1
|
Severo NC, Inês de Assumpção T, Silva Peixer MA, da Cunha Xavier M, Malard PF, Brunel HDSS, Lançoni R. Effectiveness of intraglandular allogeneic mesenchymal stem cell administration for treating chronic vesicular adenitis in bulls. Theriogenology 2025; 241:117419. [PMID: 40188642 DOI: 10.1016/j.theriogenology.2025.117419] [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: 04/22/2024] [Revised: 03/27/2025] [Accepted: 03/31/2025] [Indexed: 04/08/2025]
Abstract
This study aimed to evaluate the effects of the application of allogeneic mesenchymal stem cells (MSCs) bilaterally and intraglandularly in the vesicular glands of bulls affected by seminal vesiculitis. Twelve bulls that presented chronic vesiculitis with two or more recurrences were selected at Semen Collection and Processing Centres, based on the presence of pus in the semen, leukocytes on the motility and vigour evaluation slide, reactive to the California Mastitis Test - CMT (one cross or more) and the presence of leukocytes on a slide stained by Diff Quik staining with more than 5 polymorphonuclear cells (PMN) per field. Ultrasound examination of the vesicular glands was performed, and the clinical signs were definitive for the diagnosis. The proposed method of the intraglandular injection of MSCs involved application through the ischiorectal fossa with a long needle measuring 30-35 cm and a guide measuring 25-30 cm in length directly into the affected vesicular glands. The MSCs were cultured and frozen in the Bio Cell Cellular Therapy® laboratory (Brasilia, Brazil) and prepared by washing and centrifugation for intraglandular injection on the day of application. In total, 3x106 MSCs were injected into each vesicular gland. Data were evaluated for normality of residuals using the Shapiro-Wilk test. When the normality of the test was significant (P < 0.05), the data were transformed or outliers were removed and reevaluated. The "T-Test" was applied to identify statistical differences between variables before and after treatment. The probability of P ≤ 0.05 was considered a significant difference. Data were presented as the mean ± standard error of the mean (S.E.M.). Improvements were observed in the initial percent motility from 60.09 ± 4.8 to 69.89 ± 4.6 (P < 0.05), as well as in the post-thawing percent motility from 26.26 ± 6.77 to 42.5 ± 5 0.99 (P < 0.05). The number of doses produced increased significantly after treatment with MSCs, from 95.61 ± 23.31 units to 337.84 ± 67.75 units (P < 0.05) per ejaculate. The number of leukocytes observed per field decreased from 5.83 ± 0.48 to zero, demonstrating the recovery of the inflamed vesicular glands. Based on the results presented, it was concluded that the application of 3x106 MSCs in the vesicular glands of bulls with vesiculitis is safe and efficient, as it improved several parameters evaluated in this research, mainly the production of semen doses per ejaculate.
Collapse
Affiliation(s)
- Neimar Correa Severo
- School of Veterinary Medicine and Animal Science, Federal University of Uberlândia, Uberlândia, MG, Brazil
| | | | | | | | | | | | - Renata Lançoni
- School of Veterinary Medicine and Animal Science, Federal University of Uberlândia, Uberlândia, MG, Brazil.
| |
Collapse
|
2
|
Hoseini SM, Montazeri F. Cell origin and microenvironment: The players of differentiation capacity in human mesenchymal stem cells. Tissue Cell 2025; 93:102709. [PMID: 39765135 DOI: 10.1016/j.tice.2024.102709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 12/12/2024] [Accepted: 12/26/2024] [Indexed: 03/05/2025]
Abstract
Mesenchymal stem cells (MSCs) have several important properties that make them desirable for regenerative medicine. These properties include immunomodulatory ability, growth factor production, and differentiation into various cell types. Despite extensive research and promising results in clinical trials, our understanding of MSC biology, their mechanism of action, and their targeted and routine use in clinics is limited. Differentiation of human MSCs (hMSCs) is a complex process influenced by various elements such as growth factors, pharmaceutical compounds, microRNAs, 3D scaffolds, and mechanical and electrical stimulation. Research has shown that different culture conditions can affect the differentiation potential of hMSCs obtained from multiple fetal and adult sources. Additionally, it seems that what affects the differentiation capacities of these cells is their secretory characteristics, which are influenced by the origin of the cells and the local microenvironment where the cells are located. The review can provide insights into the microenvironment-based mechanisms involved in MSC differentiation, which can be valuable for future therapeutic applications.
Collapse
Affiliation(s)
- Seyed Mehdi Hoseini
- Biotechnology Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran; Hematology and Oncology Research Center, Non-communicable Diseases Research Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Fateme Montazeri
- Abortion Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran.
| |
Collapse
|
3
|
Li L, He Y, Zhao J, Yin H, Feng X, Fan X, Wu W, Lu Q. Mesenchymal Stromal Cell-Based Therapy: A Promising Approach for Autoimmune Diseases. Clin Rev Allergy Immunol 2025; 68:21. [PMID: 39982546 DOI: 10.1007/s12016-025-09030-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/04/2025] [Indexed: 02/22/2025]
Abstract
Autoimmune diseases are characterized by immune dysregulation, resulting in aberrant reactivity of T cells and antibodies to self-antigens, leading to various patterns of inflammation and organ dysfunction. However, current therapeutic agents exhibit broad-spectrum activity and lack disease-specific selectivity, leading to enduring adverse effects, notably severe infections, and malignancies, and patients often fail to achieve the intended clinical goals. Mesenchymal stromal cells (MSCs) are multipotent stromal cells that can be easily derived from various tissues, such as adipose tissue, umbilical cords, Wharton's jelly, placenta, and dental tissues. MSCs offer advantages due to their immunomodulatory and anti-inflammatory abilities, low immunogenicity, and a high capacity for proliferation and multipotent differentiation, making them excellent candidates for cell-based treatment in autoimmune disorders. This review will cover preclinical studies and clinical trials involving MSCs in autoimmune diseases, as well as the primary challenges associated with the clinical application of MSC therapies and strategies for maximizing their therapeutic potential.
Collapse
Affiliation(s)
- Liming Li
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
- Key Laboratory of Basic and Translational Research On Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China
| | - Yong He
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Junpeng Zhao
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
- Key Laboratory of Basic and Translational Research On Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China
- Department of Clinical Laboratory, Xiangya Hospital, Central South University, Changsha, China
| | - Huiqi Yin
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
- Key Laboratory of Basic and Translational Research On Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China
| | - Xiwei Feng
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
- Key Laboratory of Basic and Translational Research On Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China
| | - Xinyu Fan
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
- Key Laboratory of Basic and Translational Research On Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China
| | - Wei Wu
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
- Key Laboratory of Basic and Translational Research On Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China
| | - Qianjin Lu
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China.
- Key Laboratory of Basic and Translational Research On Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China.
| |
Collapse
|
4
|
Valencia J, Yáñez RM, Muntión S, Fernández-García M, Martín-Rufino JD, Zapata AG, Bueren JA, Vicente Á, Sánchez-Guijo F. Improving the therapeutic profile of MSCs: Cytokine priming reduces donor-dependent heterogeneity and enhances their immunomodulatory capacity. Front Immunol 2025; 16:1473788. [PMID: 40034706 PMCID: PMC11872697 DOI: 10.3389/fimmu.2025.1473788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 01/28/2025] [Indexed: 03/05/2025] Open
Abstract
Introduction MSCs exhibit regenerative, anti-inflammatory and immunomodulatory properties due to the large amount of cytokines, chemokines and growth factors they secrete. MSCs have been extensively evaluated in clinical trials, however, in some cases their therapeutic effects are variable. Therefore, strategies to improve their therapeutic potential, such as preconditioning with proinflammatory factors, have been proposed. Several priming approaches have provided non-conclusive results, and the duration of priming effects on MSC properties or their response to a second inflammatory stimulus have not been fully addressed. Methods We have investigated the impact of triple cytokine priming in MSCs on their characterization and viability, their transcriptomic profile, the functionality of innate and acquired immune cells, as well as the maintenance of the response to priming over time, their subsequent responsiveness to a second inflammatory stimulus. Results Priming MSCs with proinflammatory cytokines (CK-MSCs) do not modify the differentiation capacity of MSCs, nor their immunophenotype and viability. Moreover, cytokine priming enhances the anti-inflammatory and immunomodulatory properties of MSCs against NK and dendritic cells, while maintaining the same T cell immunomodulatory capacity as unstimulated MSCs. Thus, they decrease T-lymphocytes and NK cell proliferation, inhibit the differentiation and allostimulatory capacity of dendritic cells and promote the differentiation of monocytes with an immunosuppressive profile. In addition, we have shown for the first time that proinflammatory priming reduces the variability between different donors and MSC origins. Finally, the effect on CK-MSC is maintained over time and even after a secondary inflammatory stimulus. Conclusions Cytokine-priming improves the therapeutic potential of MSCs and reduces inter-donor variability.
Collapse
Affiliation(s)
- Jaris Valencia
- Department of Cell Biology, School of Medicine, Complutense University of Madrid, Madrid, Spain
- Heath Research Institute Hospital Clínico San Carlos (IdISSC), Madrid, Spain
- RICORS TERAV, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Rosa M. Yáñez
- RICORS TERAV, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Hematopoietic Innovative Therapies Division, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT) and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
- Heath Research Institute-Fundación Jiménez Díaz (IIS-FJD), Madrid, Spain
| | - Sandra Muntión
- RICORS TERAV, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Department of Medicine, University of Salamanca and Cell Therapy Area and Hematology Department, IBSAL-University Hospital of Salamanca, Salamanca, Spain
- Regenerative Medicine and Cellular Therapy Network Center of Castilla y León, Salamanca, Spain
| | - María Fernández-García
- RICORS TERAV, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Hematopoietic Innovative Therapies Division, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT) and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
- Heath Research Institute-Fundación Jiménez Díaz (IIS-FJD), Madrid, Spain
| | - Jorge Diego Martín-Rufino
- Division of Hematology/Oncology, Boston Children’s Hospital and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, United States
- Broad Institute of MIT and Harvard, Cambridge, MA, United States
| | - Agustín G. Zapata
- RICORS TERAV, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Department of Cell Biology, Faculty of Biology, Complutense University of Madrid, Madrid, Spain
- Heath Research Institute Hospital 12 de Octubre (I+12), Madrid, Spain
| | - Juan A. Bueren
- RICORS TERAV, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Hematopoietic Innovative Therapies Division, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT) and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
- Heath Research Institute-Fundación Jiménez Díaz (IIS-FJD), Madrid, Spain
| | - Ángeles Vicente
- Department of Cell Biology, School of Medicine, Complutense University of Madrid, Madrid, Spain
- RICORS TERAV, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Heath Research Institute Hospital 12 de Octubre (I+12), Madrid, Spain
| | - Fermín Sánchez-Guijo
- RICORS TERAV, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Department of Medicine, University of Salamanca and Cell Therapy Area and Hematology Department, IBSAL-University Hospital of Salamanca, Salamanca, Spain
- Regenerative Medicine and Cellular Therapy Network Center of Castilla y León, Salamanca, Spain
| |
Collapse
|
5
|
Esfehani R, Khadivi F, Valipour J, Shabani M, Ramesh M, Javanbakht P, Zarini D, Mojaverrostami S, Hoseini M. Secretome of human amniotic membrane stem cells promote recovery and testicular functions through modulating SIRT1/NRF2/TNF-α pathway in mice testicular torsion: An experimental study. Int J Reprod Biomed 2024; 22:821-836. [PMID: 39906090 PMCID: PMC11788658 DOI: 10.18502/ijrm.v22i10.17670] [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: 12/21/2023] [Revised: 05/05/2024] [Accepted: 08/17/2024] [Indexed: 02/06/2025] Open
Abstract
Background Testicular ischemia/reperfusion injury, a significant result of testicular torsion, can lead to the risk of male infertility. Objective The current study aimed to evaluate the effect of human amniotic membrane-derived mesenchymal stem cells (hAMSCs) secretome on testicular torsion/detorsion (T/D) in mice. Materials and Methods All the experiments were performed in the Anatomy Department of Tehran University of Medical Sciences, Tehran, Iran, during the period of March 2023 to December 2023. 40 male NMRI mice (5-7 wk, 25-30 gr) were randomized into: 1) the sham group: mice received sham operations with no other interventions, 2) T/D group, 3) negative control group; torsion detorsion + intratesticular injection of Dulbecco's Modified Eagle Medium/Nutrient Mixture F-12, and 4) the T/D group + hAMSCs secreted factors. Serum testosterone levels, hematoxylin and eosin staining, and sperm quality parameters were used to evaluate the therapeutic effects of hAMSCs secreted factors on the testicular structure and function. Tissue oxidative stress was measured by determining malondialdehyde, superoxide dismutase, catalase, and glutathione peroxidase-1. Nuclear factor erythroid 2-related factor 2, Kelch-like ECH-associated protein 1, NAD-dependent deacetylase sirtuin-1, tumor necrosis factor-alpha and tumor protein P53 mRNA expressions were assessed in testis via real-time polymerase chain reaction. Results The results showed that hAMSCs secreted factors alleviated testicular T/D injury by attenuating oxidative stress, inflammatory response, and apoptosis via modulating the sirtuin-1/ nuclear factor erythroid 2-related factor 2/tumor necrosis factor-alpha signaling pathway. Conclusion hAMSCs secreted factors increased antioxidative, anti-inflammatory, and antiapoptotic properties which consequently increased testosterone levels, spermatogenesis, and sperm quality parameters.
Collapse
Affiliation(s)
- Roghayeh Esfehani
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Farnaz Khadivi
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
- Department of Anatomy, School of Medicine, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Jamal Valipour
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Shabani
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahya Ramesh
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Parinaz Javanbakht
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Davood Zarini
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Sina Mojaverrostami
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Masih Hoseini
- Department of Anatomy, School of Medicine, Shahrekord University of Medical Sciences, Shahrekord, Iran
| |
Collapse
|
6
|
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
|
7
|
Kim CD, Koo KM, Kim HJ, Kim TH. Recent Advances in Nanomaterials for Modulation of Stem Cell Differentiation and Its Therapeutic Applications. BIOSENSORS 2024; 14:407. [PMID: 39194636 DOI: 10.3390/bios14080407] [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: 07/19/2024] [Revised: 08/14/2024] [Accepted: 08/20/2024] [Indexed: 08/29/2024]
Abstract
Challenges in directed differentiation and survival limit the clinical use of stem cells despite their promising therapeutic potential in regenerative medicine. Nanotechnology has emerged as a powerful tool to address these challenges and enable precise control over stem cell fate. In particular, nanomaterials can mimic an extracellular matrix and provide specific cues to guide stem cell differentiation and proliferation in the field of nanotechnology. For instance, recent studies have demonstrated that nanostructured surfaces and scaffolds can enhance stem cell lineage commitment modulated by intracellular regulation and external stimulation, such as reactive oxygen species (ROS) scavenging, autophagy, or electrical stimulation. Furthermore, nanoframework-based and upconversion nanoparticles can be used to deliver bioactive molecules, growth factors, and genetic materials to facilitate stem cell differentiation and tissue regeneration. The increasing use of nanostructures in stem cell research has led to the development of new therapeutic approaches. Therefore, this review provides an overview of recent advances in nanomaterials for modulating stem cell differentiation, including metal-, carbon-, and peptide-based strategies. In addition, we highlight the potential of these nano-enabled technologies for clinical applications of stem cell therapy by focusing on improving the differentiation efficiency and therapeutics. We believe that this review will inspire researchers to intensify their efforts and deepen their understanding, thereby accelerating the development of stem cell differentiation modulation, therapeutic applications in the pharmaceutical industry, and stem cell therapeutics.
Collapse
Affiliation(s)
- Chang-Dae Kim
- School of Integrative Engineering, Chung-Ang University, 84 Heukseuk-ro, Dongjak-gu, Seoul 06974, Republic of Korea
| | - Kyeong-Mo Koo
- School of Integrative Engineering, Chung-Ang University, 84 Heukseuk-ro, Dongjak-gu, Seoul 06974, Republic of Korea
| | - Hyung-Joo Kim
- School of Integrative Engineering, Chung-Ang University, 84 Heukseuk-ro, Dongjak-gu, Seoul 06974, Republic of Korea
| | - Tae-Hyung Kim
- School of Integrative Engineering, Chung-Ang University, 84 Heukseuk-ro, Dongjak-gu, Seoul 06974, Republic of Korea
| |
Collapse
|
8
|
Chasov V, Ganeeva I, Zmievskaya E, Davletshin D, Gilyazova E, Valiullina A, Bulatov E. Cell-Based Therapy and Genome Editing as Emerging Therapeutic Approaches to Treat Rheumatoid Arthritis. Cells 2024; 13:1282. [PMID: 39120313 PMCID: PMC11312096 DOI: 10.3390/cells13151282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 07/19/2024] [Accepted: 07/26/2024] [Indexed: 08/10/2024] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease characterized by chronic inflammation of the joints. Although much remains unknown about the pathogenesis of RA, there is evidence that impaired immune tolerance and the development of RA are related. And it is precisely the restoration of immune tolerance at the site of the inflammation that is the ultimate goal of the treatment of RA. Over the past few decades, significant progress has been made in the treatment of RA, with higher rates of disease remission and improved long-term outcomes. Unfortunately, despite these successes, the proportion of patients with persistent, difficult-to-treat disease remains high, and the task of improving our understanding of the basic mechanisms of disease development and developing new ways to treat RA remains relevant. This review focuses on describing new treatments for RA, including cell therapies and gene editing technologies that have shown potential in preclinical and early clinical trials. In addition, we discuss the opportunities and limitations associated with the use of these new approaches in the treatment of RA.
Collapse
Affiliation(s)
- Vitaly Chasov
- Laboratory of Biomedical Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, 18 Kremlyovskaya Street, Kazan 420008, Russia
| | - Irina Ganeeva
- Laboratory of Biomedical Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, 18 Kremlyovskaya Street, Kazan 420008, Russia
| | - Ekaterina Zmievskaya
- Laboratory of Biomedical Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, 18 Kremlyovskaya Street, Kazan 420008, Russia
| | - Damir Davletshin
- Laboratory of Biomedical Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, 18 Kremlyovskaya Street, Kazan 420008, Russia
| | - Elvina Gilyazova
- Laboratory of Biomedical Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, 18 Kremlyovskaya Street, Kazan 420008, Russia
| | - Aygul Valiullina
- Laboratory of Biomedical Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, 18 Kremlyovskaya Street, Kazan 420008, Russia
| | - Emil Bulatov
- Laboratory of Biomedical Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, 18 Kremlyovskaya Street, Kazan 420008, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia
- Federal State Autonomous Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow 119048, Russia
| |
Collapse
|
9
|
Liu L, Liu D. Bioengineered mesenchymal stem cell-derived exosomes: emerging strategies for diabetic wound healing. BURNS & TRAUMA 2024; 12:tkae030. [PMID: 39015252 PMCID: PMC11250359 DOI: 10.1093/burnst/tkae030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 05/10/2024] [Indexed: 07/18/2024]
Abstract
Diabetic wounds are among the most common complications of diabetes mellitus and their healing process can be delayed due to persistent inflammatory reactions, bacterial infections, damaged vascularization and impaired cell proliferation, which casts a blight on patients'health and quality of life. Therefore, new strategies to accelerate diabetic wound healing are being positively explored. Exosomes derived from mesenchymal stem cells (MSC-Exos) can inherit the therapeutic and reparative abilities of stem cells and play a crucial role in diabetic wound healing. However, poor targeting, low concentrations of therapeutic molecules, easy removal from wounds and limited yield of MSC-Exos are challenging for clinical applications. Bioengineering techniques have recently gained attention for their ability to enhance the efficacy and yield of MSC-Exos. In this review, we summarise the role of MSC-Exos in diabetic wound healing and focus on three bioengineering strategies, namely, parental MSC-Exos engineering, direct MSC-Exos engineering and MSC-Exos combined with biomaterials. Furthermore, the application of bioengineered MSC-Exos in diabetic wound healing is reviewed. Finally, we discuss the future prospects of bioengineered MSC-Exos, providing new insights into the exploration of therapeutic strategies.
Collapse
Affiliation(s)
- Lihua Liu
- Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Yongwaizheng Road, Donghu District, Nanchang, Jiangxi, P.R. China
- Huankui Academy, Nanchang University, Xuefu Road, Honggutan District, Nanchang, Jiangxi, 330006, P.R. China
| | - Dewu Liu
- Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Yongwaizheng Road, Donghu District, Nanchang, Jiangxi, P.R. China
| |
Collapse
|
10
|
Ghoneim MA, Gabr MM, El-Halawani SM, Refaie AF. Current status of stem cell therapy for type 1 diabetes: a critique and a prospective consideration. Stem Cell Res Ther 2024; 15:23. [PMID: 38281991 PMCID: PMC10823744 DOI: 10.1186/s13287-024-03636-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 01/10/2024] [Indexed: 01/30/2024] Open
Abstract
Over the past decade, there had been progress in the development of cell therapy for insulin-dependent diabetes. Nevertheless, important hurdles that need to be overcome still remain. Protocols for the differentiation of pluripotent stem cells into pancreatic progenitors or fully differentiated β-cells have been developed. The resulting insulin-producing cells can control chemically induced diabetes in rodents and were the subject of several clinical trials. However, these cells are immunogenic and possibly teratogenic for their transplantation, and an immunoisolation device and/or immunosuppression is needed. A growing number of studies have utilized genetic manipulations to produce immune evasive cells. Evidence must be provided that in addition to the expected benefit, gene manipulations should not lead to any unforeseen complications. Mesenchymal stem/stromal cells (MSCs) can provide a viable alternative. MSCs are widely available from many tissues. They can form insulin-producing cells by directed differentiation. Experimentally, evidence has shown that the transplantation of allogenic insulin-producing cells derived from MSCs is associated with a muted allogeneic response that does not interfere with their functionality. This can be explained by the immunomodulatory functions of the MSC subpopulation that did not differentiate into insulin-producing cells. Recently, exosomes derived from naive MSCs have been used in the experimental domain to treat diabetes in rodents with varying degrees of success. Several mechanisms for their beneficial functions were proposed including a reduction in insulin resistance, the promotion of autophagy, and an increase in the T regulatory population. However, euglycemia was not achieved in any of these experiments. We suggest that exosomes derived from β-cells or insulin-producing cells (educated) can provide a better therapeutic effect than those derived from undifferentiated cells.
Collapse
|
11
|
Zhao M, Liu S, Wang Y, Lv K, Lou P, Zhou P, Zhu J, Li L, Cheng J, Lu Y, Liu J. The mitochondria‒paraspeckle axis regulates the survival of transplanted stem cells under oxidative stress conditions. Theranostics 2024; 14:1517-1533. [PMID: 38389853 PMCID: PMC10879866 DOI: 10.7150/thno.88764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 01/24/2024] [Indexed: 02/24/2024] Open
Abstract
Rationale: Stem cell-based therapies have emerged as promising tools for tissue engineering and regenerative medicine, but their therapeutic efficacy is largely limited by the oxidative stress-induced loss of transplanted cells at injured tissue sites. To address this issue, we aimed to explore the underlying mechanism and protective strategy of ROS-induced MSC loss. Methods: Changes in TFAM (mitochondrial transcription factor A) signaling, mitochondrial function, DNA damage, apoptosis and senescence in MSCs under oxidative stress conditions were assessed using real-time PCR, western blotting and RNA sequencing, etc. The impact of TFAM or lncRNA nuclear paraspeckle assembly transcript 1 (NEAT1) knockdown or overexpression on mitochondrial function, DNA damage repair, apoptosis and senescence in MSCs was also analyzed. The effect of mitochondrion-targeted antioxidant (Mito-TEMPO) on the survival of transplanted MSCs was evaluated in a mouse model of renal ischemia/reperfusion (I/R) injury. Results: Mitochondrial ROS (mtROS) bursts caused defects in TFAM signaling and overall mitochondrial function, which further impaired NEAT1 expression and its mediated paraspeckle formation and DNA repair pathways in MSCs, thereby jointly promoting MSC senescence and death under oxidative stress. In contrast, targeted inhibition of the mtROS bursts is a sufficient strategy for attenuating early transplanted MSC loss at injured tissue sites, and coadministration of Mito-TEMPO improved the local retention of transplanted MSCs and reduced oxidative injury in ischemic kidneys. Conclusions: This study identified the critical role of the mitochondria‒paraspeckle axis in regulating cell survival and may provide insights into developing advanced stem cell therapies for tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Meng Zhao
- Department of General Surgery and NHC Key Laboratory of Transplant Engineering and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
- Department of Emergency, Guizhou Provincial People's Hospital, Guiyang 550002, China
| | - Shuyun Liu
- Department of General Surgery and NHC Key Laboratory of Transplant Engineering and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yizhuo Wang
- Department of General Surgery and NHC Key Laboratory of Transplant Engineering and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ke Lv
- Department of General Surgery and NHC Key Laboratory of Transplant Engineering and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Peng Lou
- Department of General Surgery and NHC Key Laboratory of Transplant Engineering and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Pingya Zhou
- Department of General Surgery and NHC Key Laboratory of Transplant Engineering and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jiaying Zhu
- Department of Emergency, Guizhou Provincial People's Hospital, Guiyang 550002, China
| | - Lan Li
- Department of General Surgery and NHC Key Laboratory of Transplant Engineering and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jingqiu Cheng
- Department of General Surgery and NHC Key Laboratory of Transplant Engineering and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yanrong Lu
- Department of General Surgery and NHC Key Laboratory of Transplant Engineering and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jingping Liu
- Department of General Surgery and NHC Key Laboratory of Transplant Engineering and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
12
|
Zendedel E, Tayebi L, Nikbakht M, Hasanzadeh E, Asadpour S. Clinical Trials of Mesenchymal Stem Cells for the Treatment of COVID 19. Curr Stem Cell Res Ther 2024; 19:1055-1071. [PMID: 37815188 DOI: 10.2174/011574888x260032230925052240] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 07/14/2023] [Accepted: 07/31/2023] [Indexed: 10/11/2023]
Abstract
Mesenchymal Stem Cells (MSCs) are being investigated as a treatment for a novel viral disease owing to their immunomodulatory, anti-inflammatory, tissue repair and regeneration characteristics, however, the exact processes are unknown. MSC therapy was found to be effective in lowering immune system overactivation and increasing endogenous healing after SARS-CoV-2 infection by improving the pulmonary microenvironment. Many studies on mesenchymal stem cells have been undertaken concurrently, and we may help speed up the effectiveness of these studies by collecting and statistically analyzing data from them. Based on clinical trial information found on clinicaltrials. gov and on 16 November 2020, which includes 63 clinical trials in the field of patient treatment with COVID-19 using MSCs, according to the trend of increasing studies in this field, and with the help of meta-analysis studies, it is possible to hope that the promise of MSCs will one day be realized. The potential therapeutic applications of MSCs for COVID-19 are investigated in this study.
Collapse
Affiliation(s)
- Elham Zendedel
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Lobat Tayebi
- Marquett University School of Dentistry, Milwaukee, WI, 53233, USA
| | - Mohammad Nikbakht
- Department of Medical Biotechnology, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Elham Hasanzadeh
- Immunogenetics Research Center, Department of Tissue Engineering & Regenerative Medicine, School of Advanced Technologies in Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Shiva Asadpour
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahrekord University of Medical Sciences, Shahrekord, Iran
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| |
Collapse
|
13
|
Kaçaroğlu D, Yaylacı S. Enhancing the Regenerative Potential of Adipose-Derived Mesenchymal Stem Cells Through TLR4-Mediated Signaling. Curr Stem Cell Res Ther 2024; 19:1514-1524. [PMID: 38204244 DOI: 10.2174/011574888x283664231219080535] [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: 09/22/2023] [Revised: 11/16/2023] [Accepted: 11/22/2023] [Indexed: 01/12/2024]
Abstract
INTRODUCTION Toll-like receptor 4 (TLR4) is a receptor that traditionally plays an important role in immunomodulation (regulation of the immune system) and the initiation of proinflammatory responses. TLR4 is used in the body to recognize molecular patterns of pathogens or damaged cells from outside. However, in recent years, it has also become clear that TLR4 can affect the immune system and the function of stem cells, especially mesenchymal stem cells. Therefore, understanding how TLR4 signaling works at the cellular and molecular level and using this knowledge in regenerative medicine could be potentially useful, especially in the treatment of adipose- derived mesenchymal stem cells (ADMSCs). How these cells can use TLR4 signaling when used to increase their regenerative potential and repair tissues is an area of research. AIMS This study aims to elucidate the multifaceted role of TLR4-mediated signaling in ADMSCs. METHODS Employing a comprehensive set of assays, including MTT for cell viability, flow cytometry for surface marker expression, and gene expression analysis, we demonstrate that TLR4 activation significantly modulates key aspects of ADMSC biology. Specifically, TLR4 signaling was found to regulate ADMSCs proliferation, surface marker expression, and regenerative capacity in a dose- and time-dependent manner. Furthermore, TLR4 activation conferred cytoprotective effects against Doxorubicin (DOX)-induced cellular apoptosis. RESULTS These findings suggest that TLR4 signaling could be used to enhance the regenerative abilities of ADMSCs and enable ADMSC-based therapies to be used more effectively for tissue engineering and therapeutic purposes. CONCLUSION However, it is important to note that research in this area needs more details and clinical studies.
Collapse
Affiliation(s)
- Demet Kaçaroğlu
- Department of Medical Biology, Faculty of Medicine, Lokman Hekim University, Ankara, Turkey
| | - Seher Yaylacı
- Department of Medical Biology, Faculty of Medicine, Lokman Hekim University, Ankara, Turkey
| |
Collapse
|
14
|
Han AR, Shin HR, Kweon J, Lee SB, Lee SE, Kim EY, Kweon J, Chang EJ, Kim Y, Kim SW. Highly efficient genome editing via CRISPR-Cas9 ribonucleoprotein (RNP) delivery in mesenchymal stem cells. BMB Rep 2024; 57:60-65. [PMID: 38053293 PMCID: PMC10828435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/18/2023] [Accepted: 08/31/2023] [Indexed: 12/07/2023] Open
Abstract
The CRISPR-Cas9 system has significantly advanced regenerative medicine research by enabling genome editing in stem cells. Due to their desirable properties, mesenchymal stem cells (MSCs) have recently emerged as highly promising therapeutic agents, which properties include differentiation ability and cytokine production. While CRISPR-Cas9 technology is applied to develop MSC-based therapeutics, MSCs exhibit inefficient genome editing, and susceptibility to plasmid DNA. In this study, we compared and optimized plasmid DNA and RNP approaches for efficient genome engineering in MSCs. The RNP-mediated approach enabled genome editing with high indel frequency and low cytotoxicity in MSCs. By utilizing Cas9 RNPs, we successfully generated B2M-knockout MSCs, which reduced T-cell differentiation, and improved MSC survival. Furthermore, this approach enhanced the immunomodulatory effect of IFN-r priming. These findings indicate that the RNP-mediated engineering of MSC genomes can achieve high efficiency, and engineered MSCs offer potential as a promising therapeutic strategy. [BMB Reports 2024; 57(1): 60-65].
Collapse
Affiliation(s)
- A Reum Han
- Department of Translational Medicine, Seoul 05505, Korea
- Department of Biochemistry and Molecular Biology, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
- Stem Cell Immunomodulation Research Center, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Ha Rim Shin
- Department of Cell and Genetic Engineering, Seoul 05505, Korea
- Center for Genomic Integrity, Institute for Basic Science, Ulsan 44919, Korea
- Stem Cell Immunomodulation Research Center, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Jiyeon Kweon
- Department of Cell and Genetic Engineering, Seoul 05505, Korea
| | - Soo Been Lee
- Department of Biochemistry and Molecular Biology, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
- Stem Cell Immunomodulation Research Center, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Sang Eun Lee
- Department of Biochemistry and Molecular Biology, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
- Stem Cell Immunomodulation Research Center, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Eun-Young Kim
- Department of Biochemistry and Molecular Biology, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
- Stem Cell Immunomodulation Research Center, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Jiyeon Kweon
- Department of Cell and Genetic Engineering, Seoul 05505, Korea
| | - Eun-Ju Chang
- Department of Biochemistry and Molecular Biology, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
- Stem Cell Immunomodulation Research Center, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Yongsub Kim
- Department of Cell and Genetic Engineering, Seoul 05505, Korea
- Stem Cell Immunomodulation Research Center, University of Ulsan College of Medicine, Seoul 05505, Korea
- Center for Cell therapy, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, Seoul 05505, Korea
| | - Seong Who Kim
- Department of Biochemistry and Molecular Biology, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
- Stem Cell Immunomodulation Research Center, University of Ulsan College of Medicine, Seoul 05505, Korea
- Center for Cell therapy, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, Seoul 05505, Korea
| |
Collapse
|
15
|
Pham DV, Nguyen TK, Nguyen BL, Kim JO, Jeong JH, Choi I, Park PH. Adiponectin restores the obesity-induced impaired immunomodulatory function of mesenchymal stromal cells via glycolytic reprogramming. Acta Pharm Sin B 2024; 14:273-291. [PMID: 38261813 PMCID: PMC10793097 DOI: 10.1016/j.apsb.2023.10.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 08/07/2023] [Accepted: 10/18/2023] [Indexed: 01/25/2024] Open
Abstract
Obesity has been known to negatively modulate the life-span and immunosuppressive potential of mesenchymal stromal cells (MSC). However, it remains unclear what drives the compromised potency of obese MSC. In this study, we examined the involvement of adiponectin, an adipose tissue-derived hormone, in obesity-induced impaired therapeutic function of MSC. Diet-induced obesity leads to a decrease in serum adiponectin, accompanied by impairment of survival and immunomodulatory effects of adipose-derived MSC (ADSC). Interestingly, priming with globular adiponectin (gAcrp) improved the immunomodulatory potential of obese ADSC. Similar effects were also observed in lean ADSC. In addition, gAcrp potentiated the therapeutic effectiveness of ADSC in a mouse model of DSS-induced colitis. Mechanistically, while obesity inhibited the glycolytic capacity of MSC, gAcrp treatment induced a metabolic shift toward glycolysis through activation of adiponectin receptor type 1/p38 MAPK/hypoxia inducible factor-1α axis. These findings suggest that activation of adiponectin signaling is a promising strategy for enhancing the therapeutic efficacy of MSC against immune-mediated disorders.
Collapse
Affiliation(s)
- Duc-Vinh Pham
- Department of Pharmacology, Hanoi University of Pharmacy, Hanoi 100000, Viet Nam
- College of Pharmacy, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Thi-Kem Nguyen
- College of Pharmacy, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Bao-Loc Nguyen
- College of Pharmacy, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Jong-Oh Kim
- College of Pharmacy, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Jee-Heon Jeong
- Department of Precision Medicine, School of Medicine, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Inho Choi
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, Republic of Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Pil-Hoon Park
- College of Pharmacy, Yeungnam University, Gyeongsan 38541, Republic of Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, Republic of Korea
| |
Collapse
|
16
|
Han AR, Shin HR, Kweon J, Lee SB, Lee SE, Kim EY, Kweon J, Chang EJ, Kim Y, Kim SW. Highly efficient genome editing via CRISPR-Cas9 ribonucleoprotein (RNP) delivery in mesenchymal stem cells. BMB Rep 2024; 57:60-65. [PMID: 38053293 PMCID: PMC10828435 DOI: 10.5483/bmbrep.2023-0113] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/18/2023] [Accepted: 08/31/2023] [Indexed: 01/30/2025] Open
Abstract
The CRISPR-Cas9 system has significantly advanced regenerative medicine research by enabling genome editing in stem cells. Due to their desirable properties, mesenchymal stem cells (MSCs) have recently emerged as highly promising therapeutic agents, which properties include differentiation ability and cytokine production. While CRISPR-Cas9 technology is applied to develop MSC-based therapeutics, MSCs exhibit inefficient genome editing, and susceptibility to plasmid DNA. In this study, we compared and optimized plasmid DNA and RNP approaches for efficient genome engineering in MSCs. The RNP-mediated approach enabled genome editing with high indel frequency and low cytotoxicity in MSCs. By utilizing Cas9 RNPs, we successfully generated B2M-knockout MSCs, which reduced T-cell differentiation, and improved MSC survival. Furthermore, this approach enhanced the immunomodulatory effect of IFN-r priming. These findings indicate that the RNP-mediated engineering of MSC genomes can achieve high efficiency, and engineered MSCs offer potential as a promising therapeutic strategy. [BMB Reports 2024; 57(1): 60-65].
Collapse
Affiliation(s)
- A Reum Han
- Department of Translational Medicine, Seoul 05505, Korea
- Department of Biochemistry and Molecular Biology, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
- Stem Cell Immunomodulation Research Center, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Ha Rim Shin
- Department of Cell and Genetic Engineering, Seoul 05505, Korea
- Center for Genomic Integrity, Institute for Basic Science, Ulsan 44919, Korea
- Stem Cell Immunomodulation Research Center, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Jiyeon Kweon
- Department of Cell and Genetic Engineering, Seoul 05505, Korea
| | - Soo Been Lee
- Department of Biochemistry and Molecular Biology, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
- Stem Cell Immunomodulation Research Center, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Sang Eun Lee
- Department of Biochemistry and Molecular Biology, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
- Stem Cell Immunomodulation Research Center, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Eun-Young Kim
- Department of Biochemistry and Molecular Biology, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
- Stem Cell Immunomodulation Research Center, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Jiyeon Kweon
- Department of Cell and Genetic Engineering, Seoul 05505, Korea
| | - Eun-Ju Chang
- Department of Biochemistry and Molecular Biology, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
- Stem Cell Immunomodulation Research Center, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Yongsub Kim
- Department of Cell and Genetic Engineering, Seoul 05505, Korea
- Stem Cell Immunomodulation Research Center, University of Ulsan College of Medicine, Seoul 05505, Korea
- Center for Cell therapy, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, Seoul 05505, Korea
| | - Seong Who Kim
- Department of Biochemistry and Molecular Biology, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
- Stem Cell Immunomodulation Research Center, University of Ulsan College of Medicine, Seoul 05505, Korea
- Center for Cell therapy, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, Seoul 05505, Korea
| |
Collapse
|
17
|
Didamoony MA, Soubh AA, Atwa AM, Ahmed LA. Innovative preconditioning strategies for improving the therapeutic efficacy of extracellular vesicles derived from mesenchymal stem cells in gastrointestinal diseases. Inflammopharmacology 2023; 31:2973-2993. [PMID: 37874430 PMCID: PMC10692273 DOI: 10.1007/s10787-023-01350-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 09/20/2023] [Indexed: 10/25/2023]
Abstract
Gastrointestinal (GI) diseases have become a global health issue and an economic burden due to their wide distribution, late prognosis, and the inefficacy of recent available medications. Therefore, it is crucial to search for new strategies for their management. In the recent decades, mesenchymal stem cells (MSCs) therapy has attracted attention as a viable option for treating a myriad of GI disorders such as hepatic fibrosis (HF), ulcerative colitis (UC), acute liver injury (ALI), and non-alcoholic fatty liver disease (NAFLD) due to their regenerative and paracrine properties. Importantly, recent studies have shown that MSC-derived extracellular vesicles (MSC-EVs) are responsible for most of the therapeutic effects of MSCs. In addition, EVs have revealed several benefits over their parent MSCs, such as being less immunogenic, having a lower risk of tumour formation, being able to cross biological barriers, and being easier to store. MSC-EVs exhibited regenerative, anti-oxidant, anti-inflammatory, anti-apoptotic, and anti-fibrotic effects in different experimental models of GI diseases. However, a key issue with their clinical application is the maintenance of their stability and efficacy following in vivo transplantation. Preconditioning of MSC-EVs or their parent cells is one of the novel methods used to improve their effectiveness and stability. Herein, we discuss the application of MSC-EVs in several GI disorders taking into account their mechanism of action. We also summarise the challenges and restrictions that need to be overcome to promote their clinical application in the treatment of various GI diseases as well as the recent developments to improve their effectiveness. A representation of the innovative preconditioning techniques that have been suggested for improving the therapeutic efficacy of MSC-EVs in GI diseases. The pathological conditions in various GI disorders (ALI, UC, HF and NAFLD) create a harsh environment for EVs and their parents, increasing the risk of apoptosis and senescence of MSCs and thereby diminishing MSC-EVs yield and restricting their large-scale applications. Preconditioning with pharmacological agents or biological mediators can improve the therapeutic efficacy of MSC-EVs through their adaption to the lethal environment to which they are subjected. This can result in establishment of a more conducive environment and activation of numerous vital trajectories that act to improve the immunomodulatory, reparative and regenerative activities of the derived EVs, as a part of MSCs paracrine system. ALI, acute liver injury; GI diseases, gastrointestinal diseases; HF, hepatic fibrosis; HSP, heat shock protein; miRNA, microRNA; mRNA, messenger RNA; MSC-EVs, mesenchymal stem cell-derived extracellular vesicles; NAFLD, non-alcoholic fatty liver disease; UC, ulcerative colitis.
Collapse
Affiliation(s)
- Manar A Didamoony
- Faculty of Pharmacy, Pharmacology and Toxicology Department, Egyptian Russian University, Cairo, 11829, Egypt.
| | - Ayman A Soubh
- Faculty of Pharmacy, Pharmacology and Toxicology Department, Ahram Canadian University, 4th Industrial Zone, Banks Complex, 6th of October City, Giza, 12451, Egypt
| | - Ahmed M Atwa
- Faculty of Pharmacy, Pharmacology and Toxicology Department, Egyptian Russian University, Cairo, 11829, Egypt
| | - Lamiaa A Ahmed
- Faculty of Pharmacy, Pharmacology and Toxicology Department, Cairo University, Cairo, 11562, Egypt.
| |
Collapse
|
18
|
Gonzalez-Uribe V, Romero-Tapia SJ, Castro-Rodriguez JA. Asthma Phenotypes in the Era of Personalized Medicine. J Clin Med 2023; 12:6207. [PMID: 37834850 PMCID: PMC10573947 DOI: 10.3390/jcm12196207] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 09/19/2023] [Accepted: 09/22/2023] [Indexed: 10/15/2023] Open
Abstract
Asthma is a widespread disease affecting approximately 300-million people globally. This condition leads to significant morbidity, mortality, and economic strain worldwide. Recent clinical and laboratory research advancements have illuminated the immunological factors contributing to asthma. As of now, asthma is understood to be a heterogeneous disease. Personalized medicine involves categorizing asthma by its endotypes, linking observable characteristics to specific immunological mechanisms. Identifying these endotypic mechanisms is paramount in accurately profiling patients and tailoring therapeutic approaches using innovative biological agents targeting distinct immune pathways. This article presents a synopsis of the key immunological mechanisms implicated in the pathogenesis and manifestation of the disease's phenotypic traits and individualized treatments for severe asthma subtypes.
Collapse
Affiliation(s)
- Victor Gonzalez-Uribe
- Alergia e Inmunología Clínica, Hospital Infantil de México Federico Gómez, Ciudad de Mexico 06720, Mexico;
- Facultad Mexicana de Medicina, Universidad La Salle México, Ciudad de Mexico 14000, Mexico
| | - Sergio J. Romero-Tapia
- Health Sciences Academic Division (DACS), Universidad Juárez Autónoma de Tabasco, Villahermosa 86040, Mexico;
| | - Jose A. Castro-Rodriguez
- Department of Pediatric Pulmonology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 7820436, Chile
| |
Collapse
|
19
|
Zhang C, Li Z, Wang J, Yao Y, Li H, Fu X. Editorial: Role of stem cell derivatives in inflammatory diseases. Front Immunol 2023; 14:1275068. [PMID: 37720207 PMCID: PMC10501792 DOI: 10.3389/fimmu.2023.1275068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 08/22/2023] [Indexed: 09/19/2023] Open
Affiliation(s)
- Cuiping Zhang
- Research Center for Tissue Repair and Regeneration affiliated to the Medical Innovation Research Department of PLA General Hospital, Beijing, China
- Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, Beijing, China
- Innovation Center for Wound Repair, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhe Li
- Burns Unit, Concord Hospital, University of Sydney Medical School, Sydney, Australia
| | - Jianghuai Wang
- National University of Ireland (NUI), Cork University Hospital, Cork, Ireland
| | - Yongming Yao
- Translational Medicine Research Center, The Center of Translational Medicine affiliated to the Medical Innovation Research Department of PLA General Hospital, Beijing, China
| | - Haihong Li
- Department of Burns and Plastic Surgery, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Xiaobing Fu
- Research Center for Tissue Repair and Regeneration affiliated to the Medical Innovation Research Department of PLA General Hospital, Beijing, China
- Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, Beijing, China
- Innovation Center for Wound Repair, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
20
|
Mendiratta M, Mendiratta M, Mohanty S, Sahoo RK, Prakash H. Breaking the graft-versus-host-disease barrier: Mesenchymal stromal/stem cells as precision healers. Int Rev Immunol 2023; 43:95-112. [PMID: 37639700 DOI: 10.1080/08830185.2023.2252007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 08/08/2023] [Accepted: 08/21/2023] [Indexed: 08/31/2023]
Abstract
Mesenchymal Stromal/Stem Cells (MSCs) are multipotent, non-hematopoietic progenitor cells with a wide range of immune modulation and regenerative potential which qualify them as a potential component of cell-based therapy for various autoimmune/chronic inflammatory ailments. Their immunomodulatory properties include the secretion of immunosuppressive cytokines, the ability to suppress T-cell activation and differentiation, and the induction of regulatory T-cells. Considering this and our interest, we here discuss the significance of MSC for the management of Graft-versus-Host-Disease (GvHD), one of the autoimmune manifestations in human. In pre-clinical models, MSCs have been shown to reduce the severity of GvHD symptoms, including skin and gut damage, which are the most common and debilitating manifestations of this disease. While initial clinical studies of MSCs in GvHD cases were promising, the results were variable in randomized studies. So, further studies are warranted to fully understand their potential benefits, safety profile, and optimal dosing regimens. Owing to these inevitable issues, here we discuss various mechanisms, and how MSCs can be employed in managing GvHD, as a cellular therapeutic approach for this disease.
Collapse
Affiliation(s)
- Mohini Mendiratta
- Department of Medical Oncology, Dr. B. R. Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India
| | | | - Sujata Mohanty
- Stem Cell Facility, All India Institute of Medical Sciences, New Delhi, India
| | - Ranjit Kumar Sahoo
- Department of Medical Oncology, Dr. B. R. Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India
| | - Hridayesh Prakash
- Amity Centre for Translational Research, Amity University, Noida, India
| |
Collapse
|
21
|
Rosado-Galindo H, Domenech M. Substrate topographies modulate the secretory activity of human bone marrow mesenchymal stem cells. Stem Cell Res Ther 2023; 14:208. [PMID: 37605275 PMCID: PMC10441765 DOI: 10.1186/s13287-023-03450-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 08/11/2023] [Indexed: 08/23/2023] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) secrete a diversity of factors with broad therapeutic potential, yet current culture methods limit potency outcomes. In this study, we used topographical cues on polystyrene films to investigate their impact on the secretory profile and potency of bone marrow-derived MSCs (hBM-MSCs). hBM-MSCs from four donors were cultured on topographic substrates depicting defined roughness, curvature, grooves and various levels of wettability. METHODS The topographical PS-based array was developed using razor printing, polishing and plasma treatment methods. hBM-MSCs from four donors were purchased from RoosterBio and used in co-culture with peripheral blood mononuclear cells (PBMCs) from Cell Applications Inc. in an immunopotency assay to measure immunosuppressive capacity. Cells were cultured on low serum (2%) for 24-48 h prior to analysis. Image-based analysis was used for cell quantification and morphology assessment. Metabolic activity of BM-hMSCs was measured as the mitochondrial oxygen consumption rate using an extracellular flux analyzer. Conditioned media samples of BM-hMSCs were used to quantify secreted factors, and the data were analyzed using R statistics. Enriched bioprocesses were identify using the Gene Ontology tool enrichGO from the clusterprofiler. One-way and two-way ANOVAs were carried out to identify significant changes between the conditions. Results were deemed statistically significant for combined P < 0.05 for at least three independent experiments. RESULTS Cell viability was not significantly affected in the topographical substrates, and cell elongation was enhanced at least twofold in microgrooves and surfaces with a low contact angle. Increased cell elongation correlated with a metabolic shift from oxidative phosphorylation to a glycolytic state which is indicative of a high-energy state. Differential protein expression and gene ontology analyses identified bioprocesses enriched across donors associated with immune modulation and tissue regeneration. The growth of peripheral blood mononuclear cells (PBMCs) was suppressed in hBM-MSCs co-cultures, confirming enhanced immunosuppressive potency. YAP/TAZ levels were found to be reduced on these topographies confirming a mechanosensing effect on cells and suggesting a potential role in the immunomodulatory function of hMSCs. CONCLUSIONS This work demonstrates the potential of topographical cues as a culture strategy to improve the secretory capacity and enrich for an immunomodulatory phenotype in hBM-MSCs.
Collapse
Affiliation(s)
- Heizel Rosado-Galindo
- Bioengineering Program, University of Puerto Rico-Mayagüez, Road 108, KM 1.1., Mayagüez, PR, 00680, USA
| | - Maribella Domenech
- Bioengineering Program, University of Puerto Rico-Mayagüez, Road 108, KM 1.1., Mayagüez, PR, 00680, USA.
- Department of Chemical Engineering, University of Puerto Rico-Mayagüez, Road 108, KM 1.1., Mayagüez, PR, 00680, USA.
| |
Collapse
|
22
|
Guo F, Pan Q, Chen T, Liao S, Li S, Li A, Chen S, Chen J, Xiao Z, Su H, Yang L, Yang C, Liu HF, Pan Q. hUC-MSC transplantation therapy effects on lupus-prone MRL/lpr mice at early disease stages. Stem Cell Res Ther 2023; 14:211. [PMID: 37605271 PMCID: PMC10441722 DOI: 10.1186/s13287-023-03432-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 07/26/2023] [Indexed: 08/23/2023] Open
Abstract
BACKGROUND The efficacy of human umbilical cord mesenchymal stem cell (hUC-MSC) transplantation in treating systemic lupus erythematosus (SLE) has been confirmed by small-scale clinical trials. However, these trials focused on severe or refractory SLE, while few studies focused on mild SLE. Therefore, this study focused on the therapeutic effects of hUC-MSC transplantation in early-stage or mild MRL/lpr lupus model mice. METHODS Commercially available hUC-MSCs were transplanted into 8-week-old MRL/lpr mice by tail vein injection. Flow cytometry was used to analyze B cells and their subsets in the peripheral blood. Further, plasma inflammatory factors, autoantibodies, and plasma biochemical indices were detected using protein chip technology and ELISA kits. In addition, pathological staining and immunofluorescence were performed to detect kidney injury in mice. RESULTS hUC-MSC transplantation did not affect the mice's body weight, and both middle and high dose hUC-MSC transplantation (MD and HD group) actually reduced spleen weight. hUC-MSC transplantation significantly decreased the proportion of plasmablasts (PB), IgG1- PB, IgG1+ PB, IgG1+ memory B (MB) cells, IgG1+ DN MB, and IgG1+ SP MB cells. The hUC-MSC transplantation had significantly reduced plasma levels of inflammatory factors, such as TNF-α, IFN-γ, IL-6, and IL-13. Pathological staining showed that the infiltration of glomerular inflammatory cells was significantly reduced and that the level of glomerular fibrosis was significantly alleviated in hUC-MSC-transplanted mice. Immunofluorescence assays showed that the deposition of IgG and IgM antibodies in the kidneys of hUC-MSC-transplanted mice was significantly lower than in the control. CONCLUSION hUC-MSC transplantation could inhibit the proliferation and differentiation of peripheral blood B cells in the early-stage of MRL/lpr mice, thereby alleviating the plasma inflammatory environment in mice, leading to kidney injury remission. The study provides a new and feasible strategy for SLE treatment.
Collapse
Affiliation(s)
- Fengbiao Guo
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, Guangdong, China
| | - Quanren Pan
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, Guangdong, China
| | - Ting Chen
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, Guangdong, China
| | - Shuzhen Liao
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, Guangdong, China
| | - Shangmei Li
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, Guangdong, China
| | - Aifen Li
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, Guangdong, China
| | - Shuxian Chen
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, Guangdong, China
| | - Jiaxuan Chen
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, Guangdong, China
| | - Zengzhi Xiao
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, Guangdong, China
| | - Hongyong Su
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, Guangdong, China
| | - Lawei Yang
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, Guangdong, China
| | - Chen Yang
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, Guangdong, China
| | - Hua-Feng Liu
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, Guangdong, China.
| | - Qingjun Pan
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, Guangdong, China.
| |
Collapse
|
23
|
Khan S, Mahgoub S, Fallatah N, Lalor PF, Newsome PN. Liver Disease and Cell Therapy: Advances Made and Remaining Challenges. Stem Cells 2023; 41:739-761. [PMID: 37052348 PMCID: PMC10809282 DOI: 10.1093/stmcls/sxad029] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 02/27/2023] [Indexed: 04/14/2023]
Abstract
The limited availability of organs for liver transplantation, the ultimate curative treatment for end stage liver disease, has resulted in a growing and unmet need for alternative therapies. Mesenchymal stromal cells (MSCs) with their broad ranging anti-inflammatory and immunomodulatory properties have therefore emerged as a promising therapeutic agent in treating inflammatory liver disease. Significant strides have been made in exploring their biological activity. Clinical application of MSC has shifted the paradigm from using their regenerative potential to one which harnesses their immunomodulatory properties. Reassuringly, MSCs have been extensively investigated for over 30 years with encouraging efficacy and safety data from translational and early phase clinical studies, but questions remain about their utility. Therefore, in this review, we examine the translational and clinical studies using MSCs in various liver diseases and their impact on dampening immune-mediated liver damage. Our key observations include progress made thus far with use of MSCs for clinical use, inconsistency in the literature to allow meaningful comparison between different studies and need for standardized protocols for MSC manufacture and administration. In addition, the emerging role of MSC-derived extracellular vesicles as an alternative to MSC has been reviewed. We have also highlighted some of the remaining clinical challenges that should be addressed before MSC can progress to be considered as therapy for patients with liver disease.
Collapse
Affiliation(s)
- Sheeba Khan
- National Institute for Health Research, Biomedical Research Centre at University Hospitals Birmingham NHS Foundation Trust and the University of Birmingham, Birmingham, West Midlands, UK
- Centre for Liver and Gastrointestinal Research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, West Midlands, UK
- Liver Unit, University Hospitals Birmingham NHS Foundation Trust, Birmingham, Birmingham, West Midlands, UK
| | - Sara Mahgoub
- National Institute for Health Research, Biomedical Research Centre at University Hospitals Birmingham NHS Foundation Trust and the University of Birmingham, Birmingham, West Midlands, UK
- Centre for Liver and Gastrointestinal Research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, West Midlands, UK
- Liver Unit, University Hospitals Birmingham NHS Foundation Trust, Birmingham, Birmingham, West Midlands, UK
| | - Nada Fallatah
- National Institute for Health Research, Biomedical Research Centre at University Hospitals Birmingham NHS Foundation Trust and the University of Birmingham, Birmingham, West Midlands, UK
- Centre for Liver and Gastrointestinal Research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, West Midlands, UK
- Department of Laboratory Medicine, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Patricia F Lalor
- National Institute for Health Research, Biomedical Research Centre at University Hospitals Birmingham NHS Foundation Trust and the University of Birmingham, Birmingham, West Midlands, UK
- Centre for Liver and Gastrointestinal Research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, West Midlands, UK
| | - Philip N Newsome
- National Institute for Health Research, Biomedical Research Centre at University Hospitals Birmingham NHS Foundation Trust and the University of Birmingham, Birmingham, West Midlands, UK
- Centre for Liver and Gastrointestinal Research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, West Midlands, UK
- Liver Unit, University Hospitals Birmingham NHS Foundation Trust, Birmingham, Birmingham, West Midlands, UK
| |
Collapse
|
24
|
Adolpho LF, Ribeiro LMS, Freitas GP, Lopes HB, Gomes MPO, Ferraz EP, Gimenes R, Beloti MM, Rosa AL. Mesenchymal Stem Cells Combined with a P(VDF-TrFE)/BaTiO 3 Scaffold and Photobiomodulation Therapy Enhance Bone Repair in Rat Calvarial Defects. J Funct Biomater 2023; 14:306. [PMID: 37367270 DOI: 10.3390/jfb14060306] [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: 05/04/2023] [Revised: 05/24/2023] [Accepted: 05/30/2023] [Indexed: 06/28/2023] Open
Abstract
BACKGROUND Tissue engineering and cell therapy have been the focus of investigations on how to treat challenging bone defects. This study aimed to produce and characterize a P(VDF-TrFE)/BaTiO3 scaffold and evaluate the effect of mesenchymal stem cells (MSCs) combined with this scaffold and photobiomodulation (PBM) on bone repair. METHODS AND RESULTS P(VDF-TrFE)/BaTiO3 was synthesized using an electrospinning technique and presented physical and chemical properties suitable for bone tissue engineering. This scaffold was implanted in rat calvarial defects (unilateral, 5 mm in diameter) and, 2 weeks post-implantation, MSCs were locally injected into these defects (n = 12/group). Photobiomodulation was then applied immediately, and again 48 and 96 h post-injection. The μCT and histological analyses showed an increment in bone formation, which exhibited a positive correlation with the treatments combined with the scaffold, with MSCs and PBM inducing more bone repair, followed by the scaffold combined with PBM, the scaffold combined with MSCs, and finally the scaffold alone (ANOVA, p ≤ 0.05). CONCLUSIONS The P(VDF-TrFE)/BaTiO3 scaffold acted synergistically with MSCs and PBM to induce bone repair in rat calvarial defects. These findings emphasize the need to combine a range of techniques to regenerate large bone defects and provide avenues for further investigations on innovative tissue engineering approaches.
Collapse
Affiliation(s)
- Leticia Faustino Adolpho
- Bone Research Lab, School of Dentistry of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040-904, SP, Brazil
| | | | - Gileade Pereira Freitas
- Bone Research Lab, School of Dentistry of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040-904, SP, Brazil
- School of Dentistry, Federal University of Goiás, Goiânia 74605-020, GO, Brazil
| | - Helena Bacha Lopes
- Bone Research Lab, School of Dentistry of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040-904, SP, Brazil
| | - Maria Paula Oliveira Gomes
- Bone Research Lab, School of Dentistry of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040-904, SP, Brazil
| | - Emanuela Prado Ferraz
- Bone Research Lab, School of Dentistry of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040-904, SP, Brazil
| | - Rossano Gimenes
- Institute of Physics and Chemistry, University of Itajubá, Itajubá 37500-903, MG, Brazil
| | - Marcio Mateus Beloti
- Bone Research Lab, School of Dentistry of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040-904, SP, Brazil
| | - Adalberto Luiz Rosa
- Bone Research Lab, School of Dentistry of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040-904, SP, Brazil
| |
Collapse
|
25
|
Phyo H, Aburza A, Mellanby K, Esteves CL. Characterization of canine adipose- and endometrium-derived Mesenchymal Stem/Stromal Cells and response to lipopolysaccharide. Front Vet Sci 2023; 10:1180760. [PMID: 37275605 PMCID: PMC10237321 DOI: 10.3389/fvets.2023.1180760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 05/02/2023] [Indexed: 06/07/2023] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) are used for regenerative therapy in companion animals. Their potential was initially attributed to multipotency, but subsequent studies in rodents, humans and veterinary species evidenced that MSCs produce factors that are key mediators of immune, anti-infective and angiogenic responses, which are essential in tissue repair. MSCs preparations have been classically obtained from bone marrow and adipose tissue (AT) in live animals, what requires the use of surgical procedures. In contrast, the uterus, which is naturally exposed to external insult and infection, can be accessed nonsurgically to obtain samples, or tissues can be taken after neutering. In this study, we explored the endometrium (EM) as an alternative source of MSCs, which we compared with AT obtained from canine paired samples. Canine AT- and EM-MSCs, formed CFUs when seeded at low density, underwent tri-lineage differentiation into adipocytes, osteocytes and chondrocytes, and expressed the CD markers CD73, CD90 and CD105, at equivalent levels. The immune genes IL8, CCL2 and CCL5 were equally expressed at basal levels by both cell types. However, in the presence of the inflammatory stimulus lipopolysaccharide (LPS), expression of IL8 was higher in EM- than in AT-MSCs (p < 0.04) while the other genes were equally elevated in both cell types (p < 0.03). This contrasted with the results for CD markers, where the expression was unaltered by exposing the MSCs to LPS. Overall, the results indicate that canine EM-MSCs could serve as an alternative cell source to AT-MSCs in therapeutic applications.
Collapse
|
26
|
Valsecchi C, Croce S, Lenta E, Acquafredda G, Comoli P, Avanzini MA. TITLE: New therapeutic approaches in pediatric diseases: Mesenchymal stromal cell and mesenchymal stromal cell-derived extracellular vesicles as new drugs. Pharmacol Res 2023; 192:106796. [PMID: 37207738 DOI: 10.1016/j.phrs.2023.106796] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 05/12/2023] [Accepted: 05/16/2023] [Indexed: 05/21/2023]
Abstract
Mesenchymal Stromal Cell (MSC) clinical applications have been widely reported and their therapeutic potential has been documented in several diseases. MSCs can be isolated from several human tissues and easily expanded in vitro, they are able to differentiate in a variety of cell lineages, and they are known to interact with most immunological cells, showing immunosuppressive and tissue repair properties. Their therapeutic efficacy is closely associated with the release of bioactive molecules, namely Extracellular Vesicles (EVs), effective as their parental cells. EVs isolated from MSCs act by fusing with target cell membrane and releasing their content, showing a great potential for the treatment of injured tissues and organs, and for the modulation of the host immune system. EV-based therapies provide, as major advantages, the possibility to cross the epithelium and blood barrier and their activity is not influenced by the surrounding environment. In the present review, we deal with pre-clinical reports and clinical trials to provide data in support of MSC and EV clinical efficacy with particular focus on neonatal and pediatric diseases. Considering pre-clinical and clinical data so far available, it is likely that cell-based and cell-free therapies could become an important therapeutic approach for the treatment of several pediatric diseases.
Collapse
Affiliation(s)
- Chiara Valsecchi
- Pediatric Hematology Oncology Unit and Cell Factory, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy.
| | - Stefania Croce
- Cell Factory, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy.
| | - Elisa Lenta
- Cell Factory, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy.
| | - Gloria Acquafredda
- Pediatric Hematology Oncology Unit and Cell Factory, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy.
| | - Patrizia Comoli
- Pediatric Hematology Oncology Unit and Cell Factory, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy.
| | - Maria Antonietta Avanzini
- Pediatric Hematology Oncology Unit and Cell Factory, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy.
| |
Collapse
|
27
|
de Pedro MÁ, Pulido M, Álvarez V, Marinaro F, Marchena AM, Sánchez-Margallo FM, Casado JG, López E. Menstrual blood-derived stromal cells: insights into their secretome in acute hypoxia conditions. Mol Med 2023; 29:48. [PMID: 37016307 PMCID: PMC10074862 DOI: 10.1186/s10020-023-00646-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 03/23/2023] [Indexed: 04/06/2023] Open
Abstract
BACKGROUND Despite constant advances in regenerative medicine, the closure of chronic wounds is still challenging. Therapeutic approaches using locally administered MSCs have been considered a promising option. However, the viability of these cells is seriously threatened by acute hypoxic stress linked to wound healing. In this work, we aimed to study the tolerance of Menstrual blood-derived stromal cells (MenSCs) to acute hypoxia and their therapeutic paracrine effect. METHODS Isolated MenSCs were phenotypically characterized and evaluated in terms of proliferation, viability, and gene expression, under acute hypoxia (AH) compared with conventional cultured condition or normoxia (N). A step further, the secretome of MenSCs under acute hypoxia was analyzed with respect to their miRNAs content and by in vitro functional assays. For the analysis of differences between the two groups, Student's t-test was performed and one-way ANOVA and Tukey's multiple comparisons test for multiple groups were used. RESULTS Our results revealed that the viability of MenSCs was not affected under acute hypoxia, although proliferation rate slowed down. Gene analysis revealed 5 up-regulated (BNIP3, ANGPTL4, IL6, IL1B, and PDK1) and 4 down-regulated genes (IDO1, HMOX1, ANGPTL2, and HGF) in AH compared to N. Global gene expression analysis revealed a decrease in the gene ontology functions of migration and wound response with respect to the normoxic condition. In contrast, functions such as angiogenesis were enriched under the AH condition. Regarding the secretome analysis, two miRNAs involved in angiogenic processes (hsa-miR-148a-3p and hsa-miR-378a-3p), were significantly up-expressed when compared to the normoxic condition, being MYC gene, the unique target of both. Functional assays on HUVECs revealed a potential pro-angiogenic capacity of MenSCs cultured in both oxygen conditions (N and AH) based on the wound closure and tube formation results of their released paracrine factors. However, when compared to normoxia, the paracrine factors of MenSCs under acute hypoxia slightly reduced the proliferation, migration, and in vitro wound closure of HUVECs. CONCLUSIONS MenSC exhibited a good survival capacity under acute hypoxic conditions as well as beneficial properties applicable in the field of tissue regeneration through their secretome, which makes them a potential cell source for wound healing interventions.
Collapse
Affiliation(s)
- María Ángeles de Pedro
- Stem Cell Therapy Unit, Jesús Usón Minimally Invasive Surgery Centre, 10071, Cáceres, Spain
- RICORS-TERAV Network, ISCIII, 28029, Madrid, Spain
| | - María Pulido
- Stem Cell Therapy Unit, Jesús Usón Minimally Invasive Surgery Centre, 10071, Cáceres, Spain
| | - Verónica Álvarez
- Stem Cell Therapy Unit, Jesús Usón Minimally Invasive Surgery Centre, 10071, Cáceres, Spain
| | - Federica Marinaro
- Stem Cell Therapy Unit, Jesús Usón Minimally Invasive Surgery Centre, 10071, Cáceres, Spain
| | - Ana María Marchena
- Stem Cell Therapy Unit, Jesús Usón Minimally Invasive Surgery Centre, 10071, Cáceres, Spain
| | - Francisco Miguel Sánchez-Margallo
- Stem Cell Therapy Unit, Jesús Usón Minimally Invasive Surgery Centre, 10071, Cáceres, Spain.
- RICORS-TERAV Network, ISCIII, 28029, Madrid, Spain.
| | - Javier G Casado
- RICORS-TERAV Network, ISCIII, 28029, Madrid, Spain
- Immunology Unit, University of Extremadura, 10003, Cáceres, Spain
- Institute of Molecular Pathology Biomarkers, University of Extremadura, 10003, Cáceres, Spain
| | - Esther López
- Stem Cell Therapy Unit, Jesús Usón Minimally Invasive Surgery Centre, 10071, Cáceres, Spain
- RICORS-TERAV Network, ISCIII, 28029, Madrid, Spain
| |
Collapse
|
28
|
Ozel C, Apaydin E, Sariboyaci AE, Tamayol A, Avci H. A multifunctional sateen woven dressings for treatment of skin injuries. Colloids Surf B Biointerfaces 2023; 224:113197. [PMID: 36822118 DOI: 10.1016/j.colsurfb.2023.113197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 01/30/2023] [Accepted: 02/02/2023] [Indexed: 02/05/2023]
Abstract
Cutaneous wounds with impaired healing such as diabetic ulcers and burns constitute major and rapidly growing threat to healthcare systems worldwide. Accelerating wound healing requires the delivery of biological factors that induce angiogenesis, support cellular proliferation, and modulate inflammation while minimizing infection. In this study, we engineered a dressing made by weaving of composite fibers (CFs) carrying mesenchymal stem cells (MSCs) and a model antibiotic using a scalable sateen textile technique. In this regard, two different sets of CFs carrying MSCs or an antimicrobial agent were used to generate a multifunctional dressing. According to cell viability and metabolic activity as CCK-8 and live/dead with qRT-PCR results, more than %90 the encapsulated MSCs remain viable for 28 days and their expression levels of the wound repair factors including ECM remodeling, angiogenesis and immunomodulatory maintained in MSCs post dressing manufacturing for 14 days. Post 10 days culture of the dressing, MSCs within CFs had 10-fold higher collagen synthesis (p < 0.0001) determined by hydroxyproline assay which indicates the enhanced healing properties. According to in vitro antimicrobial activity results determined by disk diffusion and broth microdilution tests, the first day and the total amount of release gentamicin loaded dressing samples during the 28 days were higher than determined minimal inhibition concentration (MIC) values for S. aureus and K. pneumonia without negatively impacting the viability and functionality of encapsulated MSCs within the dressing. The dressing is also flexible and can conform to skin curvatures making the dressing suitable for the treatment of different skin injuries such as burns and diabetic ulcers.
Collapse
Affiliation(s)
- Ceren Ozel
- Department of Stem Cell, Institute of Health Sciences, Eskisehir Osmangazi University, Eskisehir 26040, Turkey; Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, Eskisehir 26040, Turkey
| | - Elif Apaydin
- Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, Eskisehir 26040, Turkey; Department of Biochemistry, Institute of Health Sciences, Anadolu University, Eskişehir 26470, Turkey
| | - Ayla Eker Sariboyaci
- Department of Stem Cell, Institute of Health Sciences, Eskisehir Osmangazi University, Eskisehir 26040, Turkey; Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, Eskisehir 26040, Turkey
| | - Ali Tamayol
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT 06269, USA.
| | - Huseyin Avci
- Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, Eskisehir 26040, Turkey; Department of Metallurgical and Materials Engineering, Eskişehir Osmangazi University, Eskişehir 26040, Turkey; Translational Medicine Research and Clinical Center (TATUM), Eskişehir Osmangazi University, Eskişehir 26040, Turkey.
| |
Collapse
|
29
|
CD73-Positive Cell Spheroid Transplantation Attenuates Colonic Atrophy. Pharmaceutics 2023; 15:pharmaceutics15030845. [PMID: 36986706 PMCID: PMC10051511 DOI: 10.3390/pharmaceutics15030845] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 02/27/2023] [Accepted: 03/02/2023] [Indexed: 03/08/2023] Open
Abstract
The incidence of inflammatory bowel diseases (IBD) is increasing worldwide. Mesenchymal stem/stromal cells (MSCs) have immunomodulatory functions and are a promising source for cell transplantation therapy for IBD. However, owing to their heterogeneous nature, their therapeutic efficacy in colitis is controversial and depends on the delivery route and form of transplanted cells. Cluster of differentiation (CD) 73 is widely expressed in MSCs and used to obtain a homogeneous MSC population. Herein, we determined the optimal method for MSC transplantation using CD73+ cells in a colitis model. mRNA sequencing analysis showed that CD73+ cells exhibited a downregulation of inflammatory gene expression and an upregulation of extracellular matrix-related gene expression. Furthermore, three-dimensional CD73+ cell spheroids showed enhanced engraftment at the injured site through the enteral route, facilitated extracellular matrix remodeling, and downregulated inflammatory gene expression in fibroblasts, leading to the attenuation of colonic atrophy. Therefore, the interaction between intestinal fibroblasts and exogenous MSCs via tissue remodeling is one mechanism that can be exploited for colitis prevention. Our results highlight that the transplantation of homogeneous cell populations with well-characterized properties is beneficial for IBD treatment.
Collapse
|
30
|
Dilogo IH, Lubis AMT, Perwida NG, Sani SA, Rasyidah RA, Hartanto BR. The Efficacy of Intra-articular Umbilical Cord-Mesenchymal Stem Cell Injection for Knee Osteoarthritis: a Systematic Review. CURRENT STEM CELL REPORTS 2023. [DOI: 10.1007/s40778-023-00223-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
|
31
|
Interferon-γ-Treated Mesenchymal Stem Cells Modulate the T Cell-Related Chemokines and Chemokine Receptors in an Animal Model of Experimental Autoimmune Encephalomyelitis. Drug Res (Stuttg) 2023; 73:213-223. [PMID: 36754055 DOI: 10.1055/a-1995-6365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) modulate immune responses, and their immunomodulatory potential can be enhanced using inflammatory cytokines. Here, the modulatory effects of IFN-γ-licensed MSCs on expression of T cell-related chemokines and chemokine receptors were evaluated using an experimental autoimmune encephalomyelitis (EAE) model. MATERIAL AND METHODS EAE was induced in 3 groups of C57bl/6 mice and then treated with PBS, MSCs and IFN-γ-treated MSCs. The EAE manifestations were registered daily and finally, the brain and spinal cords were isolated for histopathological and gene expression studies. RESULTS The clinical scores were lowered in MSCs and IFN-γ-licensed MSCs groups, however, mice treated with IFN-γ-licensed MSCs exhibited lower clinical scores than MSCs-treated mice. Leukocyte infiltration into the brain was reduced after treatment with MSCs or IFN-γ-licensed MSCs compared to untreated group (P<0.05 and P<0.01, respectively). In comparison with untreated EAE mice, treatment with MSCs reduced CCL20 expression (P<0.001) and decreased CXCR3 and CCR6 expression (P<0.02 and P<0.04, respectively). In comparison with untreated EAE mice, treatment with IFN-γ-licensed MSCs reduced CXCL10, CCL17 and CCL20 expression (P<0.05, P<0.05, and P<0.001, respectively) as well as decreased CXCR3 and CCR6 expression (P<0.002 and P<0.02, respectively), whilst promoting expression of CCL22 and its receptor CCR4 (P<0.0001 and P<0.02, respectively). In comparison with MSC-treated group, mice treated with IFN-γ-licensed MSCs exhibited lower CXCL10 and CCR6 expression (P<0.002 and P<0.01, respectively), whereas greater expression of CCL22 and CCR4 (P<0.0001 and P<0.01, respectively). CONCLUSION Priming the MSC with IFN-γ can be an efficient approach to enhance the immunomodulatory potential of MSCs.
Collapse
|
32
|
Porous composite hydrogels with improved MSC survival for robust epithelial sealing around implants and M2 macrophage polarization. Acta Biomater 2023; 157:108-123. [PMID: 36435441 DOI: 10.1016/j.actbio.2022.11.029] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 11/12/2022] [Accepted: 11/15/2022] [Indexed: 11/25/2022]
Abstract
The application of mesenchymal stem cell (MSC)-based therapy is expected to make a significant contribution to the improvement of epithelial sealing around implants. However, there is currently no optimal MSC delivery biomaterial for clinical application in peri-implant epithelium (PIE) integration. In this study, we show that injectable photo-cross-linkable porous gelatin methacryloyl (GelMA)/silk fibroin glycidyl methacrylate (SilMA) hydrogels encapsulating gingival tissue-derived MSCs (GMSCs) are a simple and practical approach for re-epithelization applications. The hydrogels played a prominent role in supporting the proliferation, survival, and spread of GMSCs. Moreover, it was found that GMSCs-laden Porous GelMA/SilMA hydrogels could significantly upregulate the hemidesmosomes (HDs)-related genes and proteins expression and promote M2 polarization while inhibiting M1 polarization in vitro. Based on a rat model of early implant placement, application of the MSC-loaded hydrogels could enhance the protein expression of LAMA3 and BP180 (COL17A1) at the implant-PIE interface and reduce horseradish peroxidase (HRP) penetration between the implants and PIE. Noticeably, hydrogel-based MSC therapy contributed to augmenting M2 macrophage infiltration at two time points in the gingival connective tissue around implants. These findings demonstrated that GMSCs-laden Porous GelMA/SilMA hydrogels could facilitate epithelial sealing around implants and M2-polarized macrophages and may be a novel and facile therapeutic strategy for implant-PIE integration. STATEMENT OF SIGNIFICANCE: In the case of poor integration between the implant and gingival epithelium, peri-implantitis can develop, which is one of the main causes of implant failure. While stem cell therapy has tremendous potential for addressing this issue, poor cell survival and engraftment compromise the effectiveness of the therapy. Due to the excellent modifiable and tunable properties of gelatin and silk fibroin, injectable photo-cross-linkable porous hydrogels were developed using gelatin methacryloyl (GelMA) and silk fibroin glycidyl methacrylate (SilMA) as delivery vehicles for gingiva-derived MSCs (GMSCs). Porous GelMA/SilMA not only enhanced the proliferation and viability of GMSCs but also promoted their immunomodulatory capability for favorable epithelial sealing around implants. Overall, GMSCs-seeded porous hydrogels could be promising strategies for re-epithelization treatment.
Collapse
|
33
|
Sadeghi S, Tehrani FR, Tahmasebi S, Shafiee A, Hashemi SM. Exosome engineering in cell therapy and drug delivery. Inflammopharmacology 2023; 31:145-169. [PMID: 36609717 PMCID: PMC9823267 DOI: 10.1007/s10787-022-01115-7] [Citation(s) in RCA: 111] [Impact Index Per Article: 55.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 12/09/2022] [Indexed: 01/09/2023]
Abstract
Cell-derived exosomes have opened new horizons in modern therapy for advanced drug delivery and therapeutic applications, due to their key features such as low immunogenicity, high physicochemical stability, capacity to penetrate into tissues, and the innate capacity to communicate with other cells over long distances. Exosome-based liquid biopsy has been potentially used for the diagnosis and prognosis of a range of disorders. Exosomes deliver therapeutic agents, including immunological modulators, therapeutic drugs, and antisense oligonucleotides to certain targets, and can be used as vaccines, though their clinical application is still far from reality. Producing exosomes on a large-scale is restricted to their low circulation lifetime, weak targeting capacity, and inappropriate controls, which need to be refined before being implemented in practice. Several bioengineering methods have been used for refining therapeutic applications of exosomes and promoting their effectiveness, on the one hand, and addressing the existing challenges, on the other. In the short run, new diagnostic platforms and emerging therapeutic strategies will further develop exosome engineering and therapeutic potential. This requires a thorough analysis of exosome engineering approaches along with their merits and drawbacks, as outlined in this paper. The present study is a comprehensive review of novel techniques for exosome development in terms of circulation time in the body, targeting capacity, and higher drug loading/delivery efficacies.
Collapse
Affiliation(s)
- Somaye Sadeghi
- Reproductive Endocrinology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Advanced Therapy Medicinal Product (ATMP) Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Fahimeh Ramezani Tehrani
- Reproductive Endocrinology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Safa Tahmasebi
- Student Research Committee, Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abbas Shafiee
- The University of Queensland Diamantina Institute, Faculty of Medicine, The University of Queensland, 37 Kent Street, Woolloongabba, Brisbane, QLD, 4102, Australia.
- Herston Biofabrication Institute, Metro North Hospital and Health Service, Brisbane, QLD 4029, Australia.
| | - Seyed Mahmoud Hashemi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
- Medical Nanotechnology and tissue engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
34
|
Ma H, Siu WS, Koon CM, Wu XX, Li X, Cheng W, Shum WT, Lau CBS, Wong CK, Leung PC. The Application of Adipose Tissue-Derived Mesenchymal Stem Cells (ADMSCs) and a Twin-Herb Formula to the Rodent Wound Healing Model: Use Alone or Together? Int J Mol Sci 2023; 24:ijms24021372. [PMID: 36674885 PMCID: PMC9867064 DOI: 10.3390/ijms24021372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/04/2023] [Accepted: 01/05/2023] [Indexed: 01/13/2023] Open
Abstract
Our previous study reported that mesenchymal stem cells (MSCs) accelerated the wound healing process through anti-inflammatory, anti-apoptotic, and pro-angiogenetic effects in a rodent skin excision model. NF3 is a twin-herb formula, which presents similar effects in promoting wound healing. Research focusing on the interaction of MSCs and Chinese medicine is limited. In this study, we applied MSCs and the twin-herb formula to the wound healing model and investigated their interactions. Wound healing was improved in all treatment groups (MSCs only, NF3 only, and MSCs + NF3). The combined therapy further enhanced the effect: more GFP-labelled ADMSCs, collagen I and collagen III expression, Sox9 positive cells, and CD31 positive cells, along with less ED-1 positive cells, were detected; the expressions of proinflammatory cytokine IL-6 and TNF-α were downregulated; and the expression of anti-inflammatory cytokine IL-10 was upregulated. In vitro, NF3 promoted the cell viability and proliferation ability of MSCs, and a higher concentration of protein was detected in the NF3-treated supernatant. A proteomic analysis showed there were 15 and 22 proteins in the supernatants of normal ADMSCs and NF3-treated ADMSCs, respectively. After PCR validation, the expressions of 11 related genes were upregulated. The results of a western blot suggested that the TGFβ/Smad and Wnt pathways were related to the therapeutic effects of the combined treatment. Our study suggests for the first time that NF3 enhanced the therapeutic effect of MSCs in the wound healing model and the TGFβ/Smad and Wnt pathways were related to the procedure.
Collapse
Affiliation(s)
- Hui Ma
- Institute of Chinese Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| | - Wing-Sum Siu
- Institute of Chinese Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| | - Chi-Man Koon
- Institute of Chinese Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| | - Xiao-Xiao Wu
- Institute of Chinese Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| | - Xiang Li
- Institute of Chinese Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| | - Wen Cheng
- Institute of Chinese Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| | - Wai-Ting Shum
- Institute of Chinese Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| | - Clara Bik-San Lau
- Institute of Chinese Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| | - Chun-Kwok Wong
- Institute of Chinese Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
- Department of Chemical Pathology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Ping-Chung Leung
- Institute of Chinese Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
- Correspondence: ; Tel.: +852-22528868
| |
Collapse
|
35
|
Neves BRO, de Freitas S, Borelli P, Rogero MM, Fock RA. Delphinidin-3-O-glucoside in vitro suppresses NF-κB and changes the secretome of mesenchymal stem cells affecting macrophage activation. Nutrition 2023; 105:111853. [PMID: 36335873 DOI: 10.1016/j.nut.2022.111853] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 08/17/2022] [Accepted: 09/19/2022] [Indexed: 11/25/2022]
Abstract
OBJECTIVE Anthocyanins are polyphenols that are promising chemopreventive agents. They stand out for their anti-inflammatory properties, with specific modulatory actions on the immune system. Additionally, regarding the immune system, a group of cells identified as mesenchymal stem cells (MSCs) have been attracting attention, mainly because of their capacity to migrate to sites of inflammation and produce potent immunomodulatory effects. Considering the ability of these cells to act on the immune system, as well as the properties of anthocyanins, especially delphinidin, in modulating the immune system, the aim of this study was to investigate the effects of delphinidin in influencing some immunoregulatory properties of MSCs. METHODS MSCs were cultivated in the presence of delphinidin 3-O-β-d-glycoside and cell viability, the cell cycle and the production of soluble factors (interleukin [IL]-1β, IL-6, IL-10, transforming growth factor [TGF]-β, prostaglandin E2 [PGE2] and nitric oxide [NO]) were evaluated, as was the expression of the transcription factors nuclear factor (NF)-κB and STAT3. Additionally, the effects of conditioned media from MSCs on macrophage activation were assessed. RESULTS Delphinidin at 50 µM does not affect cell viability. In association with lipopolysaccharide, delphinidin was able to induce MSC proliferation. Additionally, delphinidin modulated the MSC immune response, showing increased levels of anti-inflammatory cytokines such as IL-10 and TGF-β as well as lower expression of NF-κB. Furthermore, conditioned media from MSCs inhibited macrophage metabolism, reducing the production of IL-1β, IL-12, and TNF-α and increasing IL-10. CONCLUSIONS Overall, this work showed that delphinidin can modify the immunomodulatory properties of MSCs, increasing the IL-10 production by macrophages.
Collapse
Affiliation(s)
- Bruna Roberta Oliveira Neves
- Department of Clinical and Toxicologic Analysis, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Sumara de Freitas
- Department of Clinical and Toxicologic Analysis, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Primavera Borelli
- Department of Clinical and Toxicologic Analysis, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Marcelo Macedo Rogero
- Department of Nutrition, School of Public Health, University of São Paulo, São Paulo, Brazil
| | - Ricardo Ambrosio Fock
- Department of Clinical and Toxicologic Analysis, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil.
| |
Collapse
|
36
|
Fu SP, Chen SY, Pang QM, Zhang M, Wu XC, Wan X, Wan WH, Ao J, Zhang T. Advances in the research of the role of macrophage/microglia polarization-mediated inflammatory response in spinal cord injury. Front Immunol 2022; 13:1014013. [PMID: 36532022 PMCID: PMC9751019 DOI: 10.3389/fimmu.2022.1014013] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 11/18/2022] [Indexed: 12/04/2022] Open
Abstract
It is often difficult to regain neurological function following spinal cord injury (SCI). Neuroinflammation is thought to be responsible for this failure. Regulating the inflammatory response post-SCI may contribute to the recovery of neurological function. Over the past few decades, studies have found that macrophages/microglia are one of the primary effector cells in the inflammatory response following SCI. Growing evidence has documented that macrophages/microglia are plastic cells that can polarize in response to microenvironmental signals into M1 and M2 macrophages/microglia. M1 produces pro-inflammatory cytokines to induce inflammation and worsen tissue damage, while M2 has anti-inflammatory activities in wound healing and tissue regeneration. Recent studies have indicated that the transition from the M1 to the M2 phenotype of macrophage/microglia supports the regression of inflammation and tissue repair. Here, we will review the role of the inflammatory response and macrophages/microglia in SCI and repair. In addition, we will discuss potential molecular mechanisms that induce macrophage/microglia polarization, with emphasis on neuroprotective therapies that modulate macrophage/microglia polarization, which will provide new insights into therapeutic strategies for SCI.
Collapse
Affiliation(s)
- Sheng-Ping Fu
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China,Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Si-Yu Chen
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Qi-Ming Pang
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Meng Zhang
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Xiang-Chong Wu
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Xue Wan
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China,Collaborative Innovation Center of Chinese Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China
| | - Wei-Hong Wan
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China,Collaborative Innovation Center of Chinese Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China
| | - Jun Ao
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Tao Zhang
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China,Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China,Collaborative Innovation Center of Chinese Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China,The Clinical Stem Cell Research Institute, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China,*Correspondence: Tao Zhang,
| |
Collapse
|
37
|
Noughabi MK, Matin MM, Farshchian M, Bahrami AR. Immunomodulatory Properties of Mouse Mesenchymal Stromal/Stem Cells Upon Ectopic Expression of Immunoregulator Nanos2. Stem Cell Rev Rep 2022; 19:734-753. [PMID: 36348161 DOI: 10.1007/s12015-022-10451-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/07/2022] [Indexed: 11/11/2022]
Abstract
BACKGROUND Mesenchymal stromal/stem cells (MSCs) are known for their involvement in modulating the immune system of mammals. This potency could be enhanced by different strategies, including regulation of key proteins, in order to meet desirable therapeutic properties. Nanos2, encoding an RNA-binding protein involved in regulation of key spermatogonial signaling pathways, has been demonstrated to downregulate a range of immune related genes in mouse embryonic fibroblasts (MEFs). Accordingly, it was hypothesized that Nanos2 functions as a potent immunosuppressing factor. This study was aimed to measure the expression profile of the immune-related genes in mouse mesenchymal stromal/stem cells (mMSCs) and assess their functional properties after Nanos2 ectopic expression. METHODS As inflammatory mediators, interferon (IFN-γ) and poly(I:C) were used to provoke an immune response. The interactions between the control and engineered mMSCs overexpressing Nanos2, with mouse peripheral blood mononuclear cells (mPBMCs) were then compared. The sensitivity of these cells to an inflammatory environment was assessed by using a conditioned medium containing high levels of inflammatory cytokines. Finally, the functional properties of the cells were investigated both in vivo and in vitro in presence of tumor and immune cells. RESULTS Deep transcriptome analysis indicated that numerous genes were downregulated as a result of higher Nanos2 expression. Most of the genes subjected to gene expression alteration, were responsible for controlling responses to external stimuli, cell-cell adhesion, and wound healing. In comparison to the control cells, Nanos2-overexpressing cells showed lower expression of several immune-related genes after pretreatment with IFN-γ and poly(I:C). They also exhibited inhibitory effects against mPBMCs proliferation. Tumor growth rate, in B16-F0 administered mice was obviously increased upon their treatment with the Nanos2-mMSCs, while no tumor or very small ones were developed in the control group. In addition, the cytotoxic environment had no significant effects on Nanos2-mMSCs. CONCLUSIONS According to the literature, MSCs are believed to be tuned very precisely by their internal and external conditions to act as either pro-inflammatory or anti-inflammatory agents. We show here that Nanos2 plays a significant role in promoting anti-inflammatory properties when expressed at higher levels by MSCs. This approach could be adopted for controlling the excessive inflammatory conditions in clinical programs, however more experiments are required to confirm it. In Brief Viral transduction was used to over express Nanos2 in mouse mesenchymal stromal/stem cells (mMSCs). Induced expression of Nanos2 downregulated the expression of immune-related genes and proteins. These modified mMSCs switched to an immunosuppressive state, even in the presence of pro-inflammatory cytokines; and could also contribute to tumor progression in a mouse model.
Collapse
Affiliation(s)
| | - Maryam M Matin
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
- Novel Diagnostics and Therapeutics Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Moein Farshchian
- Stem Cells and Regenerative Medicine Research Group, Academic Center for Education, Culture and Research (ACECR)-Khorasan Razavi, Mashhad, Iran
| | - Ahmad Reza Bahrami
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran.
- Industrial Biotechnology Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran.
| |
Collapse
|
38
|
Divband S, Tasharrofi N, Abroun S, Zomorrod MS. Human Umbilical Cord Mesenchymal Stem Cells-Derived Small Extracellular Vesicles Can Be Considered as Cell-Free Therapeutics for Angiogenesis Promotion. CELL JOURNAL 2022; 24:689-696. [PMID: 36377219 PMCID: PMC9663965 DOI: 10.22074/cellj.2022.8275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Indexed: 12/01/2022]
Abstract
OBJECTIVE Angiogenesis has critical roles in several physiological processes. Restoring angiogenesis in some pathological conditions such as a few vascular diseases can be a therapeutic approach to controlling this issue. Mesenchymal stem cells (MSCs) secrete specific intracellular products known as extracellular vesicles (EVs) with high therapeutic potential which compared to their source cells, do not have the limitations of cell therapy. The angiogenic effect of the human umbilical cord MSCs (hUCMSCs)-derived small EVs are evaluated in the present work. Aim of this research is to show that hUCMSCs-derived small EVs cause differentiation of genes involved in angiogenesis like FGFR-1, FGF, VEGF, and VEGFR-2. MATERIALS AND METHODS In this experimental study, MSCs were isolated from the human umbilical cord, and after confirming their identities, their secreted EVs (including exosomes) were extracted by ultracentrifugation. The isolated small EVs were characterized by dynamic light scattering (DLS), transmission electron microscopy (TEM), bicinchoninic acid assay (BCA), and Western Blotting. Then, the human umbilical vein endothelial cells (HUVECs) were treated with derived small EVs for 72 hours, and the expression of the angiogenic factors including FGFR-1, FGF, VEGF, and VEGFR-2 was evaluated by quantitative real-time-polymerase chain reaction (qPCR). Angiogenesis was also evaluated via a tube formation assay. RESULTS The results demonstrated that FGFR-1, FGF, VEGF, and VEGFR-2 could be elevated 2, 2, 3.5, and 2 times, respectively, in EVs treated HUVECs, and derivative EVs can encourage tube formation in HUVECs. CONCLUSION These findings imply that hUCMSCs-derived small EVs are valuable resources in promoting angiogenesis and are very promising in cell-free therapy.
Collapse
Affiliation(s)
- Somayeh Divband
- Department of Hematology and Cell Therapy, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Nooshin Tasharrofi
- Faculty of Pharmacy, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Saeid Abroun
- Department of Hematology and Cell Therapy, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mina Soufi Zomorrod
- Department of Hematology and Cell Therapy, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran,P.O.Box: 14115-111Department of Hematology and Cell TherapyFaculty of Medical SciencesTarbiat Modares
UniversityTehranIran
| |
Collapse
|
39
|
Shi MY, Liu L, Yang FY. Strategies to improve the effect of mesenchymal stem cell therapy on inflammatory bowel disease. World J Stem Cells 2022; 14:684-699. [PMID: 36188115 PMCID: PMC9516464 DOI: 10.4252/wjsc.v14.i9.684] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 07/07/2022] [Accepted: 09/07/2022] [Indexed: 02/07/2023] Open
Abstract
Inflammatory bowel disease (IBD) includes Crohn’s disease and ulcerative colitis and is an idiopathic, chronic inflammatory disease of the colonic mucosa. The occurrence of IBD, causes irreversible damage to the colon and increases the risk of carcinoma. The routine clinical treatment of IBD includes drug treatment, endoscopic treatment and surgery. The vast majority of patients are treated with drugs and biological agents, but the complete cure of IBD is difficult. Mesenchymal stem cells (MSCs) have become a new type of cell therapy for the treatment of IBD due to their immunomodulatory and nutritional functions, which have been confirmed in many clinical trials. This review discusses some potential mechanisms of MSCs in the treatment of IBD, summarizes the experimental results, and provides new insights to enhance the therapeutic effects of MSCs in future applications.
Collapse
Affiliation(s)
- Meng-Yue Shi
- School of Medicine, Yangtze University, Jingzhou 434023, Hubei Province, China
| | - Lian Liu
- Department of Pharmacology, Medical School of Yangtze University, Yangtze University, Jingzhou 434023, Hubei Province, China
| | - Fu-Yuan Yang
- Health Science Center, Yangtze University, Jingzhou 434020, Hubei Province, China
| |
Collapse
|
40
|
Sarsenova M, Kim Y, Raziyeva K, Kazybay B, Ogay V, Saparov A. Recent advances to enhance the immunomodulatory potential of mesenchymal stem cells. Front Immunol 2022; 13:1010399. [PMID: 36211399 PMCID: PMC9537745 DOI: 10.3389/fimmu.2022.1010399] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 09/07/2022] [Indexed: 11/19/2022] Open
Abstract
Considering the unique therapeutic potential of mesenchymal stem cells (MSCs), including their immunosuppressive and immunomodulatory properties as well as their ability to improve tissue regeneration, these cells have attracted the attention of scientists and clinicians for the treatment of different inflammatory and immune system mediated disorders. However, various clinical trials using MSCs for the therapeutic purpose are conflicting and differ from the results of promising preclinical studies. This inconsistency is caused by several factors such as poor migration and homing capacities, low survival rate, low level of proliferation and differentiation, and donor-dependent variation of the cells. Enhancement and retention of persistent therapeutic effects of the cells remain a challenge to overcome in MSC-based therapy. In this review, we summarized various approaches to enhance the clinical outcomes of MSC-based therapy as well as revised current and future perspectives for the creation of cellular products with improved potential for diverse clinical applications.
Collapse
Affiliation(s)
- Madina Sarsenova
- Department of Medicine, School of Medicine, Nazarbayev University, Nur-Sultan, Kazakhstan
| | - Yevgeniy Kim
- Department of Medicine, School of Medicine, Nazarbayev University, Nur-Sultan, Kazakhstan
| | - Kamila Raziyeva
- Department of Medicine, School of Medicine, Nazarbayev University, Nur-Sultan, Kazakhstan
| | - Bexultan Kazybay
- Department of Medicine, School of Medicine, Nazarbayev University, Nur-Sultan, Kazakhstan
| | - Vyacheslav Ogay
- Laboratory of Stem Cells, National Center for Biotechnology, Nur-Sultan, Kazakhstan
| | - Arman Saparov
- Department of Medicine, School of Medicine, Nazarbayev University, Nur-Sultan, Kazakhstan
- *Correspondence: Arman Saparov,
| |
Collapse
|
41
|
Zhuang Z, Zhang Y, Yang X, Yu T, Zhang Y, Sun K, Zhang Y, Cheng F, Zhang L, Wang H. Matrix stiffness regulates the immunomodulatory effects of mesenchymal stem cells on macrophages via AP1/TSG-6 signaling pathways. Acta Biomater 2022; 149:69-81. [PMID: 35820593 DOI: 10.1016/j.actbio.2022.07.010] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 07/05/2022] [Accepted: 07/05/2022] [Indexed: 11/28/2022]
Abstract
It is well-recognized that the matrix stiffness as an important stem cell niche can mediate stem cell behavior such as attachment, proliferation and differentiation, but how matrix stiffness affects the immunomodulatory efficacy of stem cells has been little explored, which, however, is of significant importance in determining the outcomes of stem cell-based therapies and engineered tissue mimics. We herein studied the immunomodulatory efficacy of mesenchymal stem cells (MSCs) in response to matrix stiffness by the evaluation of macrophage polarization in vitro and inflammatory response in vivo by subcutaneous implantation of MSC-laden hydrogels. Remarkably, we found that soft matrix enabled MSCs to produce significantly higher levels of immunomodulatory factors compared to stiff matrix, and induced the presence of more anti-inflammatory macrophages in vitro and attenuated macrophages-mediated inflammatory response in vivo. More importantly, we revealed stiffness-mediated immunoregulatory effect of MSCs was mainly attributed to tumor necrosis factor-α-stimulated protein 6 (TSG-6), which was mechanosensitively regulated by the MAPK and Hippo signaling pathway and downstream AP1 complex, and which in turn exerted an effect on macrophages through CD44 receptor to inhibit NF-κB pathway. To conclude, our results for the first time identify TSG-6 as the key factor in regulating immunomodulatory efficacy of MSCs in mechanical response, and can be potentially utilized to empower stem cell-based therapy and tissue engineering strategy in regenerative medicine. STATEMENT OF SIGNIFICANCE: Matrix stiffness as an important stem cell niche can mediate stem cell behavior such as attachment and differentiation, but how matrix stiffness affects the immunomodulatory efficacy of stem cells has been little explored, which, however, is of significant importance in determining the outcomes of stem cell-based therapies and engineered tissue mimics. Our results for the first time identify TSG-6 as the key factor in regulating the immunomodulatory efficacy of MSCs in mechanical response, which was regulated by the MAPK and Hippo signaling pathways and downstream AP1 complex, and which in turn exerted an effect on macrophages through CD44 receptor to inhibit NF-κB pathway, and can be potentially utilized to empower stem cell-based therapy and tissue engineering strategy in regenerative medicine.
Collapse
Affiliation(s)
- Zhumei Zhuang
- Key State Laboratory of Fine Chemicals, School of Bioengineering, Dalian University of Technology, No.2 Linggong Road, High-tech District, Dalian, 116024, China
| | - Yang Zhang
- School of Stomatology, Health Science Center, Shenzhen University, Shenzhen, 518037, China; School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen 518060, China
| | - Xueying Yang
- Key State Laboratory of Fine Chemicals, School of Bioengineering, Dalian University of Technology, No.2 Linggong Road, High-tech District, Dalian, 116024, China
| | - Taozhao Yu
- School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen 518060, China
| | - Yue Zhang
- Key State Laboratory of Fine Chemicals, School of Bioengineering, Dalian University of Technology, No.2 Linggong Road, High-tech District, Dalian, 116024, China
| | - Kai Sun
- Key State Laboratory of Fine Chemicals, School of Bioengineering, Dalian University of Technology, No.2 Linggong Road, High-tech District, Dalian, 116024, China
| | - Yonggang Zhang
- Key State Laboratory of Fine Chemicals, School of Bioengineering, Dalian University of Technology, No.2 Linggong Road, High-tech District, Dalian, 116024, China
| | - Fang Cheng
- Key State Laboratory of Fine Chemicals, School of Chemical Engineering, Dalian University of Technology, No.2 Linggong Road, High-tech District, Dalian, 116024, China
| | - Lijun Zhang
- Third People's Hospital of Dalian, Dalian Eye Hospital, No.40 Qianshan Road, Ganjingzi District, Dalian, 116024, China
| | - Huanan Wang
- Key State Laboratory of Fine Chemicals, School of Bioengineering, Dalian University of Technology, No.2 Linggong Road, High-tech District, Dalian, 116024, China.
| |
Collapse
|
42
|
Liu G, He G, Zhang J, Zhang Z, Wang L. Identification of SCRG1 as a Potential Therapeutic Target for Human Synovial Inflammation. Front Immunol 2022; 13:893301. [PMID: 35720295 PMCID: PMC9204521 DOI: 10.3389/fimmu.2022.893301] [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: 03/10/2022] [Accepted: 04/26/2022] [Indexed: 01/15/2023] Open
Abstract
Synovial inflammation of joint tissue is the most important cause of tissue damage, joint destruction, and disability and is associated with higher morbidity or mortality. Therefore, this study aims to identify key genes in osteoarthritis synovitis tissue to increase our understanding of the underlying mechanisms of osteoarthritis and identify new therapeutic targets. Five GEO datasets with a total of 41 normal synovial membrane tissues and 45 osteoarthritis synovial membrane samples were used for analysis, and seven common differential genes were identified. The classification model constructed by LASSO analysis showed that six genes including CDKN1A, FOSB, STMN2, SLC2A3, TAC, and SCRG1 can be used as biomarkers of osteoarthritis, and the SCRG1 gene shows importance in osteoarthritis. Furthermore, drug database enrichment found that these six DEGs may be the drug targets of synovitis in osteoarthritis, and Valproic Acid CTD 00006977 may be a potential targeted therapeutic drug of SCRG1. Spearman correlation analysis was performed on the SCRG1 gene, and 27 genes with consistent expression were obtained. Functional analysis showed that 27 genes were mainly involved in metabolism, complement, antigen presentation, apoptosis, and regulation of immune pathways. The co-regulatory network of TFs-miRNA suggested that the SCRG1 gene may be regulated by hsa-miR-363-3p miRNA. In conclusion, SCRG1, as a diagnostic marker of osteoarthritis, co-regulates immune-related pathways through the interaction of related proteins, playing an important role in the occurrence and development of osteoarthritis, which may be a novel drug target.
Collapse
Affiliation(s)
- Guoqiang Liu
- Department of Orthopedics, Academy of Orthopedics, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Guisong He
- Department of Orthopedics, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jie Zhang
- Department of Orthopedics, Academy of Orthopedics, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Zhongmin Zhang
- Department of Orthopedics, Academy of Orthopedics, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China.,Division of Spine Surgery, Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Liang Wang
- Department of Orthopedics, Academy of Orthopedics, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
43
|
Hertel FC, da Silva AS, Sabino ADP, Valente FL, Reis ECC. Preconditioning Methods to Improve Mesenchymal Stromal Cell-Derived Extracellular Vesicles in Bone Regeneration—A Systematic Review. BIOLOGY 2022; 11:biology11050733. [PMID: 35625461 PMCID: PMC9138769 DOI: 10.3390/biology11050733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 04/20/2022] [Accepted: 05/07/2022] [Indexed: 12/09/2022]
Abstract
Simple Summary The evidence of the therapeutic effects of mesenchymal stromal cells (MSCs), so-called stem cells, in several diseases relies mostly on the substances they secrete, including their extracellular vesicles (EVs). EVs are an important component of cell communication and they carry a cargo that is similar to their parent cell. Cells respond differently based on their microenvironment, and so it is expected that the therapeutic potential of these vesicles can be modulated by the enrichment of their parent cell microenvironment. With this in mind, we conducted a systematic search for papers that preconditioned MSCs and collected their EVs to assess their potential to favor bone formation. The results showed different methods for MSC preconditioning, including chemical induction, culture conditions, and genetic modifications. All methods were able to improve the therapeutic effects of the derived EVs for bone formation. However, the heterogeneity among studies—regarding the type of cell, EV concentration, and scaffolds—made it difficult to compare fairly the types of preconditioning methods. In summary, the microenvironment greatly influences MSCs, and using preconditioning methods can potentially improve the therapeutic effects of their derived EVs in bone regeneration and other bone diseases. Abstract Mesenchymal stromal cells (MSCs) have long been used in research for bone regeneration, with evidence of their beneficial properties. In the segmental area of MSC-based therapies, MSC-derived extracellular vesicles (EVs) have also shown great therapeutic effects in several diseases, including bone healing. This study aimed to assess whether the conditioning of MSCs improves the therapeutic effects of their derived extracellular vesicles for bone regeneration. Electronic research was performed until February 2021 to recover the studies in the following databases: PubMed, Scopus, and Web of Science. The studies were screened based on the inclusion criteria. Relevant information was extracted, including in vitro and in vivo experiments, and the animal studies were evaluated for risk of bias by the SYRCLE tool. A total of 463 studies were retrieved, and 18 studies met the inclusion criteria (10 studies for their in vitro analysis, and 8 studies for their in vitro and in vivo analysis). The conditioning methods reported included: osteogenic medium; dimethyloxalylglycine; dexamethasone; strontium-substituted calcium silicate; hypoxia; 3D mechanical microenvironment; and the overexpression of miR-375, bone morphogenetic protein-2, and mutant hypoxia-inducible factor-1α. The conditioning methods of MSCs in the reported studies generate exosomes able to significantly promote bone regeneration. However, heterogeneity regarding cell source, conditioning method, EV isolation and concentration, and defect model was observed among the studies. The different conditioning methods reported in this review do improve the therapeutic effects of MSC-derived EVs for bone regeneration, but they still need to be addressed in larger animal models for further clinical application.
Collapse
Affiliation(s)
- Fernanda Campos Hertel
- Veterinary Department, Federal University of Viçosa, Vicosa 36570-900, Brazil; (F.C.H.); (A.S.d.S.); (F.L.V.)
| | - Aline Silvestrini da Silva
- Veterinary Department, Federal University of Viçosa, Vicosa 36570-900, Brazil; (F.C.H.); (A.S.d.S.); (F.L.V.)
| | - Adriano de Paula Sabino
- Department of Clinical and Toxicological Analysis, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil;
| | - Fabrício Luciani Valente
- Veterinary Department, Federal University of Viçosa, Vicosa 36570-900, Brazil; (F.C.H.); (A.S.d.S.); (F.L.V.)
| | - Emily Correna Carlo Reis
- Veterinary Department, Federal University of Viçosa, Vicosa 36570-900, Brazil; (F.C.H.); (A.S.d.S.); (F.L.V.)
- Correspondence:
| |
Collapse
|
44
|
Li M, Jiang Y, Hou Q, Zhao Y, Zhong L, Fu X. Potential pre-activation strategies for improving therapeutic efficacy of mesenchymal stem cells: current status and future prospects. Stem Cell Res Ther 2022; 13:146. [PMID: 35379361 PMCID: PMC8981790 DOI: 10.1186/s13287-022-02822-2] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 03/20/2022] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cell (MSC)-based therapy has been considered as a promising approach targeting a variety of intractable diseases due to remarkable multiple effect of MSCs, such as multilineage differentiation, immunomodulatory property, and pro-regenerative capacity. However, poor engraftment, low survival rate of transplanted MSC, and impaired donor-MSC potency under host age/disease result in unsatisfactory therapeutic outcomes. Enhancement strategies, including genetic manipulation, pre-activation, and modification of culture method, have been investigated to generate highly functional MSC, and approaches for MSC pre-activation are highlighted. In this review, we summarized the current approaches of MSC pre-activation and further classified, analysed the scientific principles and main characteristics of these manipulations, and described the pros and cons of individual pre-activation strategies. We also discuss the specialized tactics to solve the challenges in this promising field so that it improves MSC therapeutic functions to serve patients better.
Collapse
Affiliation(s)
- Meirong Li
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Division and 4th Medical Center, PLA General Hospital and PLA Medical College, Beijing, China. .,PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, Beijing, China. .,Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences 2019RU051, Beijing, China.
| | - Yufeng Jiang
- Wound Repairing Department, PLA Strategic Support Force Characteristic Medical Center, Beijing, 100101, China
| | - Qian Hou
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Division and 4th Medical Center, PLA General Hospital and PLA Medical College, Beijing, China.,PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, Beijing, China.,Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences 2019RU051, Beijing, China
| | - Yali Zhao
- Central Laboratory, Trauma Treatment Center, Chinese PLA General Hospital, Hainan Hospital, Sanya, China
| | - Lingzhi Zhong
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Division and 4th Medical Center, PLA General Hospital and PLA Medical College, Beijing, China.,PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, Beijing, China.,Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences 2019RU051, Beijing, China
| | - Xiaobing Fu
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Division and 4th Medical Center, PLA General Hospital and PLA Medical College, Beijing, China. .,PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, Beijing, China. .,Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences 2019RU051, Beijing, China.
| |
Collapse
|
45
|
Uberti B, Plaza A, Henríquez C. Pre-conditioning Strategies for Mesenchymal Stromal/Stem Cells in Inflammatory Conditions of Livestock Species. Front Vet Sci 2022; 9:806069. [PMID: 35372550 PMCID: PMC8974404 DOI: 10.3389/fvets.2022.806069] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 02/16/2022] [Indexed: 12/20/2022] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) therapy has been a cornerstone of regenerative medicine in humans and animals since their identification in 1968. MSCs can interact and modulate the activity of practically all cellular components of the immune response, either through cell-cell contact or paracrine secretion of soluble mediators, which makes them an attractive alternative to conventional therapies for the treatment of chronic inflammatory and immune-mediated diseases. Many of the mechanisms described as necessary for MSCs to modulate the immune/inflammatory response appear to be dependent on the animal species and source. Although there is evidence demonstrating an in vitro immunomodulatory effect of MSCs, there are disparate results between the beneficial effect of MSCs in preclinical models and their actual use in clinical diseases. This discordance might be due to cells' limited survival or impaired function in the inflammatory environment after transplantation. This limited efficacy may be due to several factors, including the small amount of MSCs inoculated, MSC administration late in the course of the disease, low MSC survival rates in vivo, cryopreservation and thawing effects, and impaired MSC potency/biological activity. Multiple physical and chemical pre-conditioning strategies can enhance the survival rate and potency of MSCs; this paper focuses on hypoxic conditions, with inflammatory cytokines, or with different pattern recognition receptor ligands. These different pre-conditioning strategies can modify MSCs metabolism, gene expression, proliferation, and survivability after transplantation.
Collapse
Affiliation(s)
- Benjamin Uberti
- Instituto de Ciencias Clínicas, Facultad de Ciencias Veterinarias, Universidad Austral de Chile, Valdivia, Chile
| | - Anita Plaza
- Instituto de Patología Animal, Facultad de Ciencias Veterinarias, Universidad Austral de Chile, Valdivia, Chile
| | - Claudio Henríquez
- Instituto de Farmacología y Morfofisiología, Facultad de Ciencias Veterinarias, Universidad Austral de Chile, Valdivia, Chile
- *Correspondence: Claudio Henríquez
| |
Collapse
|
46
|
New Perspectives to Improve Mesenchymal Stem Cell Therapies for Drug-Induced Liver Injury. Int J Mol Sci 2022; 23:ijms23052669. [PMID: 35269830 PMCID: PMC8910533 DOI: 10.3390/ijms23052669] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 02/23/2022] [Accepted: 02/24/2022] [Indexed: 02/06/2023] Open
Abstract
Drug-induced liver injury (DILI) is one of the leading causes of acute liver injury. Many factors may contribute to the susceptibility of patients to this condition, making DILI a global medical problem that has an impact on public health and the pharmaceutical industry. The use of mesenchymal stem cells (MSCs) has been at the forefront of regenerative medicine therapies for many years, including MSCs for the treatment of liver diseases. However, there is currently a huge gap between these experimental approaches and their application in clinical practice. In this concise review, we focus on the pathophysiology of DILI and highlight new experimental approaches conceived to improve cell-based therapy by the in vitro preconditioning of MSCs and/or the use of cell-free products as treatment for this liver condition. Finally, we discuss the advantages of new approaches, but also the current challenges that must be addressed in order to develop safer and more effective procedures that will allow cell-based therapies to reach clinical practice, enhancing the quality of life and prolonging the survival time of patients with DILI.
Collapse
|
47
|
Pochon C, Notarantonio AB, Laroye C, Reppel L, Bensoussan D, Bertrand A, Rubio MT, D'Aveni M. Wharton's jelly-derived stromal cells and their cell therapy applications in allogeneic haematopoietic stem cell transplantation. J Cell Mol Med 2022; 26:1339-1350. [PMID: 35088933 PMCID: PMC8899189 DOI: 10.1111/jcmm.17105] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 10/30/2021] [Accepted: 11/23/2021] [Indexed: 12/16/2022] Open
Abstract
For decades, mesenchymal stromal cells (MSCs) have been of great interest in the fields of regenerative medicine, tissue engineering and immunomodulation. Their tremendous potential makes it desirable to cryopreserve and bank MSCs to increase their accessibility and availability. Postnatally derived MSCs seem to be of particular interest because they are harvested after delivery without ethical controversy, they have the capacity to expand at a higher rate than adult‐derived MSCs, in which expansion decreases with ageing, and they have demonstrated immunological and haematological supportive properties similar to those of adult‐derived MSCs. In this review, we focus on MSCs obtained from Wharton's jelly (the mucous connective tissue of the umbilical cord between the amniotic epithelium and the umbilical vessels). Wharton's jelly MSCs (WJ‐MSCs) are a good candidate for cellular therapy in haematology, with accumulating data supporting their potential to sustain haematopoietic stem cell engraftment and to modulate alloreactivity such as Graft Versus Host Disease (GVHD). We first present an overview of their in‐vitro properties and the results of preclinical murine models confirming the suitability of WJ‐MSCs for cellular therapy in haematology. Next, we focus on clinical trials and discuss tolerance, efficacy and infusion protocols reported in haematology for GVHD and engraftment.
Collapse
Affiliation(s)
- Cécile Pochon
- Pediatric Oncohematology Department, CHRU Nancy, Université de Lorraine, Nancy, France.,UMR 7365 CNRS, IMoPA, Université de Lorraine, Nancy, France
| | - Anne-Béatrice Notarantonio
- UMR 7365 CNRS, IMoPA, Université de Lorraine, Nancy, France.,Hematology Department, CHRU Nancy, Université de Lorraine, Nancy, France
| | - Caroline Laroye
- Pediatric Oncohematology Department, CHRU Nancy, Université de Lorraine, Nancy, France.,Cell Therapy Unit, CHRU Nancy, Université de Lorraine, Nancy, France
| | - Loic Reppel
- UMR 7365 CNRS, IMoPA, Université de Lorraine, Nancy, France.,Cell Therapy Unit, CHRU Nancy, Université de Lorraine, Nancy, France
| | - Danièle Bensoussan
- UMR 7365 CNRS, IMoPA, Université de Lorraine, Nancy, France.,Cell Therapy Unit, CHRU Nancy, Université de Lorraine, Nancy, France
| | - Allan Bertrand
- UMR 7365 CNRS, IMoPA, Université de Lorraine, Nancy, France
| | - Marie-Thérèse Rubio
- UMR 7365 CNRS, IMoPA, Université de Lorraine, Nancy, France.,Hematology Department, CHRU Nancy, Université de Lorraine, Nancy, France
| | - Maud D'Aveni
- UMR 7365 CNRS, IMoPA, Université de Lorraine, Nancy, France.,Hematology Department, CHRU Nancy, Université de Lorraine, Nancy, France
| |
Collapse
|
48
|
Russo V, El Khatib M, Prencipe G, Citeroni MR, Faydaver M, Mauro A, Berardinelli P, Cerveró-Varona A, Haidar-Montes AA, Turriani M, Di Giacinto O, Raspa M, Scavizzi F, Bonaventura F, Stöckl J, Barboni B. Tendon Immune Regeneration: Insights on the Synergetic Role of Stem and Immune Cells during Tendon Regeneration. Cells 2022; 11:434. [PMID: 35159244 PMCID: PMC8834336 DOI: 10.3390/cells11030434] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/19/2022] [Accepted: 01/25/2022] [Indexed: 12/11/2022] Open
Abstract
Tendon disorders represent a very common pathology in today's population, and tendinopathies that account 30% of tendon-related injuries, affect yearly millions of people which in turn cause huge socioeconomic and health repercussions worldwide. Inflammation plays a prominent role in the development of tendon pathologies, and advances in understanding the underlying mechanisms during the inflammatory state have provided additional insights into its potential role in tendon disorders. Different cell compartments, in combination with secreted immune modulators, have shown to control and modulate the inflammatory response during tendinopathies. Stromal compartment represented by tenocytes has shown to display an important role in orchestrating the inflammatory response during tendon injuries due to the interplay they exhibit with the immune-sensing and infiltrating compartments, which belong to resident and recruited immune cells. The use of stem cells or their derived secretomes within the regenerative medicine field might represent synergic new therapeutical approaches that can be used to tune the reaction of immune cells within the damaged tissues. To this end, promising opportunities are headed to the stimulation of macrophages polarization towards anti-inflammatory phenotype together with the recruitment of stem cells, that possess immunomodulatory properties, able to infiltrate within the damaged tissues and improve tendinopathies resolution. Indeed, the comprehension of the interactions between tenocytes or stem cells with the immune cells might considerably modulate the immune reaction solving hence the inflammatory response and preventing fibrotic tissue formation. The purpose of this review is to compare the roles of distinct cell compartments during tendon homeostasis and injury. Furthermore, the role of immune cells in this field, as well as their interactions with stem cells and tenocytes during tendon regeneration, will be discussed to gain insights into new ways for dealing with tendinopathies.
Collapse
Affiliation(s)
- Valentina Russo
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (M.R.C.); (M.F.); (A.M.); (P.B.); (A.C.-V.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Mohammad El Khatib
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (M.R.C.); (M.F.); (A.M.); (P.B.); (A.C.-V.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Giuseppe Prencipe
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (M.R.C.); (M.F.); (A.M.); (P.B.); (A.C.-V.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Maria Rita Citeroni
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (M.R.C.); (M.F.); (A.M.); (P.B.); (A.C.-V.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Melisa Faydaver
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (M.R.C.); (M.F.); (A.M.); (P.B.); (A.C.-V.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Annunziata Mauro
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (M.R.C.); (M.F.); (A.M.); (P.B.); (A.C.-V.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Paolo Berardinelli
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (M.R.C.); (M.F.); (A.M.); (P.B.); (A.C.-V.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Adrián Cerveró-Varona
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (M.R.C.); (M.F.); (A.M.); (P.B.); (A.C.-V.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Arlette A. Haidar-Montes
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (M.R.C.); (M.F.); (A.M.); (P.B.); (A.C.-V.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Maura Turriani
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (M.R.C.); (M.F.); (A.M.); (P.B.); (A.C.-V.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Oriana Di Giacinto
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (M.R.C.); (M.F.); (A.M.); (P.B.); (A.C.-V.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Marcello Raspa
- National Research Council (CNR), Campus International Development (EMMA-INFRAFRONTIER-IMPC), Institute of Biochemistry and Cellular Biology (IBBC), 00015 Monterotondo Scalo, Italy; (M.R.); (F.S.); (F.B.)
| | - Ferdinando Scavizzi
- National Research Council (CNR), Campus International Development (EMMA-INFRAFRONTIER-IMPC), Institute of Biochemistry and Cellular Biology (IBBC), 00015 Monterotondo Scalo, Italy; (M.R.); (F.S.); (F.B.)
| | - Fabrizio Bonaventura
- National Research Council (CNR), Campus International Development (EMMA-INFRAFRONTIER-IMPC), Institute of Biochemistry and Cellular Biology (IBBC), 00015 Monterotondo Scalo, Italy; (M.R.); (F.S.); (F.B.)
| | - Johannes Stöckl
- Centre for Pathophysiology, Infectiology and Immunology, Institute of Immunology, Medical University of Vienna, 1090 Vienna, Austria;
| | - Barbara Barboni
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (M.R.C.); (M.F.); (A.M.); (P.B.); (A.C.-V.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| |
Collapse
|
49
|
Zhang Y, Zhang X, Zhang H, Song P, Pan W, Xu P, Wang G, Hu P, Wang Z, Huang K, Zhang X, Wang H, Zhang J. Mesenchymal Stem Cells Derived Extracellular Vesicles Alleviate Traumatic Hemorrhagic Shock Induced Hepatic Injury via IL-10/PTPN22-Mediated M2 Kupffer Cell Polarization. Front Immunol 2022; 12:811164. [PMID: 35095903 PMCID: PMC8790700 DOI: 10.3389/fimmu.2021.811164] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 12/20/2021] [Indexed: 12/19/2022] Open
Abstract
Traumatic hemorrhagic shock (THS) is a major cause of mortality and morbidity worldwide in severely injured patients. Mesenchymal stem cells (MSCs) possess immunomodulatory properties and tissue repair potential mainly through a paracrine pathway mediated by MSC-derived extracellular vesicles (MSC-EVs). Interleukin 10 (IL-10) is a potent anti-inflammatory cytokine that plays a crucial role during the inflammatory response, with a broad range of effects on innate and adaptive immunity, preventing damage to the host and maintaining normal tissue homeostasis. However, the function and mechanism of IL-10 in MSC-mediated protective effect in THS remain obscure. Here, we show that MSCs significantly attenuate hepatic injury and inflammation from THS in mice. Notably, these beneficial effects of MSCs disappeared when IL-10 was knocked out in EVs or when recombinant IL-10 was administered to mice. Mechanistically, MSC-EVs function to carry and deliver IL-10 as cargo. WT MSC-EVs restored the function of IL-10 KO MSCs during THS injury. We further demonstrated that EVs containing IL-10 mainly accumulated in the liver during THS, where they were captured by Kupffer cells and induced the expression of PTPN22. These effects subsequently shifted Kupffer cells to an anti-inflammatory phenotype and mitigated liver inflammation and injury. Therefore, our study indicates that MSC-EVs containing IL-10 alleviate THS-induced hepatic injury and may serve as a cell-free therapeutic approach for THS.
Collapse
Affiliation(s)
- Yunwei Zhang
- Department of Emergency, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaofei Zhang
- Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongji Zhang
- Department of Surgery, The Ohio State University, Columbus, OH, United States
| | - Peng Song
- Department of Breast and Thyroid Surgery, Ningxia hui Autonomous Region People’s Hospital, Yinchuan, China
| | - Wenming Pan
- Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Peng Xu
- Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guoliang Wang
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ping Hu
- Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zixuan Wang
- Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kunpeng Huang
- Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaodong Zhang
- College of Life Sciences, Wuhan University, Wuhan, China
| | - Hui Wang
- Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Medical Genetics, Basic School of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jinxiang Zhang
- Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
50
|
Ekram S, Khalid S, Salim A, Khan I. Regulating the fate of stem cells for regenerating the intervertebral disc degeneration. World J Stem Cells 2021; 13:1881-1904. [PMID: 35069988 PMCID: PMC8727226 DOI: 10.4252/wjsc.v13.i12.1881] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/12/2021] [Accepted: 11/15/2021] [Indexed: 02/06/2023] Open
Abstract
Lower back pain is a leading cause of disability and is one of the reasons for the substantial socioeconomic burden. The etiology of intervertebral disc (IVD) degeneration is complicated, and its mechanism is still not completely understood. Factors such as aging, systemic inflammation, biochemical mediators, toxic environmental factors, physical injuries, and genetic factors are involved in the progression of its pathophysiology. Currently, no therapy for restoring degenerated IVD is available except pain management, reduced physical activities, and surgical intervention. Therefore, it is imperative to establish regenerative medicine-based approaches to heal and repair the injured disc, repopulate the cell types to retain water content, synthesize extracellular matrix, and strengthen the disc to restore normal spine flexion. Cellular therapy has gained attention for IVD management as an alternative therapeutic option. In this review, we present an overview of the anatomical and molecular structure and the surrounding pathophysiology of the IVD. Modern therapeutic approaches, including proteins and growth factors, cellular and gene therapy, and cell fate regulators are reviewed. Similarly, small molecules that modulate the fate of stem cells for their differentiation into chondrocytes and notochordal cell types are highlighted.
Collapse
Affiliation(s)
- Sobia Ekram
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Sindh, Pakistan
| | - Shumaila Khalid
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Sindh, Pakistan
| | - Asmat Salim
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Sindh, Pakistan
| | - Irfan Khan
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Sindh, Pakistan.
| |
Collapse
|