1
|
Afkhami H, Yarahmadi A, Bostani S, Yarian N, Haddad MS, Lesani SS, Aghaei SS, Zolfaghari MR. Converging frontiers in cancer treatment: the role of nanomaterials, mesenchymal stem cells, and microbial agents-challenges and limitations. Discov Oncol 2024; 15:818. [PMID: 39707033 DOI: 10.1007/s12672-024-01590-0] [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] [Received: 09/23/2024] [Accepted: 11/14/2024] [Indexed: 12/23/2024] Open
Abstract
Globally, people widely recognize cancer as one of the most lethal diseases due to its high mortality rates and lack of effective treatment options. Ongoing research into cancer therapies remains a critical area of inquiry, holding significant social relevance. Currently used treatment, such as chemotherapy, radiation, or surgery, often suffers from other problems like damaging side effects, inaccuracy, and the lack of ability to clear tumors. Conventional cancer therapies are usually imprecise and ineffective and usually develop resistance to treatments and cancer recurs. Cancer patients need fresh and innovative treatment that can reduce side effects while maximizing effectiveness. In recent decades several breakthroughs in these, and other areas of medical research, have paved the way for new avenues of fighting cancer including more focused and more effective alternatives. This study reviews exciting possibilities for mesenchymal stem cells (MSCs), nanomaterials, and microbial agents in the modern realm of cancer treatment. Nanoparticles (NPs) have demonstrated surprisingly high potential. They improve drug delivery systems (DDS) significantly, enhance imaging techniques remarkably, and target cancer cells selectively while protecting healthy tissues. MSCs play a double role in tissue repair and are a vehicle for novel cancer treatments such as gene treatments or NPs loaded with therapeutic agents. Additionally, therapies utilizing microbial agents, particularly those involving bacteria, offer an inventive approach to cancer treatment. This review investigates the potential of nanomaterials, MSCs, and microbial agents in addressing the shortcomings of conventional cancer therapies. We will also discuss the challenges and limitations of using these therapeutic approaches.
Collapse
Affiliation(s)
- 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
| | - Aref Yarahmadi
- Department of Biology, Khorramabad Branch, Islamic Azad University, Khorramabad, Iran
| | - Shoroq Bostani
- Department of Microbiology, Qom Branch, Islamic Azad University, Qom, Iran
| | - Nahid Yarian
- Department of Microbiology, Qom Branch, Islamic Azad University, Qom, Iran
| | | | - Shima Sadat Lesani
- Department of Microbiology, Qom Branch, Islamic Azad University, Qom, Iran
| | | | | |
Collapse
|
2
|
Soltanmohammadi F, Mahmoudi Gharehbaba A, Alizadeh E, Javadzadeh Y. Innovative approaches to tissue engineering: Utilizing decellularized extracellular matrix hydrogels for mesenchymal stem cell transport. Int J Biol Macromol 2024; 290:138893. [PMID: 39706433 DOI: 10.1016/j.ijbiomac.2024.138893] [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: 09/24/2024] [Revised: 12/07/2024] [Accepted: 12/16/2024] [Indexed: 12/23/2024]
Abstract
In recent years, the realm of tissue regeneration experienced significant advancements, leading to the development of innovative therapeutic agents. The systemic delivery of mesenchymal stem cells (MSCs) emerged as a promising strategy for promoting tissue regeneration. However, this approach is hindered by hurdles such as poor cell survival, limited cell propagation, and inadequate cell integration. Decellularized extracellular matrix (dECM) hydrogel serves as an innovative carrier that protects MSCs from the detrimental effects of the hostile microenvironment, facilitates their localization and retention at the injection site, and preserves their viability. Regarding its low immunogenicity, low cytotoxicity, high biocompatibility, and its ability to mimic natural extracellular matrix (ECM), this natural hydrogel offers a new avenue for systemic delivery of MSCs. This review digs into the properties of dECM hydrogels (dECMHs), the methods employed for decellularization and the utilization of dECMH as carriers for various types of MSCs for tissue regeneration purposes. This review also sheds light on the benefits of hybrid hydrogels composed of dECMH and other components such as proteins and polysaccharides. By addressing the limitations of conventional hydrogels and enhancing efficacy of cell therapy, dECMH opens new pathways for the future of tissue regeneration.
Collapse
Affiliation(s)
- Fatemeh Soltanmohammadi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Adel Mahmoudi Gharehbaba
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Effat Alizadeh
- Endocrin Research Center and Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Yousef Javadzadeh
- Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran; Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
3
|
Salah B, Shahin D, Sarhan M, Al-Karmi J, Al-Kurdi B, Al-Atoom R, Ismail MA, Hammad N, Jafar H, Awidi A, Ababneh NA. Effect of cigarette smoke on the proliferation, viability, gene expression, and cellular functions of adipose-derived mesenchymal stem cells from smoking and non-smoking donors. Biol Open 2024; 13:bio061665. [PMID: 39625294 PMCID: PMC11646114 DOI: 10.1242/bio.061665] [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: 08/04/2024] [Accepted: 10/11/2024] [Indexed: 12/16/2024] Open
Abstract
Cigarette smoking negatively impacts mesenchymal stem cell functionality, including proliferation, viability, and differentiation potential. Adipose-derived mesenchymal stem cells (ADMSCs) are increasingly used for therapeutic purposes, but the specific effects of smoking in vivo on these cells are poorly understood. This study investigates the effects of cigarette smoke on the proliferation, viability, gene expression, and cellular functions of ADMSCs from smoking and non-smoking donors. In this study, ADMSCs were isolated from healthy smokers and non-smokers, and cell proliferation was assessed using the MTT assay, viability with apoptosis assays, mitochondrial membrane potential (MMP), and gene expression related to oxidative stress and cellular functions. Cell cycle analysis was also conducted. Our findings reveal a significant decrease in the proliferation of ADMSCs from smokers. Apoptosis assays showed reduced viable cells in smokers without a significant change in MMP, suggesting alternative pathways contributing to decreased viability. Gene expression analysis indicated the upregulation of genes associated with oxidative stress response and cellular defense mechanisms and the downregulation of genes related to inflammatory signaling, detoxification, and cellular metabolism. Cell cycle analysis indicates cycle arrest or delay in smokers, possibly due to stress and potential DNA damage. Smoking negatively affects ADMSCs' proliferation, viability, and function through oxidative stress and gene expression alterations. These findings highlight the importance of considering smoking status in ADMSC therapies and the need for further research to mitigate the effect of smoking on stem cells.
Collapse
Affiliation(s)
- Bareqa Salah
- General Surgery Department/Plastic & Reconstructive, Jordan University Hospital, the University of Jordan, 11942
| | - Diana Shahin
- Cell Therapy Center, the University of Jordan, Amman, Jordan, 11942
| | - Momen Sarhan
- Cell Therapy Center, the University of Jordan, Amman, Jordan, 11942
- School of Medicine, the University of Jordan, Amman, Jordan, 11942
| | - Joud Al-Karmi
- School of Medicine, the University of Jordan, Amman, Jordan, 11942
| | - Ban Al-Kurdi
- Cell Therapy Center, the University of Jordan, Amman, Jordan, 11942
| | - Renata Al-Atoom
- Cell Therapy Center, the University of Jordan, Amman, Jordan, 11942
| | | | - Nouran Hammad
- School of Medicine, Jordan University of Science and Technology, Al-Ramtha, Jordan, 22110
| | - Hanan Jafar
- Cell Therapy Center, the University of Jordan, Amman, Jordan, 11942
| | - Abdalla Awidi
- Cell Therapy Center, the University of Jordan, Amman, Jordan, 11942
- Hemostasis and Thrombosis Laboratory, School of Medicine, the University of Jordan, Amman, Jordan, 11942
- Department of Hematology and Oncology, Jordan University Hospital, Amman, Jordan, 11492
| | - Nidaa A. Ababneh
- Cell Therapy Center, the University of Jordan, Amman, Jordan, 11942
| |
Collapse
|
4
|
Zou X, Brigstock D. Extracellular Vesicles from Mesenchymal Stem Cells: Potential as Therapeutics in Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD). Biomedicines 2024; 12:2848. [PMID: 39767754 PMCID: PMC11673942 DOI: 10.3390/biomedicines12122848] [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: 11/11/2024] [Revised: 12/06/2024] [Accepted: 12/12/2024] [Indexed: 01/03/2025] Open
Abstract
Background/Objectives: Metabolic dysfunction-associated steatotic liver disease (MASLD) is characterized by the accumulation of triglycerides within hepatocytes, which can progress to more severe conditions, such as metabolic dysfunction-associated steatohepatitis (MASH), which may include progressive fibrosis, leading to cirrhosis, cancer, and death. This goal of this review is to highlight recent research showing the potential of mesenchymal stem cell-derived extracellular vesicles (MSC-EVs) in reducing the key pathogenic pathways of MASLD or MASH. Methods: Relevant published studies were identified using PubMed with one or more of the following search terms: MASLD, MASH, NAFLD, NASH, exosome, extracellular vesicle (EV), therapy, and/or mesenchymal stem cells (MSC). The primary literature were subsequently downloaded and summarized. Results: Using in vitro or in vivo models, MSC-EVs have been found to counteract oxidative stress, a significant contributor to liver injury in MASH, and to suppress disease progression, including steatosis, inflammation, and, in a few instances, fibrosis. Some of these outcomes have been attributed to specific EV cargo components including microRNAs and proteins. Thus, MSC-EVs enriched with these types of molecules may have improved the therapeutic efficacy for MASLD/MASH and represent a novel approach to potentially halt or reverse the disease process. Conclusions: MSC-EVs are attractive therapeutic agents for treating MASLD/MASH. Further studies are necessary to validate the clinical applicability and efficacy of MSC-EVs in human MASH patients, focusing on optimizing delivery strategies and identifying the pathogenic pathways that are targeted by specific EV components.
Collapse
Affiliation(s)
- Xue Zou
- Center for Clinical and Translational Research, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA;
| | - David Brigstock
- Center for Clinical and Translational Research, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA;
- Department of Surgery, Wexner Medical Center, The Ohio State University, Columbus, OH 43212, USA
| |
Collapse
|
5
|
Semitela A, Marques PAAP, Completo A. Strategies to engineer articular cartilage with biomimetic zonal features: a review. Biomater Sci 2024; 12:5961-6005. [PMID: 39463257 DOI: 10.1039/d4bm00579a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Articular cartilage (AC) is a highly specialized tissue with restricted ability for self-regeneration, given its avascular and acellular nature. Although a considerable number of surgical treatments is available for the repair, reconstruction, and regeneration of AC defects, most of them do not prioritize the development of engineered cartilage with zonal stratification derived from biomimetic biochemical, biomechanical and topographic cues. In the absence of these zonal elements, engineered cartilage will exhibit increased susceptibility to failure and will neither be able to withstand the mechanical loading to which AC is subjected nor will it integrate well with the surrounding tissue. In this regard, new breakthroughs in the development of hierarchical stratified engineered cartilage are highly sought after. Initially, this review provides a comprehensive analysis of the composition and zonal organization of AC, aiming to enhance our understanding of the significance of the structure of AC for its function. Next, we direct our attention towards the existing in vitro and in vivo studies that introduce zonal elements in engineered cartilage to elicit appropriate AC regeneration by employing tissue engineering strategies. Finally, the advantages, challenges, and future perspectives of these approaches are presented.
Collapse
Affiliation(s)
- Angela Semitela
- Centre of Mechanical Technology and Automation (TEMA), Department of Mechanical Engineering, University of Aveiro, 3810-193 Aveiro, Portugal.
| | - Paula A A P Marques
- Centre of Mechanical Technology and Automation (TEMA), Department of Mechanical Engineering, University of Aveiro, 3810-193 Aveiro, Portugal.
| | - António Completo
- Centre of Mechanical Technology and Automation (TEMA), Department of Mechanical Engineering, University of Aveiro, 3810-193 Aveiro, Portugal.
| |
Collapse
|
6
|
Sun L, Xiao M, Chen L, Ni L, Chen X, Zhang L, Yao J, Shao Z, Zhao B, Chen X, Liu Y. Enhanced Tissue Regeneration Through Immunomodulatory Extracellular Vesicles Encapsulated in Diversified Silk-Based Injectable Hydrogels. Adv Healthc Mater 2024; 13:e2401460. [PMID: 39011805 DOI: 10.1002/adhm.202401460] [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: 04/21/2024] [Revised: 06/26/2024] [Indexed: 07/17/2024]
Abstract
Mesenchymal stem cells (MSCs) immunologically trained using lipopolysaccharide (LPS) display enhanced immunomodulatory capabilities. Extracellular vesicles (EVs) derived from MSCs are widely used in regenerative medicine owing to their bioactive properties without the drawbacks of cell therapy. However, it remains unclear whether EVs derived from LPS-stimulated (trained) MSCs (L-EVs) inherit the enhanced reparative potential from their parent cells. Thus, this study first aims to explore the effect of immunological training on the bioactivity of L-EVs. LPS-trained bone marrow-derived MSCs (BMSCs) secrete more EVs, and these EVs significantly promote M2 macrophage polarization. Subsequently, hydrogel systems based on thixotropic injectable silk fibroin are prepared for in vivo EV delivery. These hydrogels have controllable gelation time and exhibit outstanding reparative effects on rat skin wounds and alveolar bone defects. Finally, it is revealed that L-EVs promote M2 macrophage polarization by inhibiting the nuclear translocation of PKM2. Overall, this study shows that the immunological training of BMSCs effectively improves the therapeutic effects of their EVs and provides a convenient and diversified EV delivery strategy using an injectable silk fibroin hydrogel. This strategy has broad clinical application prospects for tissue regeneration.
Collapse
Affiliation(s)
- Liangyan Sun
- Department of Orthodontics, Department of Macromolecular Science, Multidisciplinary Consultant Center, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai, 200001, China
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, 200001, China
| | - Menglin Xiao
- Department of Orthodontics, Department of Macromolecular Science, Multidisciplinary Consultant Center, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai, 200001, China
- State Key Laboratory of Molecular Engineering of Polymers, Laboratory of Advanced Materials, Fudan University, Shanghai, 200433, China
| | - Ling Chen
- Department of Orthodontics, Department of Macromolecular Science, Multidisciplinary Consultant Center, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai, 200001, China
- State Key Laboratory of Molecular Engineering of Polymers, Laboratory of Advanced Materials, Fudan University, Shanghai, 200433, China
| | - Lingyue Ni
- Department of Orthodontics, Department of Macromolecular Science, Multidisciplinary Consultant Center, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai, 200001, China
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, 200001, China
| | - Xiaoxuan Chen
- Department of Orthodontics, Department of Macromolecular Science, Multidisciplinary Consultant Center, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai, 200001, China
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, 200001, China
| | - Lina Zhang
- Department of Biostatistics, Clinical Research Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jinrong Yao
- Department of Orthodontics, Department of Macromolecular Science, Multidisciplinary Consultant Center, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai, 200001, China
- State Key Laboratory of Molecular Engineering of Polymers, Laboratory of Advanced Materials, Fudan University, Shanghai, 200433, China
| | - Zhengzhong Shao
- Department of Orthodontics, Department of Macromolecular Science, Multidisciplinary Consultant Center, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai, 200001, China
- State Key Laboratory of Molecular Engineering of Polymers, Laboratory of Advanced Materials, Fudan University, Shanghai, 200433, China
| | - Bingjiao Zhao
- Department of Orthodontics, Department of Macromolecular Science, Multidisciplinary Consultant Center, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai, 200001, China
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, 200001, China
| | - Xin Chen
- Department of Orthodontics, Department of Macromolecular Science, Multidisciplinary Consultant Center, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai, 200001, China
- State Key Laboratory of Molecular Engineering of Polymers, Laboratory of Advanced Materials, Fudan University, Shanghai, 200433, China
| | - Yuehua Liu
- Department of Orthodontics, Department of Macromolecular Science, Multidisciplinary Consultant Center, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai, 200001, China
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, 200001, China
| |
Collapse
|
7
|
Shamsul Kamal AA, Fakiruddin KS, Bobbo KA, Ling KH, Vidyadaran S, Abdullah S. Engineered Mesenchymal Stem Cells as Treatment for Cancers: Opportunities, Clinical Applications and Challenges. Malays J Med Sci 2024; 31:56-82. [PMID: 39416732 PMCID: PMC11477465 DOI: 10.21315/mjms2024.31.5.5] [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: 02/13/2024] [Accepted: 06/27/2024] [Indexed: 10/19/2024] Open
Abstract
The insufficient and unspecific target of classical chemotherapies often leads to therapy resistance and cancer recurrence. Over the past decades, discoveries about mesenchymal stem cell (MSC) biology have provided new potential approaches to improve cancer therapy. Researchers have utilised the multipotent, regenerative and immunosuppressive qualities of MSCs and tropisms towards inflammatory, hypoxic and malignant sites in various therapeutic applications. Although MSC-based therapies have generally been demonstrated safe, their effectiveness remains limited when these cells are used alone. However, through genetic engineering, researchers have proven that MSCs can be modified to have specialised delivery roles to increase their therapeutic efficacy in cancer treatment. They can be made to overexpress therapeutic proteins through viral or non-viral genetic modification, which enhances their innate properties. Nevertheless, these engineering strategies must be optimised to increase therapeutic efficacy and targeting effectiveness while minimising any loss of MSC function. This review underscores the cutting-edge methods for engineering MSCs, discusses their promise and the difficulties in translating them into clinical settings, and offers some prospective suggestions for the future on achieving their full therapeutic potential.
Collapse
Affiliation(s)
- Aishah Amirah Shamsul Kamal
- UPM-MAKNA Cancer Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, Selangor, Malaysia
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Selangor, Malaysia
| | - Kamal Shaik Fakiruddin
- Haematology Unit, Cancer Research Centre, Institute for Medical Research, National Institutes of Health, Ministry of Health Malaysia, Selangor, Malaysia
| | - Khadijat Abubakar Bobbo
- UPM-MAKNA Cancer Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, Selangor, Malaysia
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Selangor, Malaysia
| | - King Hwa Ling
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Selangor, Malaysia
- Malaysian Research Institute on Ageing, Universiti Putra Malaysia, Selangor, Malaysia
| | - Sharmili Vidyadaran
- Department of Pathology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Selangor, Malaysia
| | - Syahril Abdullah
- UPM-MAKNA Cancer Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, Selangor, Malaysia
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Selangor, Malaysia
- Malaysia Genome and Vaccine Institute, National Institutes of Biotechnology Malaysia, Selangor, Malaysia
| |
Collapse
|
8
|
Maeda S, Matsumoto M, Segawa K, Iwamoto K, Nakamura N. Development of scaffold-free tissue-engineered constructs derived from mesenchymal stem cells with serum-free media for cartilage repair and long-term preservation. Cytotechnology 2024; 76:595-612. [PMID: 39188648 PMCID: PMC11344744 DOI: 10.1007/s10616-024-00637-y] [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: 01/11/2024] [Accepted: 06/18/2024] [Indexed: 08/28/2024] Open
Abstract
Synovial mesenchymal stem cells (sMSCs) have great potential for cartilage repair, but their therapeutic design to avoid adverse effects associated with unknown factors remains a challenge. In addition, because long-term preservation is indispensable to maintain high quality levels until implantation, it is necessary to reduce their fluctuations. This study aimed to investigate the properties and feasibility of novel scaffold-free tissue-engineered constructs using serum-free media and to develop long-term preservation methods. sMSCs were cultured in serum-free media, seeded at high density in a monolayer, and finally developed as a sheet-like construct called "gMSC1". The properties of frozen gMSC1 (Fro-gMSC1) were compared with those of refrigerated gMSC1 (Ref-gMSC1) and then examined by their profile. Chondrogenic differentiation potential was analyzed by quantitative real-time polymerase chain reaction and quantification of glycosaminoglycan content. Xenografts into the cartilage defect model in rats were evaluated by histological staining. gMSC1 showed nearly similar properties independent of the preservation conditions. The animal experiment demonstrated that the defect could be filled with cartilage-like tissue with good integration to the adjacent tissue, suggesting that gMSC1 was formed and replaced the cartilage. Furthermore, several chondrogenesis-related factors were significantly secreted inside and outside gMSC1. Morphological analysis of Fro-gMSC1 revealed comparable quality levels to those of fresh gMSC1. Thus, if cryopreserved, gMSC1, with no complicated materials or processes, could have sustained cartilage repair capacity. gMSC1 is a prominent candidate in novel clinical practice for cartilage repair, allowing for large quantities to be manufactured at one time and preserved for a long term by freezing. Supplementary Information The online version contains supplementary material available at 10.1007/s10616-024-00637-y.
Collapse
Affiliation(s)
- Satoshi Maeda
- TWOCELLS Co., Ltd, 1–6-10 Deshio, Minami-ku, Hiroshima, 734–0001 Japan
| | - Masaya Matsumoto
- TWOCELLS Co., Ltd, 1–6-10 Deshio, Minami-ku, Hiroshima, 734–0001 Japan
| | - Kotaro Segawa
- TWOCELLS Co., Ltd, 1–6-10 Deshio, Minami-ku, Hiroshima, 734–0001 Japan
| | - Kaori Iwamoto
- TWOCELLS Co., Ltd, 1–6-10 Deshio, Minami-ku, Hiroshima, 734–0001 Japan
| | - Norimasa Nakamura
- Department of Orthopaedics, Osaka University Graduate School of Medicine, 2–2 Yamadaoka, Suita, Osaka, 565–0871 Japan
| |
Collapse
|
9
|
Chen B, Zhu Q, Duan M, Li Q, Wang G, Guan X, Yu P, Xu X, He Y, Xu Y. Optimal Treatment Parameters for Ultrasound-Stimulated Microbubbles in Upregulating Proliferation and Stemness of Bone Marrow Mesenchymal Stem Cells. JOURNAL OF ULTRASOUND IN MEDICINE : OFFICIAL JOURNAL OF THE AMERICAN INSTITUTE OF ULTRASOUND IN MEDICINE 2024; 43:1333-1342. [PMID: 38563453 DOI: 10.1002/jum.16457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 03/16/2024] [Accepted: 03/23/2024] [Indexed: 04/04/2024]
Abstract
OBJECTIVES Ultrasound-targeted microbubble disruption (UTMD) is a widely used technique to improve the differentiation and proliferation capacity of mesenchymal stem cells (MSCs), but the optimal therapeutic parameters for UTMD are unclear. In this study, we aimed to find the appropriate peak negative pressure (PNP), which is a key parameter for enhancing the stemness properties and proliferation of MSCs. METHODS Experiments were performed in UTMD group, ultrasound (US) group under different PNP exposure conditions (0.5, 1.0, and 1.5 MPa), and control group. Apoptosis safety was analyzed by flow cytometry and MSC proliferation was measured at 12, 24, and 36 hours after irradiation by cell counting kit 8. The expression of the stemness genes NANOG, OCT-4, and SOX-2 were determined by enzyme-linked immunosorbent assay (ELISA) or reverse transcription polymerase chain reaction. RESULTS The results showed that the 1.5 MPa UTMD-treated group had the highest proliferation capacity of MSCs at 24 hours. ELISA or quantitative reverse transcription polymerase chain reaction results showed that UTMD treatment of the 1.5 MPa group significantly upregulated the expression of the stemness genes NANOG, SOX-2, and OCT-4. CONCLUSIONS In conclusion, the appropriate peak PNP value of UTMD was 1.5 MPa, and 1.5 MPa-mediated UTMD group obviously promoted MSCs proliferation and maintained stemness by upregulating the expression of stemness genes.
Collapse
Affiliation(s)
- Beibei Chen
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
- Department of Ultrasound, Postgraduate Training Basement of Jinzhou Medical University, The PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Qiong Zhu
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
- Department of Ultrasound, 953th Hospital, Shigatse Branch, Xinqiao Hospital, Army Medical University, Shigatse, China
| | - Mao Duan
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Qinglong Li
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Gong Wang
- Department of Ultrasound, Postgraduate Training Basement of Jinzhou Medical University, The PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Xue Guan
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Pu Yu
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Xiaoxun Xu
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Ying He
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Yali Xu
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
10
|
Marquez-Curtis LA, Elliott JAW. Mesenchymal stromal cells derived from various tissues: Biological, clinical and cryopreservation aspects: Update from 2015 review. Cryobiology 2024; 115:104856. [PMID: 38340887 DOI: 10.1016/j.cryobiol.2024.104856] [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: 11/28/2023] [Revised: 01/26/2024] [Accepted: 01/30/2024] [Indexed: 02/12/2024]
Abstract
Mesenchymal stromal cells (MSCs) have become one of the most investigated and applied cells for cellular therapy and regenerative medicine. In this update of our review published in 2015, we show that studies continue to abound regarding the characterization of MSCs to distinguish them from other similar cell types, the discovery of new tissue sources of MSCs, and the confirmation of their properties and functions that render them suitable as a therapeutic. Because cryopreservation is widely recognized as the only technology that would enable the on-demand availability of MSCs, here we show that although the traditional method of cryopreserving cells by slow cooling in the presence of 10% dimethyl sulfoxide (Me2SO) continues to be used by many, several novel MSC cryopreservation approaches have emerged. As in our previous review, we conclude from these recent reports that viable and functional MSCs from diverse tissues can be recovered after cryopreservation using a variety of cryoprotectants, freezing protocols, storage temperatures, and periods of storage. We also show that for logistical reasons there are now more studies devoted to the cryopreservation of tissues from which MSCs are derived. A new topic included in this review covers the application in COVID-19 of MSCs arising from their immunomodulatory and antiviral properties. Due to the inherent heterogeneity in MSC populations from different sources there is still no standardized procedure for their isolation, identification, functional characterization, cryopreservation, and route of administration, and not likely to be a "one-size-fits-all" approach in their applications in cell-based therapy and regenerative medicine.
Collapse
Affiliation(s)
- Leah A Marquez-Curtis
- Department of Chemical and Materials Engineering, University of Alberta, Edmonton, AB, Canada, T6G 1H9; Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, Canada, T6G 1C9
| | - Janet A W Elliott
- Department of Chemical and Materials Engineering, University of Alberta, Edmonton, AB, Canada, T6G 1H9; Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, Canada, T6G 1C9.
| |
Collapse
|
11
|
Akbar N, Razzaq SS, Salim A, Haneef K. Mesenchymal Stem Cell-Derived Exosomes and Their MicroRNAs in Heart Repair and Regeneration. J Cardiovasc Transl Res 2024; 17:505-522. [PMID: 37875715 DOI: 10.1007/s12265-023-10449-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 10/06/2023] [Indexed: 10/26/2023]
Abstract
Mesenchymal stem cells (MSCs) can be differentiated into cardiac, endothelial, and smooth muscle cells. Therefore, MSC-based therapeutic approaches have the potential to deal with the aftermaths of cardiac diseases. However, transplanted stem cells rarely survive in damaged myocardium, proposing that paracrine factors other than trans-differentiation may involve in heart regeneration. Apart from cytokines/growth factors, MSCs secret small, single-membrane organelles named exosomes. The MSC-secreted exosomes are enriched in lipids, proteins, nucleic acids, and microRNA (miRNA). There has been an increasing amount of data that confirmed that MSC-derived exosomes and their active molecule microRNA (miRNAs) regulate signaling pathways involved in heart repair/regeneration. In this review, we systematically present an overview of MSCs, their cardiac differentiation, and the role of MSC-derived exosomes and exosomal miRNAs in heart regeneration. In addition, biological functions regulated by MSC-derived exosomes and exosomal-derived miRNAs in the process of heart regeneration are reviewed.
Collapse
Affiliation(s)
- Nukhba Akbar
- Dr. Zafar H. Zaidi Center for Proteomics, University of Karachi, Karachi, 75270, Pakistan
| | - Syeda Saima Razzaq
- Dr. Zafar H. Zaidi Center for Proteomics, University of Karachi, Karachi, 75270, Pakistan
| | - Asmat Salim
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Kanwal Haneef
- Dr. Zafar H. Zaidi Center for Proteomics, University of Karachi, Karachi, 75270, Pakistan.
| |
Collapse
|
12
|
Liu J, Qi L, Bao S, Yan F, Chen J, Yu S, Dong C. The acute spinal cord injury microenvironment and its impact on the homing of mesenchymal stem cells. Exp Neurol 2024; 373:114682. [PMID: 38199509 DOI: 10.1016/j.expneurol.2024.114682] [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: 11/07/2023] [Revised: 12/08/2023] [Accepted: 01/02/2024] [Indexed: 01/12/2024]
Abstract
Spinal cord injury (SCI) is a highly debilitating condition that inflicts devastating harm on the lives of affected individuals, underscoring the urgent need for effective treatments. By activating inflammatory cells and releasing inflammatory factors, the secondary injury response creates an inflammatory microenvironment that ultimately determines whether neurons will undergo necrosis or regeneration. In recent years, mesenchymal stem cells (MSCs) have garnered increasing attention for their therapeutic potential in SCI. MSCs not only possess multipotent differentiation capabilities but also have homing abilities, making them valuable as carriers and mediators of therapeutic agents. The inflammatory microenvironment induced by SCI recruits MSCs to the site of injury through the release of various cytokines, chemokines, adhesion molecules, and enzymes. However, this mechanism has not been previously reported. Thus, a comprehensive exploration of the molecular mechanisms and cellular behaviors underlying the interplay between the inflammatory microenvironment and MSC homing is crucial. Such insights have the potential to provide a better understanding of how to harness the therapeutic potential of MSCs in treating inflammatory diseases and facilitating injury repair. This review aims to delve into the formation of the inflammatory microenvironment and how it influences the homing of MSCs.
Collapse
Affiliation(s)
- Jinyi Liu
- Department of Anatomy, Medical College of Nantong University, Nantong, China
| | - Longju Qi
- Affiliated Nantong Hospital 3 of Nantong University, Nantong, China
| | - Shengzhe Bao
- Department of Anatomy, Medical College of Nantong University, Nantong, China
| | - Fangsu Yan
- Department of Anatomy, Medical College of Nantong University, Nantong, China
| | - Jiaxi Chen
- Department of Anatomy, Medical College of Nantong University, Nantong, China
| | - Shumin Yu
- Department of Anatomy, Medical College of Nantong University, Nantong, China
| | - Chuanming Dong
- Department of Anatomy, Medical College of Nantong University, Nantong, China; Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China.
| |
Collapse
|
13
|
Slautin V, Konyshev K, Gavrilov I, Beresneva O, Maklakova I, Grebnev D. Fucoxanthin Enhances the Antifibrotic Potential of Placenta-derived Mesenchymal Stem Cells in a CCl4-induced Mouse Model of Liver. Curr Stem Cell Res Ther 2024; 19:1484-1496. [PMID: 38204245 DOI: 10.2174/011574888x279940231206100902] [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: 10/04/2023] [Revised: 10/31/2023] [Accepted: 11/03/2023] [Indexed: 01/12/2024]
Abstract
BACKGROUND The effectiveness of fucoxanthin (Fx) in liver diseases has been reported due to its anti-inflammatory and antifibrotic effects. Mesenchymal stem cells (MSCs)-based therapy has also been proposed as a promising strategy for liver fibrosis treatment. Recent studies have shown that the co-administration of MSCs and drugs demonstrates a pronounced effect on liver fibrosis. AIM This study aimed to determine the therapeutic potential of placenta-derived MSCs (PD-MSCs) in combination with Fx to treat liver fibrosis and evaluate their impact on the main links of liver fibrosis pathogenesis. METHODS After PD-MSCs isolation and identification, outbred ICR/CD1 mice were divided into five groups: Control group, CCl4 group (CCl4), Fx group (CCl4+Fx), PD-MSCs group (CCl4+MSCs) and cotreatment group (CCl4+MSCs+Fx). Biochemical histopathological investigations were performed. Semiquantitative analysis of the alpha-smooth muscle actin (α-SMA+), matrix metalloproteinases (MMP-9+, MMP-13+), tissue inhibitor of matrix metalloproteinases-1 (TIMP-1+) areas, and the number of positive cells in them were studied by immunohistochemical staining. Transforming growth factor-beta (TGF-β), hepatic growth factor (HGF), procollagen-1 (COL1α1) in liver homogenate and proinflammatory cytokines in blood serum were determined using an enzyme immunoassay. RESULTS Compared to the single treatment with PD-MSCs or Fx, their combined administration significantly reduced liver enzyme activity, the severity of liver fibrosis, the proinflammatory cytokine levels, TGF-β level, α-SMA+, TIMP-1+ areas and the number of positive cells in them, and increased HGF level, MMP-13+, and MMP-9+ areas. CONCLUSION Fx enhanced the therapeutic potential of PD-MSCs in CCl4-induced liver fibrosis, but more investigations are necessary to understand the mutual impact of PD-MSCs and Fx.
Collapse
Affiliation(s)
- Vasilii Slautin
- Department of Pathophysiology , Ural State Medical University, 3, Repin Street, 620028, Yekaterinburg, Russia
| | - Konstantin Konyshev
- Department of Pathophysiology , Ural State Medical University, 3, Repin Street, 620028, Yekaterinburg, Russia
- Institute of Medical Cell Technologies, 22a, Karl Marx Street, 620026, Yekaterinburg, Russia
| | - Ilya Gavrilov
- Department of Pathophysiology , Ural State Medical University, 3, Repin Street, 620028, Yekaterinburg, Russia
- Institute of Medical Cell Technologies, 22a, Karl Marx Street, 620026, Yekaterinburg, Russia
| | - Olga Beresneva
- Department of Pathophysiology , Ural State Medical University, 3, Repin Street, 620028, Yekaterinburg, Russia
| | - Irina Maklakova
- Department of Pathophysiology , Ural State Medical University, 3, Repin Street, 620028, Yekaterinburg, Russia
- Institute of Medical Cell Technologies, 22a, Karl Marx Street, 620026, Yekaterinburg, Russia
| | - Dmitry Grebnev
- Department of Pathophysiology , Ural State Medical University, 3, Repin Street, 620028, Yekaterinburg, Russia
- Institute of Medical Cell Technologies, 22a, Karl Marx Street, 620026, Yekaterinburg, Russia
| |
Collapse
|
14
|
Garcia Gómez-Heras S, Garcia-Arranz M, Vega-Clemente L, Olivera-Salazar R, Vélez Pinto JF, Fernández-García M, Guadalajara H, Yáñez R, Garcia-Olmo D. Study of the Effect of Wild-Type and Transiently Expressing CXCR4 and IL-10 Mesenchymal Stromal Cells in a Mouse Model of Peritonitis. Int J Mol Sci 2023; 25:520. [PMID: 38203690 PMCID: PMC10778615 DOI: 10.3390/ijms25010520] [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: 11/21/2023] [Revised: 12/19/2023] [Accepted: 12/21/2023] [Indexed: 01/12/2024] Open
Abstract
Sepsis due to peritonitis is a process associated with an inflammatory state. Mesenchymal stromal cells (MSCs) modulate the immune system due to the paracrine factors released and may be a therapeutic alternative. Three treatment groups were developed in a murine model of peritonitis to verify the effect of human adipose mesenchymal stem cell (hASCs). Additionally, a temporary modification was carried out on them to improve their arrival in inflamed tissues (CXCR4), as well as their anti-inflammatory activity (IL-10). The capacity to reduce systemic inflammation was studied using a local application (peritoneal injection) as a treatment route. Comparisons involving the therapeutic effect of wild-type ASCs and ASCs transiently expressing CXCR4 and IL-10 were carried out with the aim of generating an improved anti-inflammatory response for sepsis in addition to standard antibiotic treatment. However, under the experimental conditions used in these studies, no differences were found between both groups with ASCs. The peritoneal administration of hASCs or genetically modified hASCs constitutes an efficient and safe therapy in our model of mouse peritonitis.
Collapse
Affiliation(s)
- Soledad Garcia Gómez-Heras
- Department of Basic Health Science, Faculty of Health Sciences, Rey Juan Carlos University, 28922 Alcorcón, Spain
| | - Mariano Garcia-Arranz
- New Therapy Laboratory, Health Research Institute Fundación Jiménez Díaz, 28033 Madrid, Spain; (L.V.-C.); (R.O.-S.); (H.G.); (D.G.-O.)
- Department of Surgery, Faculty of Medicine, Universidad Autónoma de Madrid, 28029 Madrid, Spain;
| | - Luz Vega-Clemente
- New Therapy Laboratory, Health Research Institute Fundación Jiménez Díaz, 28033 Madrid, Spain; (L.V.-C.); (R.O.-S.); (H.G.); (D.G.-O.)
| | - Rocio Olivera-Salazar
- New Therapy Laboratory, Health Research Institute Fundación Jiménez Díaz, 28033 Madrid, Spain; (L.V.-C.); (R.O.-S.); (H.G.); (D.G.-O.)
| | - Juan Felipe Vélez Pinto
- Department of Surgery, Faculty of Medicine, Universidad Autónoma de Madrid, 28029 Madrid, Spain;
| | - María Fernández-García
- Biomedical Innovation Unit, Division of Hematopoietic Innovative Therapies, Centro de Investigaciones Energéticas Medioambientales y Tecnológicas (CIEMAT), Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD, UAM), 28040 Madrid, Spain; (M.F.-G.); (R.Y.)
| | - Héctor Guadalajara
- New Therapy Laboratory, Health Research Institute Fundación Jiménez Díaz, 28033 Madrid, Spain; (L.V.-C.); (R.O.-S.); (H.G.); (D.G.-O.)
- Surgery Department, Fundación Jiménez Díaz University Hospital, 28033 Madrid, Spain
| | - Rosa Yáñez
- Biomedical Innovation Unit, Division of Hematopoietic Innovative Therapies, Centro de Investigaciones Energéticas Medioambientales y Tecnológicas (CIEMAT), Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD, UAM), 28040 Madrid, Spain; (M.F.-G.); (R.Y.)
| | - Damian Garcia-Olmo
- New Therapy Laboratory, Health Research Institute Fundación Jiménez Díaz, 28033 Madrid, Spain; (L.V.-C.); (R.O.-S.); (H.G.); (D.G.-O.)
- Department of Surgery, Faculty of Medicine, Universidad Autónoma de Madrid, 28029 Madrid, Spain;
- Surgery Department, Fundación Jiménez Díaz University Hospital, 28033 Madrid, Spain
| |
Collapse
|
15
|
Wang X, Tian H, Yang X, Zhao H, Liang X, Li Y. Mesenchymal Stem Cells‐Derived Extracellular Vesicles in Orthopedic Diseases: Recent Advances and Therapeutic Potential. ADVANCED THERAPEUTICS 2023; 6. [DOI: 10.1002/adtp.202300193] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Indexed: 01/06/2025]
Abstract
AbstractEver since the first application of mesenchymal stem cell (MSC) transplantation treating human hematologic malignancies in 1995, MSC‐based treatments have demonstrated great therapeutic potential in clinical settings. However, only a few MSC‐based cell therapy products have been clinically approved. Accumulating evidence suggests that the beneficial effects of MSCs are mainly attributed to the release of paracrine factors or extracellular vesicles (EVs) rather than their mesodermal differentiation potential. Therefore, MSC‐derived EVs (MSC‐EVs), such as exosomes and microvesicles, have merged as promising alternatives to traditional cell‐based therapeutics in clinical practice. They offer several advantages such as better safety, lower immunogenicity, protection of cargoes from degradation, and the ability to overcome biological barriers. Moreover, there have been multiple clinical studies exploring the potential of MSC‐EVs for treating various diseases, including orthopedic disorders. However, there is no definitive “cure” for conditions such as osteoporosis and other bone disorders, but MSC‐EVs have displayed significant therapeutic potential for these orthopedic ailments. Therefore, the objective of this study is to conduct a systematic review of current knowledge related to MSC‐EVs and emphasize their potential application in treating orthopedic diseases, such as bone defects, osteoarthritis, osteoporosis, intervertebral disc degeneration, osteosarcoma, and osteoradionecrosis.
Collapse
Affiliation(s)
- Xinwen Wang
- Department of Foot and Ankle Surgery, Honghui Hospital Xi'an Jiaotong University Xi'an Shaanxi Province 710054 P. R. China
| | - Haodong Tian
- Department of Foot and Ankle Surgery, Honghui Hospital Xi'an Jiaotong University Xi'an Shaanxi Province 710054 P. R. China
| | - Xinquan Yang
- Department of Foot and Ankle Surgery, Honghui Hospital Xi'an Jiaotong University Xi'an Shaanxi Province 710054 P. R. China
| | - Hongmou Zhao
- Department of Foot and Ankle Surgery, Honghui Hospital Xi'an Jiaotong University Xi'an Shaanxi Province 710054 P. R. China
| | - Xiaojun Liang
- Department of Foot and Ankle Surgery, Honghui Hospital Xi'an Jiaotong University Xi'an Shaanxi Province 710054 P. R. China
| | - Yi Li
- Department of Foot and Ankle Surgery, Honghui Hospital Xi'an Jiaotong University Xi'an Shaanxi Province 710054 P. R. China
| |
Collapse
|
16
|
He YF, Wang XL, Deng SP, Wang YL, Huang QQ, Lin S, Lyu GR. Latest progress in low-intensity pulsed ultrasound for studying exosomes derived from stem/progenitor cells. Front Endocrinol (Lausanne) 2023; 14:1286900. [PMID: 38089611 PMCID: PMC10715436 DOI: 10.3389/fendo.2023.1286900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 11/06/2023] [Indexed: 12/18/2023] Open
Abstract
Stem cells have self-renewal, replication, and multidirectional differentiation potential, while progenitor cells are undifferentiated, pluripotent or specialized stem cells. Stem/progenitor cells secrete various factors, such as cytokines, exosomes, non-coding RNAs, and proteins, and have a wide range of applications in regenerative medicine. However, therapies based on stem cells and their secreted exosomes present limitations, such as insufficient source materials, mature differentiation, and low transplantation success rates, and methods addressing these problems are urgently required. Ultrasound is gaining increasing attention as an emerging technology. Low-intensity pulsed ultrasound (LIPUS) has mechanical, thermal, and cavitation effects and produces vibrational stimuli that can lead to a series of biochemical changes in organs, tissues, and cells, such as the release of extracellular bodies, cytokines, and other signals. These changes can alter the cellular microenvironment and affect biological behaviors, such as cell differentiation and proliferation. Here, we discuss the effects of LIPUS on the biological functions of stem/progenitor cells, exosomes, and non-coding RNAs, alterations involved in related pathways, various emerging applications, and future perspectives. We review the roles and mechanisms of LIPUS in stem/progenitor cells and exosomes with the aim of providing a deeper understanding of LIPUS and promoting research and development in this field.
Collapse
Affiliation(s)
- Yi-fang He
- Department of Ultrasound, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Xia-li Wang
- Department of Ultrasound, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
- Departments of Medical Imaging, Quanzhou Medical College, Quanzhou, China
| | - Shuang-ping Deng
- Department of Ultrasound, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Yan-li Wang
- Department of Ultrasound, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Qing-qing Huang
- Department of Ultrasound, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Shu Lin
- Centre of Neurological and Metabolic Research, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
- Diabetes and Metabolism Division, Garvan Institute of Medical Research, Darlinghurst, Sydney, NSW, Australia
| | - Guo-rong Lyu
- Department of Ultrasound, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
- Departments of Medical Imaging, Quanzhou Medical College, Quanzhou, China
| |
Collapse
|
17
|
Huang F, He Y, Zhang M, Luo K, Li J, Li J, Zhang X, Dong X, Tang J. Progress in Research on Stem Cells in Neonatal Refractory Diseases. J Pers Med 2023; 13:1281. [PMID: 37623531 PMCID: PMC10455340 DOI: 10.3390/jpm13081281] [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: 06/23/2023] [Revised: 08/03/2023] [Accepted: 08/16/2023] [Indexed: 08/26/2023] Open
Abstract
With the development and progress of medical technology, the survival rate of premature and low-birth-weight infants has increased, as has the incidence of a variety of neonatal diseases, such as hypoxic-ischemic encephalopathy, intraventricular hemorrhage, bronchopulmonary dysplasia, necrotizing enterocolitis, and retinopathy of prematurity. These diseases cause severe health conditions with poor prognoses, and existing control methods are ineffective for such diseases. Stem cells are a special type of cells with self-renewal and differentiation potential, and their mechanisms mainly include anti-inflammatory and anti-apoptotic properties, reducing oxidative stress, and boosting regeneration. Their paracrine effects can affect the microenvironment in which they survive, thereby affecting the biological characteristics of other cells. Due to their unique abilities, stem cells have been used in treating various diseases. Therefore, stem cell therapy may open up the possibility of treating such neonatal diseases. This review summarizes the research progress on stem cells and exosomes derived from stem cells in neonatal refractory diseases to provide new insights for most researchers and clinicians regarding future treatments. In addition, the current challenges and perspectives in stem cell therapy are discussed.
Collapse
Affiliation(s)
- Fangjun Huang
- Department of Neonatology, West China Second Hospital, Sichuan University, Chengdu 610041, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu 610041, China
| | - Yang He
- Department of Neonatology, West China Second Hospital, Sichuan University, Chengdu 610041, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu 610041, China
| | - Meng Zhang
- Department of Neonatology, West China Second Hospital, Sichuan University, Chengdu 610041, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu 610041, China
| | - Keren Luo
- Department of Neonatology, West China Second Hospital, Sichuan University, Chengdu 610041, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu 610041, China
| | - Jiawen Li
- Department of Neonatology, West China Second Hospital, Sichuan University, Chengdu 610041, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu 610041, China
| | - Jiali Li
- Department of Neonatology, West China Second Hospital, Sichuan University, Chengdu 610041, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu 610041, China
| | - Xinyu Zhang
- Department of Neonatology, West China Second Hospital, Sichuan University, Chengdu 610041, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu 610041, China
| | - Xiaoyan Dong
- Department of Neonatology, West China Second Hospital, Sichuan University, Chengdu 610041, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu 610041, China
| | - Jun Tang
- Department of Neonatology, West China Second Hospital, Sichuan University, Chengdu 610041, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu 610041, China
| |
Collapse
|
18
|
Yan L, Li J, Zhang C. The role of MSCs and CAR-MSCs in cellular immunotherapy. Cell Commun Signal 2023; 21:187. [PMID: 37528472 PMCID: PMC10391838 DOI: 10.1186/s12964-023-01191-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 06/07/2023] [Indexed: 08/03/2023] Open
Abstract
Chimeric antigen receptors (CARs) are widely used by T cells (CAR-T cells), natural killer cells dendritic cells and macrophages, and they are of great importance in cellular immunotherapy. However, the use of CAR-related products faces several challenges, including the poor persistence of cells carrying CARs, cell dysfunction or exhaustion, relapse of disease, immune effector cell-associated neurotoxicity syndrome, cytokine release syndrome, low efficacy against solid tumors and immunosuppression by the tumor microenvironment. Another important cell therapy regimen involves mesenchymal stem cells (MSCs). Recent studies have shown that MSCs can improve the anticancer functions of CAR-related products. CAR-MSCs can overcome the flaws of cellular immunotherapy. Thus, MSCs can be used as a biological vehicle for CARs. In this review, we first discuss the characteristics and immunomodulatory functions of MSCs. Then, the role of MSCs as a source of exosomes, including the characteristics of MSC-derived exosomes and their immunomodulatory functions, is discussed. The role of MSCs in CAR-related products, CAR-related product-derived exosomes and the effect of MSCs on CAR-related products are reviewed. Finally, the use of MSCs as CAR vehicles is discussed. Video Abstract.
Collapse
Affiliation(s)
- Lun Yan
- Medical Center of Hematology, State Key Laboratory of Trauma, Burn and Combined Injury, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Jing Li
- Medical Center of Hematology, State Key Laboratory of Trauma, Burn and Combined Injury, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Cheng Zhang
- Medical Center of Hematology, State Key Laboratory of Trauma, Burn and Combined Injury, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China.
| |
Collapse
|
19
|
Yang L, Fang C, Song C, Zhang Y, Zhang R, Zhou S. Mesenchymal Stem Cell-Derived Exosomes are Effective for Radiation Enteritis and Essential for the Proliferation and Differentiation of Lgr5 + Intestinal Epithelial Stem Cells by Regulating Mir-195/Akt/β-Catenin Pathway. Tissue Eng Regen Med 2023; 20:739-751. [PMID: 37326937 PMCID: PMC10352229 DOI: 10.1007/s13770-023-00541-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 03/25/2023] [Accepted: 03/29/2023] [Indexed: 06/17/2023] Open
Abstract
BACKGROUND Radiation enteritis (RE) is a common complication of abdominal or pelvic radiotherapy, which when severe, could be life-threatening. Currently, there are no effective treatments. Studies have shown that mesenchymal stem cells (MSCs)-derived exosomes (MSC-exos) exhibit promising therapeutic effects in inflammatory diseases. However, the specific role of MSC-exos in RE and the regulatory mechanisms remain elusive. METHODS In vivo assay was carried out by injecting MSC-exos into the total abdominal irradiation (TAI)-induced RE mouse model. For in vitro assay, Lgr5-positive intestinal epithelial stem cells (Lgr5+ IESC) were extracted from mice, followed by irradiation along with MSC-exos treatment. HE staining was performed to measure histopathological changes. mRNA expression of inflammatory factors TNF-α and IL-6 and stem cell markers LGR5, and OCT4 were quantified by RT-qPCR. EdU and TUNEL staining was performed to estimate cell proliferation and apoptosis. MiR-195 expression in TAI mice and radiation-induced Lgr5+ IESC was tested. RESULTS We found that the injection of MSC-exos inhibited inflammatory reaction, increased stem cell marker expression, and maintained intestinal epithelial integrity in TAI mice. Furthermore, MSC-exos treatment increased the proliferation and simultaneously suppressed apoptosis in radiation-stimulated Lgr5+ IESC. MiR-195 expression increased by radiation exposure was decreased by MSC-exos therapy. MiR-195 overexpression facilitated the progress of RE by counteracting the effect of MSC-exos. Mechanistically, the Akt and Wnt/β-catenin pathways inhibited by MSC-exos were activated by miR-195 upregulation. CONCLUSION MSC-Exos are effective in treating RE and are essential for the proliferation and differentiation of Lgr5+ IESCs. Moreover, MSC-exos mediates its function by regulating miR-195 Akt β-catenin pathways.
Collapse
Affiliation(s)
- Leilei Yang
- Department of Gastrointestinal Surgery, Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, No. 150, Ximen Street, Linhai, Taizhou, 317000, Zhejiang, China
| | - Chengfeng Fang
- Department of Gastrointestinal Surgery, Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, No. 150, Ximen Street, Linhai, Taizhou, 317000, Zhejiang, China
| | - Caifang Song
- Department of Gastrointestinal Surgery, Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, No. 150, Ximen Street, Linhai, Taizhou, 317000, Zhejiang, China
| | - Yaya Zhang
- Department of Gastrointestinal Surgery, Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, No. 150, Ximen Street, Linhai, Taizhou, 317000, Zhejiang, China
| | - Ruili Zhang
- Department of Gastrointestinal Surgery, Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, No. 150, Ximen Street, Linhai, Taizhou, 317000, Zhejiang, China.
| | - Shenkang Zhou
- Department of Gastrointestinal Surgery, Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, No. 150, Ximen Street, Linhai, Taizhou, 317000, Zhejiang, China.
| |
Collapse
|
20
|
Baouche M, Ochota M, Locatelli Y, Mermillod P, Niżański W. Mesenchymal Stem Cells: Generalities and Clinical Significance in Feline and Canine Medicine. Animals (Basel) 2023; 13:1903. [PMID: 37370414 DOI: 10.3390/ani13121903] [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: 03/31/2023] [Revised: 05/29/2023] [Accepted: 06/02/2023] [Indexed: 06/29/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are multipotent cells: they can proliferate like undifferentiated cells and have the ability to differentiate into different types of cells. A considerable amount of research focuses on the potential therapeutic benefits of MSCs, such as cell therapy or tissue regeneration, and MSCs are considered powerful tools in veterinary regenerative medicine. They are the leading type of adult stem cells in clinical trials owing to their immunosuppressive, immunomodulatory, and anti-inflammatory properties, as well as their low teratogenic risk compared with pluripotent stem cells. The present review details the current understanding of the fundamental biology of MSCs. We focus on MSCs' properties and their characteristics with the goal of providing an overview of therapeutic innovations based on MSCs in canines and felines.
Collapse
Affiliation(s)
- Meriem Baouche
- Department of Reproduction and Clinic of Farm Animals, Wrocław University of Environmental and Life Sciences, 50-366 Wrocław, Poland
| | - Małgorzata Ochota
- Department of Reproduction and Clinic of Farm Animals, Wrocław University of Environmental and Life Sciences, 50-366 Wrocław, Poland
| | - Yann Locatelli
- Physiology of Reproduction and Behaviors (PRC), UMR085, INRAE, CNRS, University of Tours, 37380 Nouzilly, France
- Museum National d'Histoire Naturelle, Réserve Zoologique de la Haute Touche, 36290 Obterre, France
| | - Pascal Mermillod
- Physiology of Reproduction and Behaviors (PRC), UMR085, INRAE, CNRS, University of Tours, 37380 Nouzilly, France
| | - Wojciech Niżański
- Department of Reproduction and Clinic of Farm Animals, Wrocław University of Environmental and Life Sciences, 50-366 Wrocław, Poland
| |
Collapse
|
21
|
Río C, Jahn AK, Martin-Medina A, Calvo Bota AM, De Francisco Casado MT, Pont Antona PJ, Gigirey Castro O, Carvajal ÁF, Villena Portella C, Gómez Bellvert C, Iglesias A, Calvo Benito J, Gayà Puig A, Ortiz LA, Sala-Llinàs E. Mesenchymal Stem Cells from COPD Patients Are Capable of Restoring Elastase-Induced Emphysema in a Murine Experimental Model. Int J Mol Sci 2023; 24:ijms24065813. [PMID: 36982887 PMCID: PMC10054868 DOI: 10.3390/ijms24065813] [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: 01/31/2023] [Revised: 03/15/2023] [Accepted: 03/17/2023] [Indexed: 03/22/2023] Open
Abstract
COPD is a chronic lung disease that affects millions of people, declining their lung function and impairing their life quality. Despite years of research and drug approvals, we are still not capable of halting progression or restoring normal lung function. Mesenchymal stem cells (MSC) are cells with extraordinary repair capacity, and MSC-based therapy brings future hope for COPD treatment, although the best source and route of administration are unclear. MSC from adipose tissue (AD-MSC) represents an option for autologous treatment; however, they could be less effective than donor MSC. We compared in vitro behavior of AD-MSC from COPD and non-COPD individuals by migration/proliferation assay, and tested their therapeutic potential in an elastase mouse model. In addition, we tested intravenous versus intratracheal routes, inoculating umbilical cord (UC) MSC and analyzed molecular changes by protein array. Although COPD AD-MSC have impaired migratory response to VEGF and cigarette smoke, they were as efficient as non-COPD in reducing elastase-induced lung emphysema. UC-MSC reduced lung emphysema regardless of the administration route and modified the inflammatory profile in elastase-treated mice. Our data demonstrate equal therapeutic potential of AD-MSC from COPD and non-COPD subjects in the pre-clinical model, thus supporting their autologous use in disease.
Collapse
Affiliation(s)
- Carlos Río
- Inflammation, Repair and Cancer of Respiratory Diseases (i-Respire), Fundació Institut d’ Investigació Sanitària Illes Balears (IdISBa), 07120 Palma, Spain
| | - Andreas K. Jahn
- Inflammation, Repair and Cancer of Respiratory Diseases (i-Respire), Fundació Institut d’ Investigació Sanitària Illes Balears (IdISBa), 07120 Palma, Spain
| | - Aina Martin-Medina
- Inflammation, Repair and Cancer of Respiratory Diseases (i-Respire), Fundació Institut d’ Investigació Sanitària Illes Balears (IdISBa), 07120 Palma, Spain
| | - Alba Marina Calvo Bota
- Inflammation, Repair and Cancer of Respiratory Diseases (i-Respire), Fundació Institut d’ Investigació Sanitària Illes Balears (IdISBa), 07120 Palma, Spain
| | | | - Pere Joan Pont Antona
- Estabulary, Scientific-Technical Services, Universitat de les Illes Balears (UIB), 07122 Palma, Spain
| | | | | | - Cristina Villena Portella
- Inflammation, Repair and Cancer of Respiratory Diseases (i-Respire), Fundació Institut d’ Investigació Sanitària Illes Balears (IdISBa), 07120 Palma, Spain
- CIBERES Pulmonary Biobank Consortium, Hospital Universitari Son Espases, 07120 Palma, Spain
| | | | - Amanda Iglesias
- Inflammation, Repair and Cancer of Respiratory Diseases (i-Respire), Fundació Institut d’ Investigació Sanitària Illes Balears (IdISBa), 07120 Palma, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Javier Calvo Benito
- Banc de Teixits, Blood and Tissue Bank of the Balearic Islands (FBSTIB), 07120 Palma, Spain
- Cell Therapy and Tissue Engineering Group (TERCIT), Institut d’ Investigació Sanitària Illes Balears (IdISBa), 07004 Palma, Spain
| | - Antoni Gayà Puig
- Banc de Teixits, Blood and Tissue Bank of the Balearic Islands (FBSTIB), 07120 Palma, Spain
- Cell Therapy and Tissue Engineering Group (TERCIT), Institut d’ Investigació Sanitària Illes Balears (IdISBa), 07004 Palma, Spain
| | - Luis A. Ortiz
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Ernest Sala-Llinàs
- Inflammation, Repair and Cancer of Respiratory Diseases (i-Respire), Fundació Institut d’ Investigació Sanitària Illes Balears (IdISBa), 07120 Palma, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Pulmonary Medicine, Hospital Universitari Son Espases, 07120 Palma, Spain
- Correspondence: ; Tel.: +34-871-206-507
| |
Collapse
|
22
|
Rodríguez-Echeverri C, Gómez BL, González Á. Histoplasma capsulatum modulates the immune response, affects proliferation and differentiation, and induces apoptosis of mesenchymal stromal cells. Mycoses 2023; 66:157-167. [PMID: 36219488 DOI: 10.1111/myc.13537] [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: 07/24/2022] [Revised: 10/06/2022] [Accepted: 10/09/2022] [Indexed: 01/12/2023]
Abstract
Mesenchymal stromal cells (MSC) have been widely used not only for tissue regeneration but also for the treatment of various diseases; however, it has been shown that infection of MSCs by different pathogens can attenuate their intrinsic immunomodulatory properties, affecting the proliferation and differentiation of these cells. Currently, the mechanisms by which MSCs respond to pathogen invasion are poorly understood. Therefore, the objective of the present study was to determine if the infection of bone marrow-derived MSCs, with yeasts of the pathogenic fungus Histoplasma capsulatum affects the activation, differentiation and/or proliferation of the MSCs. The results indicate that MSCs have the ability to phagocytose H. capsulatum yeasts but do not exert a notable antifungal effect. On the contrary, the infection of the MSCs with this fungal pathogen not only modulates the expression of inflammatory mediators by a mechanism dependent on TLR2, TLR4 and Dectin-1 but also affects the viability and differentiation capacity of the MSCs. These findings suggest that infection of MSCs by H. capsulatum could not only affect haematopoiesis but also modulate the immune response in the infected host and, furthermore, these MSCs could provide a niche for the fungus, allowing it to persist and evade the immune response of the host.
Collapse
Affiliation(s)
- Carolina Rodríguez-Echeverri
- Basic and Applied Microbiology Group (MICROBA), School of Microbiology, Universidad de Antioquia, Medellín, Colombia
| | - Beatriz L Gómez
- Translational Microbiology and Emerging Diseases Research Group (MICROS), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Ángel González
- Basic and Applied Microbiology Group (MICROBA), School of Microbiology, Universidad de Antioquia, Medellín, Colombia
| |
Collapse
|
23
|
Bezerra AF, Alves JPM, Fernandes CCL, Cavalcanti CM, Silva MRL, Conde AJH, Tetaping GM, Ferreira ACA, Melo LM, Rodrigues APR, Rondina D. Dyslipidemia induced by lipid diet in late gestation donor impact on growth kinetics and in vitro potential differentiation of umbilical cord Wharton's Jelly mesenchymal stem cells in goats. Vet Res Commun 2022; 46:1259-1270. [PMID: 36125693 DOI: 10.1007/s11259-022-09995-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 09/04/2022] [Indexed: 10/14/2022]
Abstract
Mesenchymal stem cells (MSC) from the umbilical cord (UC) have several attractive properties for clinical use. This study aimed to verify the impact of a lipid-rich diet during late gestation of donor goats on the growth and differentiation of MSCs from UC. From the 100th day of pregnancy to delivery, 22 goats were grouped based on their diet into the donor-lipid (DLD; n = 11) and donor-baseline (DBD; n = 11) diet groups. Diets were isonitrogenous and isoenergetic, differing in fat content (2.8% vs. 6.3% on a dry matter basis). Wharton's jelly (WJ) fragments were cultured. After primary culture, samples of WJ-MSCs were characterized by the expression of CD90, CD73, CD34, CD45, CD105, and Fas genes, mitochondrial activity using MitoTracker (MT) fluorescence probe, and growth kinetics. Population doubling time (PDT) was also determined. WJ-MSCs were differentiated into chondrocytes, adipocytes and osteocytes, and the mineralized area and adipocytes were determined. The lipid diet significantly increased triglyceride and cholesterol levels during pregnancy. The DLD group showed sub-expression of the CD90 gene, a high MT intensity, and a low proliferation rate at the end of the subculture. The mean PDT was 83.9 ± 1.3 h. Mineralized area and lipid droplet stain intensity from osteogenic and adipogenic differentiations, respectively, were greater in DLD. We conclude that in donor goats, dietary dyslipidemia during late pregnancy affects the ability of UC-derived MSCs to express their developmental potential in vitro, thus limiting their possible use for therapeutic purposes.
Collapse
Affiliation(s)
| | | | | | - Camila Muniz Cavalcanti
- School of Veterinary Medicine, Ceará State University (UECE), Fortaleza, CE, 60.714.903, Brazil
| | | | | | - Gildas Mbemya Tetaping
- School of Veterinary Medicine, Ceará State University (UECE), Fortaleza, CE, 60.714.903, Brazil
| | | | - Luciana Magalhães Melo
- School of Veterinary Medicine, Centro Universitario Fametro (UNIFAMETRO), Fortaleza, CE, 60010-470, Brazil
| | | | - Davide Rondina
- School of Veterinary Medicine, Ceará State University (UECE), Fortaleza, CE, 60.714.903, Brazil.
| |
Collapse
|
24
|
Stem Cells from Human Exfoliated Deciduous Teeth Attenuate Atopic Dermatitis Symptoms in Mice through Modulating Immune Balance and Skin Barrier Function. Mediators Inflamm 2022; 2022:6206883. [PMID: 35909660 PMCID: PMC9334056 DOI: 10.1155/2022/6206883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 05/13/2022] [Accepted: 07/07/2022] [Indexed: 11/17/2022] Open
Abstract
Atopic dermatitis (AD) is a chronic skin inflammatory disease associated with immune abnormalities and disrupted skin barrier function. Mesenchymal stem cells (MSCs) have been suggested as an alternative therapeutic option in AD. Stem cells from human exfoliated deciduous teeth (SHEDs) are a unique postnatal stem cell population with high immunomodulatory properties. The aim of this study was to explore the effects of SHEDs on AD in the BALB/c mouse model induced by 2,4-dinitrochlorobenzene (DNCB). SHEDs were administrated intravenously or subcutaneously, and clinical severity, histopathological findings, skin barrier function, and organ indexes were evaluated. Skin tissue cytokine mRNA levels and serum cytokine protein levels were further analysed. SHED administration significantly alleviated AD clinical severity, including dermatitis scores, ear thickness, scratching behaviour, and infiltration of mast cells. In addition, disrupted skin barrier function and enlarged spleens were restored by SHED administration. Further, SHED treatment reduced the levels of IgE, IgG1, and thymic stromal lymphopoietin (TSLP) in the serum and the modulated expression of Th1-, Th2-, and Th17-associated cytokines in skin lesions. In conclusion, SHEDs attenuated AD-like skin lesions in mice by modulating the immune balance and skin barrier function. SHEDs could be a potential new treatment agent for AD.
Collapse
|
25
|
Chitosan chemistry review for living organisms encapsulation. Carbohydr Polym 2022; 295:119877. [DOI: 10.1016/j.carbpol.2022.119877] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 07/12/2022] [Accepted: 07/13/2022] [Indexed: 12/20/2022]
|
26
|
Silvestro S, Diomede F, Chiricosta L, Zingale VD, Marconi GD, Pizzicannella J, Valeri A, Avanzini MA, Calcaterra V, Pelizzo G, Mazzon E. The Role of Hypoxia in Improving the Therapeutic Potential of Mesenchymal Stromal Cells. A Comparative Study From Healthy Lung and Congenital Pulmonary Airway Malformations in Infants. Front Bioeng Biotechnol 2022; 10:868486. [PMID: 35774062 PMCID: PMC9237219 DOI: 10.3389/fbioe.2022.868486] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 05/20/2022] [Indexed: 11/17/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) play an important role in the field of regenerative medicine thanks to their immunomodulatory properties and their ability to secrete paracrine factors. The use of MSCs has also been tested in children with congenital lung diseases inducing fibrosis and a decrease in lung function. Congenital malformations of the pulmonary airways (CPAM) are the most frequently encountered lung lesion that results from defects in early development of airways. Despite the beneficial properties of MSCs, interventions aimed at improving the outcome of cell therapy are needed. Hypoxia may be an approach aimed to ameliorate the therapeutic potential of MSCs. In this regard, we evaluated the transcriptomic profile of MSCs collected from pediatric patients with CPAM, analyzing similarities and differences between healthy tissue (MSCs-lung) and cystic tissue (MSCs-CPAM) both in normoxia and in cells preconditioned with hypoxia (0.2%) for 24 h. Study results showed that hypoxia induces cell cycle activation, increasing in such a way the cell proliferation ability, and enhancing cell anaerobic metabolism in both MSCs-lung and MSCs-CPAM-lung. Additionally, hypoxia downregulated several pro-apoptotic genes preserving MSCs from apoptosis and, at the same time, improving their viability in both comparisons. Finally, data obtained indicates that hypoxia leads to a greater expression of genes involved in the regulation of the cytoskeleton in MSCs-lung than MSCs-CPAM.
Collapse
Affiliation(s)
| | - Francesca Diomede
- Department of Innovative Technologies in Medicine and Dentistry, University “G. D’Annunzio” Chieti-Pescara, Chieti, Italy
| | | | | | - Guya Diletta Marconi
- Department of Medical, Oral and Biotechnological Sciences, University “G. D’Annunzio” Chieti-Pescara, Chieti, Italy
| | | | - Andrea Valeri
- IRCCS Centro Neurolesi “Bonino-Pulejo”, Messina, Italy
| | - Maria Antonietta Avanzini
- Cell Factory, Pediatric Hematology Oncology Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Valeria Calcaterra
- Pediatrics and Adolescentology Unit, Department of Internal Medicine, University of Pavia, Pavia, Italy
- Pediatric Department, Children’s Hospital “Vittore Buzzi”, Milano, Italy
| | - Gloria Pelizzo
- Pediatric Surgery Department, Children’s Hospital “Vittore Buzzi”, Milano, Italy
- Department of Biomedical and Clinical Sciences-L. Sacco, University of Milan, Milan, Italy
| | | |
Collapse
|
27
|
Xia P, Shi Y, Wang X, Li X. Advances in the application of low-intensity pulsed ultrasound to mesenchymal stem cells. Stem Cell Res Ther 2022; 13:214. [PMID: 35619156 PMCID: PMC9137131 DOI: 10.1186/s13287-022-02887-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Accepted: 05/03/2022] [Indexed: 11/10/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are stem cells that exhibit self-renewal capacity and multi-directional differentiation potential. They can be extracted from the bone marrow and umbilical cord, as well as adipose, amnion, and other tissues. They are widely used in tissue engineering and are currently considered an important source of cells in the field of regenerative medicine. Since certain limitations, such as an insufficient cell source, mature differentiation, and low transplantation efficiency, are still associated with MSCs, researchers have currently focused on improving the efficacy of MSCs. Low-intensity pulsed ultrasound (LIPUS) has mechanical, cavitation, and thermal effects that can produce different biological effects on organs, tissues, and cells. It can be used for fracture treatment, cartilage repair, and stem cell applications. An in-depth study of the role and mechanism of action of LIPUS in MSC treatment would promote our understanding of LIPUS and promote research in this field. In this article, we have reviewed the progress in research on the use of LIPUS with various MSCs and comprehensively discussed the progress in the use of LIPUS for promoting the proliferation, differentiation, and migration of MSCs, as well as its future prospects.
Collapse
Affiliation(s)
- Peng Xia
- Department of Rehabilitation Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China.
| | - Yi Shi
- Department of Rehabilitation Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Xiaoju Wang
- Department of Rehabilitation Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Xueping Li
- Department of Rehabilitation Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China.
| |
Collapse
|
28
|
Gilazieva Z, Ponomarev A, Rizvanov A, Solovyeva V. The Dual Role of Mesenchymal Stromal Cells and Their Extracellular Vesicles in Carcinogenesis. BIOLOGY 2022; 11:biology11060813. [PMID: 35741334 PMCID: PMC9220333 DOI: 10.3390/biology11060813] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 05/23/2022] [Accepted: 05/24/2022] [Indexed: 02/07/2023]
Abstract
Simple Summary Extracellular vesicles (EVs) are membrane structures that play the role of intermediaries between tumor cells and the tumor microenvironment (TME) because they have the ability to transport lipids, transcription factors, mRNA, and proteins. Mesenchymal stem cells (MSCs) are a major component of the TME and may have different effects on tumor progression using EVs. This review includes information about various studies which have reported that EVs from MSCs can have either antitumor or pro-tumor effects, depending on both the tumor type and developmental stage. It provides an overview of the published data on EV MSCs and their effect on tumor cells. In addition, the use of EV MSCs for the development of new methods for treating oncological diseases is described. Abstract Mesenchymal stem cells (MSCs) are a major component of the tumor microenvironment (TME) and play an important role in tumor progression. MSCs remodel the extracellular matrix, participate in the epithelial–mesenchymal transition, promote the spread of metastases, and inhibit antitumor immune responses in the TME; however, there are also data pertaining to the antitumor effects of MSCs. MSCs activate the cell death mechanism by modulating the expression of proteins involved in the regulation of the cell cycle, angiogenesis receptors, and proapoptotic proteins. One of the main ways in which MSCs and TME interact is through the production of extracellular vesicles (EVs) by cells. Currently, data on the effects of both MSCs and their EVs on tumor cells are rather contradictory. Various studies have reported that EVs from MSCs can have either antitumor or pro-tumor effects, depending on both the tumor type and developmental stage. In this review, we discuss published data on EV MSCs and their effect on tumor cells. The molecular composition of vesicles obtained from MSCs is also presented in the review. In addition, the use of EV MSCs for the development of new methods for treating oncological diseases is described.
Collapse
|
29
|
da Silva RO, Hastreiter AA, Vivian GK, Dias CC, Santos ACA, Makiyama EN, Borelli P, Fock RA. The influence of association between aging and reduced protein intake on some immunomodulatory aspects of bone marrow mesenchymal stem cells: an experimental study. Eur J Nutr 2022; 61:3391-3406. [PMID: 35508740 DOI: 10.1007/s00394-022-02893-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 04/12/2022] [Indexed: 11/30/2022]
Abstract
PURPOSE Dietary protein deficiency is common in the elderly, compromising hematopoiesis and the immune response, and may cause a greater susceptibility to infections. Mesenchymal stem cells (MSCs) have immunomodulatory properties and are essential to hematopoiesis. Therefore, this study aimed to investigate, in an aging model subjected to malnutrition due a reduced protein intake, aspects related to the immunomodulatory capacity of MSCs. METHODS Male C57BL/6 mice from young and elderly groups were fed with normoproteic or hypoproteic diets (12% and 2% of protein, respectively) and nutritional, biochemical and hematological parameters were evaluated. MSCs from bone marrow were isolated, characterized and their secretory parameters evaluated, along with gene expression. Additionally, the effects of aging and protein malnutrition on MSC immunomodulatory properties were assessed. RESULTS Malnourished mice lost weight and demonstrated anemia, leukopenia, and bone marrow hypoplasia. MSCs from elderly animals from both groups showed reduced CD73 expression and higher senescence rate; also, the malnourished state affected CD73 expression in young animals. The production of IL-1β and IL-6 by MSCs was affected by aging and malnutrition, but the IL-10 production not. Aging also increased the expression of NFκB, reducing the expression of STAT-3. However, MSCs from malnourished groups, regardless of age, showed decreased TGF-β and PGE2 production. Evaluation of the immunomodulatory capacity of MSCs revealed that aging and malnutrition affected, mainly in lymphocytes, the production of IFN-γ and IL-10. CONCLUSION Aging and reduced protein intake are factors that, alone or together, influence the immunomodulatory properties of MSCs and provide basic knowledge that can be further investigated to explore whether MSCs' therapeutic potential may be affected.
Collapse
Affiliation(s)
- Renaira Oliveira da Silva
- Laboratory of Experimental Haematology, Department of Clinical and Toxicological Analysis, School of Pharmaceutical Sciences, University of São Paulo, Avenida Lineu Prestes, 580-Bloco 17, São Paulo, SP, 05508-900, Brazil
| | - Araceli Aparecida Hastreiter
- Laboratory of Experimental Haematology, Department of Clinical and Toxicological Analysis, School of Pharmaceutical Sciences, University of São Paulo, Avenida Lineu Prestes, 580-Bloco 17, São Paulo, SP, 05508-900, Brazil
| | - Gabriela Kodja Vivian
- Laboratory of Experimental Haematology, Department of Clinical and Toxicological Analysis, School of Pharmaceutical Sciences, University of São Paulo, Avenida Lineu Prestes, 580-Bloco 17, São Paulo, SP, 05508-900, Brazil
| | - Carolina Carvalho Dias
- Laboratory of Experimental Haematology, Department of Clinical and Toxicological Analysis, School of Pharmaceutical Sciences, University of São Paulo, Avenida Lineu Prestes, 580-Bloco 17, São Paulo, SP, 05508-900, Brazil
| | - Andressa Cristina Antunes Santos
- Laboratory of Experimental Haematology, Department of Clinical and Toxicological Analysis, School of Pharmaceutical Sciences, University of São Paulo, Avenida Lineu Prestes, 580-Bloco 17, São Paulo, SP, 05508-900, Brazil
| | - Edson Naoto Makiyama
- Laboratory of Experimental Haematology, Department of Clinical and Toxicological Analysis, School of Pharmaceutical Sciences, University of São Paulo, Avenida Lineu Prestes, 580-Bloco 17, São Paulo, SP, 05508-900, Brazil
| | - Primavera Borelli
- Laboratory of Experimental Haematology, Department of Clinical and Toxicological Analysis, School of Pharmaceutical Sciences, University of São Paulo, Avenida Lineu Prestes, 580-Bloco 17, São Paulo, SP, 05508-900, Brazil
| | - Ricardo Ambrósio Fock
- Laboratory of Experimental Haematology, Department of Clinical and Toxicological Analysis, School of Pharmaceutical Sciences, University of São Paulo, Avenida Lineu Prestes, 580-Bloco 17, São Paulo, SP, 05508-900, Brazil.
| |
Collapse
|
30
|
Zhu Y, Luo M, Bai X, Lou Y, Nie P, Jiang S, Li J, Li B, Luo P. Administration of mesenchymal stem cells in diabetic kidney disease: mechanisms, signaling pathways, and preclinical evidence. Mol Cell Biochem 2022; 477:2073-2092. [PMID: 35469057 DOI: 10.1007/s11010-022-04421-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 03/22/2022] [Indexed: 02/07/2023]
Abstract
Diabetic kidney disease (DKD) is a serious microvascular complication of diabetes. Currently, the prevalence and mortality of DKD are increasing annually. However, with no effective drugs to prevent its occurrence and development, the primary therapeutic option is to control blood sugar and blood pressure. Therefore, new and effective drugs/methods are imperative to prevent the development of DKD in patients with diabetes. Mesenchymal stem cells (MSCs) with multi-differentiation potential and paracrine function have received extensive attention as a new treatment option for DKD. However, their role and mechanism in the treatment of DKD remain unclear, and clinical applications are still being explored. Given this, we here provide an unbiased review of recent advances in MSCs for the treatment of DKD in the last decade from the perspectives of the pathogenesis of DKD, biological characteristics of MSCs, and different molecular and signaling pathways. Furthermore, we summarize information on combination therapy strategies using MSCs. Finally, we discuss the challenges and prospects for clinical application.
Collapse
Affiliation(s)
- Yuexin Zhu
- Department of Nephrology, The Second Hospital of Jilin University, 218 ZiQiang Street, Changchun, 130041, Jilin, People's Republic of China
| | - Manyu Luo
- Department of Nephrology, The Second Hospital of Jilin University, 218 ZiQiang Street, Changchun, 130041, Jilin, People's Republic of China
| | - Xue Bai
- Department of Nephrology, The Second Hospital of Jilin University, 218 ZiQiang Street, Changchun, 130041, Jilin, People's Republic of China
| | - Yan Lou
- Department of Nephrology, The Second Hospital of Jilin University, 218 ZiQiang Street, Changchun, 130041, Jilin, People's Republic of China
| | - Ping Nie
- Department of Nephrology, The Second Hospital of Jilin University, 218 ZiQiang Street, Changchun, 130041, Jilin, People's Republic of China
| | - Shan Jiang
- Department of Nephrology, The Second Hospital of Jilin University, 218 ZiQiang Street, Changchun, 130041, Jilin, People's Republic of China
| | - Jicui Li
- Department of Nephrology, The Second Hospital of Jilin University, 218 ZiQiang Street, Changchun, 130041, Jilin, People's Republic of China
| | - Bing Li
- Department of Nephrology, The Second Hospital of Jilin University, 218 ZiQiang Street, Changchun, 130041, Jilin, People's Republic of China.
| | - Ping Luo
- Department of Nephrology, The Second Hospital of Jilin University, 218 ZiQiang Street, Changchun, 130041, Jilin, People's Republic of China.
| |
Collapse
|
31
|
Cho HY, Lee S, Park JH, Kwak YH, Kweon H, Kang D. Competitive Hybridization of a Microarray Identifies CMKLR1 as an Up-Regulated Gene in Human Bone Marrow-Derived Mesenchymal Stem Cells Compared to Human Embryonic Fibroblasts. Curr Issues Mol Biol 2022; 44:1497-1512. [PMID: 35723360 PMCID: PMC9164045 DOI: 10.3390/cimb44040102] [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: 02/24/2022] [Revised: 03/21/2022] [Accepted: 03/21/2022] [Indexed: 11/28/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have been widely applied to the regeneration of damaged tissue and the modulation of immune response. The purity of MSC preparation and the delivery of MSCs to a target region are critical factors for success in therapeutic application. In order to define the molecular identity of an MSC, the gene expression pattern of a human bone marrow-derived mesenchymal stem cell (hBMSC) was compared with that of a human embryonic fibroblast (hEF) by competitive hybridization of a microarray. A total of 270 and 173 genes were two-fold up- and down-regulated with FDR < 0.05 in the hBMSC compared to the hEF, respectively. The overexpressed genes in the hBMSC over the hEF, including transcription factors, were enriched for biological processes such as axial pattern formation, face morphogenesis and skeletal system development, which could be expected from the differentiation potential of MSCs. CD70 and CD339 were identified as additional CD markers that were up-regulated in the hBMSC over the hEF. The differential expression of CD70 and CD339 might be exploited to distinguish hEF and hBMSC. CMKLR1, a chemokine receptor, was up-regulated in the hBMSC compared to the hEF. RARRES2, a CMKLR1 ligand, stimulated specific migration of the hBMSC, but not of the hEF. RARRES2 manifested as ~two-fold less effective than SDF-1α in the directional migration of the hBMSC. The expression of CMKLR1 was decreased upon the osteoblastic differentiation of the hBMSC. However, the RARRES2-loaded 10% HA-silk scaffold did not recruit endogenous cells to the scaffold in vivo. The RARRES2−CMKLR1 axis could be employed in recruiting systemically delivered or endogenous MSCs to a specific target lesion.
Collapse
Affiliation(s)
- Hee-Yeon Cho
- Ilsong Institute of Life Science, Hallym University, Beodeunaru-ro 55, Seoul 07247, Korea; (H.-Y.C.); (S.L.); (J.-H.P.)
- Department of Biomedical Gerontology, Hallym University Graduate School, Chuncheon 24252, Korea
| | - Sooho Lee
- Ilsong Institute of Life Science, Hallym University, Beodeunaru-ro 55, Seoul 07247, Korea; (H.-Y.C.); (S.L.); (J.-H.P.)
| | - Ji-Hong Park
- Ilsong Institute of Life Science, Hallym University, Beodeunaru-ro 55, Seoul 07247, Korea; (H.-Y.C.); (S.L.); (J.-H.P.)
- Department of Biomedical Gerontology, Hallym University Graduate School, Chuncheon 24252, Korea
| | - Yoon Hae Kwak
- Department of Orthopaedic Surgery, Asan Medical Center, Ulsan University College of Medicine, Seoul 05505, Korea;
| | - HaeYong Kweon
- Industrial Insect and Sericulture Division, National Institute of Agricultural Sciences, RDA, Wanju-gun 55365, Korea;
| | - Dongchul Kang
- Ilsong Institute of Life Science, Hallym University, Beodeunaru-ro 55, Seoul 07247, Korea; (H.-Y.C.); (S.L.); (J.-H.P.)
- Department of Biomedical Gerontology, Hallym University Graduate School, Chuncheon 24252, Korea
- Correspondence: ; Tel.: +82-2-6923-8230
| |
Collapse
|
32
|
Tomita S, Ishihara S, Kurita R. A polymer-based chemical tongue for the non-invasive monitoring of osteogenic stem-cell differentiation by pattern recognition of serum-supplemented spent media. J Mater Chem B 2022; 10:7581-7590. [DOI: 10.1039/d2tb00606e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The development of non-invasive techniques to characterize cultured cells is invaluable not only to ensure the reproducibility of cell research, but also for quality assurance of industrial cell products for...
Collapse
|
33
|
Chen J, Chen Y, Du X, Liu G, Fei X, Peng JR, Zhang X, Xiao F, Wang X, Yang X, Feng Z. Integrative Studies of Human Cord Blood Derived Mononuclear Cells and Umbilical Cord Derived Mesenchyme Stem Cells in Ameliorating Bronchopulmonary Dysplasia. Front Cell Dev Biol 2021; 9:679866. [PMID: 34858969 PMCID: PMC8631197 DOI: 10.3389/fcell.2021.679866] [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: 03/12/2021] [Accepted: 08/30/2021] [Indexed: 11/13/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD) is a common pulmonary complication observed in preterm infants that is composed of multifactorial pathogenesis. Current strategies, albeit successful in moderately reducing morbidity and mortality of BPD, failed to draw overall satisfactory conclusion. Here, using a typical mouse model mimicking hallmarks of BPD, we revealed that both cord blood-derived mononuclear cells (CB-MNCs) and umbilical cord-derived mesenchymal stem cells (UC-MSCs) are efficient in alleviating BPD. Notably, infusion of CB-MNCs has more prominent effects in preventing alveolar simplification and pulmonary vessel loss, restoring pulmonary respiratory functions and balancing inflammatory responses. To further elucidate the underlying mechanisms within the divergent therapeutic effects of UC-MSC and CB-MNC, we systematically investigated the long noncoding RNA (lncRNA)-microRNA (miRNA)-messenger RNA (mRNA) and circular RNA (circRNA)-miRNA-mRNA networks by whole-transcriptome sequencing. Importantly, pathway analysis integrating Gene Ontology (GO)/Kyoto Encyclopedia of Genes and Genomes (KEGG)/gene set enrichment analysis (GSEA) method indicates that the competing endogenous RNA (ceRNA) network is mainly related to the regulation of GTPase activity (GO: 0043087), extracellular signal-regulated kinase 1 (ERK1) and ERK2 signal cascade (GO: 0070371), chromosome regulation (GO: 0007059), and cell cycle control (GO: 0044770). Through rigorous selection of the lncRNA/circRNA-based ceRNA network, we demonstrated that the hub genes reside in UC-MSC- and CB-MNC-infused networks directed to the function of cell adhesion, motor transportation (Cdk13, Lrrn2), immune homeostasis balance, and autophagy (Homer3, Prkcd) relatively. Our studies illustrate the first comprehensive mRNA-miRNA-lncRNA and mRNA-miRNA-circRNA networks in stem cell-infused BPD model, which will be valuable in identifying reliable biomarkers or therapeutic targets for BPD pathogenesis and shed new light in the priming and conditioning of UC-MSCs or CB-MNCs in the treatment of neonatal lung injury.
Collapse
Affiliation(s)
- Jia Chen
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Department of Neonatology, Senior Department of Pediatrics, The Seventh Medical Center of PLA General Hospital, Beijing, China.,National Engineering Laboratory for Birth Defects Prevention and Control of Key Technology, Beijing, China.,Beijing Key Laboratory of Pediatric Organ Failure, Beijing, China
| | - Yuhan Chen
- Department of Neonatology, Senior Department of Pediatrics, The Seventh Medical Center of PLA General Hospital, Beijing, China.,National Engineering Laboratory for Birth Defects Prevention and Control of Key Technology, Beijing, China.,Beijing Key Laboratory of Pediatric Organ Failure, Beijing, China
| | - Xue Du
- Department of Neonatology, Senior Department of Pediatrics, The Seventh Medical Center of PLA General Hospital, Beijing, China.,National Engineering Laboratory for Birth Defects Prevention and Control of Key Technology, Beijing, China.,Beijing Key Laboratory of Pediatric Organ Failure, Beijing, China.,The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Guojun Liu
- Shandong Qilu Stem Cell Engineering Co., Ltd., Jinan, China
| | - Xiaowei Fei
- The First Affiliated Hospital of Dalian Medical University, Dalian, China.,Department of Neurosurgery, Xijing Hospital, Air Force Military Medical University, Xi'an, China
| | - Jian Ru Peng
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Department of Neonatology, Senior Department of Pediatrics, The Seventh Medical Center of PLA General Hospital, Beijing, China.,National Engineering Laboratory for Birth Defects Prevention and Control of Key Technology, Beijing, China.,Beijing Key Laboratory of Pediatric Organ Failure, Beijing, China
| | - Xing Zhang
- Department of Neonatology, Senior Department of Pediatrics, The Seventh Medical Center of PLA General Hospital, Beijing, China.,National Engineering Laboratory for Birth Defects Prevention and Control of Key Technology, Beijing, China.,Beijing Key Laboratory of Pediatric Organ Failure, Beijing, China
| | - Fengjun Xiao
- Department of Experimental Hematology and Biochemistry, Beijing Institute of Radiation Medicine, Beijing, China
| | - Xue Wang
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiao Yang
- Department of Neonatology, Senior Department of Pediatrics, The Seventh Medical Center of PLA General Hospital, Beijing, China.,National Engineering Laboratory for Birth Defects Prevention and Control of Key Technology, Beijing, China.,Beijing Key Laboratory of Pediatric Organ Failure, Beijing, China
| | - Zhichun Feng
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Department of Neonatology, Senior Department of Pediatrics, The Seventh Medical Center of PLA General Hospital, Beijing, China.,National Engineering Laboratory for Birth Defects Prevention and Control of Key Technology, Beijing, China.,Beijing Key Laboratory of Pediatric Organ Failure, Beijing, China.,The First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
34
|
Krstić J, Mojsilović S, Mojsilović SS, Santibanez JF. Regulation of the mesenchymal stem cell fate by interleukin-17: Implications in osteogenic differentiation. World J Stem Cells 2021; 13:1696-1713. [PMID: 34909118 PMCID: PMC8641017 DOI: 10.4252/wjsc.v13.i11.1696] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 05/14/2021] [Accepted: 10/18/2021] [Indexed: 02/06/2023] Open
Abstract
Bone regeneration is a tightly regulated process that ensures proper repair and functionality after injury. The delicate balance between bone formation and resorption is governed by cytokines and signaling molecules released during the inflammatory response. Interleukin (IL)-17A, produced in the early phase of inflammation, influences the fate of osteoprogenitors. Due to their inherent capacity to differentiate into osteoblasts, mesenchymal stem/stromal cells (MSCs) contribute to bone healing and regeneration. This review presents an overview of IL-17A signaling and the leading cellular and molecular mechanisms by which it regulates the osteogenic differentiation of MSCs. The main findings demonstrating IL-17A’s influence on osteoblastogenesis are described. To this end, divergent information exists about the capacity of IL-17A to regulate MSCs’ osteogenic fate, depending on the tissue context and target cell type, along with contradictory findings in the same cell types. Therefore, we summarize the data showing both the pro-osteogenic and anti-osteogenic roles of IL-17, which may help in the understanding of IL-17A function in bone repair and regeneration.
Collapse
Affiliation(s)
- Jelena Krstić
- Gottfried Schatz Research Center, Medical University of Graz, Graz 8010, Austria
| | - Slavko Mojsilović
- Group for Hematology and Stem Cells, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, Belgrade 11129, Serbia
| | - Sonja S Mojsilović
- Group for Immunology, Institute for Medical Research, National Institute of Republic of Serbia, Belgrade 11129, Serbia
| | - Juan F Santibanez
- Group for Molecular Oncology, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, Belgrade 11000, Serbia
- Centro Integrativo de Biología y Química Aplicada, Universidad Bernardo O’Higgins, Chile 8370993, Chile
| |
Collapse
|
35
|
SARS-CoV-2 Exposed Mesenchymal Stromal Cell from Congenital Pulmonary Airway Malformations: Transcriptomic Analysis and the Expression of Immunomodulatory Genes. Int J Mol Sci 2021; 22:ijms222111814. [PMID: 34769246 PMCID: PMC8584055 DOI: 10.3390/ijms222111814] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 10/28/2021] [Accepted: 10/29/2021] [Indexed: 12/22/2022] Open
Abstract
The inflammatory response plays a central role in the complications of congenital pulmonary airway malformations (CPAM) and severe coronavirus disease 2019 (COVID-19). The aim of this study was to evaluate the transcriptional changes induced by SARS-CoV-2 exposure in pediatric MSCs derived from pediatric lung (MSCs-lung) and CPAM tissues (MSCs-CPAM) in order to elucidate potential pathways involved in SARS-CoV-2 infection in a condition of exacerbated inflammatory response. MSCs-lung and MSCs-CPAM do not express angiotensin-converting enzyme 2 (ACE2) and transmembrane serine protease 2 (TRMPSS2). SARS-CoV-2 appears to be unable to replicate in MSCs-CPAM and MSCs-lung. MSCs-lung and MSCs-CPAM maintained the expression of stemness markers MSCs-lung show an inflammatory response (IL6, IL1B, CXCL8, and CXCL10), and the activation of Notch3 non-canonical pathway; this route appears silent in MSCs-CPAM, and cytokine genes expression is reduced. Decreased value of p21 in MSCs-lung suggested no cell cycle block, and cells did not undergo apoptosis. MSCs-lung appears to increase genes associated with immunomodulatory function but could contribute to inflammation, while MSCs-CPAM keeps stable or reduce the immunomodulatory receptors expression, but they also reduce their cytokines expression. These data indicated that, independently from their perilesional or cystic origin, the MSCs populations already present in a patient affected with CPAM are not permissive for SARS-CoV-2 entry, and they will not spread the disease in case of infection. Moreover, these MSCs will not undergo apoptosis when they come in contact with SARS-CoV-2; on the contrary, they maintain their staminality profile.
Collapse
|
36
|
Takao S, Nakashima T, Masuda T, Namba M, Sakamoto S, Yamaguchi K, Horimasu Y, Miyamoto S, Iwamoto H, Fujitaka K, Hamada H, Takahashi S, Nakashima A, Hattori N. Human bone marrow-derived mesenchymal stromal cells cultured in serum-free media demonstrate enhanced antifibrotic abilities via prolonged survival and robust regulatory T cell induction in murine bleomycin-induced pulmonary fibrosis. Stem Cell Res Ther 2021; 12:506. [PMID: 34530920 PMCID: PMC8444523 DOI: 10.1186/s13287-021-02574-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 08/27/2021] [Indexed: 12/29/2022] Open
Abstract
Background Mesenchymal stromal cells (MSCs) are a potential therapeutic tool for pulmonary fibrosis. However, ex vivo MSC expansion using serum poses risks of harmful immune responses or unknown pathogen infections in the recipients. Therefore, MSCs cultured in serum-free media (SF-MSCs) are ideal for clinical settings; however, their efficacy in pulmonary fibrosis is unknown. Here, we investigated the effects of SF-MSCs on bleomycin-induced pulmonary inflammation and fibrosis compared to those of MSCs cultured in serum-containing media (S-MSCs). Methods SF-MSCs and S-MSCs were characterized in vitro using RNA sequence analysis. The in vivo kinetics and efficacy of SF-MSC therapy were investigated using a murine model of bleomycin-induced pulmonary fibrosis. For normally distributed data, Student’s t test and one-way repeated measures analysis of variance followed by post hoc Tukey’s test were used for comparison between two groups and multiple groups, respectively. For non-normally distributed data, Kruskal–Wallis and Mann–Whitney U tests were used for comparison between groups, using e Bonferroni’s correction for multiple comparisons. All tests were two-sided, and P < 0.05 was considered statistically significant. Results Serum-free media promoted human bone marrow-derived MSC expansion and improved lung engraftment of intravenously administered MSCs in recipient mice. SF-MSCs inhibited the reduction in serum transforming growth factor-β1 and the increase of interleukin-6 in both the serum and the bronchoalveolar lavage fluid during bleomycin-induced pulmonary fibrosis. SF-MSC administration increased the numbers of regulatory T cells (Tregs) in the blood and lungs more strongly than in S-MSC administration. Furthermore, SF-MSCs demonstrated enhanced antifibrotic effects on bleomycin-induced pulmonary fibrosis, which were diminished by antibody-mediated Treg depletion. Conclusions SF-MSCs significantly suppressed BLM-induced pulmonary inflammation and fibrosis through enhanced induction of Tregs into the lungs and corrected the dysregulated cytokine balance. Therefore, SF-MSCs could be a useful tool for preventing pulmonary fibrosis progression without the demerits of serum use. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02574-5.
Collapse
Affiliation(s)
- Shun Takao
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Taku Nakashima
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, 734-8551, Japan.
| | - Takeshi Masuda
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Masashi Namba
- Department of Clinical Oncology, Hiroshima University Hospital, 1-2-3, Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Shinjiro Sakamoto
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Kakuhiro Yamaguchi
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Yasushi Horimasu
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Shintaro Miyamoto
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Hiroshi Iwamoto
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Kazunori Fujitaka
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Hironobu Hamada
- Department of Physical Analysis and Therapeutic Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Shinya Takahashi
- Department of Cardiovascular Surgery, Graduate School of Medicine, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Ayumu Nakashima
- Department of Stem Cell Biology and Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8553, Japan
| | - Noboru Hattori
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| |
Collapse
|
37
|
Chen X, Wang F, Huang Z, Wu Y, Geng J, Wang Y. Clinical applications of mesenchymal stromal cell-based therapies for pulmonary diseases: An Update and Concise Review. Int J Med Sci 2021; 18:2849-2870. [PMID: 34220313 PMCID: PMC8241779 DOI: 10.7150/ijms.59218] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 05/09/2021] [Indexed: 12/15/2022] Open
Abstract
Lung disorders are a leading cause of morbidity and death worldwide. For many disease conditions, no effective and curative treatment options are available. Mesenchymal stromal cell (MSC)-based therapy is one of the cutting-edge topics in medical research today. It offers a novel and promising therapeutic option for various acute and chronic lung diseases due to its potent and broad-ranging immunomodulatory activities, bacterial clearance, tissue regeneration, and proangiogenic and antifibrotic properties, which rely on both cell-to-cell contact and paracrine mechanisms. This review covers the sources and therapeutic potential of MSCs. In particular, a total of 110 MSC-based clinical applications, either completed clinical trials with safety and early efficacy results reported or ongoing worldwide clinical trials of pulmonary diseases, are systematically summarized following preferred reporting items for systematic reviews and meta-analyses (PRISMA) guidelines, including acute/viral pulmonary disease, community-acquired pneumonia (CAP), chronic obstructive pulmonary disease (COPD), bronchopulmonary dysplasia (BPD), interstitial lung diseases (ILD), chronic pulmonary fibrosis, bronchiolitis obliterans syndrome (BOS) and lung cancer. The results of recent clinical studies suggest that MSCs are a promising therapeutic approach for the treatment of lung diseases. Nevertheless, large-scale clinical trials and evaluation of long-term effects are necessary in further studies.
Collapse
Affiliation(s)
- Xiaobo Chen
- Unicell Life Science Development Co., Ltd, Tianjin, China
| | - Feng Wang
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Zhiwei Huang
- Department of Clinical Laboratory Medicine, the Second Hospital of Tianjin Medical University, Tianjin Institute of Urology, Tianjin, China
| | - Yan Wu
- Department of Clinical Laboratory Medicine, Tianjin TEDA Hospital, Tianjin, China
| | - Jie Geng
- Department of Clinical Laboratory Medicine, the Second Hospital of Tianjin Medical University, Tianjin Institute of Urology, Tianjin, China
| | - Yuliang Wang
- Department of Clinical Laboratory Medicine, the Second Hospital of Tianjin Medical University, Tianjin Institute of Urology, Tianjin, China
| |
Collapse
|