of angiotensin II (Ang II) in male apoe-/- mice and calcium chloride (CaCl2) in male C57/B6 mice, with subsequent tail vein injection of exosomes to evaluate their efficacy against AAA. Macrophage polarization was assessed using immunofluorescence staining and WB analysis. Mechanistic analysis was performed using 4D Label-free Proteomics analysis. RESULTS We found that intravenous administration of MSC-Exo induced M2 polarization of macrophages within an inflammatory environment, effectively impeding AAA development in Ang II or CaCl2-induced AAA model. The therapeutic efficacy of MSC-Exo treatment was dependent on the presence of macrophages. Mechanistically, MSC-Exo suppressed the levels of cluster of differentiation 74 (CD74), modulating macrophage polarization through the TSC2-mTOR-AKT pathway. These findings highlight the potential of MSC-Exo as a therapeutic strategy for AAA by modulating macrophage polarization.
Collapse
Affiliation(s)
- Jiamin Xu
- Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Jiling Zhao
- Cardiovascular Disease Center, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi Clinical College of Wuhan University, No. 158 Wuyang Avenue, Enshi, Hubei, China
| | - Haiting Chen
- Department of Cardiology, The First Affiliated Hospital of Anhui Medical University, Anhui, China
| | - Xi Tan
- Department of Cardiology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Wenfeng Zhang
- Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Zhongnan Xia
- Cardiovascular Disease Center, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi Clinical College of Wuhan University, No. 158 Wuyang Avenue, Enshi, Hubei, China
| | - Dejiang Yao
- Surgical Division III, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi Clinical College of Wuhan University, Enshi, Hubei, China
| | - Yuhua Lei
- Cardiovascular Disease Center, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi Clinical College of Wuhan University, No. 158 Wuyang Avenue, Enshi, Hubei, China.
| | - Biao Xu
- Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China.
| | - Zhonghai Wei
- Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China.
| | - Jiaxin Hu
- Cardiovascular Disease Center, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi Clinical College of Wuhan University, No. 158 Wuyang Avenue, Enshi, Hubei, China.
- Hubei Selenium and Human Health Institute, the Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, 445000, China.
| |
Collapse
|
12
|
Seow KS, Ling APK. Mesenchymal stem cells as future treatment for cardiovascular regeneration and its challenges. ANNALS OF TRANSLATIONAL MEDICINE 2024; 12:73. [PMID: 39118948 PMCID: PMC11304428 DOI: 10.21037/atm-23-1936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 12/04/2023] [Indexed: 08/10/2024]
Abstract
Cardiovascular diseases (CVDs), particularly stroke and myocardial infarction (MI) contributed to the leading cause of death annually among the chronic diseases globally. Despite the advancement of technology, the current available treatments mainly served as palliative care but not treating the diseases. However, the discovery of mesenchymal stem cells (MSCs) had gained a consideration to serve as promising strategy in treating CVDs. Recent evidence also showed that MSCs are the strong candidate to be used as stem cell therapy involving cardiovascular regeneration due to its cardiomyogenesis, anti-inflammatory and immunomodulatory properties, antifibrotic effects and neovascularization capacity. Besides, MSCs could be used for cellular cardiomyoplasty with its transdifferentiation of MSCs into cardiomyocytes, paracrine effects, microvesicles and exosomes as well as mitochondrial transfer. The safety and efficacy of utilizing MSCs have been described in well-established preclinical and clinical studies in which the accomplishment of MSCs transplantation resulted in further improvement of the cardiac function. Tissue engineering could enhance the desired properties and therapeutic effects of MSCs in cardiovascular regeneration by genome-editing, facilitating the cell delivery and retention, biomaterials-based scaffold, and three-dimensional (3D)-bioprinting. However, there are still obstacles in the use of MSCs due to the complexity and versatility of MSCs, low retention rate, route of administration and the ethical and safety issues of the use of MSCs. The aim of this review is to highlight the details of therapeutic properties of MSCs in treating CVDs, strategies to facilitate the therapeutic effects of MSCs through tissue engineering and the challenges faced using MSCs. A comprehensive review has been done through PubMed and National Center for Biotechnology Information (NCBI) from the year of 2010 to 2021 based on some specific key terms such as 'mesenchymal stem cells in cardiovascular disease', 'mesenchymal stem cells in cardiac regeneration', 'mesenchymal stem cells facilitate cardiac repairs', 'tissue engineering of MSCs' to include relevant literature in this review.
Collapse
Affiliation(s)
- Ke Sin Seow
- Division of Applied Biomedical Sciences and Biotechnology, School of Health Sciences, International Medical University, Kuala Lumpur, Malaysia
| | - Anna Pick Kiong Ling
- Division of Applied Biomedical Sciences and Biotechnology, School of Health Sciences, International Medical University, Kuala Lumpur, Malaysia
| |
Collapse
|
13
|
Masroor M, Wang Y, Zhang C, Dong N. Surgical Management of Ischemic Cardiomyopathy Patients with Severe Left Ventricular Dysfunction: Is It Time to Reconsider Revascularization Surgery? J Cardiovasc Dev Dis 2024; 11:184. [PMID: 39057607 PMCID: PMC11276879 DOI: 10.3390/jcdd11070184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Revised: 06/05/2024] [Accepted: 06/19/2024] [Indexed: 07/28/2024] Open
Abstract
Ischemic cardiomyopathy patients with severe left ventricular dysfunction are a specific group of patients with poor surgical outcomes. There are few surgical treatment options in practice for the treatment of these patients such as heart transplantation, coronary artery bypass surgery, surgical ventricular restoration, etc. Despite multiple treatment options, there are no explicit clinical guidelines available to guide surgeons in choosing the most appropriate option and ensuring that the specific patient can benefit from the selected surgical treatment. Heart transplantation is the gold standard treatment for ischemic cardiomyopathy patients with severe left ventricular dysfunction, but it is limited to very few highly equipped centers around the world due to donor shortages, complex perioperative and surgical management, and limited technological and human resources. It is evident from some studies that heart transplant-eligible candidates can benefit from alternative surgical options such as coronary artery bypass surgery alone or combined with surgical ventricular restoration. Therefore, alternative surgical options that are used for most of the population, especially in developing and underdeveloped countries, need to be discussed to improve their outcomes. A challenge in the recent era which has yet to find a solution is to determine which heart transplant candidate can benefit from simple revascularization compared to a complex heart transplantation procedure. Myocardial viability testing was one of the most important determinants in deciding whether a patient should undergo revascularization, but its role in guiding appropriate surgical options has been challenged. This review aims to discuss the available surgical management options and their long-term outcomes for patients with ischemic cardiomyopathy, which will eventually help surgeons when choosing a surgical procedure.
Collapse
Affiliation(s)
- Matiullah Masroor
- Department of Cardiovascular Surgery, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (M.M.); (Y.W.)
- Department of Cardiothoracic and Vascular Surgery, Amiri Medical Complex, Qargha Rd, Kabul 1010, Afghanistan
| | - Yixuan Wang
- Department of Cardiovascular Surgery, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (M.M.); (Y.W.)
| | - Chao Zhang
- Department of Cardiovascular Surgery, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (M.M.); (Y.W.)
| | - Nianguo Dong
- Department of Cardiovascular Surgery, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (M.M.); (Y.W.)
| |
Collapse
|
14
|
Abdelhamid MAA, Khalifa HO, Ki MR, Pack SP. Nanoengineered Silica-Based Biomaterials for Regenerative Medicine. Int J Mol Sci 2024; 25:6125. [PMID: 38892312 PMCID: PMC11172759 DOI: 10.3390/ijms25116125] [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: 03/30/2024] [Revised: 05/24/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024] Open
Abstract
The paradigm of regenerative medicine is undergoing a transformative shift with the emergence of nanoengineered silica-based biomaterials. Their unique confluence of biocompatibility, precisely tunable porosity, and the ability to modulate cellular behavior at the molecular level makes them highly desirable for diverse tissue repair and regeneration applications. Advancements in nanoengineered silica synthesis and functionalization techniques have yielded a new generation of versatile biomaterials with tailored functionalities for targeted drug delivery, biomimetic scaffolds, and integration with stem cell therapy. These functionalities hold the potential to optimize therapeutic efficacy, promote enhanced regeneration, and modulate stem cell behavior for improved regenerative outcomes. Furthermore, the unique properties of silica facilitate non-invasive diagnostics and treatment monitoring through advanced biomedical imaging techniques, enabling a more holistic approach to regenerative medicine. This review comprehensively examines the utilization of nanoengineered silica biomaterials for diverse applications in regenerative medicine. By critically appraising the fabrication and design strategies that govern engineered silica biomaterials, this review underscores their groundbreaking potential to bridge the gap between the vision of regenerative medicine and clinical reality.
Collapse
Affiliation(s)
- Mohamed A. A. Abdelhamid
- Department of Biotechnology and Bioinformatics, Korea University, Sejong-Ro 2511, Sejong 30019, Republic of Korea;
- Department of Botany and Microbiology, Faculty of Science, Minia University, Minia 61519, Egypt
| | - Hazim O. Khalifa
- Department of Veterinary Medicine, College of Agriculture and Veterinary Medicine, United Arab Emirates University, Al Ain P.O. Box 1555, United Arab Emirates;
- Department of Pharmacology, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafr El-Sheikh 33516, Egypt
| | - Mi-Ran Ki
- Department of Biotechnology and Bioinformatics, Korea University, Sejong-Ro 2511, Sejong 30019, Republic of Korea;
- Institute of Industrial Technology, Korea University, Sejong-Ro 2511, Sejong 30019, Republic of Korea
| | - Seung Pil Pack
- Department of Biotechnology and Bioinformatics, Korea University, Sejong-Ro 2511, Sejong 30019, Republic of Korea;
| |
Collapse
|
15
|
Wang M, Li C, Liu Y, Jin Y, Yu Y, Tan X, Zhang C. The effect of macrophages and their exosomes in ischemic heart disease. Front Immunol 2024; 15:1402468. [PMID: 38799471 PMCID: PMC11116575 DOI: 10.3389/fimmu.2024.1402468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Accepted: 04/29/2024] [Indexed: 05/29/2024] Open
Abstract
Ischemic heart disease (IHD) is a leading cause of disability and death worldwide, with immune regulation playing a crucial role in its pathogenesis. Various immune cells are involved, and as one of the key immune cells residing in the heart, macrophages play an indispensable role in the inflammatory and reparative processes during cardiac ischemia. Exosomes, extracellular vesicles containing lipids, nucleic acids, proteins, and other bioactive molecules, have emerged as important mediators in the regulatory functions of macrophages and hold promise as a novel therapeutic target for IHD. This review summarizes the regulatory mechanisms of different subsets of macrophages and their secreted exosomes during cardiac ischemia over the past five years. It also discusses the current status of clinical research utilizing macrophages and their exosomes, as well as strategies to enhance their therapeutic efficacy through biotechnology. The aim is to provide valuable insights for the treatment of IHD.
Collapse
Affiliation(s)
- Minrui Wang
- Department of Physiology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
| | - Chunhong Li
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Yuchang Liu
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Yuanyuan Jin
- The Key Laboratory of Medical Electrophysiology of the Ministry of Education, Southwest Medical University, Luzhou, Sichuan, China
| | - Yang Yu
- The Key Laboratory of Medical Electrophysiology of the Ministry of Education, Southwest Medical University, Luzhou, Sichuan, China
| | - Xiaoqiu Tan
- Department of Physiology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
- The Key Laboratory of Medical Electrophysiology of the Ministry of Education, Southwest Medical University, Luzhou, Sichuan, China
- Department of Cardiology, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Chunxiang Zhang
- The Key Laboratory of Medical Electrophysiology of the Ministry of Education, Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
16
|
Li S, Guan X, Yu W, Zhao Z, Sun Y, Bai Y. Effect of human periodontal ligament stem cell-derived exosomes on cementoblast activity. Oral Dis 2024; 30:2511-2522. [PMID: 37448205 DOI: 10.1111/odi.14671] [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: 03/26/2023] [Revised: 06/19/2023] [Accepted: 06/26/2023] [Indexed: 07/15/2023]
Abstract
OBJECTIVES Exosomes derived from stem cells are a potential cell-free tool for tissue regeneration with therapeutic potential. However, its application in cementum repair is unclear. This study aimed to investigate the effect of human periodontal ligament stem cell-derived exosomes on the biological activity of cementoblasts, the main effector cells in cementum synthesis. MATERIALS AND METHODS OCCM-30 cementoblasts were cultured with various human periodontal ligament stem cell-derived exosome concentrations. OCCM-30 cells proliferation, migration, and cementogenic mineralization were examined, along with the gene and protein expression of factors associated with cementoblastic mineralization. RESULTS Exosomal promoted the migration, proliferation, and mineralization of OCCM-30 cells. The exosome-treated group significantly increased the expression of cementogenic-related genes and proteins. Furthermore, the expression of p-PI3K and p-AKT was enhanced by exosome administration. Treatment with a PI3K/AKT inhibitor markedly attenuated the gene and protein expression of cementoblastic factors, and this effect was partially reversed by exosome administration. CONCLUSIONS Human periodontal ligament stem cell-derived exosomes can promote the activity of cementoblasts via the PI3K/AKT signaling pathway, providing a scientific basis for promoting the repair process in orthodontically induced inflammatory root resorption.
Collapse
Affiliation(s)
- Shengnan Li
- Department of Orthodontics, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China
| | - Xiuchen Guan
- Department of Orthodontics, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China
| | - Wenting Yu
- Department of Orthodontics, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China
| | - Zeqing Zhao
- Department of Orthodontics, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China
| | - Yaxi Sun
- Department of Orthodontics, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China
| | - Yuxing Bai
- Department of Orthodontics, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China
| |
Collapse
|
17
|
Zhu Y, Huang C, Zheng L, Li Q, Ge J, Geng S, Zhai M, Chen X, Yuan H, Li Y, Jia W, Sun K, Li Y, Ye T, Zhao Z, Liu H, Liu Z, Jiang H. Safety and efficacy of umbilical cord tissue-derived mesenchymal stem cells in the treatment of patients with aging frailty: a phase I/II randomized, double-blind, placebo-controlled study. Stem Cell Res Ther 2024; 15:122. [PMID: 38679727 PMCID: PMC11057094 DOI: 10.1186/s13287-024-03707-2] [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: 01/08/2024] [Accepted: 03/24/2024] [Indexed: 05/01/2024] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) hold a great promise for cell-based therapy in the field of regenerative medicine. In this study, we aimed to evaluate the safety and efficacy of intravenous infusion of human umbilical cord-derived MSCs (HUC-MSCs) in patients with aging frailty. METHODS In this randomized, double-blind, placebo-controlled trial, participants diagnosed with aging frailty were randomly assigned to receive intravenous administrations of HUC-MSCs or placebo. All of serious adverse events and AEs were monitored to evaluate the safety of treatment during the 6-month follow-up. The primary efficacy endpoint was alteration of physical component scores (PCS) of SF-36 qualities of life at 6 months. The secondary outcomes including physical performance tests and pro-inflammatory cytokines, were also observed and compared at each follow-up visits. All evaluations were performed at 1 week, 1, 2, 3 and 6 months following the first intravenous infusion of HUC-MSCs. RESULTS In the MSCs group, significant improvements in PCS of SF-36 were observed from first post-treatment visit and sustained throughout the follow-up period, with greater changes compared to the placebo group (p = 0.042). EQ-VAS scores of MSCs group improved significantly at 2 month (p = 0.023) and continued until the end of the 6-month visit (p = 0.002) in comparison to the placebo group. The timed up and go (TUG) physical performance test revealed significant group difference and showed continual enhancements over 6 months (p < 0.05). MSC transplantation improved the function of 4-m walking test (4MWT) compared with the placebo group with a decrease of 2.05 s at 6 months of follow-up (p = 0.21). The measurement of grip strength revealed group difference with MSCs group demonstrating better performance, particularly at 6 months (p = 0.002). Inflammatory cytokines (TNF-α, IL-17) exhibited declines in MSCs group at 6 months compared to the placebo group (p = 0.034 and 0.033, respectively). There was no difference of incidence of AEs between the two groups. CONCLUSION Intravenous transplantation of HUC-MSCs is a safe and effective therapeutic approach on aging frailty. The positive outcomes observed in improving quality of life, physical performance, and reducing chronic inflammation, suggest that HUC-MSC therapy may be a promising potential treatment option for aging frailty. TRIAL REGISTRATION Clinicaltrial.gov; NCT04314011; https://clinicaltrials.gov/ct2/show/NCT04314011 .
Collapse
Affiliation(s)
- Yingqian Zhu
- Department of Geriatrics, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
- Department of General Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Ce Huang
- Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
- Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai Municipality, 200032, China
| | - Liang Zheng
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Qingqing Li
- Department of Geriatrics, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
- Department of General Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Jianli Ge
- Department of Geriatrics, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
- Department of General Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - ShaSha Geng
- Department of Geriatrics, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
- Department of General Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Miaomiao Zhai
- Department of Geriatrics, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
- Department of General Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Xin Chen
- Department of Geriatrics, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
- Department of General Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Huixiao Yuan
- Department of Geriatrics, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
- Department of General Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Yang Li
- Department of Geriatrics, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
- Department of General Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Wenwen Jia
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, 200120, China
| | - Keping Sun
- Department of Geriatrics, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
- Department of General Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Yan Li
- Department of Geriatrics, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
- Department of General Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Tong Ye
- Department of Geriatrics, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
- Department of General Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Zhengmei Zhao
- Department of Geriatrics, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
- Department of General Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Hailiang Liu
- Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.
| | - Zhongmin Liu
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, 200120, China.
- Department of Cardiovascular Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.
- Translational Medical Center for Stem Cell Therapy, Shanghai East Hospital, Tongji University, Shanghai, 200120, China.
| | - Hua Jiang
- Department of Geriatrics, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.
- Department of General Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.
| |
Collapse
|
18
|
Liu B, Wei Y, He J, Feng B, Chen Y, Guo R, Griffin MD, Hynes SO, Shen S, Liu Y, Cui H, Ma J, O'Brien T. Human umbilical cord-derived mesenchymal stromal cells improve myocardial fibrosis and restore miRNA-133a expression in diabetic cardiomyopathy. Stem Cell Res Ther 2024; 15:120. [PMID: 38659015 PMCID: PMC11040946 DOI: 10.1186/s13287-024-03715-2] [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: 07/31/2023] [Accepted: 04/02/2024] [Indexed: 04/26/2024] Open
Abstract
BACKGROUND Diabetic cardiomyopathy (DCM) is a serious health-threatening complication of diabetes mellitus characterized by myocardial fibrosis and abnormal cardiac function. Human umbilical cord mesenchymal stromal cells (hUC-MSCs) are a potential therapeutic tool for DCM and myocardial fibrosis via mechanisms such as the regulation of microRNA (miRNA) expression and inflammation. It remains unclear, however, whether hUC-MSC therapy has beneficial effects on cardiac function following different durations of diabetes and which mechanistic aspects of DCM are modulated by hUC-MSC administration at different stages of its development. This study aimed to investigate the therapeutic effects of intravenous administration of hUC-MSCs on DCM following different durations of hyperglycemia in an experimental male model of diabetes and to determine the effects on expression of candidate miRNAs, target mRNA and inflammatory mediators. METHODS A male mouse model of diabetes was induced by multiple low-dose streptozotocin injections. The effects on severity of DCM of intravenous injections of hUC-MSCs and saline two weeks previously were compared at 10 and 18 weeks after diabetes induction. At both time-points, biochemical assays, echocardiography, histopathology, polymerase chain reaction (PCR), immunohistochemistry and enzyme-linked immunosorbent assays (ELISA) were used to analyze blood glucose, body weight, cardiac structure and function, degree of myocardial fibrosis and expression of fibrosis-related mRNA, miRNA and inflammatory mediators. RESULTS Saline-treated diabetic male mice had impaired cardiac function and increased cardiac fibrosis after 10 and 18 weeks of diabetes. At both time-points, cardiac dysfunction and fibrosis were improved in hUC-MSC-treated mice. Pro-fibrotic indicators (α-SMA, collagen I, collagen III, Smad3, Smad4) were reduced and anti-fibrotic mediators (FGF-1, miRNA-133a) were increased in hearts of diabetic animals receiving hUC-MSCs compared to saline. Increased blood levels of pro-inflammatory cytokines (IL-6, TNF, IL-1β) and increased cardiac expression of IL-6 were also observed in saline-treated mice and were reduced by hUC-MSCs at both time-points, but to a lesser degree at 18 weeks. CONCLUSION Intravenous injection of hUC-MSCs ameliorated key functional and structural features of DCM in male mice with diabetes of shorter and longer duration. Mechanistically, these effects were associated with restoration of intra-myocardial expression of miRNA-133a and its target mRNA COL1AI as well as suppression of systemic and localized inflammatory mediators.
Collapse
Affiliation(s)
- Boxin Liu
- Stem Cell Research Center, Hebei Medical University-University of Galway, Hebei Medical University, Hebei Province, 050017, China
- Hebei Research Center for Stem Cell Medical Translational Engineering, Hebei Province, 050017, China
- Hebei Technology Innovation Center for Stem Cell and Regenerative Medicine, Hebei Province, China
- Hebei International Joint Research Center for Stem Cell and Regenerative Medicine, Hebei Province, China
| | - Yan Wei
- Stem Cell Research Center, Hebei Medical University-University of Galway, Hebei Medical University, Hebei Province, 050017, China
- Hebei Research Center for Stem Cell Medical Translational Engineering, Hebei Province, 050017, China
- Hebei Technology Innovation Center for Stem Cell and Regenerative Medicine, Hebei Province, China
- Hebei International Joint Research Center for Stem Cell and Regenerative Medicine, Hebei Province, China
| | - Jingjing He
- Stem Cell Research Center, Hebei Medical University-University of Galway, Hebei Medical University, Hebei Province, 050017, China
- Hebei Research Center for Stem Cell Medical Translational Engineering, Hebei Province, 050017, China
- Hebei Technology Innovation Center for Stem Cell and Regenerative Medicine, Hebei Province, China
- Hebei International Joint Research Center for Stem Cell and Regenerative Medicine, Hebei Province, China
| | - Baofeng Feng
- Stem Cell Research Center, Hebei Medical University-University of Galway, Hebei Medical University, Hebei Province, 050017, China
- Hebei Research Center for Stem Cell Medical Translational Engineering, Hebei Province, 050017, China
- Human Anatomy Department, Hebei Medical University, Hebei Province, 050017, China
- Hebei Technology Innovation Center for Stem Cell and Regenerative Medicine, Hebei Province, China
- Hebei International Joint Research Center for Stem Cell and Regenerative Medicine, Hebei Province, China
| | - Yimeng Chen
- Stem Cell Research Center, Hebei Medical University-University of Galway, Hebei Medical University, Hebei Province, 050017, China
- Hebei Research Center for Stem Cell Medical Translational Engineering, Hebei Province, 050017, China
- Hebei Technology Innovation Center for Stem Cell and Regenerative Medicine, Hebei Province, China
- Hebei International Joint Research Center for Stem Cell and Regenerative Medicine, Hebei Province, China
| | - Ruiyun Guo
- Stem Cell Research Center, Hebei Medical University-University of Galway, Hebei Medical University, Hebei Province, 050017, China
- Hebei Research Center for Stem Cell Medical Translational Engineering, Hebei Province, 050017, China
- Hebei Technology Innovation Center for Stem Cell and Regenerative Medicine, Hebei Province, China
- Hebei International Joint Research Center for Stem Cell and Regenerative Medicine, Hebei Province, China
| | - Matthew D Griffin
- Stem Cell Research Center, Hebei Medical University-University of Galway, Hebei Medical University, Hebei Province, 050017, China
- Regenerative Medicine Institute (REMEDI) at CÚRAM SFI Research Centre for Medical Devices, School of Medicine, University of Galway, Galway, Ireland
| | - Seán O Hynes
- Discipline of Pathology, School of Medicine, University of Galway, Galway, Ireland
| | - Sanbing Shen
- Stem Cell Research Center, Hebei Medical University-University of Galway, Hebei Medical University, Hebei Province, 050017, China
- Regenerative Medicine Institute (REMEDI) at CÚRAM SFI Research Centre for Medical Devices, School of Medicine, University of Galway, Galway, Ireland
| | - Yan Liu
- Department of Endocrinology, Hebei Medical University Third Affiliated Hospital, Shijiazhuang, Hebei, 050051, China
| | - Huixian Cui
- Stem Cell Research Center, Hebei Medical University-University of Galway, Hebei Medical University, Hebei Province, 050017, China.
- Hebei Research Center for Stem Cell Medical Translational Engineering, Hebei Province, 050017, China.
- Human Anatomy Department, Hebei Medical University, Hebei Province, 050017, China.
- Hebei Technology Innovation Center for Stem Cell and Regenerative Medicine, Hebei Province, China.
- Hebei International Joint Research Center for Stem Cell and Regenerative Medicine, Hebei Province, China.
| | - Jun Ma
- Stem Cell Research Center, Hebei Medical University-University of Galway, Hebei Medical University, Hebei Province, 050017, China.
- Hebei Research Center for Stem Cell Medical Translational Engineering, Hebei Province, 050017, China.
- Human Anatomy Department, Hebei Medical University, Hebei Province, 050017, China.
- Hebei Technology Innovation Center for Stem Cell and Regenerative Medicine, Hebei Province, China.
- Hebei International Joint Research Center for Stem Cell and Regenerative Medicine, Hebei Province, China.
| | - Timothy O'Brien
- Stem Cell Research Center, Hebei Medical University-University of Galway, Hebei Medical University, Hebei Province, 050017, China.
- Regenerative Medicine Institute (REMEDI) at CÚRAM SFI Research Centre for Medical Devices, School of Medicine, University of Galway, Galway, Ireland.
| |
Collapse
|
19
|
Wang L, Jiang W, Zhao S, Xie D, Chen Q, Zhao Q, Wu H, Luo J, Yang L. Sorafenib inhibits ossification of the posterior longitudinal ligament by blocking LOXL2-mediated vascularization. Bone Res 2024; 12:24. [PMID: 38594260 PMCID: PMC11004159 DOI: 10.1038/s41413-024-00327-7] [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: 05/24/2023] [Revised: 03/10/2024] [Accepted: 03/16/2024] [Indexed: 04/11/2024] Open
Abstract
Ossification of the Posterior Longitudinal Ligament (OPLL) is a degenerative hyperostosis disease characterized by the transformation of the soft and elastic vertebral ligament into bone, resulting in limited spinal mobility and nerve compression. Employing both bulk and single-cell RNA sequencing, we elucidate the molecular characteristics, cellular components, and their evolution during the OPLL process at a single-cell resolution, and validate these findings in clinical samples. This study also uncovers the capability of ligament stem cells to exhibit endothelial cell-like phenotypes in vitro and in vivo. Notably, our study identifies LOXL2 as a key regulator in this process. Through gain-and loss-of-function studies, we elucidate the role of LOXL2 in the endothelial-like differentiation of ligament cells. It acts via the HIF1A pathway, promoting the secretion of downstream VEGFA and PDGF-BB. This function is not related to the enzymatic activity of LOXL2. Furthermore, we identify sorafenib, a broad-spectrum tyrosine kinase inhibitor, as an effective suppressor of LOXL2-mediated vascular morphogenesis. By disrupting the coupling between vascularization and osteogenesis, sorafenib demonstrates significant inhibition of OPLL progression in both BMP-induced and enpp1 deficiency-induced animal models while having no discernible effect on normal bone mass. These findings underscore the potential of sorafenib as a therapeutic intervention for OPLL.
Collapse
Affiliation(s)
- Longqing Wang
- Spine Center, Department of Orthopaedics, Shanghai Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, Shanghai, 200003, PR China
| | - Wenhao Jiang
- Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Tongji University School of Medicine, Shanghai, PR China
| | - Siyuan Zhao
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, PR China
| | - Dong Xie
- Department of Orthopaedics, No. 905 Hospital of PLA Navy, Shanghai, PR China
| | - Qing Chen
- Spine Center, Department of Orthopaedics, Shanghai Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, Shanghai, 200003, PR China
| | - Qi Zhao
- Spine Center, Department of Orthopaedics, Shanghai Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, Shanghai, 200003, PR China
| | - Hao Wu
- Spine Center, Department of Orthopaedics, Shanghai Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, Shanghai, 200003, PR China
| | - Jian Luo
- Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Tongji University School of Medicine, Shanghai, PR China.
| | - Lili Yang
- Spine Center, Department of Orthopaedics, Shanghai Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, Shanghai, 200003, PR China.
| |
Collapse
|
20
|
Friend NE, Beamish JA, Margolis EA, Schott NG, Stegemann JP, Putnam AJ. Pre-cultured, cell-encapsulating fibrin microbeads for the vascularization of ischemic tissues. J Biomed Mater Res A 2024; 112:549-561. [PMID: 37326361 PMCID: PMC10724379 DOI: 10.1002/jbm.a.37580] [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: 03/07/2023] [Revised: 05/04/2023] [Accepted: 05/31/2023] [Indexed: 06/17/2023]
Abstract
There is a significant clinical need to develop effective vascularization strategies for tissue engineering and the treatment of ischemic pathologies. In patients afflicted with critical limb ischemia, comorbidities may limit common revascularization strategies. Cell-encapsulating modular microbeads possess a variety of advantageous properties, including the ability to support prevascularization in vitro while retaining the ability to be injected in a minimally invasive manner in vivo. Here, fibrin microbeads containing human umbilical vein endothelial cells (HUVEC) and bone marrow-derived mesenchymal stromal cells (MSC) were cultured in suspension for 3 days (D3 PC microbeads) before being implanted within intramuscular pockets in a SCID mouse model of hindlimb ischemia. By 14 days post-surgery, animals treated with D3 PC microbeads showed increased macroscopic reperfusion of ischemic foot pads and improved limb salvage compared to the cellular controls. Delivery of HUVEC and MSC via microbeads led to the formation of extensive microvascular networks throughout the implants. Engineered vessels of human origins showed evidence of inosculation with host vasculature, as indicated by erythrocytes present in hCD31+ vessels. Over time, the total number of human-derived vessels within the implant region decreased as networks remodeled and an increase in mature, pericyte-supported vascular structures was observed. Our findings highlight the potential therapeutic benefit of developing modular, prevascularized microbeads as a minimally invasive therapeutic for treating ischemic tissues.
Collapse
Affiliation(s)
- Nicole E. Friend
- Department of Biomedical Engineering, University of Michigan, Ann Arbor
| | - Jeffrey A. Beamish
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor
| | - Emily A. Margolis
- Department of Biomedical Engineering, University of Michigan, Ann Arbor
| | | | - Jan P. Stegemann
- Department of Biomedical Engineering, University of Michigan, Ann Arbor
| | - Andrew J. Putnam
- Department of Biomedical Engineering, University of Michigan, Ann Arbor
| |
Collapse
|
21
|
Marchal S, Andriantsitohaina R, Martinez MC. Biotherapeutic approaches against cardio-metabolic dysfunctions based on extracellular vesicles. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167095. [PMID: 38428684 DOI: 10.1016/j.bbadis.2024.167095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/29/2024] [Accepted: 02/19/2024] [Indexed: 03/03/2024]
Abstract
Among the different pathways involved in the cell-to-cell communication, extracellular vesicles (EVs) are defined as key players in the transport of different signalling molecules, such as lipids, proteins, and RNA, from the originating cells to specific target cells. The biogenesis and composition of EVs are complex and confer them a unique ability to more effectively reach tissues and cells as compared to other types of synthetic carriers. Owing to these properties, EVs have been suggested as new therapeutic tools for personalized medicine. Since cardiometabolic diseases have reached pandemic proportions, new therapies are needed to be developed. In this context, EVs appear as promising therapeutic tools against cardiometabolic disorders associated with obesity and diabetes. This review focuses on the latest research on preclinical applications of EVs for cardiometabolic diseases, and draw primarily on our experience in this area.
Collapse
Affiliation(s)
- Stéphane Marchal
- PhyMedExp, University of Montpellier, INSERM, CNRS, Montpellier, France
| | | | - M Carmen Martinez
- PhyMedExp, University of Montpellier, INSERM, CNRS, Montpellier, France.
| |
Collapse
|
22
|
Kink JA, Bellio MA, Forsberg MH, Lobo A, Thickens AS, Lewis BM, Ong IM, Khan A, Capitini CM, Hematti P. Large-scale bioreactor production of extracellular vesicles from mesenchymal stromal cells for treatment of acute radiation syndrome. Stem Cell Res Ther 2024; 15:72. [PMID: 38475968 DOI: 10.1186/s13287-024-03688-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 02/28/2024] [Indexed: 03/14/2024] Open
Abstract
BACKGROUND Hematopoietic acute radiation syndrome (H-ARS) occurring after exposure to ionizing radiation damages bone marrow causing cytopenias, increasing susceptibility to infections and death. We and others have shown that cellular therapies like human mesenchymal stromal cells (MSCs), or monocytes/macrophages educated ex-vivo with extracellular vesicles (EVs) from MSCs were effective in a lethal H-ARS mouse model. However, given the complexity of generating cellular therapies and the potential risks of using allogeneic products, development of an "off-the-shelf" cell-free alternative like EVs may have utility in conditions like H-ARS that require rapid deployment of available therapeutics. The purpose of this study was to determine the feasibility of producing MSC-derived EVs at large scale using a bioreactor and assess critical quality control attributes like identity, sterility, and potency in educating monocytes and promoting survival in a lethal H-ARS mouse model. METHODS EVs were isolated by ultracentrifugation from unprimed and lipopolysaccharide (LPS)-primed MSCs grown at large scale using a hollow fiber bioreactor and compared to a small scale system using flasks. The physical identity of EVs included a time course assessment of particle diameter, yield, protein content and surface marker profile by flow-cytometry. Comparison of the RNA cargo in EVs was determined by RNA-seq. Capacity of EVs to generate exosome educated monocytes (EEMos) was determined by qPCR and flow cytometry, and potency was assessed in vivo using a lethal ARS model with NSG mice. RESULTS Physical identity of EVs at both scales were similar but yields by volume were up to 38-fold more using a large-scale bioreactor system. RNA-seq indicated that flask EVs showed upregulated let-7 family and miR-143 micro-RNAs. EEMos educated with LPS-EVs at each scale were similar, showing increased gene expression of IL-6, IDO, FGF-2, IL-7, IL-10, and IL-15 and immunophenotyping consistent with a PD-L1 high, CD16 low, and CD86 low cell surface expression. Treatment with LPS-EVs manufactured at both scales were effective in the ARS model, improving survival and clinical scores through improved hematopoietic recovery. EVs from unprimed MSCs were less effective than LPS-EVs, with flask EVs providing some improved survival while bioreactor EVs provide no survival benefit. CONCLUSIONS LPS-EVs as an effective treatment for H-ARS can be produced using a scale-up development manufacturing process, representing an attractive off-the-shelf, cell-free therapy.
Collapse
Affiliation(s)
- John A Kink
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
- University of Wisconsin Carbone Cancer Center, 1111 Highland Ave, WIMR 4137, Madison, WI, USA
| | - Michael A Bellio
- Interdisciplinary Stem Cell Institute, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Matthew H Forsberg
- Department of Pediatrics, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Alexandra Lobo
- Department of Biostatistics and Medical Informatics, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Anna S Thickens
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Bryson M Lewis
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Irene M Ong
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
- University of Wisconsin Carbone Cancer Center, 1111 Highland Ave, WIMR 4137, Madison, WI, USA
- Department of Biostatistics and Medical Informatics, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
- Department of Obstetrics and Gynecology, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Aisha Khan
- Interdisciplinary Stem Cell Institute, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Christian M Capitini
- University of Wisconsin Carbone Cancer Center, 1111 Highland Ave, WIMR 4137, Madison, WI, USA.
- Department of Pediatrics, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA.
| | - Peiman Hematti
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA.
- University of Wisconsin Carbone Cancer Center, 1111 Highland Ave, WIMR 4137, Madison, WI, USA.
- Medical College of Wisconsin, 9200 W. Wisconsin Ave, Milwaukee, WI, 53326, USA.
| |
Collapse
|
23
|
Choubey U, Srinivas V, Trivedi YV, Garg N, Gupta V, Jain R. Regenerating the ailing heart: Stem cell therapies for hypoplastic left heart syndrome. Ann Pediatr Cardiol 2024; 17:124-131. [PMID: 39184114 PMCID: PMC11343389 DOI: 10.4103/apc.apc_24_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 04/14/2024] [Accepted: 04/14/2024] [Indexed: 08/27/2024] Open
Abstract
Hypoplastic left heart syndrome (HLHS) is a complex congenital heart defect (CHD) characterized by a spectrum of underdeveloped left-sided cardiac structures. It is a serious defect and warrants either 3-staged surgical palliation or a heart transplant. Despite numerous surgical advancements, long-term outcomes remain challenging and still have significant morbidity and mortality. There have been notable advancements in stem cell therapy for HLHS, including developments in diverse stem cell origins and methods of administration. Clinical trials have shown safety and potential benefits, including improved ventricular function, reduced heart failure, and fewer adverse events. Younger myocardium seems particularly receptive to stem cell signals, suggesting the importance of early intervention. This review explores the potential of emerging stem cell-based therapies as an adjunctive approach to improve the outcomes for HLHS patients.
Collapse
Affiliation(s)
- Udit Choubey
- Department of General Surgery, Shyam Shah Medical College, Rewa, Madhya Pradesh, India
| | - Varsha Srinivas
- Department of Internal Medicine, PES Institute of Medical Sciences and Research, Kuppam, Andhra Pradesh, India
| | - Yash Vardhan Trivedi
- Department of Internal Medicine, Jawaharlal Nehru Medical College, Ajmer, Rajasthan, India
| | - Nikita Garg
- Department of Pediatric College, Children’s Hospital of Michigan, Detroit, MI, USA
| | - Vasu Gupta
- Department of Internal Medicine, Cleveland Clinic Akron General, Akron, OH, USA
| | - Rohit Jain
- Penn State Milton S Hershey Medical Center, Hershey, PA, USA
| |
Collapse
|
24
|
Xu Z, Mao X, Lu X, Shi P, Ye J, Yang X, Fu Q, He C, Su D, Nie Y, Liu L, Wang C, Zhou B, Luo W, Cheng F, Chen H. Dual-Targeting Nanovesicles Carrying CSF1/CD47 Identified from Single-Cell Transcriptomics of Innate Immune Cells in Heart Transplant for Alleviating Acute Rejection. Adv Healthc Mater 2024; 13:e2302443. [PMID: 37962054 DOI: 10.1002/adhm.202302443] [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: 07/29/2023] [Revised: 10/28/2023] [Indexed: 11/15/2023]
Abstract
Although immunosuppressive drugs for targeting T cells are the standard of care in acute transplantation rejection, the role of innate immune cells should not be ignored. Here, single-cell RNA sequencing (scRNA-seq) and flow cytometry are performed to reveal the dynamic changes of innate immune cells within the acute rejection time and find a significantly-increased presence of Ly6G- Ly6C+ inflammatory macrophages and decreased presence of neutrophils among all types of immune cells. Next, to further explore potential targets regulating Ly6G- Ly6C+ inflammatory macrophages, scRNA-seq is used to analyze the reciprocal signaling of both neutrophils and macrophages, along with the surface genes of macrophages. It is found that activating colony-stimulating factor 1/ colony-stimulating factor 1 receptor (CSF1/CSF1R) andcluster of differentiation 47/signal regulatory protein α (CD47/SIRPα) signaling may serve as a strategy to relieve Ly6G- Ly6C+ inflammatory macrophage-mediated early graft rejection. To investigate this hypothesis, CSF1/CD47 dual-targeting nanovesicles (NVs) derived from IFN-γ-stimulated induced pluripotent stem cell-derived mesenchymal stem cells ( iPSC-MSCs )are designed and constructed. It is confirmed that CSF1/CD47 NVs synergistically induce the differentiation of Ly6G- Ly6C- M2 inhibitory macrophages by the CSF1/CSF1R pathway, and inhibit the phagocytosis of inflammatory macrophages and inflammatory response by the CD47/SIRPα pathway, ultimately relieving immune rejection. This study highlights the power of dual-targeting CSF1/CD47 NVs as an immunosuppressant against early innate immune responses with the potential for broad clinical applications.
Collapse
Affiliation(s)
- Zhanxue Xu
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Gongchang Road, Shenzhen, Guangdong, 518107, China
- Department of Pharmacy, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong, 518107, China
| | - Xiaofan Mao
- Clinical Research Institute, The First People's Hospital of Foshan, NO. 81 North of Lingnan Avenue, Foshan, Guangdong, 528000, China
| | - Xingyu Lu
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Gongchang Road, Shenzhen, Guangdong, 518107, China
| | - Peilin Shi
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Gongchang Road, Shenzhen, Guangdong, 518107, China
| | - Jingping Ye
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Gongchang Road, Shenzhen, Guangdong, 518107, China
| | - Xinrui Yang
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Gongchang Road, Shenzhen, Guangdong, 518107, China
| | - Qingling Fu
- Centre for Stem Cell Clinical Research and Application, The First Affiliated Hospital, Sun Yat-Sen University, Zhongshan 2nd Road, Guangzhou, Guangdong, 510080, China
| | - Chao He
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Gongchang Road, Shenzhen, Guangdong, 518107, China
| | - Dandan Su
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Gongchang Road, Shenzhen, Guangdong, 518107, China
| | - Yichu Nie
- Clinical Research Institute, The First People's Hospital of Foshan, NO. 81 North of Lingnan Avenue, Foshan, Guangdong, 528000, China
| | - Longshan Liu
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Zhongshan 2nd Road, Guangzhou, Guangdong, 510080, China
| | - Changxi Wang
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Zhongshan 2nd Road, Guangzhou, Guangdong, 510080, China
| | - Benjie Zhou
- Department of Pharmacy, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong, 518107, China
| | - Wei Luo
- Clinical Research Institute, The First People's Hospital of Foshan, NO. 81 North of Lingnan Avenue, Foshan, Guangdong, 528000, China
| | - Fang Cheng
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Gongchang Road, Shenzhen, Guangdong, 518107, China
| | - Hongbo Chen
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Gongchang Road, Shenzhen, Guangdong, 518107, China
| |
Collapse
|
25
|
Mikłosz A, Chabowski A. Efficacy of adipose-derived mesenchymal stem cell therapy in the treatment of chronic micro- and macrovascular complications of diabetes. Diabetes Obes Metab 2024; 26:793-808. [PMID: 38073423 DOI: 10.1111/dom.15375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 10/23/2023] [Accepted: 11/05/2023] [Indexed: 02/06/2024]
Abstract
Diabetes mellitus is a highly prevalent disease characterized by hyperglycaemia that damages the vascular system, leading to micro- (retinopathy, neuropathy, nephropathy) and macrovascular diseases (cardiovascular disease). There are also secondary complications of diabetes (cardiomyopathy, erectile dysfunction or diabetic foot ulcers). Stem cell-based therapies have become a promising tool targeting diabetes symptoms and its chronic complications. Among all stem cells, adipose-derived mesenchymal stem cells (ADMSCs) are of great importance because of their abundance, non-invasive isolation and no ethical limitations. Characteristics that make ADMSCs good candidates for cell-based therapy are their wide immunomodulatory properties and paracrine activities through the secretion of an array of growth factors, chemokines, cytokines, angiogenic factors and anti-apoptotic molecules. Besides, after transplantation, ADMSCs show great ex vivo expansion capacity and differentiation to other cell types, including insulin-producing cells, cardiomyocytes, chondrocytes, hepatocyte-like cells, neurons, endothelial cells, photoreceptor-like cells, or astrocytes. Preclinical studies have shown that ADMSC-based therapy effectively improved visual acuity, ameliorated polyneuropathy and foot ulceration, arrested the development and progression of diabetic kidney disease, or alleviated the diabetes-induced cardiomyocyte hypertrophy. However, despite the positive results obtained in animal models, there are still several challenges that need to be overcome before the results of preclinical studies can be translated into clinical applications. To date, there are several clinical trials or ongoing trials using ADMSCs in the treatment of diabetic complications, most of them in the treatment of diabetic foot ulcers. This narrative review summarizes the most recent outcomes on the usage of ADMSCs in the treatment of long-term complications of diabetes in both animal models and clinical trials.
Collapse
Affiliation(s)
- Agnieszka Mikłosz
- Department of Physiology, Medical University of Bialystok, Bialystok, Poland
| | - Adrian Chabowski
- Department of Physiology, Medical University of Bialystok, Bialystok, Poland
| |
Collapse
|
26
|
Gong ZT, Xiong YY, Ning Y, Tang RJ, Xu JY, Jiang WY, Li XS, Zhang LL, Chen C, Pan Q, Hu MJ, Xu J, Yang YJ. Nicorandil-Pretreated Mesenchymal Stem Cell-Derived Exosomes Facilitate Cardiac Repair After Myocardial Infarction via Promoting Macrophage M2 Polarization by Targeting miR-125a-5p/TRAF6/IRF5 Signaling Pathway. Int J Nanomedicine 2024; 19:2005-2024. [PMID: 38469055 PMCID: PMC10926597 DOI: 10.2147/ijn.s441307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 02/18/2024] [Indexed: 03/13/2024] Open
Abstract
Background Exosomes derived from bone marrow mesenchymal stem cells (MSC-exo) have been considered as a promising cell-free therapeutic strategy for ischemic heart disease. Cardioprotective drug pretreatment could be an effective approach to improve the efficacy of MSC-exo. Nicorandil has long been used in clinical practice for cardioprotection. This study aimed to investigate whether the effects of exosomes derived from nicorandil pretreated MSC (MSCNIC-exo) could be enhanced in facilitating cardiac repair after acute myocardial infarction (AMI). Methods MSCNIC-exo and MSC-exo were collected and injected into the border zone of infarcted hearts 30 minutes after coronary ligation in rats. Macrophage polarization was detected 3 days post-infarction, cardiac function as well as histological pathology were measured on the 28th day after AMI. Macrophages were separated from the bone marrow of rats for in vitro model. Exosomal miRNA sequencing was conducted to identify differentially expressed miRNAs between MSCNIC-exo and MSC-exo. MiRNA mimics and inhibitors were transfected to MSCs or macrophages to explore the specific mechanism. Results Compared to MSC-exo, MSCNIC-exo showed superior therapeutic effects on cardiac functional and structural recovery after AMI and markedly elevated the ratio of CD68+ CD206+/ CD68+cells in infarcted hearts 3 days post-infarction. The notable ability of MSCNIC-exo to promote macrophage M2 polarization was also confirmed in vitro. Exosomal miRNA sequencing and both in vivo and in vitro experiments identified and verified that miR-125a-5p was an effector of the roles of MSCNIC-exo in vivo and in vitro. Furthermore, we found miR-125a-5p promoted macrophage M2 polarization by inhibiting TRAF6/IRF5 signaling pathway. Conclusion This study suggested that MSCNIC-exo could markedly facilitate cardiac repair post-infarction by promoting macrophage M2 polarization by upregulating miR-125a-5p targeting TRAF6/IRF5 signaling pathway, which has great potential for clinical translation.
Collapse
Affiliation(s)
- Zhao-Ting Gong
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100037, People’s Republic of China
| | - Yu-Yan Xiong
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100037, People’s Republic of China
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
| | - Yu Ning
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100037, People’s Republic of China
| | - Rui-Jie Tang
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100037, People’s Republic of China
| | - Jun-Yan Xu
- Department of Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, People’s Republic of China
| | - Wen-Yang Jiang
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100037, People’s Republic of China
| | - Xiao-Song Li
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100037, People’s Republic of China
| | - Li-Li Zhang
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100037, People’s Republic of China
| | - Cheng Chen
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100037, People’s Republic of China
| | - Qi Pan
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100037, People’s Republic of China
| | - Meng-Jin Hu
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100037, People’s Republic of China
| | - Jing Xu
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100037, People’s Republic of China
| | - Yue-Jin Yang
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100037, People’s Republic of China
| |
Collapse
|
27
|
Sun Z, Cai Y, Chen Y, Jin Q, Zhang Z, Zhang L, Li Y, Huang L, Wang J, Yang Y, Lv Q, Han Z, Xie M, Zhu X. Ultrasound-targeted microbubble destruction promotes PDGF-primed bone mesenchymal stem cell transplantation for myocardial protection in acute Myocardial Infarction in rats. J Nanobiotechnology 2023; 21:481. [PMID: 38102643 PMCID: PMC10725038 DOI: 10.1186/s12951-023-02204-7] [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: 06/21/2023] [Accepted: 11/07/2023] [Indexed: 12/17/2023] Open
Abstract
BACKGROUND Ultrasound-targeted microbubble destruction (UTMD) has emerged as a promising strategy for the targeted delivery of bone marrow mesenchymal stem cells (MSCs) to the ischemic myocardium. However, the limited migration capacity and poor survival of MSCs remains a major therapeutic barrier. The present study was performed to investigate the synergistic effect of UTMD with platelet-derived growth factor BB (PDGF-BB) on the homing of MSCs for acute myocardial infarction (AMI). METHODS MSCs from male donor rats were treated with PDGF-BB, and a novel microbubble formulation was prepared using a thin-film hydration method. In vivo, MSCs with or without PDGF-BB pretreatment were transplanted by UTMD after inducing AMI in experimental rats. The therapeutic efficacy of PDGF-BB-primed MSCs on myocardial apoptosis, angiogenesis, cardiac function and scar repair was estimated. The effects and molecular mechanisms of PDGF-BB on MSC migration and survival were explored in vitro. RESULTS The results showed that the biological effects of UTMD increased the local levels of stromal-derived factor-1 (SDF-1), which promoted the migration of transplanted MSCs to the ischemic region. Compared with UTMD alone, UTMD combined with PDGF-BB pretreatment significantly increased the cardiac homing of MSCs, which subsequently reduced myocardial apoptosis, promoted neovascularization and tissue repair, and increased cardiac function 30 days after MI. The vitro results demonstrated that PDGF-BB enhanced MSC migration and protected these cells from H2O2-induced apoptosis. Mechanistically, PDGF-BB pretreatment promoted MSC migration and inhibited H2O2-induced MSC apoptosis via activation of the phosphatidylinositol 3-kinase/serine-threonine kinase (PI3K/Akt) pathway. Furthermore, crosstalk between PDGF-BB and stromal-derived factor-1/chemokine receptor 4 (SDF-1/CXCR4) is involved in the PI3K/AKT signaling pathway. CONCLUSION The present study demonstrated that UTMD combined with PDGF-BB treatment could enhance the homing ability of MSCs, thus alleviating AMI in rats. Therefore, UTMD combined with PDGF-BB pretreatment may offer exciting therapeutic opportunities for strengthening MSC therapy in ischemic diseases.
Collapse
Grants
- 81701716; 82230066, 81922033, 81727805, 82171961, 81801715, 81801716 National Natural Science Foundation of China
- 81701716; 82230066, 81922033, 81727805, 82171961, 81801715, 81801716 National Natural Science Foundation of China
- 81701716; 82230066, 81922033, 81727805, 82171961, 81801715, 81801716 National Natural Science Foundation of China
- 81701716; 82230066, 81922033, 81727805, 82171961, 81801715, 81801716 National Natural Science Foundation of China
- 81701716; 82230066, 81922033, 81727805, 82171961, 81801715, 81801716 National Natural Science Foundation of China
- 2018CFB568 National Natural Science Foundation of Hubei
Collapse
Affiliation(s)
- Zhenxing Sun
- Anhui Medical University, Hefei, 230031, China
- Department of Ultrasound, The First Affiliated Hospital of Wannan Medical College, Wuhu, 241001, China
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, 430022, Wuhan, China
| | - Yu Cai
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, 430022, Wuhan, China
| | - Yihan Chen
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, 430022, Wuhan, China
| | - Qiaofeng Jin
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, 430022, Wuhan, China
| | - Ziming Zhang
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, 430022, Wuhan, China
| | - Li Zhang
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, 430022, Wuhan, China
| | - Yuman Li
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, 430022, Wuhan, China
| | - Lei Huang
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, 430022, Wuhan, China
| | - Jing Wang
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, 430022, Wuhan, China
| | - Yali Yang
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, 430022, Wuhan, China
| | - Qing Lv
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, 430022, Wuhan, China
| | - Zhengyang Han
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Province Key Laboratory of Molecular Imaging, 430022, Wuhan, China.
- Department of Ultrasound, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| | - Mingxing Xie
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Province Key Laboratory of Molecular Imaging, 430022, Wuhan, China.
| | - Xiangming Zhu
- Anhui Medical University, Hefei, 230031, China.
- Department of Ultrasound, The First Affiliated Hospital of Wannan Medical College, Wuhu, 241001, China.
| |
Collapse
|
28
|
Xu S, Xu C, Xu J, Zhang K, Zhang H. Macrophage Heterogeneity and Its Impact on Myocardial Ischemia-Reperfusion Injury: An Integrative Review. J Inflamm Res 2023; 16:5971-5987. [PMID: 38088942 PMCID: PMC10712254 DOI: 10.2147/jir.s436560] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 11/30/2023] [Indexed: 10/21/2024] Open
Abstract
The coronary reperfusion following acute myocardial infarction can paradoxically trigger myocardial ischemia-reperfusion (IR) injury. This complex phenomenon involves the intricate interplay of different subsets of macrophages. These macrophages are crucial players in the post-infarction inflammatory response and subsequent myocardial anti-inflammatory repair. However, their diverse functions can lead to both beneficial and detrimental effects. On one hand, these macrophages play a crucial role in orchestrating the inflammatory response, aiding in the clearance of cellular debris and initiating tissue repair mechanisms. On the other hand, their excessive infiltration and activation can contribute to the perpetuation of the inflammatory cascade, leading to additional myocardial injury and adverse cardiac remodeling. Multiple mechanisms contribute to the IR injury mediated by macrophages, including oxidative stress, apoptosis, and autophagy. These processes further exacerbate the damage to the already vulnerable myocardial tissue. To address this delicate balance, therapeutic strategies aiming to target and modulate macrophage polarization and function are being explored. By fine-tuning the immune inflammatory response, such interventions hold promise in mitigating post-infarction myocardial injury and fostering a more favorable environment for myocardial healing and recovery. Through advancements in this area of research, potential anti-inflammatory interventions may pave the way for improved clinical outcomes and better management of patients after acute myocardial infarction.
Collapse
Affiliation(s)
- Shuwan Xu
- Cardiovascular Department, the Eighth Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, Guangdong, People’s Republic of China
- Department of Cardiology, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Guangzhou, Guangdong, People’s Republic of China
| | - Cong Xu
- Cardiovascular Department, the Eighth Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, Guangdong, People’s Republic of China
| | - Jiahua Xu
- Cardiovascular Department, the Eighth Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, Guangdong, People’s Republic of China
| | - Kun Zhang
- Department of Cardiology, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Guangzhou, Guangdong, People’s Republic of China
| | - Huanji Zhang
- Cardiovascular Department, the Eighth Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, Guangdong, People’s Republic of China
| |
Collapse
|
29
|
Hao T, Ji G, Qian M, Li QX, Huang H, Deng S, Liu P, Deng W, Wei Y, He J, Wang S, Gao W, Li T, Cheng J, Tian J, Pan L, Gao F, Li Z, Zhao Q. Intracellular delivery of nitric oxide enhances the therapeutic efficacy of mesenchymal stem cells for myocardial infarction. SCIENCE ADVANCES 2023; 9:eadi9967. [PMID: 38019911 PMCID: PMC10686553 DOI: 10.1126/sciadv.adi9967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 10/27/2023] [Indexed: 12/01/2023]
Abstract
Cell therapy by autologous mesenchymal stem cells (MSCs) is a clinically acceptable strategy for treating various diseases. Unfortunately, the therapeutic efficacy is largely affected by the low quality of MSCs collected from patients. Here, we showed that the gene expression of MSCs from patients with diabetes was differentially regulated compared to that of MSCs from healthy controls. Then, MSCs were genetically engineered to catalyze an NO prodrug to release NO intracellularly. Compared to extracellular NO conversion, intracellular NO delivery effectively prolonged survival and enhanced the paracrine function of MSCs, as demonstrated by in vitro and in vivo assays. The enhanced therapeutic efficacy of engineered MSCs combined with intracellular NO delivery was further confirmed in mouse and rat models of myocardial infarction, and a clinically relevant cell administration paradigm through secondary thoracotomy has been attempted.
Collapse
Affiliation(s)
- Tian Hao
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials (Ministry of Education), Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Guangbo Ji
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials (Ministry of Education), Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Meng Qian
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials (Ministry of Education), Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Qiu Xuan Li
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Haoyan Huang
- Nankai University School of Medicine, Tianjin 300071, China
| | - Shiyu Deng
- The Key Laboratory of Weak-Light Nonlinear Photonics of Education Ministry, School of Physics and TEDA Institute of Applied Physics, Nankai University, Tianjin 300071, China
| | - Pei Liu
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials (Ministry of Education), Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Weiliang Deng
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials (Ministry of Education), Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Yongzhen Wei
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials (Ministry of Education), Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Ju He
- Department of Vascular Surgery, Tianjin First Central Hospital, Nankai University, Tianjin 300192, China
| | - Shusen Wang
- Organ Transplant Center, NHC Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Nankai University, Tianjin, China
| | - Wenqing Gao
- Department of Heart Center, The Third Central Hospital of Tianjin, Nankai University, Tianjin, China
| | - Tong Li
- Department of Heart Center, The Third Central Hospital of Tianjin, Nankai University, Tianjin, China
| | - Jiansong Cheng
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Nankai University, Tianjin 300353, China
| | - Jinwei Tian
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Leiting Pan
- The Key Laboratory of Weak-Light Nonlinear Photonics of Education Ministry, School of Physics and TEDA Institute of Applied Physics, Nankai University, Tianjin 300071, China
| | - Fei Gao
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Zongjin Li
- Nankai University School of Medicine, Tianjin 300071, China
| | - Qiang Zhao
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials (Ministry of Education), Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin 300071, China
| |
Collapse
|
30
|
Ranjan P, Colin K, Dutta RK, Verma SK. Challenges and future scope of exosomes in the treatment of cardiovascular diseases. J Physiol 2023; 601:4873-4893. [PMID: 36398654 PMCID: PMC10192497 DOI: 10.1113/jp282053] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 10/21/2022] [Indexed: 07/28/2023] Open
Abstract
Exosomes are nanosized vesicles that carry biologically diverse molecules for intercellular communication. Researchers have been trying to engineer exosomes for therapeutic purposes by using different approaches to deliver biologically active molecules to the various target cells efficiently. Recent technological advances may allow the biodistribution and pharmacokinetics of exosomes to be modified to meet scientific needs with respect to specific diseases. However, it is essential to determine an exosome's optimal dosage and potential side effects before its clinical use. Significant breakthroughs have been made in recent decades concerning exosome labelling and imaging techniques. These tools provide in situ monitoring of exosome biodistribution and pharmacokinetics and pinpoint targetability. However, because exosomes are nanometres in size and vary significantly in contents, a deeper understanding is required to ensure accurate monitoring before they can be applied in clinical settings. Different research groups have established different approaches to elucidate the roles of exosomes and visualize their spatial properties. This review covers current and emerging strategies for in vivo and in vitro exosome imaging and tracking for potential studies.
Collapse
Affiliation(s)
- Prabhat Ranjan
- Department of Medicine, Division of Cardiovascular Disease, The University of Alabama at Birmingham, Birmingham, AL-35233
| | - Karen Colin
- Department of Medicine, Division of Cardiovascular Disease, The University of Alabama at Birmingham, Birmingham, AL-35233
- UAB School of Health Professions, The University of Alabama at Birmingham, Birmingham, AL
| | - Roshan Kumar Dutta
- Department of Medicine, Division of Cardiovascular Disease, The University of Alabama at Birmingham, Birmingham, AL-35233
| | - Suresh Kumar Verma
- Department of Medicine, Division of Cardiovascular Disease, The University of Alabama at Birmingham, Birmingham, AL-35233
- Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
31
|
Zhang H, Wan X, Tian J, An Z, Liu L, Zhao X, Zhou Y, Zhang L, Ge C, Song X. The therapeutic efficacy and clinical translation of mesenchymal stem cell-derived exosomes in cardiovascular diseases. Biomed Pharmacother 2023; 167:115551. [PMID: 37783149 DOI: 10.1016/j.biopha.2023.115551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 09/08/2023] [Accepted: 09/18/2023] [Indexed: 10/04/2023] Open
Abstract
Exosomes, mainly derived from mesenchymal stem cells, provide a good reference for cardiac function repair and clinical application in cardiac and vascular diseases by regulating cardiomyocyte viability, inflammatory levels, angiogenesis, and ventricular remodeling after a heart injury. This review presents the cardioprotective efficacy of mesenchymal stem cell-originated exosomes and explores the underlying molecular mechanisms. Furthermore, we expound on several efficient approaches to transporting exosomes into the heart in clinical application and comment on the advantages and disadvantages of each method.
Collapse
Affiliation(s)
- Huan Zhang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, PR China
| | - Xueqi Wan
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, PR China
| | - Jinfan Tian
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, PR China
| | - Ziyu An
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, PR China
| | - Libo Liu
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, PR China; The Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong 271000, PR China
| | - Xin Zhao
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, PR China
| | - Yuquan Zhou
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, PR China
| | - Lijun Zhang
- Department of Radiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, PR China
| | - Changjiang Ge
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, PR China.
| | - Xiantao Song
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, PR China.
| |
Collapse
|
32
|
Li D, Liu L, He X, Wang N, Sun R, Li X, Yu T, Chu XM. Roles of long non-coding RNAs in angiogenesis-related diseases: Focusing on non-neoplastic aspects. Life Sci 2023; 330:122006. [PMID: 37544376 DOI: 10.1016/j.lfs.2023.122006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 06/28/2023] [Accepted: 08/03/2023] [Indexed: 08/08/2023]
Abstract
Angiogenesis is a key process in organ and tissue morphogenesis, as well as growth during human development, and is coordinated by pro- and anti-angiogenic factors. When this balance is affected, the related physiological and pathological changes lead to disease. Long non-coding RNAs (lncRNAs) are an important class of non-coding RNAs that do not encode proteins, but play a dynamic role in regulating gene expression. LncRNAs have been reported to be extensively involved in angiogenesis, particularly tumor angiogenesis. The non-tumor aspects have received relatively little attention and summary, but there is a broad space for research and exploration on lncRNA-targeted angiogenesis in this area. In this review, we focus on lncRNAs in angiogenesis-related diseases other than tumors, such as atherosclerosis, myocardial infarction, stroke, diabetic complications, hypertension, osteoporosis, dermatosis, as well as, endocrine, neurological, and other systemic disorders. Moreover, multiple cell types have been implicated in lncRNA-targeted angiogenesis, but only endothelial cells have attracted widespread attention. Thus, we explore the roles of other cells. Finally, we summarize the potential research directions in the area of lncRNAs and angiogenesis that can be undertaken by combining cutting-edge technology and interdisciplinary research, which will provide new insights into the involvement of lncRNAs in angiogenesis-related diseases.
Collapse
Affiliation(s)
- Daisong Li
- Department of Cardiology, The Affiliated Hospital of Qingdao University, No. 59 Haier Road, Qingdao 266100, People's Republic of China
| | - Lili Liu
- School of Basic Medicine, Qingdao University, No. 308 Ningxia Road, Qingdao, People's Republic of China
| | - Xiangqin He
- Department of Echocardiography, The Affiliated Hospital of Qingdao University, Qingdao, 266000, People's Republic of China
| | - Ni Wang
- Department of Cardiology, The Affiliated Hospital of Qingdao University, No. 59 Haier Road, Qingdao 266100, People's Republic of China
| | - Ruicong Sun
- Department of Cardiology, The Affiliated Hospital of Qingdao University, No. 59 Haier Road, Qingdao 266100, People's Republic of China
| | - Xiaolu Li
- Department of Echocardiography, The Affiliated Hospital of Qingdao University, Qingdao, 266000, People's Republic of China
| | - Tao Yu
- Institute for Translational Medicine, Qingdao University, No. 38 Dengzhou Road, 266021, People's Republic of China.
| | - Xian-Ming Chu
- Department of Cardiology, The Affiliated Hospital of Qingdao University, No. 59 Haier Road, Qingdao 266100, People's Republic of China; Department of Cardiology, The Affiliated Cardiovascular Hospital of Qingdao University, No. 5 Zhiquan Road, Qingdao 266000, People's Republic of China.
| |
Collapse
|
33
|
Hu HJ, Xiao XR, Li T, Liu DM, Geng X, Han M, Cui W. Integrin beta 3-overexpressing mesenchymal stromal cells display enhanced homing and can reduce atherosclerotic plaque. World J Stem Cells 2023; 15:931-946. [PMID: 37900938 PMCID: PMC10600744 DOI: 10.4252/wjsc.v15.i9.931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 06/22/2023] [Accepted: 08/23/2023] [Indexed: 09/25/2023] Open
Abstract
BACKGROUND Umbilical cord (UC) mesenchymal stem cell (MSC) transplantation is a potential therapeutic intervention for atherosclerotic vascular disease. Integrin beta 3 (ITGB3) promotes cell migration in several cell types. However, whether ITGB-modified MSCs can migrate to plaque sites in vivo and play an anti-atherosclerotic role remains unclear. AIM To investigate whether ITGB3-overexpressing MSCs (MSCsITGB3) would exhibit improved homing efficacy in atherosclerosis. METHODS UC MSCs were isolated and expanded. Lentiviral vectors encoding ITGB3 or green fluorescent protein (GFP) as control were transfected into MSCs. Sixty male apolipoprotein E-/- mice were acquired from Beijing Vital River Lab Animal Technology Co., Ltd and fed with a high-fat diet (HFD) for 12 wk to induce the formation of atherosclerotic lesions. These HFD-fed mice were randomly separated into three clusters. GFP-labeled MSCs (MSCsGFP) or MSCsITGB3 were transplanted into the mice intravenously via the tail vein. Immunofluorescence staining, Oil red O staining, histological analyses, western blotting, enzyme-linked immunosorbent assay, and quantitative real-time polymerase chain reaction were used for the analyses. RESULTS ITGB3 modified MSCs successfully differentiated into the "osteocyte" and "adipocyte" phenotypes and were characterized by positive expression (> 91.3%) of CD29, CD73, and CD105 and negative expression (< 1.35%) of CD34 and Human Leukocyte Antigen-DR. In a transwell assay, MSCsITGB3 showed significantly faster migration than MSCsGFP. ITGB3 overexpression had no effects on MSC viability, differentiation, and secretion. Immunofluorescence staining revealed that ITGB3 overexpression substantially enhanced the homing of MSCs to plaque sites. Oil red O staining and histological analyses further confirmed the therapeutic effects of MSCsITGB3, significantly reducing the plaque area. Enzyme-linked immunosorbent assay and quantitative real-time polymerase chain reaction revealed that MSCITGB3 transplantation considerably decreased the inflammatory response in pathological tissues by improving the dynamic equilibrium of pro- and anti-inflammatory cytokines. CONCLUSION These results showed that ITGB3 overexpression enhanced the MSC homing ability, providing a potential approach for MSC delivery to plaque sites, thereby optimizing their therapeutic effects.
Collapse
Affiliation(s)
- Hai-Juan Hu
- First Division, Department of Cardiology, The Second Hospital of Hebei Medical University and Institute of Cardiocerebrovascular Disease of Hebei Province, Shijiazhuang 050000, Hebei Province, China
| | - Xue-Ru Xiao
- Department of Obstetrics, Shijiazhuang People's Hospital, Shijiazhuang 050030, Hebei Province, China
| | - Tong Li
- First Division, Department of Cardiology, The Second Hospital of Hebei Medical University and Institute of Cardiocerebrovascular Disease of Hebei Province, Shijiazhuang 050000, Hebei Province, China
| | - De-Min Liu
- First Division, Department of Cardiology, The Second Hospital of Hebei Medical University and Institute of Cardiocerebrovascular Disease of Hebei Province, Shijiazhuang 050000, Hebei Province, China
| | - Xue Geng
- First Division, Department of Cardiology, The Second Hospital of Hebei Medical University and Institute of Cardiocerebrovascular Disease of Hebei Province, Shijiazhuang 050000, Hebei Province, China
| | - Mei Han
- Key Laboratory of Medical Biotechnology of Hebei Province, Department of Biochemistry and Molecular Biology, College of Basic Medicine, Cardiovascular Medical Science Center, Hebei Medical University, Shijiazhuang 050017, Hebei Province, China
| | - Wei Cui
- First Division, Department of Cardiology, The Second Hospital of Hebei Medical University and Institute of Cardiocerebrovascular Disease of Hebei Province, Shijiazhuang 050000, Hebei Province, China.
| |
Collapse
|
34
|
Bernava G, Iop L. Advances in the design, generation, and application of tissue-engineered myocardial equivalents. Front Bioeng Biotechnol 2023; 11:1247572. [PMID: 37811368 PMCID: PMC10559975 DOI: 10.3389/fbioe.2023.1247572] [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: 06/26/2023] [Accepted: 08/29/2023] [Indexed: 10/10/2023] Open
Abstract
Due to the limited regenerative ability of cardiomyocytes, the disabling irreversible condition of myocardial failure can only be treated with conservative and temporary therapeutic approaches, not able to repair the damage directly, or with organ transplantation. Among the regenerative strategies, intramyocardial cell injection or intravascular cell infusion should attenuate damage to the myocardium and reduce the risk of heart failure. However, these cell delivery-based therapies suffer from significant drawbacks and have a low success rate. Indeed, cardiac tissue engineering efforts are directed to repair, replace, and regenerate native myocardial tissue function. In a regenerative strategy, biomaterials and biomimetic stimuli play a key role in promoting cell adhesion, proliferation, differentiation, and neo-tissue formation. Thus, appropriate biochemical and biophysical cues should be combined with scaffolds emulating extracellular matrix in order to support cell growth and prompt favorable cardiac microenvironment and tissue regeneration. In this review, we provide an overview of recent developments that occurred in the biomimetic design and fabrication of cardiac scaffolds and patches. Furthermore, we sift in vitro and in situ strategies in several preclinical and clinical applications. Finally, we evaluate the possible use of bioengineered cardiac tissue equivalents as in vitro models for disease studies and drug tests.
Collapse
Affiliation(s)
| | - Laura Iop
- Department of Cardiac Thoracic Vascular Sciences and Public Health, Padua Medical School, University of Padua, Padua, Italy
| |
Collapse
|
35
|
Li N, Liu G, Gao H, Wu Q, Meng J, Wang F, Jiang S, Chen M, Xu W, Zhang Y, Wang Y, Feng Y, Liu J, Xu C, Lu H. Geriatric syndromes, chronic inflammation, and advances in the management of frailty: A review with new insights. Biosci Trends 2023; 17:262-270. [PMID: 37612125 DOI: 10.5582/bst.2023.01184] [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] [Indexed: 08/25/2023]
Abstract
As people age, geriatric syndromes characterized by frailty significantly impact both clinical practice and public health. Aging weakens people's immune functions, leading to chronic low-grade inflammation that ultimately contributes to the development of frailty. Effectively managing geriatric syndromes and frailty can help alleviate the economic burden of an aging population. This review delves into the intricate relationship among aging, infection-induced inflammation, chronic inflammation, and frailty. In addition, it analyzes various approaches and interventions to address frailty, such as smart rehabilitation programs and stem-cell treatments, offering promising solutions in this new era. Given the importance of this topic, further research into the mechanisms of frailty is crucial. Equally essential is the devising of relevant measures to delay its onset and the formulation of comprehensive clinical, research, and public health strategies to enhance the quality of life for elderly individuals.
Collapse
Affiliation(s)
- Niuniu Li
- Department of Geriatric Medicine, National Clinical Research Center for Infectious Diseases, the Third People's Hospital of Shenzhen, Shenzhen, Guangdong, China
| | - Gaolin Liu
- Washington University in St. Louis, St. Louis, United States
| | - Hong Gao
- Department of Geriatric Medicine, National Clinical Research Center for Infectious Diseases, the Third People's Hospital of Shenzhen, Shenzhen, Guangdong, China
| | - Qiang Wu
- Department of Geriatric Medicine, National Clinical Research Center for Infectious Diseases, the Third People's Hospital of Shenzhen, Shenzhen, Guangdong, China
| | - Juan Meng
- Department of Geriatric Medicine, National Clinical Research Center for Infectious Diseases, the Third People's Hospital of Shenzhen, Shenzhen, Guangdong, China
| | - Fei Wang
- Department of Geriatric Medicine, National Clinical Research Center for Infectious Diseases, the Third People's Hospital of Shenzhen, Shenzhen, Guangdong, China
| | - Siwei Jiang
- Department of Geriatric Medicine, National Clinical Research Center for Infectious Diseases, the Third People's Hospital of Shenzhen, Shenzhen, Guangdong, China
| | - Meixia Chen
- Department of Geriatric Medicine, National Clinical Research Center for Infectious Diseases, the Third People's Hospital of Shenzhen, Shenzhen, Guangdong, China
| | - Wenhui Xu
- Department of Geriatric Medicine, National Clinical Research Center for Infectious Diseases, the Third People's Hospital of Shenzhen, Shenzhen, Guangdong, China
| | - Yifan Zhang
- Department of Geriatric Medicine, National Clinical Research Center for Infectious Diseases, the Third People's Hospital of Shenzhen, Shenzhen, Guangdong, China
| | - Yanjun Wang
- Department of Geriatric Medicine, National Clinical Research Center for Infectious Diseases, the Third People's Hospital of Shenzhen, Shenzhen, Guangdong, China
| | - Yingqian Feng
- Department of Geriatric Medicine, National Clinical Research Center for Infectious Diseases, the Third People's Hospital of Shenzhen, Shenzhen, Guangdong, China
| | - Juncai Liu
- Department of Geriatric Medicine, National Clinical Research Center for Infectious Diseases, the Third People's Hospital of Shenzhen, Shenzhen, Guangdong, China
| | - Cheng Xu
- Department of Geriatric Medicine, National Clinical Research Center for Infectious Diseases, the Third People's Hospital of Shenzhen, Shenzhen, Guangdong, China
| | - Hongzhou Lu
- Department of Infectious Diseases, National Clinical Research Center for Infectious Diseases, the Third People's Hospital of Shenzhen, Shenzhen, Guangdong, China
| |
Collapse
|
36
|
Sundin A, Ionescu SI, Balkan W, Hare JM. Mesenchymal STRO-1/STRO-3 + precursor cells for the treatment of chronic heart failure with reduced ejection fraction. Future Cardiol 2023; 19:567-581. [PMID: 37933628 PMCID: PMC10652293 DOI: 10.2217/fca-2023-0081] [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: 06/01/2023] [Accepted: 08/30/2023] [Indexed: 11/08/2023] Open
Abstract
The heart is susceptible to proinflammatory and profibrotic responses after myocardial injury, leading to further worsening of cardiac dysfunction. Important developments in the management of heart failure with reduced ejection fraction have reduced morbidity and mortality; however, these therapies focus on optimizing cardiac function through hemodynamic and neurohormonal pathways and not by repairing the underlying cardiac injury. The potential of cell-based therapy to reverse cardiac injury has received substantial attention. Herein are examined the phase II and III studies of bone marrow-derived mesenchymal STRO-1+ or STRO-1/STRO-3+ precursor cells in patients with ischemic and nonischemic heart failure with reduced ejection fraction, addressing the safety and efficacy of cell-based therapy throughout multiple clinical trials, the optimal dose and the steps toward revolutionizing the treatment of heart failure.
Collapse
Affiliation(s)
- Andrew Sundin
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Simona I Ionescu
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Wayne Balkan
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Joshua M Hare
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
37
|
Mohamad Yusoff F, Higashi Y. Mesenchymal Stem/Stromal Cells for Therapeutic Angiogenesis. Cells 2023; 12:2162. [PMID: 37681894 PMCID: PMC10486439 DOI: 10.3390/cells12172162] [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: 07/13/2023] [Revised: 08/18/2023] [Accepted: 08/25/2023] [Indexed: 09/09/2023] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) are known to possess medicinal properties to facilitate vascular regeneration. Recent advances in the understanding of the utilities of MSCs in physiological/pathological tissue repair and technologies in isolation, expansion, and enhancement strategies have led to the use of MSCs for vascular disease-related treatments. Various conditions, including chronic arterial occlusive disease, diabetic ulcers, and chronic wounds, cause significant morbidity in patients. Therapeutic angiogenesis by cell therapy has led to the possibilities of treatment options in promoting angiogenesis, treating chronic wounds, and improving amputation-free survival. Current perspectives on the options for the use of MSCs for therapeutic angiogenesis in vascular research and in medicine, either as a monotherapy or in combination with conventional interventions, for treating patients with peripheral artery diseases are discussed in this review.
Collapse
Affiliation(s)
- Farina Mohamad Yusoff
- Department of Regenerative Medicine, Division of Radiation Medical Science, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima 734-8553, Japan;
| | - Yukihito Higashi
- Department of Regenerative Medicine, Division of Radiation Medical Science, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima 734-8553, Japan;
- Division of Regeneration and Medicine, Hiroshima University Hospital, Hiroshima 734-8551, Japan
| |
Collapse
|
38
|
Xu L, Mu J, Ma Z, Lin P, Xia F, Hu X, Wu J, Cao J, Liu S, Huang T, Ling D, Gao J, Li F. Nanozyme-Integrated Thermoresponsive In Situ Forming Hydrogel Enhances Mesenchymal Stem Cell Viability and Paracrine Effect for Efficient Spinal Cord Repair. ACS APPLIED MATERIALS & INTERFACES 2023; 15:37193-37204. [PMID: 37493513 DOI: 10.1021/acsami.3c06189] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/27/2023]
Abstract
Mesenchymal stem cell (MSC)-based therapy has emerged as a promising strategy for the treatment of spinal cord injury (SCI). However, the hostile microenvironment of SCI, which can adversely affect the survival and paracrine effect of the implanted MSCs, severely limits the therapeutic efficacy of this approach. Here, we report on a ceria nanozyme-integrated thermoresponsive in situ forming hydrogel (CeNZ-gel) that can enable dual enhancement of MSC viability and paracrine effect, leading to highly efficient spinal cord repair. The sol-gel transition property of the CeNZ-gel at body temperature ensures uniform coverage of the hydrogel in injured spinal cord tissues. Our results demonstrate that the CeNZ-gel significantly increases the viability of transplanted MSCs in the microenvironment by attenuating oxidative stress and, more importantly, promotes the secretion of angiogenic factors from MSCs by inducing autophagy of MSCs. The synergy between the oxidative stress-relieving effect of CeNZs and the paracrine effect of MSCs accelerates angiogenesis, nerve repair, and motor function recovery after SCI, providing an efficient strategy for MSC-based SCI therapy.
Collapse
Affiliation(s)
- Lilan Xu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jiafu Mu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Zhiyuan Ma
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Peihua Lin
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Fan Xia
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xi Hu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Jiahe Wu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jian Cao
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Shanbiao Liu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Tianchen Huang
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Daishun Ling
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Frontiers Science Center for Transformative Molecules, School of Chemistry and Chemical Engineering, National Center for Translational Medicine, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University, Shanghai 200240, China
- World Laureates Association (WLA) Laboratories, Shanghai 201203, China
| | - Jianqing Gao
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou 310058, China
- Department of Pharmacy, The First Affiliated Hospital of Ningbo University, Ningbo University, Zhejiang 315010, China
| | - Fangyuan Li
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Frontiers Science Center for Transformative Molecules, School of Chemistry and Chemical Engineering, National Center for Translational Medicine, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
39
|
Clavellina D, Balkan W, Hare JM. Stem cell therapy for acute myocardial infarction: Mesenchymal Stem Cells and induced Pluripotent Stem Cells. Expert Opin Biol Ther 2023; 23:951-967. [PMID: 37542462 PMCID: PMC10837765 DOI: 10.1080/14712598.2023.2245329] [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: 06/13/2023] [Revised: 08/01/2023] [Accepted: 08/03/2023] [Indexed: 08/07/2023]
Abstract
INTRODUCTION Acute myocardial infarction (AMI) remains a leading cause of death in the United States. The limited capacity of cardiomyocytes to regenerate and the restricted contractility of scar tissue after AMI are not addressed by current pharmacologic interventions. Mesenchymal stem/stromal cells (MSCs) have emerged as a promising therapeutic approach due to their low antigenicity, ease of harvesting, and efficacy and safety in preclinical and clinical studies, despite their low survival and engraftment rates. Other stem cell types, such as induced pluripotent stem cells (iPSCs) also show promise, and optimizing cardiac repair requires integrating emerging technologies and strategies. AREAS COVERED This review offers insights into advancing cell-based therapies for AMI, emphasizing meticulously planned trials with a standardized definition of AMI, for a bench-to-bedside approach. We critically evaluate fundamental studies and clinical trials to provide a comprehensive overview of the advances, limitations and prospects for cell-based therapy in AMI. EXPERT OPINION MSCs continue to show potential promise for treating AMI and its sequelae, but addressing their low survival and engraftment rates is crucial for clinical success. Integrating emerging technologies such as pluripotent stem cells and conducting well-designed trials will harness the full potential of cell-based therapy in AMI management. Collaborative efforts are vital to developing effective stem cell therapies for AMI patients.
Collapse
Affiliation(s)
- Diana Clavellina
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Wayne Balkan
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Joshua M Hare
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
40
|
Almahasneh F, Abu-El-Rub E, Khasawneh RR. Mechanisms of analgesic effect of mesenchymal stem cells in osteoarthritis pain. World J Stem Cells 2023; 15:196-208. [PMID: 37181003 PMCID: PMC10173815 DOI: 10.4252/wjsc.v15.i4.196] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/25/2023] [Accepted: 03/27/2023] [Indexed: 04/26/2023] Open
Abstract
Osteoarthritis (OA) is the most common musculoskeletal disease, and it is a major cause of pain, disability and health burden. Pain is the most common and bothersome presentation of OA, but its treatment is still suboptimal, due to the short-term action of employed analgesics and their poor adverse effect profile. Due to their regenerative and anti-inflammatory properties, mesenchymal stem cells (MSCs) have been extensively investigated as a potential therapy for OA, and numerous preclinical and clinical studies found a significant improvement in joint pathology and function, pain scores and/or quality of life after administration of MSCs. Only a limited number of studies, however, addressed pain control as the primary end-point or investigated the potential mechanisms of analgesia induced by MSCs. In this paper, we review the evidence reported in literature that support the analgesic action of MSCs in OA, and we summarize the potential mechanisms of these antinociceptive effects.
Collapse
Affiliation(s)
- Fatimah Almahasneh
- Basic Medical Sciences, Faculty of Medicine -Yarmouk University, Irbid 21163, Jordan
| | - Ejlal Abu-El-Rub
- Basic Medical Sciences, Faculty of Medicine -Yarmouk University, Irbid 21163, Jordan
| | - Ramada R Khasawneh
- Basic Medical Sciences, Faculty of Medicine -Yarmouk University, Irbid 21163, Jordan
| |
Collapse
|
41
|
Zeng CW. Multipotent Mesenchymal Stem Cell-Based Therapies for Spinal Cord Injury: Current Progress and Future Prospects. BIOLOGY 2023; 12:biology12050653. [PMID: 37237467 DOI: 10.3390/biology12050653] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 04/25/2023] [Accepted: 04/25/2023] [Indexed: 05/28/2023]
Abstract
Spinal cord injury (SCI) represents a significant medical challenge, often resulting in permanent disability and severely impacting the quality of life for affected individuals. Traditional treatment options remain limited, underscoring the need for novel therapeutic approaches. In recent years, multipotent mesenchymal stem cells (MSCs) have emerged as a promising candidate for SCI treatment due to their multifaceted regenerative capabilities. This comprehensive review synthesizes the current understanding of the molecular mechanisms underlying MSC-mediated tissue repair in SCI. Key mechanisms discussed include neuroprotection through the secretion of growth factors and cytokines, promotion of neuronal regeneration via MSC differentiation into neural cell types, angiogenesis through the release of pro-angiogenic factors, immunomodulation by modulating immune cell activity, axonal regeneration driven by neurotrophic factors, and glial scar reduction via modulation of extracellular matrix components. Additionally, the review examines the various clinical applications of MSCs in SCI treatment, such as direct cell transplantation into the injured spinal cord, tissue engineering using biomaterial scaffolds that support MSC survival and integration, and innovative cell-based therapies like MSC-derived exosomes, which possess regenerative and neuroprotective properties. As the field progresses, it is crucial to address the challenges associated with MSC-based therapies, including determining optimal sources, intervention timing, and delivery methods, as well as developing standardized protocols for MSC isolation, expansion, and characterization. Overcoming these challenges will facilitate the translation of preclinical findings into clinical practice, providing new hope and improved treatment options for individuals living with the devastating consequences of SCI.
Collapse
Affiliation(s)
- Chih-Wei Zeng
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
42
|
He R, Xu Y, Yu L, Meng N, Wang H, Cui Y, Yam JWP. Extracellular Vesicles Act as Carriers for Cargo Delivery and Regulate Wnt Signaling in the Hepatocellular Carcinoma Tumor Microenvironment. Cancers (Basel) 2023; 15:cancers15072088. [PMID: 37046749 PMCID: PMC10093647 DOI: 10.3390/cancers15072088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/22/2023] [Accepted: 03/29/2023] [Indexed: 04/03/2023] Open
Abstract
As the primary type of liver cancer, hepatocellular carcinoma (HCC) causes a large number of deaths every year. Despite extensive research conducted on this disease, the prognosis of HCC remains unclear. Recently, research has largely focused on extracellular vesicles (EVs), and they have been found to participate in various ways in the development of various diseases, including HCC, such as by regulating cell signaling pathways. However, recent studies have reported the mechanisms underlying the regulation of Wnt signaling by EVs in HCC, primarily focusing on the regulation of the canonical pathways. This review summarizes the current literature on the regulation of Wnt signaling by EVs in HCC and their underlying mechanisms. In addition, we also present future research directions in this field. This will deepen the understanding of HCC and provide new ideas for its treatment.
Collapse
Affiliation(s)
- Risheng He
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Yi Xu
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
- Key Laboratory of Basic Pharmacology of Ministry of Education, Zunyi Medical University, Zunyi 563006, China
- Key Laboratory of Functional and Clinical Translational Medicine, Fujian Province University, Xiamen Medical College, Xiamen 361000, China
- Jiangsu Province Engineering Research Center of Tumor Targeted Nano Diagnostic and Therapeutic Materials, Yancheng Teachers University, Yancheng 224007, China
- Key Laboratory of Biomarkers and In Vitro Diagnosis Translation of Zhejiang Province, Hangzhou 310063, China
- Key Laboratory of Gastrointestinal Cancer, Ministry of Education, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350122, China
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen 518055, China
- Key Laboratory of Intelligent Pharmacy and Individualized Therapy of Huzhou, Department of Pharmacy, Changxing People’s Hospital, Changxing 313000, China
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR 999077, China
| | - Liang Yu
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Nanfeng Meng
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Hang Wang
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Yunfu Cui
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Judy Wai Ping Yam
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR 999077, China
| |
Collapse
|
43
|
Gil-Cabrerizo P, Scaccheti I, Garbayo E, Blanco-Prieto MJ. Cardiac tissue engineering for myocardial infarction treatment. Eur J Pharm Sci 2023; 185:106439. [PMID: 37003408 DOI: 10.1016/j.ejps.2023.106439] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 03/26/2023] [Accepted: 03/28/2023] [Indexed: 04/03/2023]
Abstract
Myocardial infarction is one of the major causes of morbidity and mortality worldwide. Current treatments can relieve the symptoms of myocardial ischemia but cannot repair the necrotic myocardial tissue. Novel therapeutic strategies based on cellular therapy, extracellular vesicles, non-coding RNAs and growth factors have been designed to restore cardiac function while inducing cardiomyocyte cycle re-entry, ensuring angiogenesis and cardioprotection, and preventing ventricular remodeling. However, they face low stability, cell engraftment issues or enzymatic degradation in vivo, and it is thus essential to combine them with biomaterial-based delivery systems. Microcarriers, nanocarriers, cardiac patches and injectable hydrogels have yielded promising results in preclinical studies, some of which are currently being tested in clinical trials. In this review, we cover the recent advances made in cellular and acellular therapies used for cardiac repair after MI. We present current trends in cardiac tissue engineering related to the use of microcarriers, nanocarriers, cardiac patches and injectable hydrogels as biomaterial-based delivery systems for biologics. Finally, we discuss some of the most crucial aspects that should be addressed in order to advance towards the clinical translation of cardiac tissue engineering approaches.
Collapse
Affiliation(s)
- Paula Gil-Cabrerizo
- Department of Pharmaceutical Technology and Chemistry, Faculty of Pharmacy and Nutrition, University of Navarra, Pamplona, C/Irunlarrea 1, E-31080, Spain.; Navarra Institute for Health Research, IdiSNA, Pamplona, C/Irunlarrea 3, E-31008 Pamplona, Spain
| | - Ilaria Scaccheti
- Department of Pharmaceutical Technology and Chemistry, Faculty of Pharmacy and Nutrition, University of Navarra, Pamplona, C/Irunlarrea 1, E-31080, Spain
| | - Elisa Garbayo
- Department of Pharmaceutical Technology and Chemistry, Faculty of Pharmacy and Nutrition, University of Navarra, Pamplona, C/Irunlarrea 1, E-31080, Spain.; Navarra Institute for Health Research, IdiSNA, Pamplona, C/Irunlarrea 3, E-31008 Pamplona, Spain..
| | - María J Blanco-Prieto
- Department of Pharmaceutical Technology and Chemistry, Faculty of Pharmacy and Nutrition, University of Navarra, Pamplona, C/Irunlarrea 1, E-31080, Spain.; Navarra Institute for Health Research, IdiSNA, Pamplona, C/Irunlarrea 3, E-31008 Pamplona, Spain..
| |
Collapse
|
44
|
Fang B, Wang X, Sun Y, Xiong X, Meng X, Li W, Yi Z. Hypoxia-induced CCL2/CCR2 axis in adipose-derived stem cells (ADSCs) promotes angiogenesis by human dermal microvascular endothelial cells (HDMECs) in flap tissues. J Physiol Biochem 2023:10.1007/s13105-023-00944-6. [PMID: 36786974 DOI: 10.1007/s13105-023-00944-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Accepted: 01/09/2023] [Indexed: 02/15/2023]
Abstract
Flap expansion has become an important method widely used in wound repair and organ reconstruction. However, distal skin flap ischemic necrosis remains a problematic complication. In this study, integrative bioinformatics analyses indicated the upregulation of C-C motif chemokine ligand 2 (CCL2) and C-C motif chemokine receptor 2 (CCR2) in reperfusion-exposed skin flap tissues. In adipose-derived stem cells (ADSCs, CD90-positive, CD29-positive, CD34-negative, and CD106-negative) exposed to hypoxia, HIF-1α and CCL2 levels were significantly elevated. Conditioned medium (CM) from hypoxia-stimulated ADSCs promoted HDMEC proliferation, migration, and tube formation, partially inhibited by sh-CCL2-induced CCL2 knockdown or neutralized antibody-induced CCL2 depletion in ADSCs. Consistently, CCL2, CCR2, TNF-α, TLR2, and TLR4 protein levels in HDMECs were significantly increased by hypoxia-treated ADSCs CM, and partially decreased by sh-CCL2-induced CCL2 knockdown or neutralizing antibody-induced CCL2 knockdown in ADSCs. In the flap expansion model, ADSCs transplantation significantly improved flap survival and angiogenesis by endothelial cells in flap tissues, whereas CCL2 knockdown in ADSCs partially eliminated the improvement by ADSCs transplantation; overexpression of CCL2 in ADSCs further promoted the effects of ADSCs transplantation on skin flap. In conclusion, the CCL2/CCR2 axis in ADSCs could be induced by hypoxia, promoting HDMEC proliferation, migration, and tube formation and improving flap survival and angiogenesis in flap tissues.
Collapse
Affiliation(s)
- Bairong Fang
- Department of Plastic and Aesthetic (Burn) Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Xiancheng Wang
- Department of Plastic and Aesthetic (Burn) Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Yang Sun
- Department of Plastic and Aesthetic (Burn) Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.
| | - Xiang Xiong
- Department of Plastic and Aesthetic (Burn) Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Xianxi Meng
- Department of Plastic and Aesthetic (Burn) Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Wenbo Li
- Department of Plastic and Aesthetic (Burn) Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Zhongjie Yi
- Department of Plastic and Aesthetic (Burn) Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| |
Collapse
|
45
|
Castillo MG, Peralta TM, Locatelli P, Velazquez C, Herrero Y, Crottogini AJ, Olea FD, Cuniberti LA. Promoting early neovascularization by allotransplanted adipose-derived Muse cells in an ovine model of acute myocardial infarction. PLoS One 2023; 18:e0277442. [PMID: 36662847 PMCID: PMC9858827 DOI: 10.1371/journal.pone.0277442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 10/26/2022] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Recent preclinical studies have demonstrated that bone marrow (BM)-derived Muse cells have a homing mechanism to reach damaged cardiac tissue while also being able to reduce myocardial infarct size and improve cardiac function; however, the potential of BM-Muse cells to foster new blood-vessel formation has not been fully assessed. Up to date, adipose tissue (AT)-derived Muse cells remain to be studied in acute myocardial infarction (AMI). The aim of the present study was to analyze in vitro and in vivo the neovascularization capacity of AT-Muse cells while exploring their biodistribution and differentiation potential in a translational ovine model of AMI. METHODS AND RESULTS AT-Muse cells were successfully isolated from ovine adipose tissue. In adult sheep, one or more diagonal branches of the left anterior descending coronary artery were permanently ligated for thirty minutes. Sheep were randomized in two groups and treated with intramyocardial injections: Vehicle (PBS, n = 4) and AT-Muse (2x107 AT-Muse cells labeled with PKH26 Red Fluorescent Dye, n = 4). Molecular characterization showed higher expression of angiogenic genes (VEGF, PGF and ANG) and increased number of tube formation in AT-Muse cells group compared to Adipose-derived mesenchymal stromal cells (ASCs) group. At 7 days post-IAM, the AT-Muse group showed significantly more arterioles and capillaries than the Vehicle group. Co-localization of PKH26+ cells with desmin, sarcomeric actin and troponin T implied the differentiation of Muse cells to a cardiac fate; moreover, PKH26+ cells also co-localized with a lectin marker, suggesting a possible differentiation to a vascular lineage. CONCLUSION Intramyocardially administered AT-Muse cells displayed a significant neovascularization activity and survival capacity in an ovine model of AMI.
Collapse
Affiliation(s)
- Martha G. Castillo
- Instituto de Medicina Traslacional, Trasplante y Bioingeniería (IMETTYB)—Universidad Favaloro—CONICET, Ciudad de Buenos Aires, Buenos Aires, Argentina
| | - Tomás M. Peralta
- Instituto de Medicina Traslacional, Trasplante y Bioingeniería (IMETTYB)—Universidad Favaloro—CONICET, Ciudad de Buenos Aires, Buenos Aires, Argentina
| | - Paola Locatelli
- Instituto de Medicina Traslacional, Trasplante y Bioingeniería (IMETTYB)—Universidad Favaloro—CONICET, Ciudad de Buenos Aires, Buenos Aires, Argentina
| | - Candela Velazquez
- Instituto de Biología y Medicina Experimental—CONICET, Ciudad de Buenos Aires, Buenos Aires, Argentina
| | - Yamila Herrero
- Instituto de Biología y Medicina Experimental—CONICET, Ciudad de Buenos Aires, Buenos Aires, Argentina
| | - Alberto J. Crottogini
- Instituto de Medicina Traslacional, Trasplante y Bioingeniería (IMETTYB)—Universidad Favaloro—CONICET, Ciudad de Buenos Aires, Buenos Aires, Argentina
| | - Fernanda D. Olea
- Instituto de Medicina Traslacional, Trasplante y Bioingeniería (IMETTYB)—Universidad Favaloro—CONICET, Ciudad de Buenos Aires, Buenos Aires, Argentina
| | - Luis A. Cuniberti
- Instituto de Medicina Traslacional, Trasplante y Bioingeniería (IMETTYB)—Universidad Favaloro—CONICET, Ciudad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
46
|
[ 68Ga]Ga-NODAGA-E[(cRGDyK)] 2 and [ 64Cu]Cu-DOTATATE PET Predict Improvement in Ischemic Cardiomyopathy. Diagnostics (Basel) 2023; 13:diagnostics13020268. [PMID: 36673078 PMCID: PMC9857952 DOI: 10.3390/diagnostics13020268] [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: 11/30/2022] [Revised: 01/05/2023] [Accepted: 01/07/2023] [Indexed: 01/12/2023] Open
Abstract
An increasing number of patients are living with chronic ischemic cardiomyopathy (ICM) and/or heart failure. Treatment options and prognostic tools are lacking for many of these patients. Our aim was to investigate the prognostic value of imaging angiogenesis and macrophage activation via positron emission tomography (PET) in terms of functional improvement after cell therapy. Myocardial infarction was induced in rats. Animals were scanned with [18F]FDG PET and echocardiography after four weeks and randomized to allogeneic adipose tissue-derived stromal cells (ASCs, n = 18) or saline (n = 9). Angiogenesis and macrophage activation were assessed before and after treatment by [68Ga]Ga-RGD and [64Cu]Cu-DOTATATE. There was no overall effect of the treatment. Rats that improved left ventricular ejection fraction (LVEF) had higher uptake of both [68Ga]Ga-RGD and [64Cu]Cu-DOTATATE at follow-up (p = 0.006 and p = 0.008, respectively). The uptake of the two tracers correlated with each other (r = 0.683, p = 0.003 pre-treatment and r = 0.666, p = 0.004 post-treatment). SUVmax at follow-up could predict improvement in LVEF (p = 0.016 for [68Ga]Ga-RGD and p = 0.045 for [64Cu]Cu-DOTATATE). High uptake of [68Ga]Ga-RGD and [64Cu]Cu-DOTATATE PET after injection of ASCs or saline preceded improvement in LVEF. The use of these tracers could improve the monitoring of heart failure patients in treatment.
Collapse
|
47
|
Harding D, Chong MHA, Lahoti N, Bigogno CM, Prema R, Mohiddin SA, Marelli-Berg F. Dilated cardiomyopathy and chronic cardiac inflammation: Pathogenesis, diagnosis and therapy. J Intern Med 2023; 293:23-47. [PMID: 36030368 DOI: 10.1111/joim.13556] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Dilated cardiomyopathy (DCM) is typically defined by left ventricular dilation and systolic dysfunction in the absence of a clear precipitant. Idiopathic disease is common; up to 50% of patients with DCM have no cause found despite imaging, genetic and biopsy assessments. Treatment remains focused on managing symptoms, reducing the risk of sudden cardiac death and ameliorating the structural and electrical complications of disease progression. In the absence of aetiology-specific treatments, the condition remains associated with a poor prognosis; mortality is approximately 40% at 10 years. The role of immune-mediated inflammatory injury in the development and progression of DCM was first proposed over 30 years ago. Despite the subsequent failures of three large clinical trials of immunosuppressive treatment (ATTACH, RENEWAL and the Myocarditis Treatment Trial), evidence for an abnormal adaptive immune response in DCM remains significant. In this review, we summarise and discuss available evidence supporting immune dysfunction in DCM, with a specific focus on cellular immunity. We also highlight current clinical and experimental treatments. We propose that the success of future immunosuppressive treatment trials in DCM will be dependent on the deep immunophenotyping of patients, to identify those with active inflammation and/or an abnormal immune response who are most likely to respond to therapy.
Collapse
Affiliation(s)
- Daniel Harding
- Centre for Biochemical Pharmacology, William Harvey Research Institute, London, UK
| | - Ming H A Chong
- Barts and The London School of Medicine and Dentistry, London, UK
| | - Nishant Lahoti
- Conquest Hospital, East Sussex Healthcare NHS Trust, St Leonards-on-Sea, UK
| | - Carola M Bigogno
- Barts and The London School of Medicine and Dentistry, London, UK
| | - Roshni Prema
- University Hospital, University Hospitals Coventry and Warwickshire NHS Trust, Coventry, UK
| | | | | |
Collapse
|
48
|
Extracellular vesicles DJ-1 derived from hypoxia-conditioned hMSCs alleviate cardiac hypertrophy by suppressing mitochondria dysfunction and preventing ATRAP degradation. Pharmacol Res 2023; 187:106607. [PMID: 36509316 DOI: 10.1016/j.phrs.2022.106607] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 12/06/2022] [Accepted: 12/09/2022] [Indexed: 12/13/2022]
Abstract
BACKGROUND As a pathological myocardial remodeling process in a variety of cardiovascular diseases, cardiac hypertrophy still has no effective treatment. Human mesenchymal stem cells (hMSCs) derived extracellular vesicles (EVs) has been recognized as a promising treatment strategy for cardiac disease. METHODS In this study, the inhibitory effects on cardiac hypertrophy are compared between normoxia-conditioned hMSC-derived EVs (Nor-EVs) and hypoxia-conditioned hMSC-derived EVs (Hypo-EVs) in neonatal rat cardiomyocytes (NRCMs) after angiotensin II (Ang II) stimulation and in a mouse model of transverse aortic constriction (TAC). RESULTS We demonstrate that Hypo-EVs exert an increased inhibitory effect on cardiac hypertrophy compared with Nor-EVs. Parkinson disease protein 7 (PARK7/DJ-1) is identify as a differential protein between Nor-EVs and Hypo-EVs by quantitative proteomics analysis. Results show that DJ-1, which is rich in Hypo-EVs, alleviates mitochondrial dysfunction and excessive mitochondrial reactive oxygen species (mtROS) production as an antioxidant. Mechanistic studies demonstrate for the first time that DJ-1 may suppress cardiac hypertrophy by inhibiting the activity of proteasome subunit beta type 10 (PSMB10) through a direct physical interaction. This interaction can inhibit angiotensin II type 1 receptor (AT1R)-mediated signaling pathways resulting in cardiac hypertrophy through alleviating ubiquitination degradation of AT1R-associated protein (ATRAP). CONCLUSIONS When taken together, our study suggests that Hypo-EVs have significant potential as a novel therapeutic agent for the treatment of cardiac hypertrophy.
Collapse
|
49
|
Papastamos C, Antonopoulos AS, Simantiris S, Koumallos N, Theofilis P, Sagris M, Tsioufis K, Androulakis E, Tousoulis D. Stem Cell-based Therapies in Cardiovascular Diseases: From Pathophysiology to Clinical Outcomes. Curr Pharm Des 2023; 29:2795-2801. [PMID: 37641986 DOI: 10.2174/1381612829666230828102130] [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: 05/10/2023] [Revised: 06/18/2023] [Accepted: 07/19/2023] [Indexed: 08/31/2023]
Abstract
Over 20 years of intensified research in the field of stem cells brought about unprecedented possibilities in treating heart diseases. The investigators were initially fascinated by the idea of regenerating the lost myocardium and replacing it with new functional cardiomyocytes, but this was extremely challenging. However, the multifactorial effects of stem cell-based therapies beyond mere cardiomyocyte generation, caused by paracrine signaling, would open up new possibilities in treating cardiovascular diseases. To date, there is a strong body of evidence that the anti-inflammatory, anti-apoptotic, and immunomodulatory effects of stem cell therapy may alleviate atherosclerosis progression. In the present review, our objective is to provide a brief overview of the stem cell-based therapeutic options. We aim to delineate the pathophysiological mechanisms of their beneficial effects in cardiovascular diseases especially in coronary artery disease and to highlight some conclusions from important clinical studies in the field of regenerative medicine in cardiovascular diseases and how we could further move onwards.
Collapse
Affiliation(s)
- Charalampos Papastamos
- 1st Cardiology Department, Hippokration General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Alexios S Antonopoulos
- 1st Cardiology Department, Hippokration General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Spyridon Simantiris
- 1st Cardiology Department, Hippokration General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Nikolaos Koumallos
- 1st Cardiology Department, Hippokration General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Panagiotis Theofilis
- 1st Cardiology Department, Hippokration General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Marios Sagris
- 1st Cardiology Department, Hippokration General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Konstantinos Tsioufis
- 1st Cardiology Department, Hippokration General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Dimitris Tousoulis
- 1st Cardiology Department, Hippokration General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
50
|
Development of an injectable alginate-collagen hydrogel for cardiac delivery of extracellular vesicles. Int J Pharm 2022; 629:122356. [DOI: 10.1016/j.ijpharm.2022.122356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 10/07/2022] [Accepted: 10/27/2022] [Indexed: 11/07/2022]
|