1
|
Saikia B, Dhanushkodi A. Engineered exosome therapeutics for neurodegenerative diseases. Life Sci 2024; 356:123019. [PMID: 39209250 DOI: 10.1016/j.lfs.2024.123019] [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: 05/03/2024] [Revised: 08/14/2024] [Accepted: 08/25/2024] [Indexed: 09/04/2024]
Abstract
An increase in life expectancy comes with a higher risk for age-related neurological and cognitive dysfunctions. Given the psycho-socioeconomic burden due to unhealthy aging in the coming decades, the United Nations has declared 2021-2030 as a decade of healthy aging. In this line, multipotent mesenchymal stromal cell-based therapeutics received special interest from the research community. Based on decades of research on cell therapy, a consensus has emerged that the therapeutic effects of cell therapy are due to the paracrine mechanisms rather than cell replacement. Exosomes, a constituent of the secretome, are nano-sized vesicles that have been a focus of intense research in recent years as a possible therapeutic agent or as a cargo to deliver drugs of interest into the central nervous system to induce neurogenesis, reduce neuroinflammation, confer neuroregeneration/neuroprotection, and improve cognitive and motor functions. In this review, we have discussed the neuroprotective properties of exosomes derived from adult mesenchymal stem cells, with a special focus on the role of exosomal miRNAs. We also reviewed various strategies to improve exosome production and their content for better therapeutic effects. Further, we discussed the utilization of ectomesenchymal stem cells like dental pulp stem cells and their exosomes in treating neurodegenerative diseases.
Collapse
Affiliation(s)
- Biplob Saikia
- Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education, Manipal, India
| | - Anandh Dhanushkodi
- Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education, Manipal, India.
| |
Collapse
|
2
|
Kato Y, Aburakawa D, Tashiro R, Zhou Y, Rashad S, Endo H, Tominaga T, Niizuma K. Intravenous administration of muse cells improves cerebral ischemia outcome via immunomodulation in the spleen. J Cereb Blood Flow Metab 2024:271678X241290363. [PMID: 39397400 DOI: 10.1177/0271678x241290363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
Ischemic stroke is a leading cause of disability and death globally. Stem cell therapies are emerging as a frontier for enhancing post-stroke recovery, with Muse cells-a subclass of pluripotent stem cells-demonstrating considerable promise. Muse cells are notable not only for their potential in cell replacement but also for their role in modulating immune responses following cerebral infarction. In the present study, we administered Muse cells intravenously to mice after inducing a stroke via distal middle cerebral artery occlusion. We evaluated motor outcomes, splenocyte populations, cytokine profiles, and gene expression 2 weeks after inducing stroke. Additionally, comparisons were drawn between outcomes in splenectomized mice and those receiving adoptive splenocyte transfer to discern the specific influence of the spleen on treatment efficacy. Our findings revealed that Muse cell therapy facilitates motor recovery, an effect that is compromised in the absence of the spleen. Spleens in treated mice exhibited a shift in neutrophil counts, increased cytokine activity, and a notable uptick in the expression of genes related to protein folding. These insights affirm the potential therapeutic effect of Muse cells in post-stroke treatment strategies, with their efficacy attributed, at least in part, to immunomodulatory pathways involving the spleen.
Collapse
Affiliation(s)
- Yuya Kato
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Daiki Aburakawa
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Ryosuke Tashiro
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yuan Zhou
- Department of Neurosurgical Engineering and Translational Neuroscience, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Sherif Rashad
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Sendai, Japan
- Department of Neurosurgical Engineering and Translational Neuroscience, Tohoku University Graduate School of Medicine, Sendai, Japan
- Department of Neurosurgical Engineering and Translational Neuroscience, Graduate School of Biomedical Engineering, Tohoku University, Sendai, Japan
| | - Hidenori Endo
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Teiji Tominaga
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kuniyasu Niizuma
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Sendai, Japan
- Department of Neurosurgical Engineering and Translational Neuroscience, Tohoku University Graduate School of Medicine, Sendai, Japan
- Department of Neurosurgical Engineering and Translational Neuroscience, Graduate School of Biomedical Engineering, Tohoku University, Sendai, Japan
| |
Collapse
|
3
|
Guo Q, Zhai Q, Ji P. The Role of Mitochondrial Homeostasis in Mesenchymal Stem Cell Therapy-Potential Implications in the Treatment of Osteogenesis Imperfecta. Pharmaceuticals (Basel) 2024; 17:1297. [PMID: 39458939 PMCID: PMC11510265 DOI: 10.3390/ph17101297] [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: 06/20/2024] [Revised: 09/11/2024] [Accepted: 09/18/2024] [Indexed: 10/28/2024] Open
Abstract
Osteogenesis imperfecta (OI) is a hereditary disorder characterized by bones that are fragile and prone to breaking. The efficacy of existing therapies for OI is limited, and they are associated with potentially harmful side effects. OI is primarily due to a mutation of collagen type I and hence impairs bone regeneration. Mesenchymal stem cell (MSC) therapy is an attractive strategy to take advantage of the potential benefits of these multipotent stem cells to address the underlying molecular defects of OI by differentiating osteoblasts, paracrine effects, or immunomodulation. The maintenance of mitochondrial homeostasis is an essential component for improving the curative efficacy of MSCs in OI by affecting the differentiation, signaling, and immunomodulatory functions of MSCs. In this review, we highlight the MSC-based therapy pathway in OI and introduce the MSC regulation mechanism by mitochondrial homeostasis. Strategies aiming to modulate the metabolism and reduce the oxidative stress, as well as innovative strategies based on the use of compounds (resveratrol, NAD+, α-KG), antioxidants, and nanomaterials, are analyzed. These findings may enable the development of new strategies for the treatment of OI, ultimately resulting in improved patient outcomes.
Collapse
Affiliation(s)
- Qingling Guo
- College of Stomatology, Chongqing Medical University, Chongqing 401147, China;
- Chongqing Key Laboratory of Oral Diseases, Chongqing 401147, China
| | - Qiming Zhai
- College of Stomatology, Chongqing Medical University, Chongqing 401147, China;
- Chongqing Key Laboratory of Oral Diseases, Chongqing 401147, China
| | - Ping Ji
- College of Stomatology, Chongqing Medical University, Chongqing 401147, China;
- Chongqing Key Laboratory of Oral Diseases, Chongqing 401147, China
| |
Collapse
|
4
|
Akbarzadeh A, Gerami MH, Farrokhi MR, Shapoori S, Jafarinia M. Therapeutic prospects of microRNAs derived from mesenchymal stem cell extracellular vesicles in rheumatoid arthritis: a comprehensive overview. Mol Cell Biochem 2024:10.1007/s11010-024-05082-1. [PMID: 39105963 DOI: 10.1007/s11010-024-05082-1] [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: 04/17/2024] [Accepted: 07/29/2024] [Indexed: 08/07/2024]
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disorder characterized by inflammatory joint damage. Recent studies have focused on the significance of microRNAs (miRNAs) in the pathogenesis of RA. Mesenchymal stem cells (MSCs) have emerged as a potential therapeutic option for RA based on their regenerative and immunomodulatory properties. MSCs release extracellular vesicles (EVs) containing miRNAs that can modulate immune and inflammatory responses. This article provides a comprehensive overview of the current evidence on the existence of various MSCs-derived miRNAs involved in the pathophysiology, characterization, and treatment of RA. An overview of the miRNA profiles in MSC-EVs is provided, along with an examination of their impact on various cell types implicated in RA pathogenesis, including synovial fibroblasts, macrophages, and T cells. Furthermore, the therapeutic capability of MSC-EVs for miRNA-based therapies in RA is discussed. In total, this review can present an extensive view of the complex interaction between EVs and MSC-derived miRNAs in RA and thus suggest valuable strategies for developing new therapeutic approaches to target this debilitating disease.
Collapse
Affiliation(s)
- Armin Akbarzadeh
- Department of Orthopedic Surgery, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Hadi Gerami
- Department of Orthopedic Surgery, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Majid Reza Farrokhi
- Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Neurosurgery, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Shima Shapoori
- Center for Research in Medical Devices (CÚRAM), University of Galway, Galway, Ireland
| | - Morteza Jafarinia
- Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
5
|
Kumar S, Yadav V, Sharma N, Sethi A. HypoxamiR-210-3p regulates mesenchymal stem cells proliferation via P53 & Akt. Mol Cell Biochem 2024; 479:2119-2129. [PMID: 37620743 DOI: 10.1007/s11010-023-04834-9] [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] [Received: 05/05/2023] [Accepted: 08/14/2023] [Indexed: 08/26/2023]
Abstract
Transplanted stem cells (˃95%) into ischemic myocardium die because of unfavourable conditions. Moreover, hypoxia role in the cell cycle regulation has been studied in transformed/immortalized cell lines which may have altered cell cycle regulators and/or mutated and, can't be transplanted in patients. We quest to find out the mechanism of cell cycle regulation in mesenchymal stem cells (MSC) to regulate its survival and proliferation in repair processes. Additionally, critically analysed role of hypoxamiR-210-3p, and cell cycle regulators that can regulate cell proliferation under hypoxic conditions. Bone marrow-derived MSC (BM-MSC) isolated from young male Fischer-344 rats by flushing the cavity of femur and propagated in vitro under 1% hypoxia for 72 h showed an increased in cell proliferation ( > 30%, p < 0.05) compared to normoxia. miR-210-3p, role in cell proliferation under hypoxic condition was confirmed by knockdown. Loss of function studies with transfection of anti-mir-210-3p, we observed decrease in proliferation of BM-MSC under hypoxia. Furthermore, BM-MSC proliferation due to miR-210-3p was confirmed using CFSE assay and flow cytometry, in which more cells were observed in S-phase. Mechanistically, western blot analysis showed miR-210-3p inhibition upregulates p53 and p21 expression and subsequent decrease in pAkt under hypoxia. On contrary, CFSE and Western blot under normoxic conditions showed downregulation of p53 and p21 whilst upregulation of pAkt indicated the key role of miR-210-3p in BM-MSC proliferation. Our results demonstrate the role of miR-210-3p in BM-MSC proliferation under both hypoxic and normoxic conditions and illustrate the potential mechanism via the regulation of pAkt, p53 and p21.
Collapse
Affiliation(s)
- Sanjay Kumar
- Department of Biochemistry, Medical College, All India Institute of Medical Sciences, Bathinda, India.
- Division of Regenerative Medicine, Department of Pathology and Laboratory Medicine, Center of Excellence (CoE) Cardiovascular Diseases, College of Medicine, University of Cincinnati, 231 Albert Sabin Way, Cincinnati, OH, 45229, USA.
- Department of Biochemistry, Medical College, All India Institute of Medical Sciences, Bathinda, Punjab, 151001, India.
| | - Varsha Yadav
- Department of Biochemistry, Medical College, All India Institute of Medical Sciences, Bathinda, India
| | - Namrta Sharma
- Department of Biochemistry, Medical College, All India Institute of Medical Sciences, Bathinda, India
| | - Anshika Sethi
- Department of Biochemistry, Medical College, All India Institute of Medical Sciences, Bathinda, India
| |
Collapse
|
6
|
Gnaiger E. Complex II ambiguities-FADH 2 in the electron transfer system. J Biol Chem 2024; 300:105470. [PMID: 38118236 PMCID: PMC10772739 DOI: 10.1016/j.jbc.2023.105470] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 11/01/2023] [Accepted: 11/06/2023] [Indexed: 12/22/2023] Open
Abstract
The prevailing notion that reduced cofactors NADH and FADH2 transfer electrons from the tricarboxylic acid cycle to the mitochondrial electron transfer system creates ambiguities regarding respiratory Complex II (CII). CII is the only membrane-bound enzyme in the tricarboxylic acid cycle and is part of the electron transfer system of the mitochondrial inner membrane feeding electrons into the coenzyme Q-junction. The succinate dehydrogenase subunit SDHA of CII oxidizes succinate and reduces the covalently bound prosthetic group FAD to FADH2 in the canonical forward tricarboxylic acid cycle. However, several graphical representations of the electron transfer system depict FADH2 in the mitochondrial matrix as a substrate to be oxidized by CII. This leads to the false conclusion that FADH2 from the β-oxidation cycle in fatty acid oxidation feeds electrons into CII. In reality, dehydrogenases of fatty acid oxidation channel electrons to the Q-junction but not through CII. The ambiguities surrounding Complex II in the literature and educational resources call for quality control, to secure scientific standards in current communications of bioenergetics, and ultimately support adequate clinical applications. This review aims to raise awareness of the inherent ambiguity crisis, complementing efforts to address the well-acknowledged issues of credibility and reproducibility.
Collapse
|
7
|
Giallongo S, Duminuco A, Dulcamare I, Zuppelli T, La Spina E, Scandura G, Santisi A, Romano A, Di Raimondo F, Tibullo D, Palumbo GA, Giallongo C. Engagement of Mesenchymal Stromal Cells in the Remodeling of the Bone Marrow Microenvironment in Hematological Cancers. Biomolecules 2023; 13:1701. [PMID: 38136573 PMCID: PMC10741414 DOI: 10.3390/biom13121701] [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: 10/30/2023] [Revised: 11/17/2023] [Accepted: 11/22/2023] [Indexed: 12/24/2023] Open
Abstract
Mesenchymal stromal cells (MSCs) are a subset of heterogeneous, non-hematopoietic fibroblast-like cells which play important roles in tissue repair, inflammation, and immune modulation. MSCs residing in the bone marrow microenvironment (BMME) functionally interact with hematopoietic stem progenitor cells regulating hematopoiesis. However, MSCs have also emerged in recent years as key regulators of the tumor microenvironment. Indeed, they are now considered active players in the pathophysiology of hematologic malignancies rather than passive bystanders in the hematopoietic microenvironment. Once a malignant event occurs, the BMME acquires cellular, molecular, and epigenetic abnormalities affecting tumor growth and progression. In this context, MSC behavior is affected by signals coming from cancer cells. Furthermore, it has been shown that stromal cells themselves play a major role in several hematological malignancies' pathogenesis. This bidirectional crosstalk creates a functional tumor niche unit wherein tumor cells acquire a selective advantage over their normal counterparts and are protected from drug treatment. It is therefore of critical importance to unveil the underlying mechanisms which activate a protumor phenotype of MSCs for defining the unmasked vulnerabilities of hematological cancer cells which could be pharmacologically exploited to disrupt tumor/MSC coupling. The present review focuses on the current knowledge about MSC dysfunction mechanisms in the BMME of hematological cancers, sustaining tumor growth, immune escape, and cancer progression.
Collapse
Affiliation(s)
- Sebastiano Giallongo
- Department of Medical, Surgical Sciences and Advanced Technologies “G.F. Ingrassia”, University of Catania, 95123 Catania, Italy; (S.G.); (G.A.P.); (C.G.)
| | - Andrea Duminuco
- Division of Hematology, AOU Policlinico, 95123 Catania, Italy; (A.D.); (A.S.)
| | - Ilaria Dulcamare
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy;
| | - Tatiana Zuppelli
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (T.Z.); (E.L.S.)
| | - Enrico La Spina
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (T.Z.); (E.L.S.)
| | - Grazia Scandura
- Department of General Surgery and Medical-Surgical Specialties, University of Catania, 95123 Catania, Italy; (G.S.); (A.R.); (F.D.R.)
| | - Annalisa Santisi
- Division of Hematology, AOU Policlinico, 95123 Catania, Italy; (A.D.); (A.S.)
| | - Alessandra Romano
- Department of General Surgery and Medical-Surgical Specialties, University of Catania, 95123 Catania, Italy; (G.S.); (A.R.); (F.D.R.)
| | - Francesco Di Raimondo
- Department of General Surgery and Medical-Surgical Specialties, University of Catania, 95123 Catania, Italy; (G.S.); (A.R.); (F.D.R.)
| | - Daniele Tibullo
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (T.Z.); (E.L.S.)
| | - Giuseppe A. Palumbo
- Department of Medical, Surgical Sciences and Advanced Technologies “G.F. Ingrassia”, University of Catania, 95123 Catania, Italy; (S.G.); (G.A.P.); (C.G.)
| | - Cesarina Giallongo
- Department of Medical, Surgical Sciences and Advanced Technologies “G.F. Ingrassia”, University of Catania, 95123 Catania, Italy; (S.G.); (G.A.P.); (C.G.)
| |
Collapse
|
8
|
Chang C, Cai RP, Su YM, Wu Q, Su Q. Mesenchymal Stem Cell-Derived Exosomal Noncoding RNAs as Alternative Treatments for Myocardial Ischemia-Reperfusion Injury: Current Status and Future Perspectives. J Cardiovasc Transl Res 2023; 16:1085-1098. [PMID: 37286924 PMCID: PMC10246878 DOI: 10.1007/s12265-023-10401-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 05/22/2023] [Indexed: 06/09/2023]
Abstract
Ischemic cardiomyopathy is treated mainly with thrombolytic drugs, percutaneous coronary intervention, and coronary artery bypass grafting to recanalize blocked vessels. Myocardial ischemia-reperfusion injury (MIRI) is an unavoidable complication of obstructive revascularization. Compared with those of myocardial ischemic injury, few effective therapeutic options are available for MIRI treatment. The pathophysiological mechanisms of MIRI involve the inflammatory response, the immune response, oxidative stress, apoptosis, intracellular Ca2+ overload, and cardiomyocyte energy metabolism. These mechanisms exacerbate MIRI. Mesenchymal stem cell-derived exosomes (MSC-EXOs) can alleviate MIRI through these mechanisms and, to some extent, prevent the limitations caused by direct MSC administration. Therefore, using MSC-EXOs instead of MSCs to treat MIRI is a potentially beneficial cell-free treatment strategy. In this review, we describe the mechanism of action of MSC-EXO-derived noncoding RNAs in the treatment of MIRI and discuss the advantages and limitations of this strategy, as well as possible future research directions.
Collapse
Affiliation(s)
- Chen Chang
- Department of Cardiology, Affiliated Hospital of Guilin Medical University, 15 Lequn Road, Guilin, 541000, China
| | - Ru-Ping Cai
- Department of Rehabilitation Medicine, The Third Affiliated Hospital of Guangxi Medical University, Nanning, 530000, China
| | - Ying-Man Su
- Department of Cardiology, Affiliated Hospital of Guilin Medical University, 15 Lequn Road, Guilin, 541000, China
| | - Qiang Wu
- Department of Cardiology, the Sixth Medical Centre, Chinese PLA General Hospital, Beijing, 100048, China.
- Journal of Geriatric Cardiology Editorial Office, Chinese PLA General Hospital, Beijing, 100853, China.
| | - Qiang Su
- Department of Cardiology, Affiliated Hospital of Guilin Medical University, 15 Lequn Road, Guilin, 541000, China.
| |
Collapse
|
9
|
Daneste H, Mohammadzadeh Boukani L, Ramezani N, Asadi F, Zaidan HK, Sadeghzade A, Ehsannia M, Azarashk A, Gholizadeh N. Combination therapy along with mesenchymal stem cells in wound healing; the state of the art. Adv Med Sci 2023; 68:441-449. [PMID: 37924749 DOI: 10.1016/j.advms.2023.10.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 06/23/2023] [Accepted: 10/17/2023] [Indexed: 11/06/2023]
Abstract
Mesenchymal stem cells (MSCs) are being increasingly used in various therapeutic applications including skin tissue repair and wound healing. The positive effects of the MSCs therapy are largely elicited by immunomodulation, increasing angiogenesis, supporting extracellular matrix (ECM) and thus favoring skin structure. However, the therapeutic competences of MSC-based therapies are somewhat hindered by their apparent modest clinical merits, conferring the need for methods that would rise the efficacy of such therapies. A plethora of reports have shown that therapeutic properties of MSCs could be enhanced with other strategies and compounds like biomaterial and platelet-rich plasma (PRP) to target key possessions of MSCs and properties of adjacent tissues concurrently. Manipulation of cellular stress-response mechanisms to improve cell resistance to oxidative stress prior to or during MSC injection could also improve therapeutic efficacy of MSCs. In the current review, we shed light on the recent advances in MSCs combination therapy with other ingredients and procedures to sustain MSCs-mediated effects in wound healing.
Collapse
Affiliation(s)
- Hossein Daneste
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Narges Ramezani
- Department of Biology, Damghan Branch, Islamic Azad University, Damghan, Iran
| | - Fatemeh Asadi
- Department of Genetics, Izeh Branch, Islamic Azad University, Izeh, Iran
| | - Haider Kamil Zaidan
- Department of Medical Laboratories Techniques, Al-Mustaqbal University College, Hillah, Babylon, Iraq
| | - Azita Sadeghzade
- Department of Oral and Maxillofacial Medicine, School of Dentistry, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Maedeh Ehsannia
- Faculty of Basic Sciences, Islamic Azad University, Tehran East Branch, Tehran, Iran
| | - Ali Azarashk
- Department of Pharmaceutics, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Nasim Gholizadeh
- Department of Dermatology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.
| |
Collapse
|
10
|
Slama Y, Ah-Pine F, Khettab M, Arcambal A, Begue M, Dutheil F, Gasque P. The Dual Role of Mesenchymal Stem Cells in Cancer Pathophysiology: Pro-Tumorigenic Effects versus Therapeutic Potential. Int J Mol Sci 2023; 24:13511. [PMID: 37686315 PMCID: PMC10488262 DOI: 10.3390/ijms241713511] [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: 08/02/2023] [Revised: 08/29/2023] [Accepted: 08/30/2023] [Indexed: 09/10/2023] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) are multipotent cells involved in numerous physiological events, including organogenesis, the maintenance of tissue homeostasis, regeneration, or tissue repair. MSCs are increasingly recognized as playing a major, dual, and complex role in cancer pathophysiology through their ability to limit or promote tumor progression. Indeed, these cells are known to interact with the tumor microenvironment, modulate the behavior of tumor cells, influence their functions, and promote distant metastasis formation through the secretion of mediators, the regulation of cell-cell interactions, and the modulation of the immune response. This dynamic network can lead to the establishment of immunoprivileged tissue niches or the formation of new tumors through the proliferation/differentiation of MSCs into cancer-associated fibroblasts as well as cancer stem cells. However, MSCs exhibit also therapeutic effects including anti-tumor, anti-proliferative, anti-inflammatory, or anti-oxidative effects. The therapeutic interest in MSCs is currently growing, mainly due to their ability to selectively migrate and penetrate tumor sites, which would make them relevant as vectors for advanced therapies. Therefore, this review aims to provide an overview of the double-edged sword implications of MSCs in tumor processes. The therapeutic potential of MSCs will be reviewed in melanoma and lung cancers.
Collapse
Affiliation(s)
- Youssef Slama
- Unité de Recherche Études Pharmaco-Immunologiques (EPI), Université de La Réunion, CHU de La Réunion, Allée des Topazes, 97400 Saint-Denis, La Réunion, France; (F.A.-P.); (M.K.); (P.G.)
- Service de Radiothérapie, Clinique Sainte-Clotilde, Groupe Clinifutur, 127 Route de Bois de Nèfles, 97400 Saint-Denis, La Réunion, France; (M.B.); (F.D.)
- Laboratoire Interdisciplinaire de Recherche en Santé (LIRS), RunResearch, Clinique Sainte-Clotilde, 127 Route de Bois de Nèfles, 97400 Saint-Denis, La Réunion, France;
| | - Franck Ah-Pine
- Unité de Recherche Études Pharmaco-Immunologiques (EPI), Université de La Réunion, CHU de La Réunion, Allée des Topazes, 97400 Saint-Denis, La Réunion, France; (F.A.-P.); (M.K.); (P.G.)
- Service d’Anatomie et Cytologie Pathologiques, CHU de La Réunion sites SUD—Saint-Pierre, Avenue François Mitterrand, 97448 Saint-Pierre Cedex, La Réunion, France
| | - Mohamed Khettab
- Unité de Recherche Études Pharmaco-Immunologiques (EPI), Université de La Réunion, CHU de La Réunion, Allée des Topazes, 97400 Saint-Denis, La Réunion, France; (F.A.-P.); (M.K.); (P.G.)
- Service d’Oncologie Médicale, CHU de La Réunion sites SUD—Saint-Pierre, Avenue François Mitterrand, 97448 Saint-Pierre Cedex, La Réunion, France
| | - Angelique Arcambal
- Laboratoire Interdisciplinaire de Recherche en Santé (LIRS), RunResearch, Clinique Sainte-Clotilde, 127 Route de Bois de Nèfles, 97400 Saint-Denis, La Réunion, France;
| | - Mickael Begue
- Service de Radiothérapie, Clinique Sainte-Clotilde, Groupe Clinifutur, 127 Route de Bois de Nèfles, 97400 Saint-Denis, La Réunion, France; (M.B.); (F.D.)
- Laboratoire Interdisciplinaire de Recherche en Santé (LIRS), RunResearch, Clinique Sainte-Clotilde, 127 Route de Bois de Nèfles, 97400 Saint-Denis, La Réunion, France;
| | - Fabien Dutheil
- Service de Radiothérapie, Clinique Sainte-Clotilde, Groupe Clinifutur, 127 Route de Bois de Nèfles, 97400 Saint-Denis, La Réunion, France; (M.B.); (F.D.)
- Laboratoire Interdisciplinaire de Recherche en Santé (LIRS), RunResearch, Clinique Sainte-Clotilde, 127 Route de Bois de Nèfles, 97400 Saint-Denis, La Réunion, France;
| | - Philippe Gasque
- Unité de Recherche Études Pharmaco-Immunologiques (EPI), Université de La Réunion, CHU de La Réunion, Allée des Topazes, 97400 Saint-Denis, La Réunion, France; (F.A.-P.); (M.K.); (P.G.)
| |
Collapse
|
11
|
Sukmana BI, Margiana R, Almajidi YQ, Almalki SG, Hjazi A, Shahab S, Romero-Parra RM, Alazbjee AAA, Alkhayyat A, John V. Supporting wound healing by mesenchymal stem cells (MSCs) therapy in combination with scaffold, hydrogel, and matrix; State of the art. Pathol Res Pract 2023; 248:154575. [PMID: 37285734 DOI: 10.1016/j.prp.2023.154575] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 05/24/2023] [Accepted: 05/25/2023] [Indexed: 06/09/2023]
Abstract
Non-healing wounds impose a huge annual cost on the survival of different countries and large populations in the world. Wound healing is a complex and multi-step process, the speed and quality of which can be changed by various factors. To promote wound healing, compounds such as platelet-rich plasma, growth factors, platelet lysate, scaffolds, matrix, hydrogel, and cell therapy, in particular, with mesenchymal stem cells (MSCs) are suggested. Nowadays, the use of MSCs has attracted a lot of attention. These cells can induce their effect by direct effect and secretion of exosomes. On the other hand, scaffolds, matrix, and hydrogels provide suitable conditions for wound healing and the growth, proliferation, differentiation, and secretion of cells. In addition to generating suitable conditions for wound healing, the combination of biomaterials and MSCs increases the function of these cells at the site of injury by favoring their survival, proliferation, differentiation, and paracrine activity. In addition, other compounds such as glycol, sodium alginate/collagen hydrogel, chitosan, peptide, timolol, and poly(vinyl) alcohol can be used along with these treatments to increase the effectiveness of treatments in wound healing. In this review article, we take a glimpse into the merging scaffolds, hydrogels, and matrix application with MSCs therapy to favor wound healing.
Collapse
Affiliation(s)
- Bayu Indra Sukmana
- Oral Biology Department, Lambung Mangkurat University, Banjarmasin, Indonesia
| | - Ria Margiana
- Department of Anatomy, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia; Master's Programme Biomedical Sciences, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia; Dr. Soetomo General Academic Hospital, Surabaya, Indonesia.
| | - Yasir Qasim Almajidi
- Department of Pharmacy (Pharmaceutics), Baghdad College of Medical Sciences, Baghdad, Iraq
| | - Sami G Almalki
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Majmaah 11952, Saudi Arabia
| | - Ahmed Hjazi
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Sana Shahab
- Department of Business Administration, College of Business Administration, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | | | | | - Afa Alkhayyat
- College of Pharmacy, the Islamic University, 54001 Najaf, Iraq
| | - Vivek John
- Uttaranchal Institute of Technology, Uttaranchal University, Dehradun 248007, India
| |
Collapse
|
12
|
Combinatorial Effect of Mesenchymal Stem Cells and Extracellular Vesicles in a Hydrogel on Cartilage Regeneration. Tissue Eng Regen Med 2022; 20:143-154. [PMID: 36482140 PMCID: PMC9852407 DOI: 10.1007/s13770-022-00509-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 10/29/2022] [Accepted: 11/01/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) are used for tissue regeneration due to their wide differentiation capacity and anti-inflammatory effects. Extracellular vesicles (EVs) derived from MSCs are also known for their regenerative effects as they contain nucleic acids, proteins, lipids, and cytokines similar to those of parental cells. There are several studies on the use of MSCs or EVs for tissue regeneration. However, the combinatorial effect of human MSCs (hMSCs) and EVs is not clear. In this study, we investigated the combinatorial effect of hMSCs and EVs on cartilage regeneration via co-encapsulation in a hyaluronic-acid (HA)-based hydrogel. METHODS A methacrylic-acid-based HA hydrogel was prepared to encapsulate hMSCs and EVs in hydrogels. Through in vitro and in vivo analyses, we investigated the chondrogenic potential of the HA hydrogel-encapsulated with hMSCs and EVs. RESULTS Co-encapsulation of hMSCs with EVs in the HA hydrogel increased the chondrogenic differentiation of hMSCs and regeneration of damaged cartilage tissue compared with that of the HA hydrogel loaded with hMSCs only. CONCLUSION Co-encapsulation of hMSCs and EVs in the HA hydrogel effectively enhances cartilage tissue regeneration due to the combinatorial therapeutic effect of hMSCs and EVs. Thus, in addition to cartilage tissue regeneration for the treatment of osteoarthritis, this approach would be a useful strategy to improve other types of tissue regeneration.
Collapse
|
13
|
The Role of Extracellular Vesicles in Optic Nerve Injury: Neuroprotection and Mitochondrial Homeostasis. Cells 2022; 11:cells11233720. [PMID: 36496979 PMCID: PMC9738450 DOI: 10.3390/cells11233720] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/16/2022] [Accepted: 11/17/2022] [Indexed: 11/25/2022] Open
Abstract
Stem cell therapies hold great promise as alternative treatments for incurable optic nerve disorders. Although mesenchymal stem cells exhibit various tissue regeneration and recovery capabilities that may serve as valuable therapies, the clinical applications remain limited. Thus, we investigated the utility of extracellular vesicles (EVs) from human placenta-derived mesenchymal stem cells (hPSCs) in this context. Hypoxically preconditioned hPSCs (HPPSCs) were prepared via short-term incubation under 2.2% O2 and 5.5% CO2. The EVs were then isolated. R28 cells (retinal precursor cells) were exposed to CoCl2 and treated with EVs for 24 h. Cell proliferation and regeneration were measured using a BrdU assay and immunoblotting; ATP quantification revealed the extent of the mitochondrial function. The proteome was determined via liquid chromatography-tandem mass spectroscopy. Differentially expressed proteins (DEPs) were detected and their interactions identified. HPPSC_EVs functions were explored using animal models of optic nerve compression. HPPSC_EVs restored cell proliferation and mitochondrial quality control in R28 cells damaged by CoCl2. We identified DEPs (p < 0.05) that aided recovery. The mitochondrial DEPs included LONP1; PARK7; VDAC1, 2, and 3; HSPD1; and HSPA9. EVs regulated the levels of mitophagic proteins in R28 cells injured by hypoxia; the protein levels did not increase in LONP1 knockdown cells. LONP1 is a key mediator of the mitophagy that restores mitochondrial function after hypoxia-induced optic nerve injury.
Collapse
|
14
|
Alcaraz MJ, Guillén MI. Cellular and Molecular Targets of Extracellular Vesicles from Mesenchymal Stem/Stromal Cells in Rheumatoid Arthritis. Stem Cells Transl Med 2022; 11:1177-1185. [PMID: 36318277 PMCID: PMC9801303 DOI: 10.1093/stcltm/szac075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 09/29/2022] [Indexed: 11/05/2022] Open
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease that causes progressive joint destruction. Despite the advances in the treatment of this condition there remains a clinical need for safe therapies leading to clinical remission. Mesenchymal stem/stromal cells (MSCs) play immunomodulatory and regenerative roles which can be partly mediated by their secretome. In recent years, the important contribution of extracellular vesicles (EVs) to MSC actions has received an increasing interest as a new therapeutic approach. We provide an extensive overview of the immunomodulatory properties of MSC EVs and their effects on articular cells such as fibroblast-like synoviocytes that play a central role in joint destruction. This review discusses the anti-arthritic effects of MSC EVs in vitro and in animal models of RA as well as their potential mechanisms. Recent preclinical data suggest that transfer of non-coding RNAs by MSC EVs regulates key signaling pathways involved in the pathogenesis of RA. We also examine a number of EV modifications for improving their anti-arthritic efficacy and carrier ability for drug delivery.
Collapse
Affiliation(s)
- María José Alcaraz
- Corresponding author: María José Alcaraz, PhD, Interuniversity Research Institute for Molecular Recognition and Technological Development (IDM), University of Valencia, Polytechnic University of Valencia, Burjassot, Valencia, Spain. E-mail:
| | - María Isabel Guillén
- Interuniversity Research Institute for Molecular Recognition and Technological Development (IDM), University of Valencia, Polytechnic University of Valencia, Av. Vicent A. Estellés s/n, Burjassot, Valencia, Spain,Department of Pharmacy, Faculty of Health Sciences, Cardenal Herrera-CEU University, Alfara del Patriarca, Valencia, Spain
| |
Collapse
|
15
|
Lu Y, Yang Y, Liu S, Ge S. Biomaterials constructed for MSC-derived extracellular vesicle loading and delivery—a promising method for tissue regeneration. Front Cell Dev Biol 2022; 10:898394. [PMID: 36092710 PMCID: PMC9454000 DOI: 10.3389/fcell.2022.898394] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 07/19/2022] [Indexed: 11/13/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have become the preferred seed cells for tissue regeneration. Nevertheless, due to their immunogenicity and tumorigenicity, MSC transplantation remains questionable. Extracellular vesicles (EVs) derived from MSCs are becoming a promising substitute for MSCs. As a route of the MSC paracrine, EVs have a nano-sized and bilayer lipid-enclosed structure, which can guarantee the integrity of their cargoes, but EVs cannot obtain full function in vivo because of the rapid biodegradation and clearance by phagocytosis. To improve the efficacy and targeting of EVs, methods have been proposed and put into practice, especially engineered vesicles and EV-controlled release systems. In particular, EVs can be cell or tissue targeting because they have cell-specific ligands on their surfaces, but their targeting ability may be eliminated by the biodegradation of the phagocytic system during circulation. Novel application strategies have been proposed beyond direct injecting. EV carriers such as biodegradable hydrogels and other loading systems have been applied in tissue regeneration, and EV engineering is also a brand-new method for higher efficacy. In this review, we distinctively summarize EV engineering and loading system construction methods, emphasizing targeting modification methods and controlled release systems for EVs, which few literature reviews have involved.
Collapse
Affiliation(s)
- Yu Lu
- Shandong Key Laboratory of Oral Tissue Regeneration, Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Department of Biomaterials, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yu Yang
- Shandong Key Laboratory of Oral Tissue Regeneration, Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Department of Biomaterials, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Shiyu Liu
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi’an, China
| | - Shaohua Ge
- Shandong Key Laboratory of Oral Tissue Regeneration, Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Department of Biomaterials, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, China
- *Correspondence: Shaohua Ge,
| |
Collapse
|
16
|
Zorova LD, Kovalchuk SI, Popkov VA, Chernikov VP, Zharikova AA, Khutornenko AA, Zorov SD, Plokhikh KS, Zinovkin RA, Evtushenko EA, Babenko VA, Pevzner IB, Shevtsova YA, Goryunov KV, Plotnikov EY, Silachev DN, Sukhikh GT, Zorov DB. Do Extracellular Vesicles Derived from Mesenchymal Stem Cells Contain Functional Mitochondria? Int J Mol Sci 2022; 23:ijms23137408. [PMID: 35806411 PMCID: PMC9266972 DOI: 10.3390/ijms23137408] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/28/2022] [Accepted: 06/30/2022] [Indexed: 02/06/2023] Open
Abstract
Extracellular vesicles (EV) derived from stem cells have become an effective complement to the use in cell therapy of stem cells themselves, which has led to an explosion of research into the mechanisms of vesicle formation and their action. There is evidence demonstrating the presence of mitochondrial components in EV, but a definitive conclusion about whether EV contains fully functional mitochondria has not yet been made. In this study, two EV fractions derived from mesenchymal stromal stem cells (MSC) and separated by their size were examined. Flow cytometry revealed the presence of mitochondrial lipid components capable of interacting with mitochondrial dyes MitoTracker Green and 10-nonylacridine orange; however, the EV response to the probe for mitochondrial membrane potential was negative. Detailed analysis revealed components from all mitochondria compartments, including house-keeping mitochondria proteins and DNA as well as energy-related proteins such as membrane-localized proteins of complexes I, IV, and V, and soluble proteins from the Krebs cycle. When assessing the functional activity of mitochondria, high variability in oxygen consumption was noted, which was only partially attributed to mitochondrial respiratory activity. Our findings demonstrate that the EV contain all parts of mitochondria; however, their independent functionality inside EV has not been confirmed, which may be due either to the absence of necessary cofactors and/or the EV formation process and, probably the methodology of obtaining EV.
Collapse
Affiliation(s)
- Ljubava D. Zorova
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia; (L.D.Z.); (V.A.P.); (S.D.Z.); (R.A.Z.); (V.A.B.); (I.B.P.); (E.Y.P.)
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, Moscow 117997, Russia; (A.A.K.); (Y.A.S.); (K.V.G.); (G.T.S.)
| | - Sergei I. Kovalchuk
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia;
| | - Vasily A. Popkov
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia; (L.D.Z.); (V.A.P.); (S.D.Z.); (R.A.Z.); (V.A.B.); (I.B.P.); (E.Y.P.)
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, Moscow 117997, Russia; (A.A.K.); (Y.A.S.); (K.V.G.); (G.T.S.)
| | | | - Anastasia A. Zharikova
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow 119992, Russia;
| | - Anastasia A. Khutornenko
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, Moscow 117997, Russia; (A.A.K.); (Y.A.S.); (K.V.G.); (G.T.S.)
| | - Savva D. Zorov
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia; (L.D.Z.); (V.A.P.); (S.D.Z.); (R.A.Z.); (V.A.B.); (I.B.P.); (E.Y.P.)
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow 119992, Russia;
| | | | - Roman A. Zinovkin
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia; (L.D.Z.); (V.A.P.); (S.D.Z.); (R.A.Z.); (V.A.B.); (I.B.P.); (E.Y.P.)
| | | | - Valentina A. Babenko
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia; (L.D.Z.); (V.A.P.); (S.D.Z.); (R.A.Z.); (V.A.B.); (I.B.P.); (E.Y.P.)
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, Moscow 117997, Russia; (A.A.K.); (Y.A.S.); (K.V.G.); (G.T.S.)
| | - Irina B. Pevzner
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia; (L.D.Z.); (V.A.P.); (S.D.Z.); (R.A.Z.); (V.A.B.); (I.B.P.); (E.Y.P.)
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, Moscow 117997, Russia; (A.A.K.); (Y.A.S.); (K.V.G.); (G.T.S.)
| | - Yulia A. Shevtsova
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, Moscow 117997, Russia; (A.A.K.); (Y.A.S.); (K.V.G.); (G.T.S.)
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow 119992, Russia;
| | - Kirill V. Goryunov
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, Moscow 117997, Russia; (A.A.K.); (Y.A.S.); (K.V.G.); (G.T.S.)
| | - Egor Y. Plotnikov
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia; (L.D.Z.); (V.A.P.); (S.D.Z.); (R.A.Z.); (V.A.B.); (I.B.P.); (E.Y.P.)
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, Moscow 117997, Russia; (A.A.K.); (Y.A.S.); (K.V.G.); (G.T.S.)
| | - Denis N. Silachev
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia; (L.D.Z.); (V.A.P.); (S.D.Z.); (R.A.Z.); (V.A.B.); (I.B.P.); (E.Y.P.)
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, Moscow 117997, Russia; (A.A.K.); (Y.A.S.); (K.V.G.); (G.T.S.)
- Correspondence: (D.N.S.); (D.B.Z.); Tel.: +7-(495)939-59-44 (D.N.S.); +7-(495)939-59-44 (D.B.Z.)
| | - Gennady T. Sukhikh
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, Moscow 117997, Russia; (A.A.K.); (Y.A.S.); (K.V.G.); (G.T.S.)
| | - Dmitry B. Zorov
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia; (L.D.Z.); (V.A.P.); (S.D.Z.); (R.A.Z.); (V.A.B.); (I.B.P.); (E.Y.P.)
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, Moscow 117997, Russia; (A.A.K.); (Y.A.S.); (K.V.G.); (G.T.S.)
- Correspondence: (D.N.S.); (D.B.Z.); Tel.: +7-(495)939-59-44 (D.N.S.); +7-(495)939-59-44 (D.B.Z.)
| |
Collapse
|
17
|
Therapeutic Strategy of Mesenchymal-Stem-Cell-Derived Extracellular Vesicles as Regenerative Medicine. Int J Mol Sci 2022; 23:ijms23126480. [PMID: 35742923 PMCID: PMC9224400 DOI: 10.3390/ijms23126480] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 06/07/2022] [Accepted: 06/08/2022] [Indexed: 12/11/2022] Open
Abstract
Extracellular vesicles (EVs) are lipid bilayer membrane particles that play critical roles in intracellular communication through EV-encapsulated informative content, including proteins, lipids, and nucleic acids. Mesenchymal stem cells (MSCs) are pluripotent stem cells with self-renewal ability derived from bone marrow, fat, umbilical cord, menstruation blood, pulp, etc., which they use to induce tissue regeneration by their direct recruitment into injured tissues, including the heart, liver, lung, kidney, etc., or secreting factors, such as vascular endothelial growth factor or insulin-like growth factor. Recently, MSC-derived EVs have been shown to have regenerative effects against various diseases, partially due to the post-transcriptional regulation of target genes by miRNAs. Furthermore, EVs have garnered attention as novel drug delivery systems, because they can specially encapsulate various target molecules. In this review, we summarize the regenerative effects and molecular mechanisms of MSC-derived EVs.
Collapse
|
18
|
Extracellular Vesicle-Mediated Mitochondrial Reprogramming in Cancer. Cancers (Basel) 2022; 14:cancers14081865. [PMID: 35454774 PMCID: PMC9032679 DOI: 10.3390/cancers14081865] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 04/01/2022] [Accepted: 04/02/2022] [Indexed: 02/08/2023] Open
Abstract
Simple Summary Mitochondria are important organelles involved in several key cellular processes including energy production and cell death regulation. For this reason, it is unsurprising that mitochondrial function and structure are altered in several pathological states including cancer. Cancer cells present variate strategies to generate sufficient energy to sustain their high proliferation rates. These adaptative strategies can be mediated by extracellular signals such as extracellular vesicles. These vesicles can alter recipient cellular behavior by delivering their molecular cargo. This review explores the different EV-mediated mitochondrial reprogramming mechanisms supporting cancer survival and progression. Abstract Altered metabolism is a defining hallmark of cancer. Metabolic adaptations are often linked to a reprogramming of the mitochondria due to the importance of these organelles in energy production and biosynthesis. Cancer cells present heterogeneous metabolic phenotypes that can be modulated by signals originating from the tumor microenvironment. Extracellular vesicles (EVs) are recognized as key players in intercellular communications and mediate many of the hallmarks of cancer via the delivery of their diverse biological cargo molecules. Firstly, this review introduces the most characteristic changes that the EV-biogenesis machinery and mitochondria undergo in the context of cancer. Then, it focuses on the EV-driven processes which alter mitochondrial structure, composition, and function to provide a survival advantage to cancer cells in the context of the hallmarks of cancers, such as altered metabolic strategies, migration and invasiveness, immune surveillance escape, and evasion of apoptosis. Finally, it explores the as yet untapped potential of targeting mitochondria using EVs as delivery vectors as a promising cancer therapeutic strategy.
Collapse
|
19
|
Maanaoui M, Kerr-Conte J. Pushing the boundaries of organs before it's too late: pre-emptive regeneration. Transpl Int 2021; 34:1761-1769. [PMID: 34532871 DOI: 10.1111/tri.13969] [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: 04/13/2021] [Revised: 07/05/2021] [Accepted: 07/06/2021] [Indexed: 11/28/2022]
Abstract
Solid organ transplantation is marked by accelerated aging and inexorable fibrosis. It is crucial to promote strategies to attenuate, or to reverse, damage before organ failure. Hence, the objective of this article is to provide insight into strategies, which aim to regenerate or rejuvenate the transplanted organs. Cell therapy with mesenchymal stromal cells is currently under investigation because of their antifibrotic properties. Their ability to promote mitochondrial biogenesis, and to transfer mitochondria to wounded cells, is another approach to boost the organ regeneration. Other teams have investigated bioengineered organs, which consists of decellularization of the damaged organ followed by recellularization. Lastly, the development of CAR-T cell-based technologies may revolutionize the field of transplantation, as recent preclinical studies showed that CAR-T cells could efficiently clear senescent cells from an organ and reverse fibrosis. Ultimately, these cutting-edge strategies may bring the holy grail of a pre-emptive regenerated organ closer to reality.
Collapse
Affiliation(s)
- Mehdi Maanaoui
- Department of Nephrology, CHU Lille, Lille, France.,Inserm, CHU Lille, Institut Pasteur Lille, U1190 - EGID, Univ. Lille, Lille, France
| | - Julie Kerr-Conte
- Inserm, CHU Lille, Institut Pasteur Lille, U1190 - EGID, Univ. Lille, Lille, France
| |
Collapse
|
20
|
Hart DA. What Molecular Recognition Systems Do Mesenchymal Stem Cells/Medicinal Signaling Cells (MSC) Use to Facilitate Cell-Cell and Cell Matrix Interactions? A Review of Evidence and Options. Int J Mol Sci 2021; 22:ijms22168637. [PMID: 34445341 PMCID: PMC8395489 DOI: 10.3390/ijms22168637] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/03/2021] [Accepted: 08/09/2021] [Indexed: 12/12/2022] Open
Abstract
Mesenchymal stem cells, also called medicinal signaling cells (MSC), have been studied regarding their potential to facilitate tissue repair for >30 years. Such cells, derived from multiple tissues and species, are capable of differentiation to a number of lineages (chondrocytes, adipocytes, bone cells). However, MSC are believed to be quite heterogeneous with regard to several characteristics, and the large number of studies performed thus far have met with limited or restricted success. Thus, there is more to understand about these cells, including the molecular recognition systems that are used by these cells to perform their functions, to enhance the realization of their potential to effect tissue repair. This perspective article reviews what is known regarding the recognition systems available to MSC, the possible systems that could be looked for, and alternatives to enhance their localization to specific injury sites and increase their subsequent facilitation of tissue repair. MSC are reported to express recognition molecules of the integrin family. However, there are a number of other recognition molecules that also could be involved such as lectins, inducible lectins, or even a MSC-specific family of molecules unique to these cells. Finally, it may be possible to engineer expression of recognition molecules on the surface of MSC to enhance their function in vivo artificially. Thus, improved understanding of recognition molecules on MSC could further their success in fostering tissue repair.
Collapse
Affiliation(s)
- David A. Hart
- Department of Surgery and Faculty of Kinesiology, University of Calgary, Calgary, AB T2N 4N1, Canada;
- McCaig Institute for Bone & Joint Health, University of Calgary, Calgary, AB T2N 4N1, Canada
- Alberta Health Services Bone & Joint Health Strategic Clinical Network, Edmonton, AB T5H 3E4, Canada
- Centre for Hip Health & Mobility, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| |
Collapse
|