1
|
Kumar A, Ramesh S, Kumar V, Mathews JE, Madhuri V. Human second-trimester fetal liver-derived mesenchymal stromal cells are more effective than adult bone marrow MSCs for their superior growth kinetics, immunomodulatory, and osteogenic potential. Tissue Cell 2025; 95:102859. [PMID: 40101501 DOI: 10.1016/j.tice.2025.102859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 03/06/2025] [Accepted: 03/11/2025] [Indexed: 03/20/2025]
Abstract
BACKGROUND Mesenchymal stromal cells (MSCs) are promising candidates for cell therapy. Most of the therapeutic applications have used adult bone marrow MSCs, adipose MSCs and perinatal tissue-derived MSCs. Recent evidence suggests that MSCs from mid-gestational fetal tissues are more primitive, grow faster and are biologically more closely related to embryonic stem cells than other sources of MSCs. However, the expression of pluripotency genes raises the question of whether these genes are safe for clinical application. In this study, we demonstrated that second-trimester fetal liver-derived MSCs lack the expression of pluripotent markers and maintain their proliferative and osteogenic differentiation potential beyond passage 12. Compared to other sources, FL-MSCs exhibit characteristics that are promising for use in skeletal regeneration. METHODS MSCs were isolated from the second-trimester fetal liver and characterized for surface antigen expression, pluripotency marker expression and multilineage differentiation. The growth kinetics, population doubling, and number of colony-forming units were analyzed at the 3rd, 5th, 8th and 10th passages of FLMSCs and compared with those of BMMSCs. The immunomodulatory properties of FLMSCs were analyzed by a T-cell proliferation assay. The osteogenic differentiation potential of FL-MSCs was assessed at passages 3, 5, 8 and 12 and compared with that of BMMSCs. RESULTS We demonstrated that second-trimester fetal liver-derived MSCs exhibited a distinct fibroblast-like spindle-shaped morphology and expressed typical MSC surface antigens. Unlike first-trimester fetal MSCs, second-trimester FL-MSCs did not express pluripotent markers and showed significantly greater self-renewal and proliferative potential at higher passages and a lower apoptotic rate than BM-MSCs. Additionally, the osteogenic differentiation potential of FL-MSCs was 4-6 times greater than that of BM-MSCs at both early and late passages CONCLUSION: Our findings underscore the robust self-renewal and proliferative potential of second-trimester fetal liver-derived MSCs, which notably lack pluripotent markers. The ability of FL-MSCs to sustain osteogenic potential through multiple passages makes them promising candidates for bone tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Ashis Kumar
- Centre for Stem Cell Research, a unit of inStem, Bengaluru, Christian Medical College, Vellore, India; Department of Paediatric Orthopedics, Christian Medical College, Vellore, India; Sree Chitra Tirunal Institute for Medical Sciences & Technology, Thiruvananthapuram, Kerala 695011, India
| | - Sowmya Ramesh
- Centre for Stem Cell Research, a unit of inStem, Bengaluru, Christian Medical College, Vellore, India; Department of Paediatric Orthopedics, Christian Medical College, Vellore, India
| | - Vignesh Kumar
- Centre for Stem Cell Research, a unit of inStem, Bengaluru, Christian Medical College, Vellore, India; Department of Paediatric Orthopedics, Christian Medical College, Vellore, India
| | | | - Vrisha Madhuri
- Centre for Stem Cell Research, a unit of inStem, Bengaluru, Christian Medical College, Vellore, India; Department of Paediatric Orthopedics, Christian Medical College, Vellore, India; Sree Chitra Tirunal Institute for Medical Sciences & Technology, Thiruvananthapuram, Kerala 695011, India; Department of Orthopedics, Amara Hospital, Tirupati, Andhra Pradesh-517520, India.
| |
Collapse
|
2
|
Peng X, Li G, Zhao J, Liu H, Wu C, Su Z, Liu Z, Fan S, Chen Y, Wu Y, Liu W, Shen H, Zheng G. Promotion of quiescence and maintenance of function of mesenchymal stem cells on substrates with surface potential. Bioelectrochemistry 2025; 164:108920. [PMID: 39904300 DOI: 10.1016/j.bioelechem.2025.108920] [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: 10/16/2024] [Revised: 01/20/2025] [Accepted: 01/26/2025] [Indexed: 02/06/2025]
Abstract
The widespread use of human mesenchymal stem cells(hMSCs) is impeded by functional loss during prolonged expansion. Although multiple approaches have been attempted to preserve hMSCs stemness, a suitable culture system remains to be modified. The interaction between electrical signals and stem cells is expected to better maintain the function of stem cells. However, it remains unclear whether the surface potential of substrates has the potential to preserve stem cell function during in vitro expansion. In our study, hMSCs cultured on materials with different surface potentials could be induced into a reversible quiescent state, and we demonstrated that quiescent hMSCs could be reactivated and transitioned back into the proliferation cell cycle. hMSCs cultured under appropriate potential displayed superior differentiation and proliferation abilities within the same generation compared to conventional conditions. These findings underscore the importance of surface potential as a critical physical factor regulating hMSCs stemness. Manipulating the surface potential of hMSCs culture substrates holds promise for optimising preservation and culture conditions, thereby enhancing their application in tissue repair and regeneration engineering.
Collapse
Affiliation(s)
- Xiaoshuai Peng
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, 3025# Shennan Road, Shenzhen, 518000, PR China
| | - Guojian Li
- Department of Spine Orthopedics, Zhuhai People's Hospital, Zhuhai Hospital affiliated with Jinan University, Zhuhai 519000, PR China
| | - Jiu Zhao
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, 3025# Shennan Road, Shenzhen, 518000, PR China
| | - Huatao Liu
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, 3025# Shennan Road, Shenzhen, 518000, PR China
| | - Changhua Wu
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, 3025# Shennan Road, Shenzhen, 518000, PR China
| | - Zepeng Su
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, 3025# Shennan Road, Shenzhen, 518000, PR China
| | - Zhidong Liu
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, 3025# Shennan Road, Shenzhen, 518000, PR China
| | - Shuai Fan
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, 3025# Shennan Road, Shenzhen, 518000, PR China
| | - Yuanquan Chen
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, 3025# Shennan Road, Shenzhen, 518000, PR China
| | - Yanfeng Wu
- Center for Biotherapy, The Eighth Affiliated Hospital, Sun Yat-sen University, 3025# Shennan Road, Shenzhen 518000, PR China
| | - Wenjie Liu
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, 3025# Shennan Road, Shenzhen, 518000, PR China.
| | - Huiyong Shen
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, 3025# Shennan Road, Shenzhen, 518000, PR China.
| | - Guan Zheng
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, 3025# Shennan Road, Shenzhen, 518000, PR China.
| |
Collapse
|
3
|
Yu JL, Yang C, Liu L, Lin A, Guo SJ, Tian WD. Optimal good manufacturing practice-compliant production of dental follicle stem cell sheet and its application in Sprague-Dawley rat periodontitis. World J Stem Cells 2025; 17:104116. [DOI: 10.4252/wjsc.v17.i5.104116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 02/27/2025] [Accepted: 04/18/2025] [Indexed: 05/26/2025] Open
Abstract
BACKGROUND Dental follicle stem cell (DFSC) sheets demonstrate strong extracellular secretion capabilities and efficacy in periodontal regeneration. However, existing methods for producing DFSC sheets lack a comprehensive discussion on the most efficient and cost-effective approaches at the good manufacturing practice (GMP) level.
AIM To investigate the culture condition of GMP-compliant DFSC sheets and to compare the properties of DFSC sheets and cell suspensions.
METHODS This study explored the optimal conditions for culturing GMP-compliant DFSC sheets, focusing on four key factors: Cell passage, cell concentration, L-ascorbic acid content, and culture duration. We evaluated the characteristics of the cell sheets under varying culture conditions, including cell viability, cell count, appearance, osteogenesis, chondrogenesis, odontogenesis, aging, relative telomere length, and extracellular matrix secretion. A comparison was also made between the periodontal regeneration, osteogenesis, and paracrine capacity of cell sheets cultured under optimal conditions and those of the cell suspensions.
RESULTS The GMP-compliant DFSC sheets cultured from passage 4 cells exhibited the highest viability (≥ 99%, P < 0.05) and optimal osteogenic differentiation capacity (optical density ≥ 0.126, P < 0.05). When cultured for 10 days, DFSC sheets demonstrated maximal expression of osteogenic, chondrogenic and periostin genes [alkaline phosphatase, Runt-related transcription factor 2, collagen type I, osteopontin, cartilage associated protein, and PERIOSTN (P < 0.001); osteocalcin (P < 0.01)]. Concurrently, they showed the lowest senescent cell count (P < 0.01) with no progression to late-stage senescence. At a seeding density of 2500 cells/cm2, GMP-compliant DFSC sheets achieved better osteogenic differentiation (P < 0.01) and maximal osteogenic, chondrogenic and periostin gene expression (P < 0.001), coupled with the highest hydroxyproline secretion (P < 0.001) and moderate sulfated glycosaminoglycan production. No statistically significant difference in senescent cell count was observed compared to DFSC sheets at a seeding density of 5000 cells/cm2. Supplementation with 25 μg/mL L-ascorbic acid significantly enhanced osteogenic gene expression (P < 0.001) and elevated hydroxyproline (P < 0.01) and sulfated glycosaminoglycan secretion to high ranges. Compared with the cell suspension, the cell sheet demonstrated improved osteogenic, paracrine, and periodontal regenerative capacities in Sprague-Dawley rats. The optimized DFSC sheets demonstrated significantly higher levels of vascular endothelial growth factor and angiopoietin-1 (P < 0.001) compared to DFSC suspensions, along with enhanced osteogenic induction outcomes (optical density = 0.1333 ± 0.01270 vs 0.1007 ± 0.0005774 in suspensions, P < 0.05). Following implantation into the rat periodontal defect model, micro-computed tomography analysis revealed superior bone regeneration metrics in the cell sheet group compared to both the cell suspension group and control group (percent bone volume, trabecular thickness, trabecular number), while trabecular spacing exhibited an inverse pattern.
CONCLUSION Optimized DFSC sheets cultured under the identified conditions outperform DFSC suspensions. This study contributes to the industrial-scale production of DFSC sheets and establishes a foundation for cell therapy applications.
Collapse
Affiliation(s)
- Jia-Lu Yu
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Chao Yang
- Department of Product Development, Chengdu Shiliankangjian Biotechnology Co., Ltd, Chengdu 610041, Sichuan Province, China
| | - Li Liu
- Engineering Research Center of Oral Translational Medicine, National Clinical Research Center for Oral Diseases, Departments of 5 Periodontics and 6 Oral and Maxillofacial Surgery, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - An Lin
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Shu-Juan Guo
- Department of Periodontics, West China School of Stomatology, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Wei-Dong Tian
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Engineering Research Center of Oral Translational Medicine, National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan Province, China
| |
Collapse
|
4
|
Souza ATP, Freitas GP, Lopes HB, Weffort D, Adolpho LF, Gomes MPO, Oliveira FS, Almeida ALG, Beloti MM, Rosa AL. Diabetes Mellitus Impairs the Bone Regeneration Capacity of Mesenchymal Stromal Cell-Based Therapy. Arch Med Res 2025; 56:103234. [PMID: 40398343 DOI: 10.1016/j.arcmed.2025.103234] [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: 01/29/2025] [Revised: 03/15/2025] [Accepted: 04/23/2025] [Indexed: 05/23/2025]
Abstract
BACKGROUND Diabetes mellitus (DM) negatively impacts bone tissue, leading to bone loss and increased fracture risk. Many individuals need additional treatments, and therapy based on mesenchymal stromal cells (MSCs) represents a promising treatment for bone defects in patients with diabetes. AIMS The present study explored the effects of interactions between MSCs from normoglycemic (NG-MSCs) and diabetic (DM-MSCs) donors on osteoblast differentiation and the effects of cell therapy using NG-MSCs on bone regeneration in defects created in diabetic rats. METHODS After inducing DM with streptozotocin, we evaluated the morphometric parameters of rat femurs and the osteoblast differentiation of MSCs, as well as the effects of the interaction between NG-MSCs and DM-MSCs on their osteoblast differentiation. The efficacy of cell therapy was measured by evaluating the bone repair in calvarial defects of diabetic rats treated with local injections of either NG-MSCs or a vehicle. RESULTS DM induced bone loss and impaired the osteoblast differentiation of MSCs, which was partially restored by NG-MSCs, while the bone formation observed in defects treated with NG-MSCs and the vehicle was similar. CONCLUSION These results indicate that the beneficial effect of NG-MSCs on DM-MSCs did not translate into enhanced bone repair, mainly due to a hostile environment created by hyperglycemia, which compromised the ability of MSCs to induce bone formation.
Collapse
Affiliation(s)
- Alann Thaffarell Portilho Souza
- Bone Research Lab, Ribeirão Preto School of Dentistry, University of São Paulo, Ribeirão Preto, SP, Brazil; School of Dentistry, Metropolitan University Center of the Amazon, Belém, Brazil; Postgraduate Dentistry Program, CEUMA University, São Luís, Brazil
| | - Gileade Pereira Freitas
- Bone Research Lab, Ribeirão Preto School of Dentistry, University of São Paulo, Ribeirão Preto, SP, Brazil; School of Dentistry, Federal University of Goiás, Goiânia, Brazil
| | - Helena Bacha Lopes
- Bone Research Lab, Ribeirão Preto School of Dentistry, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Denise Weffort
- Bone Research Lab, Ribeirão Preto School of Dentistry, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Leticia Faustino Adolpho
- Bone Research Lab, Ribeirão Preto School of Dentistry, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Maria Paula Oliveira Gomes
- Bone Research Lab, Ribeirão Preto School of Dentistry, University of São Paulo, Ribeirão Preto, SP, Brazil
| | | | | | - Marcio Mateus Beloti
- Bone Research Lab, Ribeirão Preto School of Dentistry, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Adalberto Luiz Rosa
- Bone Research Lab, Ribeirão Preto School of Dentistry, University of São Paulo, Ribeirão Preto, SP, Brazil.
| |
Collapse
|
5
|
Wu J, Ge Y, Huang W, Zhang L, Huang J, Huang N, Luo Y. Natural bioactive compounds modified with mesenchymal stem cells: new hope for regenerative medicine. Front Bioeng Biotechnol 2025; 13:1446537. [PMID: 40416310 PMCID: PMC12098461 DOI: 10.3389/fbioe.2025.1446537] [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: 06/10/2024] [Accepted: 04/25/2025] [Indexed: 05/27/2025] Open
Abstract
Mesenchymal stem cells (MSCs) have the potential to differentiate into various cell types, providing important sources of cells for the development of regenerative medicine. Although MSCs have various advantages, there are also various problems, such as the low survival rate of transplanted cells and poor migration and homing; therefore, determining how to reform MSCs to improve their utilization is particularly important. Although many natural bioactive compounds have shown great potential for improving MSCs, many mechanisms and pathways are involved; however, in the final analysis, natural bioactive compounds promoted MSC proliferation, migration and homing and promoted differentiation and antiaging. This article reviews the regulatory effects of natural bioactive compounds on MSCs to provide new ideas for the therapeutic effects of modified MSCs on diseases.
Collapse
Affiliation(s)
- Jingjing Wu
- Department of Neurology, Third Affiliated Hospital of Zunyi Medical University (The First People’s Hospital of Zunyi), Zunyi, Guizhou, China
| | - Ying Ge
- Department of Neurology, Third Affiliated Hospital of Zunyi Medical University (The First People’s Hospital of Zunyi), Zunyi, Guizhou, China
| | - Wendi Huang
- Department of Neurology, Third Affiliated Hospital of Zunyi Medical University (The First People’s Hospital of Zunyi), Zunyi, Guizhou, China
| | - Li Zhang
- Department of Neurology, Third Affiliated Hospital of Zunyi Medical University (The First People’s Hospital of Zunyi), Zunyi, Guizhou, China
| | - Juan Huang
- Key Laboratory of Basic Pharmacology and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China
| | - Nanqu Huang
- National Drug Clinical Trial Institution, Third Affiliated Hospital of Zunyi Medical University (The First People’s Hospital of Zunyi), Zunyi, Guizhou, China
- Department of Gerontology, Third Affiliated Hospital of Zunyi Medical University (The First People’s Hospital of Zunyi), Zunyi, Guizhou, China
| | - Yong Luo
- Department of Neurology, Third Affiliated Hospital of Zunyi Medical University (The First People’s Hospital of Zunyi), Zunyi, Guizhou, China
- Department of Gerontology, Third Affiliated Hospital of Zunyi Medical University (The First People’s Hospital of Zunyi), Zunyi, Guizhou, China
| |
Collapse
|
6
|
Hirono K, Hayashi Y, A Udugama I, Gaddem MR, Tanaka K, Takemoto Y, Kato R, Kino-Oka M, Sugiyama H. Determination and validation of design space for mesenchymal stem cell cultivation processes using prediction intervals. Commun Biol 2025; 8:657. [PMID: 40341300 PMCID: PMC12062477 DOI: 10.1038/s42003-025-08063-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 04/09/2025] [Indexed: 05/10/2025] Open
Abstract
In regenerative medicine, mesenchymal stem cells (MSCs) constitute a promising therapeutic route for many diseases. The current quality-by-design guidelines do not clearly define a framework for MSC production. Here, we suggest and experimentally validate a model-based method to determine design spaces (DSs) for MSC cultivation. A kinetic model used in previous work was employed; part of the experimental data was used to re-estimate the maximum specific growth rate in the kinetic model and then calculate the prediction intervals of this parameter. Subsequently, regions of seeding density and harvesting time where both the upper and lower limits of growth predictions met the acceptable number of cells and confluency with given risk levels were defined as DSs. Finally, the established DS was validated with the remaining data; it allowed better predictions of the cell numbers and confluency under specific cultivation conditions and improved the overall robustness of MSC cultivation processes.
Collapse
Affiliation(s)
- Keita Hirono
- Department of Chemical System Engineering, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Yusuke Hayashi
- Department of Chemical System Engineering, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Isuru A Udugama
- Department of Chemical System Engineering, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Mohamed Rami Gaddem
- Department of Chemical System Engineering, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Kenjiro Tanaka
- Department of Basic Medicinal Sciences, Graduate School of Pharmaceutical Sciences, Nagoya University, Tokai National Higher Education and Research System, Nagoya, Aichi, Japan
| | - Yuto Takemoto
- Department of Basic Medicinal Sciences, Graduate School of Pharmaceutical Sciences, Nagoya University, Tokai National Higher Education and Research System, Nagoya, Aichi, Japan
| | - Ryuji Kato
- Department of Basic Medicinal Sciences, Graduate School of Pharmaceutical Sciences, Nagoya University, Tokai National Higher Education and Research System, Nagoya, Aichi, Japan
- Institute of Nano-Life-Systems, Institutes of Innovation for Future Society, Nagoya University, Tokai National Higher Education and Research System, Nagoya, Aichi, Japan
| | - Masahiro Kino-Oka
- Department of Biotechnology, The University of Osaka, Suita, Osaka, Japan
| | - Hirokazu Sugiyama
- Department of Chemical System Engineering, The University of Tokyo, Bunkyo-ku, Tokyo, Japan.
| |
Collapse
|
7
|
Westhauser F, Jacobsen V, Zheng K, Merle C, Boccaccini AR, Renkawitz T, Kunisch E. Insights into ionic medicine: Cerium reduces the presence of reactive oxygen species and favors osteogenic over adipogenic differentiation in human mesenchymal stromal cells. J Trace Elem Med Biol 2025; 89:127668. [PMID: 40345102 DOI: 10.1016/j.jtemb.2025.127668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Revised: 04/21/2025] [Accepted: 04/30/2025] [Indexed: 05/11/2025]
Abstract
The guided application of metallic ions in bone tissue engineering (BTE) has recently gained popularity being described as one important example of ionic medicine (IM). BTE aims to enhance osteogenic differentiation of precursor cells like bone-marrow-derived mesenchymal stromal cells (BMSCs) and, by that, regenerate bone tissue. BMSCs however can also differentiate into adipogenic lineage. It is known that elevated levels of reactive oxygen species (ROS) stimulate BMSC towards (undesired) adipogenic differentiation. One ion, that is particularly interesting for application in IM-guided BTE is cerium (Ce) since it acts as a self-regenerating ROS-scavenger and has already been successfully incorporated in biomaterials. Ce has demonstrated pro-osteogenic, anti-adipogenic and anti-oxidative effects before, however, so far, there is no direct comparative study available that analyzes these effects on human BMSCs in one and the same setting. Therefore, in this study, the influence of Ce nitrate (CeN) on the expression of osteogenic, adipogenic and ROS-scavenging genes in BMSCs was evaluated as well as its impact on formation of an osseous extracellular matrix (ECM), lipid formation and physical ROS presence. The presence of CeN improved BMSCs viability, enhanced proliferation, and reduced ROS-levels. Furthermore, CeN suppressed adipogenesis while osteogenic differentiation and the formation and maturation of the ECM were enhanced. The presence of CeN reduced the physical presence of ROS and the gene expression patterns shifted towards an anti-oxidant profile. Ce therefore constitutes an attractive ion for application in IM-guided BTE. Further research is necessary to clarify the biological mechanisms and pathways that are involved in the Ce-mediated modulation of BMSC differentiation.
Collapse
Affiliation(s)
- F Westhauser
- Department of Orthopaedics, Regensburg University Medical Center, Asklepios Klinikum Bad Abbach, Kaiser-Karl V.-Allee 3, Bad Abbach 93077, Germany; Department of Orthopedics, Heidelberg University Hospital, Schlierbacher Landstraße 200a, Heidelberg 69118, Germany.
| | - V Jacobsen
- Department of Orthopedics, Heidelberg University Hospital, Schlierbacher Landstraße 200a, Heidelberg 69118, Germany
| | - K Zheng
- Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, 136 Hanzhong Rd., Nanjing 210029, China
| | - C Merle
- Joint Replacement Centre, Orthopedic Surgery Paulinenhilfe, Diakonie-Klinikum Stuttgart, Rosenbergstraße 38, Stuttgart 70176, Germany
| | - A R Boccaccini
- Institute of Biomaterials, University of Erlangen-Nuremberg, Cauerstraße 6, Erlangen 91058, Germany
| | - T Renkawitz
- Department of Orthopaedics, Regensburg University Medical Center, Asklepios Klinikum Bad Abbach, Kaiser-Karl V.-Allee 3, Bad Abbach 93077, Germany; Department of Orthopedics, Heidelberg University Hospital, Schlierbacher Landstraße 200a, Heidelberg 69118, Germany
| | - E Kunisch
- Department of Orthopedics, Heidelberg University Hospital, Schlierbacher Landstraße 200a, Heidelberg 69118, Germany
| |
Collapse
|
8
|
Rossello-Gelabert M, Igartua M, Santos-Vizcaino E, Hernandez RM. Fine-tuning licensing strategies to boost MSC-based immunomodulatory secretome. Stem Cell Res Ther 2025; 16:183. [PMID: 40247371 PMCID: PMC12004826 DOI: 10.1186/s13287-025-04315-4] [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: 02/22/2025] [Accepted: 04/04/2025] [Indexed: 04/19/2025] Open
Abstract
BACKGROUND Immune-mediated inflammatory diseases (IMIDs) are a major global health challenge, affecting millions of people and often lacking effective treatments. The mesenchymal stromal cell (MSC)-derived secretome has emerged as a promising therapeutic approach owing to its potent immunomodulatory properties. However, progress has been hindered by the lack of standardized protocols for inducing a robust immunomodulatory MSC phenotype. METHODS In this study, we focused on optimizing the MSC-derived secretome to enhance its ability to suppress activated immune cells. Specifically, we examined (1) the effects of IFN-γ and TNF-α, individually and in combination, to uncover potential synergy; (2) the ideal cytokine ratio and (3) concentration; (4) the best production time for the secretome; and (5) the impact of cellular confluence. These factors were systematically evaluated to assess their influence on cell behavior, viability, cytosolic content release, and the secretion of key immunomodulatory and regenerative factors. RESULTS Our results demonstrate that overnight licensing with a 1:1 ratio of IFN-γ and TNF-α at 60 ng/mL, followed by 48 h of incubation at 90% confluence, yields an optimized conditioned media (CM) with significantly enhanced immunomodulatory properties. Functional assays showed that this CM can inhibit human peripheral blood mononuclear cell (PBMC) activation with more than twice the effectiveness of suboptimal protocols. Additionally, we found that direct cell-cell contact was critical for inducing regulatory T cells (Tregs), highlighting the complex dynamics of immune regulation. CONCLUSIONS These findings establish a robust and standardized MSC licensing protocol, paving the way for the development of innovative and effective therapies to combat IMIDs. CLINICAL TRIAL NUMBER Not applicable.
Collapse
Affiliation(s)
- Maria Rossello-Gelabert
- NanoBioCel Research Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, Vitoria Gasteiz, 01006, Spain
- Bioaraba, NanoBioCel Research Group, Vitoria Gasteiz, Spain
| | - Manoli Igartua
- NanoBioCel Research Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, Vitoria Gasteiz, 01006, Spain
- Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, Madrid, Spain
- Bioaraba, NanoBioCel Research Group, Vitoria Gasteiz, Spain
| | - Edorta Santos-Vizcaino
- NanoBioCel Research Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, Vitoria Gasteiz, 01006, Spain.
- Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, Madrid, Spain.
- Bioaraba, NanoBioCel Research Group, Vitoria Gasteiz, Spain.
| | - Rosa Maria Hernandez
- NanoBioCel Research Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, Vitoria Gasteiz, 01006, Spain.
- Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, Madrid, Spain.
- Bioaraba, NanoBioCel Research Group, Vitoria Gasteiz, Spain.
| |
Collapse
|
9
|
Zhang L, Yuan X, Song R, Yuan Z, Zhao Y, Zhang Y. Engineered 3D mesenchymal stem cell aggregates with multifunctional prowess for bone regeneration: Current status and future prospects. J Adv Res 2025:S2090-1232(25)00227-9. [PMID: 40220897 DOI: 10.1016/j.jare.2025.04.008] [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: 11/17/2024] [Revised: 03/29/2025] [Accepted: 04/05/2025] [Indexed: 04/14/2025] Open
Abstract
BACKGROUND Impaired efficacy of in vitro expanded mesenchymal stem cells (MSCs) is a universal and thorny situation, which cast a shadow on further clinical translation of exogenous MSCs. Moreover, the relatively lengthy healing process, host metabolic heterogeneity and the sophisticated cell recognition and crosstalk pose rigorous challenges towards MSC-based bone regeneration strategies. Three-dimensional (3D) cell aggregates facilitate more robust intercellular communications and cell-extracellular matrix (ECM) interactions, providing a better mimicry of microarchitectures and biochemical milieus in vivo, which is conducive for stemness maintenance and downstream bone formation. AIM OF REVIEW This review enunciates the phenotypic features of MSCs in aggregates, which deepens the knowledge of the MSC fate determination in 3D microenvironment. By summarizing current empowerment methods and biomaterial-combined techniques for establishing functionalized MSC aggregates, this review aims to spark innovative and promising solutions for exalting the translational value of MSCs and improve their therapeutic applications in bone tissue repair. KEY SCIENTIFIC CONCEPTS OF REVIEW 3D aggregates optimize regenerative behaviors of in vitro cultured MSCs including cell adhesion, viability, proliferation, pluripotency and immunoregulation capacity, etc. Biomaterials hybridization endows MSC aggregates with tailored mechanical and biological properties, which offers more possibilities in adapting various clinical scenarios.
Collapse
Affiliation(s)
- Linxue Zhang
- Department of Pediatrics, Peking University School and Hospital of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials, Beijing 100081, PR China
| | - Xiaojing Yuan
- Department of Pediatrics, Peking University School and Hospital of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials, Beijing 100081, PR China
| | - Rui Song
- Department of Pediatrics, Peking University School and Hospital of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials, Beijing 100081, PR China
| | - Zuoying Yuan
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, PR China; Medical Innovation and Research, Peking University Third Hospital, Beijing 100191, PR China.
| | - Yuming Zhao
- Department of Pediatrics, Peking University School and Hospital of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials, Beijing 100081, PR China.
| | - Yunfan Zhang
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials, 22 Zhongguancun South Avenue, Haidian District, Beijing, PR China.
| |
Collapse
|
10
|
Maldonado VV, Jensen H, Barnes CL, Samsonraj RM. Phenotypic changes associated with continuous long term in vitro expansion of bone marrow-derived mesenchymal stem cells. Biochimie 2025; 234:62-75. [PMID: 40209891 DOI: 10.1016/j.biochi.2025.04.002] [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: 01/28/2025] [Revised: 03/26/2025] [Accepted: 04/07/2025] [Indexed: 04/12/2025]
Abstract
In vitro expansion of mesenchymal stem cells is necessary to obtain a higher cell number for clinical applications. However, long-term expansion can produce significant phenotypic changes on these cells, decreasing their therapeutic utility. Therefore, understanding the phenotypic changes that long-term expansion triggers in mesenchymal stem cells will allow for better and more consistent cell therapy results. Here, we evaluate the phenotypic changes caused by continuous passaging through colony forming unit-fibroblast assay, senescence beta-galactosidase staining, morphology examination, secretome analysis, surface marker expression, protein quantification, osteogenic and adipogenic differentiation, and CD4+ T lymphocyte immunosuppressive potential. Long-term in vitro culture decreases mesenchymal stem cell osteogenic potential and self-renewal, increases cell size, and senescence, but does not consistently affect adipogenic differentiation. Surface marker expression remains similar for positive and negative markers, while secretory phenotype shifts with decreased p14ARF, MMP-3, p21 Waf1/Cip1,ENA-78, GCP-2, GROα, IL-3, IL-7, IL-8, RANTES, TNFβ, and VEGF-A expression, and increased p53, p16 INK4a, MCP-1, and SDF-1 expression. Immunomodulatory potential remains unchanged. These findings can help better understand the phenotypic changes that mesenchymal stem cells undergo while expanded in vitro.
Collapse
Affiliation(s)
- Vitali V Maldonado
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR, 72701, USA
| | - Hanna Jensen
- Department of Surgery, University of Arkansas for Medical Sciences, Northwest Regional Campus, Fayetteville, AR, 72701, USA
| | - C Lowry Barnes
- Department of Orthopedic Surgery, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Rebekah M Samsonraj
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR, 72701, USA; Department of Orthopedic Surgery, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA.
| |
Collapse
|
11
|
Skamagki M, Zhang C, Hacisuleyman E, Galleti G, Wu C, Vinagolu RK, Cha H, Ata D, Kim J, Weiskittel T, Diop M, Aung T, Del Latto M, Kim AS, Li Z, Miele M, Zhao R, Tang LH, Hendrickson RC, Romesser PB, Smith JJ, Giannakakou P, Darnell RB, Bott MJ, Li H, Kim K. Aging-dependent dysregulation of EXOSC2 is maintained in cancer as a dependency. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.04.647279. [PMID: 40236131 PMCID: PMC11996493 DOI: 10.1101/2025.04.04.647279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Reprogramming of aged donor tissue cells into induced pluripotent stem cells (A-iPSC) preserved the epigenetic memory of aged-donor tissue, defined as genomic instability and poor tissue differentiation in our previous study. The unbalanced expression of RNA exosome subunits affects the RNA degradation complex function and is associated with geriatric diseases including premature aging and cancer progression. We hypothesized that the age-dependent progressive subtle dysregulation of EXOSC2 (exosome component 2) causes the aging traits (abnormal cell cycle and poor tissue differentiation). We used embryonic stem cells as a tool to study EXOSC2 function as the aging trait epigenetic memory determined in A-iPSC because these aging traits could not be studied in senesced aged cells or immortalized cancer cells. We found that the regulatory subunit of PP2A phosphatase, PPP2R5E, is a key target of EXOSC2 and this regulation is preserved in stem cells and cancer.
Collapse
|
12
|
Simpkins LLC, Tsai T, Egun E, Adams TNG. Electrical Phenotyping of Aged Human Mesenchymal Stem Cells Using Dielectrophoresis. MICROMACHINES 2025; 16:435. [PMID: 40283310 PMCID: PMC12029641 DOI: 10.3390/mi16040435] [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: 03/02/2025] [Revised: 03/22/2025] [Accepted: 03/28/2025] [Indexed: 04/29/2025]
Abstract
Human mesenchymal stem cells (hMSCs) are widely used in regenerative medicine, but large-scale in vitro expansion alters their function, impacting proliferation and differentiation potential. Currently, a predictive marker to assess these changes is lacking. Here, we used dielectrophoresis (DEP) to characterize the electrical phenotype of hMSCs derived from bone marrow (BM), adipose tissue (AT), and umbilical cord (UC) as they aged in vitro from passage 4 (P4) to passage 9 (P9). The electrical phenotype was defined by the DEP spectra, membrane capacitance, and cytoplasm conductivity. Cell morphology and size, growth characteristics, adipogenic differentiation potential, and osteogenic differentiation potential were assessed alongside label-free biomarker membrane capacitance and cytoplasm conductivity. Differentiation was confirmed by histological staining and RT-qPCR. All hMSCs exhibited typical morphology, though cell size varied, with UC-hMSCs displaying the largest variability across all size metrics. Growth analysis revealed that UC-hMSCs proliferated the fastest. The electrical phenotype varied with cell source and in vitro age, with high passage hMSCs showing noticeable shifts in DEP spectra, membrane capacitance, and cytoplasm conductivity. Correlation analysis revealed that population doubling level (PDL) correlated with membrane capacitance and cytoplasm conductivity, indicating PDL as a more precise marker of in vitro aging than passage number. Additionally, we demonstrate that membrane capacitance correlates with the osteogenic marker COL1A1 and that cytoplasm conductivity correlates with the adipogenic markers ADIPOQ and FABP4, suggesting that DEP-derived electrical properties serve as label-free biomarkers of differentiation potential. While DEP has previously been applied to BM-hMSCs and AT-hMSCs, and more recently to UC-hMSCs, few studies have provided a direct comparison across all three sources or tracked changes across continuous expansion. These findings underscore the utility of DEP as a label-free approach for assessing hMSC aging and function, offering practical applications for optimizing stem cell expansion and stem cell banking in clinical settings.
Collapse
Affiliation(s)
- Lexi L. C. Simpkins
- Department of Chemical and Biomolecular Engineering, University of California Irvine, Irvine, CA 92697, USA; (L.L.C.S.); (T.T.); (E.E.)
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA 92697, USA
| | - Tunglin Tsai
- Department of Chemical and Biomolecular Engineering, University of California Irvine, Irvine, CA 92697, USA; (L.L.C.S.); (T.T.); (E.E.)
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA 92697, USA
| | - Emmanuel Egun
- Department of Chemical and Biomolecular Engineering, University of California Irvine, Irvine, CA 92697, USA; (L.L.C.S.); (T.T.); (E.E.)
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA 92697, USA
| | - Tayloria N. G. Adams
- Department of Chemical and Biomolecular Engineering, University of California Irvine, Irvine, CA 92697, USA; (L.L.C.S.); (T.T.); (E.E.)
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA 92697, USA
- Department of Materials Science Engineering, University of California, Irvine, CA 92697, USA
- Department of Biomedical Engineering, University of California, Irvine, CA 92697, USA
| |
Collapse
|
13
|
Palamà MEF, Gorgun C, Rovere M, Shaw GM, Reverberi D, Formica M, Quarto E, Barry F, Murphy M, Gentili C. Batch variability and anti-inflammatory effects of iPSC-derived mesenchymal stromal cell extracellular vesicles in osteoarthritis in vitro model. Front Bioeng Biotechnol 2025; 13:1536843. [PMID: 40242358 PMCID: PMC11999995 DOI: 10.3389/fbioe.2025.1536843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 03/21/2025] [Indexed: 04/18/2025] Open
Abstract
Mesenchymal stromal cell-derived extracellular vesicles (MSC-EVs) hold promise as a cell-free therapy for osteoarthritis (OA), due to their immunomodulatory and anti-inflammatory properties. However, the need for large-scale expansion to obtain MSC-EVs for clinical use can lead to senescence-related changes and loss of stem-like properties. In this scenario, induced pluripotent stem cell (iPSC)-derived MSCs (iMSCs) offer the unique opportunity to address obstacles associated with traditional MSC-based therapies. This study used a xeno-free (XFS) medium for long-term expansion of both MSCs and iMSCs, and their EVs comparison. Characterization of both cells and EVs was conducted across different passages, and the anti-inflammatory potential of EVs and iEVs was assessed using an in vitro model of osteoarthritis. Long-term expansion of MSCs resulted in cellular senescence and a reduction in trilineage differentiation capacity by passage five, accompanied by diminished anti-inflammatory properties of EVs. On the other hand, iMSCs exhibited batch-to-batch variability in differentiation and EV biological properties. However, the effects of iMSC-EVs were prolonged compared to MSC-EVs, providing a wider window of activity for therapeutic purposes. Despite this, the variability among iMSC batches poses challenges for their reliability in OA treatment. Further work is needed to overcome these limitations for clinical application.
Collapse
Affiliation(s)
| | - Cansu Gorgun
- Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy
| | - Matteo Rovere
- Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy
| | - Georgina M. Shaw
- Regenerative Medicine Institute (REMEDI), University of Galway (UoG), Galway, Ireland
| | - Daniele Reverberi
- UOC Research-Scientific Direction, Istituto di Scientifico Ricovero e Cura a Carattere, Ospedale Policlinico San Martino, Genoa, Italy
| | - Matteo Formica
- UOC Clinica Ortopedica, Istituto di Scientifico Ricovero e Cura a Carattere, Ospedale Policlinico San Martino, Genoa, Italy
| | - Emanuele Quarto
- UOC Clinica Ortopedica, Istituto di Scientifico Ricovero e Cura a Carattere, Ospedale Policlinico San Martino, Genoa, Italy
| | - Frank Barry
- Regenerative Medicine Institute (REMEDI), University of Galway (UoG), Galway, Ireland
| | - Mary Murphy
- Regenerative Medicine Institute (REMEDI), University of Galway (UoG), Galway, Ireland
| | - Chiara Gentili
- Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy
- UOC Oncologia Cellulare, Istituto di Scientifico Ricovero e Cura a Carattere, Ospedale Policlinico San Martino, Genoa, Italy
| |
Collapse
|
14
|
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
|
15
|
Lin H, Zhou C, Li Q, Xie Q, Xia L, Liu L, Bao W, Xiong X, Zhang H, Zheng Z, Zhao J, Liang W. Nanotechnology-Assisted mesenchymal stem cells treatment for improved cartilage regeneration: A review of current practices. Biochem Pharmacol 2025; 237:116895. [PMID: 40154890 DOI: 10.1016/j.bcp.2025.116895] [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: 11/18/2024] [Revised: 02/26/2025] [Accepted: 03/24/2025] [Indexed: 04/01/2025]
Abstract
Cartilage tissue does not promptly elicit an inflammatory response upon injury, hence constraining its capacity for healing and self-regeneration. Mesenchymal Stem Cells (MSC) therapy, enhanced by nanotechnology, offers promising advancements in cartilage repair. Injuries to cartilage often cause chronic pain, where current treatments are inadequate. As MSCs can readily differentiate into chondrocytes and secrete soluble factors, they are essential components in tissue engineering of cartilage repair. Although, like other stem cell applications, clinical applications are restricted by poor post implantation survival and differentiation. Recent studies show that nanoparticles (NPs) can further improve MSC outcomes by promoting cell adhesion, and chondrogenic differentiation allowing for sustained growth factor release. In addition, nanomaterials can improve the biological activity of MSCs, by also facilitating the composition of a conducive microenvironment for cartilage repair. In this review, the application of nanofibrous scaffolds, hydrogels and nanoscale particulate matter to improve mechanical properties in cartilage tissue engineering, are discussed. Moreover, the MSCs and nanotechnology synergistic effects present hope of overcoming the limitations of conventional treatments. Nanotechnology greatly enhances the MSC based cartilage regeneration strategies and could provide better treatment for cartilage related diseases in the future. Future research should be aimed at standardizing MSC harvesting and culturing protocols and contrasting their long-term efficacy.
Collapse
Affiliation(s)
- Hongming Lin
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan 316000 Zhejiang Province, China
| | - Chao Zhou
- Department of Orthopedics, Zhoushan Guanghua hospital, Zhoushan 316000 Zhejiang Province, China
| | - Qingping Li
- Medical Research Center, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan 316000 Zhejiang Province, China
| | - Qiong Xie
- Medical Research Center, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan 316000 Zhejiang Province, China
| | - Linying Xia
- Medical Research Center, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan 316000 Zhejiang Province, China
| | - Lu Liu
- Medical Research Center, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan 316000 Zhejiang Province, China
| | - Wenwen Bao
- Medical Research Center, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan 316000 Zhejiang Province, China
| | - Xiaochun Xiong
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan 316000 Zhejiang Province, China
| | - Hao Zhang
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan 316000 Zhejiang Province, China
| | - Zeping Zheng
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan 316000 Zhejiang Province, China
| | - Jiayi Zhao
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan 316000 Zhejiang Province, China.
| | - Wenqing Liang
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan 316000 Zhejiang Province, China.
| |
Collapse
|
16
|
Zapata-Linares N, Toillon I, Wanherdrick K, Pigenet A, Duhalde F, Binvignat M, Martin-Uriz PS, Louvet L, Calleja-Cervantes ME, Ghali Mhenni O, Guibert C, Nourissat G, Nogier A, Leterme D, Broux O, Magneron P, Prosper F, Chauveau C, Landoulsi J, Berenbaum F, Rodriguez-Madoz JR, Lafage-Proust MH, Lucas S, Houard X. Implication of bone marrow adipose tissue in bone homeostasis during osteoarthritis. Osteoarthritis Cartilage 2025:S1063-4584(25)00870-2. [PMID: 40154729 DOI: 10.1016/j.joca.2025.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 03/16/2025] [Accepted: 03/19/2025] [Indexed: 04/01/2025]
Abstract
OBJECTIVE To explore the role of bone marrow adipocytes (BMAds) in osteoarthritis (OA). METHODS Male and female C57BL/6 mice (n=4/group) underwent meniscectomy (MNX) or SHAM surgery. OA was determined using Osteoarthritis Research Society International (OARSI) score, and the number of perilipin+ adipocytes was quantified. Mesenchymal Stromal Cells (MSCs) from MNX and SHAM mice were differentiated into osteoblasts and adipocytes. Human adipocytes and MSCs (n=8) were enzymatically isolated from epiphyseal and metaphyseal marrow, and from subcutaneous adipose tissue (SCAT) of hip OA patients. Human OA MSCs were differentiated into osteoblasts and adipocytes (OA-Diff-hAdipo). Gene expression patterns of epiphyseal and metaphyseal BMAds, SCAT adipocytes and OA-Diff-hAdipo were evaluated by RNAseq (n=4). The effect conditioned media from OA epiphyseal bone (n=5) on the alkaline phosphatase (ALP) activity and mineralization kinetics was assessed in vitro. RESULTS Increase in BMAd density was positively correlated with cartilage degradation in MNX mice. OA modified the differentiation capacity of MSCs, accelerating adipocyte differentiation and failing to produce osteoblasts in both human and mice. Human epiphyseal, metaphyseal and SCAT adipocytes from the same OA patients each displayed a specific transcriptome, suggesting different functions. Enrichment analysis defined metaphyseal OA-BMAds as cells implicated in hematopoietic stem cell differentiation. On the other hand, epiphyseal OA-BMAds were considered as osteogenic cells showing an up-regulation of genes related to bone mineralization and remodeling. Specifically, OA epiphysis-secreted molecules decreased ALP activity and altered in vitro the mineralization process. CONCLUSION All these results support the emergence of BMAds as new cell partners in OA, opening new venues for therapeutic approaches.
Collapse
Affiliation(s)
- Natalia Zapata-Linares
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, F-75012 Paris, France
| | - Indira Toillon
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, F-75012 Paris, France
| | - Kristell Wanherdrick
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, F-75012 Paris, France
| | - Audrey Pigenet
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, F-75012 Paris, France
| | - Fanny Duhalde
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, F-75012 Paris, France; Sorbonne Université, CNRS, Laboratoire de Réactivité de Surface, LRS, F-75005, Paris, France
| | - Marie Binvignat
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, F-75012 Paris, France
| | | | - Loïc Louvet
- Marrow Adiposity and Bone Lab (MABLab) ULR4490, Université du Littoral Côte d'Opale, F-62200 Boulogne sur Mer, Univ. Lille, F-59000 Lille, CHU Lille, F-59000 Lille, France
| | - Maria E Calleja-Cervantes
- Hemato-Oncology Program. CIMA Universidad de Navarra-IdiSNA, Pamplona, Spain; Computational Biology Program, CIMA Universidad de Navarra-IdiSNA, Pamplona, Spain
| | - Olfa Ghali Mhenni
- Marrow Adiposity and Bone Lab (MABLab) ULR4490, Université du Littoral Côte d'Opale, F-62200 Boulogne sur Mer, Univ. Lille, F-59000 Lille, CHU Lille, F-59000 Lille, France
| | - Clément Guibert
- Sorbonne Université, CNRS, Laboratoire de Réactivité de Surface, LRS, F-75005, Paris, France
| | - Geoffroy Nourissat
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, F-75012 Paris, France
| | | | - Damien Leterme
- Marrow Adiposity and Bone Lab (MABLab) ULR4490, Université du Littoral Côte d'Opale, F-62200 Boulogne sur Mer, Univ. Lille, F-59000 Lille, CHU Lille, F-59000 Lille, France
| | - Odile Broux
- Marrow Adiposity and Bone Lab (MABLab) ULR4490, Université du Littoral Côte d'Opale, F-62200 Boulogne sur Mer, Univ. Lille, F-59000 Lille, CHU Lille, F-59000 Lille, France
| | - Paul Magneron
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, F-75012 Paris, France
| | - Felipe Prosper
- Hemato-Oncology Program. CIMA Universidad de Navarra-IdiSNA, Pamplona, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain; Hematology and Cell Therapy Department, Clinica Universidad de Navarra, IdiSNA, Pamplona, Spain; Cancer Center Universidad de Navarra (CCUN), Pamplona, Spain
| | - Christophe Chauveau
- Marrow Adiposity and Bone Lab (MABLab) ULR4490, Université du Littoral Côte d'Opale, F-62200 Boulogne sur Mer, Univ. Lille, F-59000 Lille, CHU Lille, F-59000 Lille, France
| | - Jessem Landoulsi
- Sorbonne Université, CNRS, Laboratoire de Réactivité de Surface, LRS, F-75005, Paris, France
| | - Francis Berenbaum
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, F-75012 Paris, France; Rheumatology Department, AP-HP Saint-Antoine Hospital, 184, Rue du Faubourg Saint-Antoine, 75012 Paris, France
| | - Juan R Rodriguez-Madoz
- Hemato-Oncology Program. CIMA Universidad de Navarra-IdiSNA, Pamplona, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain; Cancer Center Universidad de Navarra (CCUN), Pamplona, Spain
| | - Marie-Hélène Lafage-Proust
- Université de Lyon - Université Jean Monnet, INSERM U1059, Faculté de Médecine, F-42270 Saint-Priest en Jarez, France
| | - Stéphanie Lucas
- Marrow Adiposity and Bone Lab (MABLab) ULR4490, Université du Littoral Côte d'Opale, F-62200 Boulogne sur Mer, Univ. Lille, F-59000 Lille, CHU Lille, F-59000 Lille, France
| | - Xavier Houard
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, F-75012 Paris, France.
| |
Collapse
|
17
|
Wang YC, Cheng JB, Feng ML. Requirements for standardizing the assessment of mesenchymal stem cell therapy and its effects on osteoarthritis. World J Orthop 2025; 16:104451. [PMID: 40124721 PMCID: PMC11924026 DOI: 10.5312/wjo.v16.i3.104451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 02/06/2025] [Accepted: 02/27/2025] [Indexed: 03/12/2025] Open
Abstract
Publications of Soufan et al and Kristjánsson et al in the World Journal of Orthopedics on mesenchymal stem cell (MSC) therapy for osteoarthritis (OA) represent a significant exploration of regenerative medicine's potential in OA treatment. In their research, it is highlighted that MSCs can alleviate OA symptoms and even regenerate cartilage, potentially reversing the disease. They also compared the efficacy of three MSC subtypes, emphasizing the therapeutic advantages of adipose-derived MSCs. MSC injections, a novel and less invasive alternative to traditional treatments such as chondrocyte transplantation or arthroplasty, have a low cost, low risks, and favorable outcomes, presenting a promising approach for OA patients. Additionally, we stressed that the efficacy evaluation criteria, heterogeneity, safety, and other factors must be carefully considered to further advance the clinical translation of MSC therapy for OA.
Collapse
Affiliation(s)
- Yu-Chen Wang
- Department of Orthopaedic Surgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Jing-Bo Cheng
- Department of Orthopaedic Surgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Ming-Li Feng
- Department of Orthopaedic Surgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| |
Collapse
|
18
|
Zhang P, Liu M, Pei S, Huang H, Zhao Z, Yang L, Pan W, Li S, Bai Q, Zhang R, Zhou P. Efficient Differentiation of hiPSCs into hMSC-like Cells under Chemically Defined Conditions on Temperature-Sensitive Micropatterned Surfaces. ACS APPLIED MATERIALS & INTERFACES 2025; 17:13358-13374. [PMID: 39976673 DOI: 10.1021/acsami.4c13686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2025]
Abstract
The fairness of long-term self-renewal and robust cell proliferation limits the applications of human mesenchymal stem cells (hMSCs) in regenerative medicine. Inducing hMSCs from human-induced pluripotent stem cells (hiPSCs), which have the advantages of autogenous and no cell number issues, is highly valuable. However, current induction methods using FBS-containing mesenchymal culture medium have problems, including immunogenicity, microbial contamination, and low efficiency. To solve these problems, we propose a chemically defined induction protocol incorporating transforming growth factor-beta 1 and retinoic acid (RA) additives in serum-free E6 medium for the suspension induction of embryoid bodies in hiPSCs. Additionally, microgroove-patterned polydimethylsiloxane surfaces coated with temperature-sensitive N-isopropylacrylamide (PNIPAAm) were prepared for efficient harvesting and purification of induced hiPSC-derived hMSCs (hiPSC-MSCs). The results showed that both supplementation with RA and patterned microgrooves with a width of 20 μm could accelerate the induction of hiPSC-MSCs, realizing the promising scalable production of homogeneous cells. This study successfully established a chemically defined induction protocol and utilized patterned surfaces to obtain clinically applicable hiPSC-MSCs, which show great promise in tissue engineering, gene therapy, and cell transplantation.
Collapse
Affiliation(s)
- Pengxia Zhang
- School of Stomatology, Lanzhou University, No. 222 Tianshui South Road, Chengguan District, Lanzhou 730000, China
| | - Maoying Liu
- School of Basic Medical Sciences, Lanzhou University, No. 222 Tianshui South Road, Chengguan District, Lanzhou 730000, China
| | - Suying Pei
- School of Stomatology, Lanzhou University, No. 222 Tianshui South Road, Chengguan District, Lanzhou 730000, China
| | - Hongxin Huang
- School of Stomatology, Lanzhou University, No. 222 Tianshui South Road, Chengguan District, Lanzhou 730000, China
| | - Zhengyan Zhao
- School of Stomatology, Lanzhou University, No. 222 Tianshui South Road, Chengguan District, Lanzhou 730000, China
| | - Ling Yang
- School of Stomatology, Lanzhou University, No. 222 Tianshui South Road, Chengguan District, Lanzhou 730000, China
| | - Wen Pan
- School of Life Science, Lanzhou University, No. 222 Tianshui South Road, Chengguan District, Lanzhou 730000, China
| | - Siyi Li
- School of Stomatology, Lanzhou University, No. 222 Tianshui South Road, Chengguan District, Lanzhou 730000, China
| | - Qifeng Bai
- School of Basic Medical Sciences, Lanzhou University, No. 222 Tianshui South Road, Chengguan District, Lanzhou 730000, China
| | - Rui Zhang
- School of Stomatology, Lanzhou University, No. 222 Tianshui South Road, Chengguan District, Lanzhou 730000, China
| | - Ping Zhou
- School of Stomatology, Lanzhou University, No. 222 Tianshui South Road, Chengguan District, Lanzhou 730000, China
| |
Collapse
|
19
|
Golebiowska AA, Intravaia JT, Sathe V, Kumbar SG, Nukavarapu SP. Engineered Osteochondral Scaffolds with Bioactive Cartilage Zone for Enhanced Articular Cartilage Regeneration. Ann Biomed Eng 2025; 53:597-611. [PMID: 39602036 PMCID: PMC11835937 DOI: 10.1007/s10439-024-03655-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 11/18/2024] [Indexed: 11/29/2024]
Abstract
Despite progress, osteochondral (OC) tissue engineering strategies face limitations in terms of articular cartilage layer development and its integration with the underlying bone tissue. The main objective of this study is to develop a zonal OC scaffold with native biochemical signaling in the cartilage zone to promote articular cartilage development devoid of cells and growth factors. Herein, we report the development and in vivo assessment of a novel gradient and zonal-structured scaffold for OC defect regeneration. The scaffold system is composed of a mechanically supportive 3D-printed template containing decellularized cartilage extracellular matrix (ECM) biomaterial in the cartilage zone that possesses bioactive characteristics, such as chemotactic activity and native tissue biochemical composition. OC scaffolds with a bioactive cartilage zone were implanted in vivo in a rabbit osteochondral defect model and assessed for gross morphology, matrix deposition, cellular distribution, and overall tissue regeneration. The scaffold system supported recruitment and infiltration of host cells into the cartilage zone of the graft, which led to increased ECM deposition and physiologically relevant articular cartilage tissue formation. Semi-quantitative ICRS scoring (overall score double for OC scaffold with bioactive cartilage zone compared to PLA scaffold) further confirm the bioactive scaffold enhanced articular cartilage engineering. This strategy of designing bioactive scaffolds to promote endogenous cellular infiltration can be a much simpler and effective approach for OC tissue repair and regeneration.
Collapse
Affiliation(s)
- Aleksandra A Golebiowska
- Department of Biomedical Engineering, University of Connecticut, 260 Glenbrook Road, Unit 3247, Storrs, CT, 06269, USA
| | - Jonathon T Intravaia
- Department of Biomedical Engineering, University of Connecticut, 260 Glenbrook Road, Unit 3247, Storrs, CT, 06269, USA
| | - Vinayak Sathe
- Department of Orthopaedic Surgery, University of Connecticut Health, Farmington, CT, 06032, USA
| | - Sangamesh G Kumbar
- Department of Biomedical Engineering, University of Connecticut, 260 Glenbrook Road, Unit 3247, Storrs, CT, 06269, USA
- Department of Materials Science & Engineering, University of Connecticut, Storrs, CT, 06269, USA
- Department of Orthopaedic Surgery, University of Connecticut Health, Farmington, CT, 06032, USA
| | - Syam P Nukavarapu
- Department of Biomedical Engineering, University of Connecticut, 260 Glenbrook Road, Unit 3247, Storrs, CT, 06269, USA.
- Department of Materials Science & Engineering, University of Connecticut, Storrs, CT, 06269, USA.
- Department of Orthopaedic Surgery, University of Connecticut Health, Farmington, CT, 06032, USA.
| |
Collapse
|
20
|
Guillaumin S, Rossoni A, Zeugolis D. State-of the-art and future perspective in co-culture systems for tendon engineering. BIOMATERIALS AND BIOSYSTEMS 2025; 17:100110. [PMID: 40130022 PMCID: PMC11932666 DOI: 10.1016/j.bbiosy.2025.100110] [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: 08/08/2024] [Revised: 11/05/2024] [Accepted: 03/04/2025] [Indexed: 03/26/2025] Open
Abstract
Tendon is a connective tissue that links bone to muscle, allowing for maintenance of skeleton posture, joint movement, energy storage and transmission of muscle force to bone. Tendon is a hypocellular and hypovascular tissue of poor self-regeneration capacity. Current surgical treatments are of limited success, frequently resulting in reinjury. Upcoming cell therapies are primarily based on tenocytes, a cell population of limited self-renewal capacity in vitro or mesenchymal stromal cells, a cell population prone to ectopic bone formation in vivo. Over the years mono- or multi- factorial cell culture technologies have failed to effectively maintain tenocyte phenotype in culture during expansion or to prime mesenchymal stromal cells towards tenogenic lineage prior to implantation. Upon these limitations the concept of co-culture was conceived. Here, we comprehensively review and discuss tenogenic differentiation of mesenchymal stromal cells through direct or indirect culture with tenocytes in an attempt to generate a tenocyte or a tendon-like cell population for regenerative medicine purposes.
Collapse
Affiliation(s)
- Salomé Guillaumin
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL) and Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, University of Galway, Galway, Ireland
| | - Andrea Rossoni
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Charles Institute of Dermatology, Conway Institute of Biomolecular and Biomedical Research and School of Mechanical and Materials Engineering, University College Dublin (UCD), Dublin, Ireland
| | - Dimitrios Zeugolis
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL) and Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, University of Galway, Galway, Ireland
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Charles Institute of Dermatology, Conway Institute of Biomolecular and Biomedical Research and School of Mechanical and Materials Engineering, University College Dublin (UCD), Dublin, Ireland
| |
Collapse
|
21
|
Watanabe M, Asawa Y, Riu D, Sakamoto T, Hoshi K, Hikita A. Identification of mesenchymal stem cell populations with high osteogenic potential using difference in cell division rate. Regen Ther 2025; 28:498-508. [PMID: 39991510 PMCID: PMC11846930 DOI: 10.1016/j.reth.2025.01.020] [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: 11/16/2024] [Revised: 01/13/2025] [Accepted: 01/25/2025] [Indexed: 02/25/2025] Open
Abstract
Introduction In bone regenerative medicine, mesenchymal stem cells (MSCs) have been widely investigated for their potential in bone regeneration. However, MSCs are a heterogeneous cell population containing a variety of cell types, making it difficult to obtain a homogeneous MSC population sufficient for tissue regeneration. Our group previously reported that by selecting rapidly dividing human auricular chondrocytes, it was possible to enrich for more chondrogenic cells. In this study, we aimed to identify a highly osteogenic MSC population by using a similar approach for mouse bone marrow MSCs. Methods Mouse bone marrow MSCs were fluorescently labeled with carboxyfluorescein succinimidyl ester (CFSE) and sorted according to the fluorescence intensity using flow cytometry on day 3 after labeling. To compare the ability to produce bone matrix in vitro, osteogenic differentiation cultures were performed and mineral deposition was confirmed by alizarin red staining. Real-time qPCR was also performed to examine the differences in gene expression between the fast- and slow-dividing cell groups immediately after aliquoting and after osteogenic differentiation. Results Differences in the growth rate of the fractionated cells were maintained after culture. Results of osteogenic differentiation culture and alizarin red staining showed more extensive mineral deposition in the slow cell group than in the fast cell group. Calcium quantification also showed higher absorbance in the slow cell group compared to the fast cell group, indicating higher osteogenic differentiation potential in the slow cell group. Furthermore, real-time qPCR analysis showed that osteocalcin expression was higher in the slow cell group in cells immediately after preparative differentiation. In addition, the expression of osteocalcin and sclerostin were higher in the slow cells after osteogenic differentiation. Conclusion The slow cell population contains many highly differentiated cells that are already more deeply committed to the bone lineage, suggesting that they have higher osteogenic differentiation potential than the fast cell population. This study will contribute to the realization of better bone regenerative medicine by utilizing the high osteogenic differentiation potential of the slow cell population.
Collapse
Affiliation(s)
- Maya Watanabe
- Department of Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Yukiyo Asawa
- Division of Tissue Engineering, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Dan Riu
- Division of Tissue Engineering, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Tomoaki Sakamoto
- Division of Tissue Engineering, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Kazuto Hoshi
- Department of Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
- Division of Tissue Engineering, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
- Department of Oral-maxillofacial Surgery, Dentistry and Orthodontics, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Atsuhiko Hikita
- Division of Tissue Engineering, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| |
Collapse
|
22
|
Xu T, Peng Y, Xu Y, Zhu J, Yang Q, Liu Y, Yang H. Exploring the therapeutic potential of small extracellular vesicles derived from induced pluripotent stem cell in periodontal regeneration. J Oral Biosci 2025; 67:100621. [PMID: 39892783 DOI: 10.1016/j.job.2025.100621] [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: 12/02/2024] [Revised: 01/27/2025] [Accepted: 01/28/2025] [Indexed: 02/04/2025]
Abstract
OBJECTIVES To investigate the role of small extracellular vesicles derived from induced pluripotent stem cells (iPSC-sEVs) in periodontal tissue regeneration, elucidate their potential molecular mechanisms, and provide theoretical guidance for the clinical application of iPSC-sEVs as a cell-free therapeutic strategy for periodontal tissue regeneration. METHODS We investigated the effects of iPSC-sEVs on the proliferation, migration, and osteogenic differentiation of periodontal ligament stem cells (PDLSCs) in vitro. The regenerative potential of iPSC-sEVs was evaluated in vivo, using a periodontal defect model. Bulk RNA sequencing was performed to elucidate the underlying molecular mechanisms. RESULTS iPSC-sEVs were isolated, characterized, and systemically evaluated for regenerative potential. The results revealed that treatment with iPSC-sEVs significantly enhanced the proliferation, migration, and osteogenic differentiation of PDLSCs. In situ treatment with iPSC-sEVs loaded onto collagen sponges was performed in a rat model of periodontal defects. Micro-CT and histological analyses indicated that iPSC-sEV treatment markedly promoted alveolar bone repair and periodontal ligament regeneration. Mechanistically, the analysis of bulk RNA sequencing data coupled with experimental validation revealed that iPSC-sEV treatment significantly activated the mitogen-activated protein kinase (MAPK) signaling pathway in PDLSCs. Further investigation showed that the inhibition of this pathway completely abolished the proliferative effects of iPSC-sEVs on PDLSCs. CONCLUSIONS iPSC-sEVs promote PDLSC proliferation through MAPK signaling pathway activation, while also enhancing PDLSC migratory and osteogenic differentiation capacities, facilitates the repair and regeneration of damaged periodontal tissue and presents a potential novel therapeutic strategy for clinical periodontal tissue regeneration.
Collapse
Affiliation(s)
- Tingting Xu
- Yunnan Key Laboratory of Stomatology, Kunming, 650106, China; Department of Periodontics, Kunming Medical University School and Hospital of Stomatology, Kunming, 650106, China.
| | - Yi Peng
- Yunnan Key Laboratory of Stomatology, Kunming, 650106, China; Department of Periodontics, Kunming Medical University School and Hospital of Stomatology, Kunming, 650106, China.
| | - Yanan Xu
- Yunnan Key Laboratory of Stomatology, Kunming, 650106, China; Department of Periodontics, Kunming Medical University School and Hospital of Stomatology, Kunming, 650106, China.
| | - Jing Zhu
- Yunnan Key Laboratory of Stomatology, Kunming, 650106, China; Department of Periodontics, Kunming Medical University School and Hospital of Stomatology, Kunming, 650106, China.
| | - Qiao Yang
- Yunnan Key Laboratory of Stomatology, Kunming, 650106, China; Department of Periodontics, Kunming Medical University School and Hospital of Stomatology, Kunming, 650106, China.
| | - Yali Liu
- Yunnan Key Laboratory of Stomatology, Kunming, 650106, China; Department of Orthodontics, Kunming Medical University School and Hospital of Stomatology, Kunming, 650106, China.
| | - Hefeng Yang
- Yunnan Key Laboratory of Stomatology, Kunming, 650106, China; Department of Prosthodontics, Kunming Medical University School and Hospital of Stomatology, Kunming, 650106, China.
| |
Collapse
|
23
|
Xie XX, Sun JD, Zang MX, Zhang G, Li CX, Zhai XW, Shen W, Ge W, Cheng SF. LPA reduces the apoptosis of cryopreserved porcine skin-derived stem cells by inhibiting the regulatory factor TNF-α. Cryobiology 2025; 118:105189. [PMID: 39706283 DOI: 10.1016/j.cryobiol.2024.105189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 12/17/2024] [Accepted: 12/17/2024] [Indexed: 12/23/2024]
Abstract
Preserving the viability and functionality of stem cells during cryopreservation is crucial for their successful application in regenerative medicine. The aim of this study is to investigate the effect of lysophosphatidic acid (LPA) on reducing the apoptosis of cryopreserved porcine skin-derived stem cells (pSDSCs). Our findings revealed that LPA, at a concentration of 5 μM, significantly improved viability and reduced apoptosis in cryopreserved pSDSCs. Furthermore, our data indicated that LPA enters pSDSCs through receptor type 1 (LPAR1). In cryopreserved pSDSCs, after LPA treatment, the expression level of tumor necrosis factor alpha (TNF-α) protein decreased, suggesting TNF-α involvement in the regulation of the anti-apoptotic process. Additionally, we found that resiquimod (R848), a TNF-α activator, increased the level of apoptosis in cryopreserved pSDSCs. When cryopreserved pSDSCs were treated with both LPA and R848, the protective effect of LPA against apoptosis was decreased. In conclusion, our study demonstrates that LPA could effectively counteract the effect of TNF-α-induced apoptosis, thereby enhancing the survival rates of cryopreserved pSDSCs. Importantly, this study explored a novel mechanism of reducing apoptosis in cryopreserved stem cells.
Collapse
Affiliation(s)
- Xin-Xiang Xie
- College of Life Sciences, Qingdao Agricultural University, Qingdao, 266109, China
| | - Jia-Dong Sun
- College of Life Sciences, Qingdao Agricultural University, Qingdao, 266109, China
| | - Ming-Xin Zang
- College of Life Sciences, Qingdao Agricultural University, Qingdao, 266109, China
| | - Geng Zhang
- College of Life Sciences, Qingdao Agricultural University, Qingdao, 266109, China
| | - Chun-Xiao Li
- College of Life Sciences, Qingdao Agricultural University, Qingdao, 266109, China
| | - Xiang-Wei Zhai
- Animal Husbandry General Station of Shandong Province, Jinan, 250010, China
| | - Wei Shen
- College of Life Sciences, Qingdao Agricultural University, Qingdao, 266109, China
| | - Wei Ge
- College of Life Sciences, Qingdao Agricultural University, Qingdao, 266109, China
| | - Shun-Feng Cheng
- College of Life Sciences, Qingdao Agricultural University, Qingdao, 266109, China.
| |
Collapse
|
24
|
Xia X, Zhou K, An LY, Zhao M, Tian BL, Zhao JY, Zhou ZG, Tong Y. Nicotinamide adenine dinucleotide rejuvenates septic bone marrow mesenchymal stem cells. World J Stem Cells 2025; 17:96893. [DOI: 10.4252/wjsc.v17.i2.96893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 11/19/2024] [Accepted: 01/16/2025] [Indexed: 02/24/2025] Open
Abstract
BACKGROUND Sepsis is a severe illness characterized by systemic and multiorgan reactive responses and damage. However, the impact of sepsis on the bone marrow, particularly on bone marrow mesenchymal stem cells (BMSCs), is less reported. BMSCs are critical stromal cells in the bone marrow microenvironment that maintain bone stability and hematopoietic homeostasis; however, the impairment caused by sepsis remains unknown.
AIM To investigate the effects of sepsis on BMSCs and the underlying mechanisms.
METHODS BMSCs were obtained from healthy donors and patients with sepsis. We compared the self-renewal capacity, differentiation potential, and hematopoietic supportive ability in vitro. Senescence of septic BMSCs was assessed using β-galactosidase staining, senescence-associated secretory phenotype, intracellular reactive oxygen species levels, and the expression of P16 and P21. Finally, the changes in septic BMSCs after nicotinamide adenine dinucleotide (NAD) treatment were evaluated.
RESULTS Septic BMSCs showed decreased proliferation and self-renewal, bias towards adipogenic differentiation, and weakened osteogenic differentiation. Additionally, hematopoietic supportive capacity declines in sepsis. The levels of aging markers were significantly higher in the septic BMSCs. After NAD treatment, the proliferation capacity of septic BMSCs showed a recovery trend, with increased osteogenic and hematopoietic supportive capacities. Sepsis resulted in decreased expression of sirtuin 3 (SIRT3) in BMSCs, whereas NAD treatment restored SIRT3 expression, enhanced superoxide dismutase enzyme activity, reduced intracellular reactive oxygen species levels, maintained mitochondrial stability and function, and ultimately rejuvenated septic BMSCs.
CONCLUSION Sepsis accelerates the aging of BMSCs, as evidenced by a decline in self-renewal and osteogenic capabilities, as well as weakened hematopoietic support functions. These deficiencies can be effectively reversed via the NAD/SIRT3/superoxide dismutase pathway.
Collapse
Affiliation(s)
- Xin Xia
- Department of Hematology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Kun Zhou
- Department of Hematology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Lin-Ying An
- Department of Hematology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Min Zhao
- Department of Hematology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Bin-Le Tian
- Cancer Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Jin-Yan Zhao
- Department of Laboratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Zhi-Gang Zhou
- Department of Critical Care Medicine, School of Medicine, Shanghai Jiao Tong University, Shanghai 201620, China
| | - Yin Tong
- Department of Hematology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| |
Collapse
|
25
|
Wang YJ, Chen ZH, Shen YT, Wang KX, Han YM, Zhang C, Yang XM, Chen BQ. Stem cell therapy: A promising therapeutic approach for skeletal muscle atrophy. World J Stem Cells 2025; 17:98693. [DOI: 10.4252/wjsc.v17.i2.98693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 12/09/2024] [Accepted: 01/23/2025] [Indexed: 02/24/2025] Open
Abstract
Skeletal muscle atrophy results from disruptions in the growth and metabolism of striated muscle, leading to a reduction or loss of muscle fibers. This condition not only significantly impacts patients’ quality of life but also imposes substantial socioeconomic burdens. The complex molecular mechanisms driving skeletal muscle atrophy contribute to the absence of effective treatment options. Recent advances in stem cell therapy have positioned it as a promising approach for addressing this condition. This article reviews the molecular mechanisms of muscle atrophy and outlines current therapeutic strategies, focusing on mesenchymal stem cells, induced pluripotent stem cells, and their derivatives. Additionally, the challenges these stem cells face in clinical applications are discussed. A deeper understanding of the regenerative potential of various stem cells could pave the way for breakthroughs in the prevention and treatment of muscle atrophy.
Collapse
Affiliation(s)
- Ying-Jie Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Nantong University, Nantong 226000, Jiangsu Province, China
| | - Ze-Hao Chen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Nantong University, Nantong 226000, Jiangsu Province, China
| | - Yun-Tian Shen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Nantong University, Nantong 226000, Jiangsu Province, China
| | - Ke-Xin Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Nantong University, Nantong 226000, Jiangsu Province, China
| | - Yi-Min Han
- Medical College, Nantong University, Nantong 226000, Jiangsu Province, China
| | - Chen Zhang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Nantong University, Nantong 226000, Jiangsu Province, China
| | - Xiao-Ming Yang
- Co-Innovation Center of Neuroregeneration, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong 226000, Jiangsu Province, China
- Research and Development Center for E-Learning, Ministry of Education, Beijing 100816, China
| | - Bing-Qian Chen
- Department of Orthopaedics, Changshu Hospital Affiliated to Soochow University, Changshu 215500, Jiangsu Province, China
| |
Collapse
|
26
|
Wang J, Jian K, Yang Q, Gu C, Sheng J, Zhou Y, Yin H, Zhang Z, Hua K, Zhang C. Retarding human adipose-derived MSCs senescence and promoting tendon repair using cell sheet engineering with a histone methyltransferase inhibitor. Sci Rep 2025; 15:6198. [PMID: 39979391 PMCID: PMC11842574 DOI: 10.1038/s41598-025-89234-3] [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: 10/29/2024] [Accepted: 02/04/2025] [Indexed: 02/22/2025] Open
Abstract
Mesenchymal stem cell (MSC) holds immense potential as candidates for cell therapy in the treatment of tendon injuries due to their remarkable ability for multiple cell differentiation. However, the proliferative and differentiation capacity of MSCs has been limited by cellular senescence during the process of expanding culture. Therefore, in this study, our aim was to maintain the beneficial properties of MSCs. We found that SETD7, a histone methyltransferase, was upregulated during ex vivo expansion of human adipose-derived mesenchymal stem cells (hAD-MSCs). Pharmacological inhibition of SETD7 with PFI-2 in hAD-MSCs cultures delayed their senescence, as evident by the diminished expression of senescent-associated genes and the maintenance of their proliferation and differentiation capacity. Upon transplantation, cell sheets derived from hAD-MSCs expanded with PFI-2 were better able to accelerate tendon repair. Therefore, the present findings reveal that SETD7 is an important target to improve the expansion of hAD-MSCs by delaying senescence, which is importance for the development of efficient stem cell-based therapeutic approaches.
Collapse
Affiliation(s)
- Junjuan Wang
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, 310000, China
| | - Ke Jian
- Department of Biomedical Engineering, College of Biology, Hunan University, Changsha, 410082, China
| | - Qing Yang
- Department of Biomedical Engineering, College of Biology, Hunan University, Changsha, 410082, China
| | - Chunyi Gu
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, 310000, China
| | - Jiajun Sheng
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, 310000, China
| | - Yan Zhou
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, 310000, China
| | - Hantian Yin
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, 310000, China
| | - Zhihan Zhang
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, 310000, China
| | - Kouzhen Hua
- Department of Anatomy, School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, 311300, China.
| | - Can Zhang
- Department of Biomedical Engineering, College of Biology, Hunan University, Changsha, 410082, China.
| |
Collapse
|
27
|
Tian RC, Zhang RY, Ma CF. Rejuvenation of Bone Marrow Mesenchymal Stem Cells: Mechanisms and Their Application in Senile Osteoporosis Treatment. Biomolecules 2025; 15:276. [PMID: 40001580 PMCID: PMC11853522 DOI: 10.3390/biom15020276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 02/01/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
Bone marrow mesenchymal stromal cells (BM-MSCs) are multipotent cells present in bone marrow; they play a crucial role in the process of bone formation. Cellular senescence is defined as a stable state of cell cycle arrest that impairs the functioning of cells. Research has shown that aging triggers a state of senescence in BM-MSCs, leading to a reduced capacity for osteogenic differentiation and the accumulation of senescent cells, which can accelerate the onset of various diseases. Therefore, it is essential to explore mechanisms and strategies for the rejuvenation of senescent BM-MSCs. Senile osteoporosis (SOP) is a metabolic bone disease characterized by reduced bone formation. The senescence of BM-MSCs is considered one of the most important factors in the occurrence and development of SOP. Therefore, the rejuvenation of BM-MSCs for the treatment of SOP represents a promising strategy. This work provides a summary of the functional alterations observed in senescent BM-MSCs and a systematic review of the mechanisms that facilitate the rejuvenation of senescent BM-MSCs. Additionally, we analyze the progress in and the limitations associated with the application of rejuvenated senescent BM-MSCs to treat SOP, with the aim of providing new insights for the prevention and treatment of SOP.
Collapse
Affiliation(s)
- Rui-Chuan Tian
- Department of Stomatology, Air Force Medical Center, The Fourth Military Medical University, Beijing 100142, China;
- Graduate School, China Medical University, Shenyang 110002, China
| | - Ru-Ya Zhang
- Department of Emergency and Oral Medicine, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang 110002, China;
| | - Chu-Fan Ma
- Department of Stomatology, Air Force Medical Center, The Fourth Military Medical University, Beijing 100142, China;
- Graduate School, China Medical University, Shenyang 110002, China
| |
Collapse
|
28
|
Huang S, Xu X, Guo J, Li Z, Wu Y, Liu Y, Sun Q, Wang S, Yan H, Su Y, Guo W. Single-Cell Transcriptome Decoding Umbilical Cord-Derived Mesenchymal Stem Cell Heterogeneity Reveals a Unique IL1R1 HighPDGFRA High Ultroser-G-MSC With Osteogenesis and Chondrogenesis Signatures. J Cell Physiol 2025; 240:e70004. [PMID: 39956958 DOI: 10.1002/jcp.70004] [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: 07/29/2024] [Revised: 01/07/2025] [Accepted: 01/13/2025] [Indexed: 02/18/2025]
Abstract
The heterogeneity of human umbilical cord mesenchymal stem cells (hUC-MSCs) is culturing-dependent, resulting in functional non-uniformness. To achieve the best clinical benefit, a comprehensive understanding of the origin of the heterogeneity in different culture systems can identify functional subgroups to direct the precise application of hUC-MSCs. Here, we create a single-cell transcriptome atlas of hUC-MSC in different culture systems for the identification of a subgroup of Ultroser-G-MSCs with high osteogenic and chondrogenic potentials featured by high expressions of IL1R1 and PDGFRA. Further experimental validations surprisingly reveal that IL1R1highPDGFRAhigh Ultroser-G-MSCs possess advantages over "traditional" hUC-MSCs in the treatments of modeled osteoarthritis, leading to a cell-cell communication network centered in Clusters 0 and 2. Moreover, we found that Wnt5 signaling is the key pathway for the dynamic transformation of osteogenic and chondrogenic phenotypes in hUC-MSC. Overall, the present study paves the way for the clarification of heterogenetic nature of hUC-MSC in different culture systems for the selection of optimal MSC types to achieve the precision on clinical treatments.
Collapse
Affiliation(s)
- Shihao Huang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Xinyu Xu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Jiaqi Guo
- Jiangsu Renocell Biotech Co. Ltd., Nanjing, China
| | - Zhuolan Li
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Yanlin Wu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Yuanyuan Liu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Qinyi Sun
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Sihan Wang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Huilin Yan
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Yueyan Su
- Jiangsu Renocell Biotech Co. Ltd., Nanjing, China
| | - Wei Guo
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
29
|
Strecanska M, Sekelova T, Smolinska V, Kuniakova M, Nicodemou A. Automated Manufacturing Processes and Platforms for Large-scale Production of Clinical-grade Mesenchymal Stem/ Stromal Cells. Stem Cell Rev Rep 2025; 21:372-389. [PMID: 39546186 PMCID: PMC11872983 DOI: 10.1007/s12015-024-10812-5] [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: 10/24/2024] [Indexed: 11/17/2024]
Abstract
Mesenchymal stem/stromal cells (MSCs) hold immense potential for regenerative medicine due to their remarkable regenerative and immunomodulatory properties. However, their therapeutic application requires large-scale production under stringent regulatory standards and Good Manufacturing Practice (GMP) guidelines, presenting significant challenges. This review comprehensively evaluates automated manufacturing processes and platforms for the scalable production of clinical-grade MSCs. Various large-scale culture vessels, including multilayer flasks and bioreactors, are analyzed for their efficacy in MSCs expansion. Furthermore, automated MSCs production platforms, such as Quantum® Cell Expansion System, CliniMACS Prodigy®, NANT001/ XL, CellQualia™, Cocoon® Platform, and Xuri™ Cell Expansion System W25 are reviewed and compared as well. We also underscore the importance of optimizing culture media specifically emphasizing the shift from fetal bovine serum to humanized or serum-free alternatives to meet GMP standards. Moreover, advances in alternative cryopreservation methods and controlled-rate freezing systems, that offer promising improvements in MSCs preservation, are discussed as well. In conclusion, advancing automated manufacturing processes and platforms is essential for realizing the full potential of MSCs-based regenerative medicine and accomplishing the increasing demand for cell-based therapies. Collaborative initiatives involving industry, academia, and regulatory bodies are emphasized to accelerate the translation of MSCs-based therapies into clinical practice.
Collapse
Affiliation(s)
- Magdalena Strecanska
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, Sasinkova 4, Bratislava, Bratislava, 811 08, Slovakia
- National Institute of Rheumatic Diseases, Nabrezie I. Krasku 4, Piestany, 921 12, Slovakia
| | - Tatiana Sekelova
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, Sasinkova 4, Bratislava, Bratislava, 811 08, Slovakia
- National Institute of Rheumatic Diseases, Nabrezie I. Krasku 4, Piestany, 921 12, Slovakia
| | - Veronika Smolinska
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, Sasinkova 4, Bratislava, Bratislava, 811 08, Slovakia
- National Institute of Rheumatic Diseases, Nabrezie I. Krasku 4, Piestany, 921 12, Slovakia
| | - Marcela Kuniakova
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, Sasinkova 4, Bratislava, Bratislava, 811 08, Slovakia
| | - Andreas Nicodemou
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, Sasinkova 4, Bratislava, Bratislava, 811 08, Slovakia.
- National Institute of Rheumatic Diseases, Nabrezie I. Krasku 4, Piestany, 921 12, Slovakia.
- GAMMA-ZA, Kollarova 8, Trencin, 911 01, Slovakia.
| |
Collapse
|
30
|
Zhong J, Li W, Li H, Zhang J, Hou Z, Wang X, Zhou E, Lu K, Zhuang W, Sang H. A self-forming bone membrane generated by periosteum-derived stem cell spheroids enhances the repair of bone defects. Acta Biomater 2025; 193:185-201. [PMID: 39742905 DOI: 10.1016/j.actbio.2024.12.058] [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: 09/19/2024] [Revised: 12/24/2024] [Accepted: 12/30/2024] [Indexed: 01/04/2025]
Abstract
The periosteum, a highly specialized thin tissue, is instrumental in contributing to as much as 70 % of early bone formation. Recognizing the periosteum's vital physiological roles, the fabrication of a biomimetic periosteum has risen as an auspicious strategy for addressing extensive bone defects. In the study, we obtained such biomimetic periosteum by utilizing periosteum-derived stem cells (PDSCs) spheroids. These spheroids are induced to spontaneously generate a bioactive membrane on a delicate 3D-printed polycaprolactone (PCL) substrate. This process yields a biomimetic periosteum rich in the resources needed for bone repair. The in vitro evaluations demonstrated that this membrane can act as a repository for growth factors and stem cells. The release kinetics confirmed a sustained delivery of BMP-2 and VEGF, which promoted enhanced osteogenesis and angiogenesis in vitro, respectively. The in vivo results further highlighted robust bone regeneration from critical cranial defects upon the application of this biomimetic periosteum. The biomimetic periosteum, easily harvested and potent in bioactivity, presents substantial clinical potential, particularly for the treatment of critical-sized bone defects. STATEMENT OF SIGNIFICANCE: PDSC theoretically demonstrates substantial potential in membrane construction, a value we've harnessed in this pioneering application. By employing cell spheroids, we've successfully integrated a substantial number of cells into the membrane framework. PDSC spheroids exhibit the remarkable ability to self-assemble into functional membranes, endowing them with robust biological capabilities that enhance their performance in biological systems. The in vitro evaluations demonstrated that this membrane can act as a repository for growth factors and stem cells. The in vivo bone repair facilitated by this membrane is notably effective, characterized by superior bone quality and accelerated formation rates. This process mirrors the natural intramembrane ossification, offering a promising approach to bone integration and regeneration.
Collapse
Affiliation(s)
- Jintao Zhong
- Department of Orthopedics, Shenzhen Hospital, Southern Medical University, Shenzhen 518000, PR China; The Third School of Clinical Medicine, Southern Medical University, Guangzhou 510000, PR China.
| | - Wenhua Li
- Department of Orthopedics, Shenzhen Hospital, Southern Medical University, Shenzhen 518000, PR China; The Third School of Clinical Medicine, Southern Medical University, Guangzhou 510000, PR China.
| | - Hetong Li
- Department of Orthopedics, Shenzhen Hospital, Southern Medical University, Shenzhen 518000, PR China.
| | - Jin Zhang
- Department of Orthopedics, Shenzhen Hospital, Southern Medical University, Shenzhen 518000, PR China; The Third School of Clinical Medicine, Southern Medical University, Guangzhou 510000, PR China.
| | - Zuoxu Hou
- Department of Orthopedics, Shenzhen Hospital, Southern Medical University, Shenzhen 518000, PR China.
| | - Xiao Wang
- Department of Orthopedics, Shenzhen Hospital, Southern Medical University, Shenzhen 518000, PR China.
| | - Enhui Zhou
- Department of Orthopedics, Shenzhen Hospital, Southern Medical University, Shenzhen 518000, PR China.
| | - Ke Lu
- Department of Orthopedics, Shenzhen Hospital, Southern Medical University, Shenzhen 518000, PR China.
| | - Weida Zhuang
- Department of Orthopedics, Shenzhen Hospital, Southern Medical University, Shenzhen 518000, PR China.
| | - Hongxun Sang
- Department of Orthopedics, Shenzhen Hospital, Southern Medical University, Shenzhen 518000, PR China; The Third School of Clinical Medicine, Southern Medical University, Guangzhou 510000, PR China.
| |
Collapse
|
31
|
Lúcio KA, Mendonça IC, Mendonça RA, Dos Santos VO. Thawing, Growth, and Maintenance of Vero Cell Lines. Methods Mol Biol 2025; 2913:211-222. [PMID: 40249438 DOI: 10.1007/978-1-0716-4458-4_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/19/2025]
Abstract
Vero cell line is derived from the kidney of an African green monkey and is one of the most common mammalian continuous cell lines used in research. Its deficiency in the production of type 1 interferons allows this cell line to be susceptible and permissible to replication of several viruses, including the yellow fever virus. Continuous cell culture in the laboratory can increase the risk of contamination, as well as loss of characteristics of interest in the strain. To minimize these risks a cultivation standard should be established. This chapter presents useful protocols for the maintenance of Vero cells for laboratorial routine, such as thawing vials from a cell bank stock, maintenance by subcultivation procedures, adaptation of the cells to grow in serum-free media, and production of cell culture plates for laboratory routine assays.
Collapse
Affiliation(s)
- Kelly Araujo Lúcio
- Laboratory of Virological Technology (LATEV), Institute of Technology in Immunobiologicals (Bio-Manguinhos), Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro, RJ, Brazil.
| | - Ivanielle Curvello Mendonça
- Laboratory of Virological Technology (LATEV), Institute of Technology in Immunobiologicals (Bio-Manguinhos), Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro, RJ, Brazil
| | - Rafael Araújo Mendonça
- Laboratory of Virological Technology (LATEV), Institute of Technology in Immunobiologicals (Bio-Manguinhos), Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro, RJ, Brazil
| | - Vanessa Oliveira Dos Santos
- Laboratory of Virological Technology (LATEV), Institute of Technology in Immunobiologicals (Bio-Manguinhos), Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro, RJ, Brazil
| |
Collapse
|
32
|
Specht SJ, Rohringer S, Hager P, Grasl C, Schmitt AM, Pach VJC, Ehrmann K, Baudis S, Liska R, Kiss H, Schneider KH, Podesser BK, Bergmeister H. Decellularized Extracellular Matrix and Polyurethane Vascular Grafts Have Positive Effects on the Inflammatory and Pro-Thrombotic State of Aged Endothelial Cells. J Biomed Mater Res A 2025; 113:e37830. [PMID: 39610352 DOI: 10.1002/jbm.a.37830] [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: 06/07/2024] [Revised: 10/14/2024] [Accepted: 10/19/2024] [Indexed: 11/30/2024]
Abstract
In vitro assessment of small-diameter synthetic vascular grafts usually uses standard cell culture conditions with early-passage cells. However, these conduits are mainly implanted in elderly patients and are subject to complex cellular interactions influenced by age and inflammation. Understanding these factors is central to the development of vascular grafts tailored to the specific needs of patients. In this study, the effects of aged endothelial cells subjected to pro- and anti-inflammatory agents and cultivated on a newly developed biodegradable electrospun thermoplastic polyurethane/poly(urethane-urea) blend (TPU/TPUU), on clinically available expanded polytetrafluorethylene (ePTFE), and on decellularized extracellular matrix (dECM) grafts were investigated. Young and aged endothelial cells were exposed to pro- and anti-inflammatory agents and characterized by morphology, migration capacity, and gene expression. In addition, the cells were seeded onto the various graft materials and examined microscopically alongside gene expression analyses. When exposed to pro-inflammatory cytokines, young and aged cells demonstrated signs of endothelial activation. Cells seeded on ePTFE showed reduced attachment and increased expression of pro-inflammatory genes compared with the other materials. dECM and TPU/TPUU substrates provided better support for endothelialization with aged cells under inflammatory conditions compared with ePTFE. Moreover, TPU/TPUU showed positive effects on reducing pro-thrombotic and pro-inflammatory gene expression in endothelial cells. Our results thus emphasize the importance of developing new synthetic graft materials as an alternative for clinically used ePTFE.
Collapse
Affiliation(s)
- Sophie J Specht
- Center for Biomedical Research and Translational Surgery, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Cardiovascular Research, Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Sabrina Rohringer
- Center for Biomedical Research and Translational Surgery, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Cardiovascular Research, Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Pia Hager
- Center for Biomedical Research and Translational Surgery, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Cardiovascular Research, Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Christian Grasl
- Center for Biomedical Research and Translational Surgery, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Cardiovascular Research, Vienna, Austria
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Vienna, Austria
| | - Anna-Maria Schmitt
- Center for Biomedical Research and Translational Surgery, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Cardiovascular Research, Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Virginia J C Pach
- Center for Biomedical Research and Translational Surgery, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Cardiovascular Research, Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Katharina Ehrmann
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
- Institute of Applied Synthetic Chemistry, Technical University of Vienna, Vienna, Austria
| | - Stefan Baudis
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
- Institute of Applied Synthetic Chemistry, Technical University of Vienna, Vienna, Austria
| | - Robert Liska
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
- Institute of Applied Synthetic Chemistry, Technical University of Vienna, Vienna, Austria
| | - Herbert Kiss
- Department of Obstetrics and Gynecology, Division of Obstetrics and Feto-Maternal Medicine, Medical University of Vienna, Vienna, Austria
| | - Karl H Schneider
- Center for Biomedical Research and Translational Surgery, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Cardiovascular Research, Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Bruno K Podesser
- Center for Biomedical Research and Translational Surgery, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Cardiovascular Research, Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Helga Bergmeister
- Center for Biomedical Research and Translational Surgery, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Cardiovascular Research, Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| |
Collapse
|
33
|
Lee J, Park SE, Kim M, Kim H, Kwon JY, Jeon HB, Chang JW, Lee J. Safety and Tolerability of Wharton's Jelly-Derived Mesenchymal Stem Cells for Patients With Duchenne Muscular Dystrophy: A Phase 1 Clinical Study. J Clin Neurol 2025; 21:40-52. [PMID: 39778566 PMCID: PMC11711273 DOI: 10.3988/jcn.2024.0299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 10/15/2024] [Accepted: 10/31/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND AND PURPOSE This study was an open-label, dose-escalation, phase 1 clinical trial to determine the safety and dose of EN001 for patients with Duchenne muscular dystrophy (DMD). EN001, developed by ENCell, are allogeneic early-passage Wharton's jelly-derived mesenchymal stem cells that originate at the umbilical cord, with preclinical studies demonstrating their high therapeutic efficacy for DMD. METHODS This phase 1 clinical trial explored the safety and tolerability of EN001 as a potential treatment option for patients with DMD. Six pediatric participants with DMD were divided into two subgroups of equal size: low-dose EN001 (5.0×10⁵ cells/kg) and high-dose EN001 (2.5×10⁶ cells/kg). All participants were monitored for 12 weeks after EN001 administration to assess its safety. Dose-limiting toxicity (DLT) was evaluated across 2 weeks post administration. Exploratory efficacy was evaluated by measuring serum creatine kinase levels, and functional evaluations-including spirometry, myometry, the North Star Ambulatory Assessment, and the 6-minute walk test-were conducted at week 12 and compared with the baseline values. RESULTS No participants experienced serious adverse events related to EN001 injection during the 12-week follow-up period. Mild adverse events included injection-related local erythema, edema, parosmia, and headache, but DLT was not observed. Functional evaluations at week 12 revealed no significant changes from baseline. CONCLUSIONS These results demonstrated that EN001 are safe and well tolerated for patients with DMD, and did not cause serious adverse events. The efficacy of EN001 could be confirmed through larger-scale future studies that incorporate repeated dosing and have a randomized controlled trial design.
Collapse
Affiliation(s)
- Jiwon Lee
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Sang Eon Park
- Cell and Gene Therapy Research Institute, ENCell Co. Ltd., Seoul, Korea
- Cell & Gene Therapy Institute, Samsung Medical Center, Seoul, Korea
| | - Mira Kim
- Clinical Development Department, ENCell Co. Ltd., Seoul, Korea
| | - Hyeongseop Kim
- Cell and Gene Therapy Research Institute, ENCell Co. Ltd., Seoul, Korea
- Cell & Gene Therapy Institute, Samsung Medical Center, Seoul, Korea
| | - Jeong-Yi Kwon
- Department of Physical and Rehabilitation Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hong Bae Jeon
- Cell and Gene Therapy Research Institute, ENCell Co. Ltd., Seoul, Korea
| | - Jong Wook Chang
- Cell and Gene Therapy Research Institute, ENCell Co. Ltd., Seoul, Korea
- Cell & Gene Therapy Institute, Samsung Medical Center, Seoul, Korea
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Korea
| | - Jeehun Lee
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
- Cell & Gene Therapy Institute, Samsung Medical Center, Seoul, Korea.
| |
Collapse
|
34
|
Atta H, Kassem DH, Kamal MM, Hamdy NM. Harnessing the ubiquitin proteasome system as a key player in stem cell biology. Biofactors 2025; 51:e2157. [PMID: 39843166 DOI: 10.1002/biof.2157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Accepted: 12/20/2024] [Indexed: 01/24/2025]
Abstract
Intracellular proteins take part in almost every body function; thus, protein homeostasis is of utmost importance. The ubiquitin proteasome system (UPS) has a fundamental role in protein homeostasis. Its main role is to selectively eradicate impaired or misfolded proteins, thus halting any damage that could arise from the accumulation of these malfunctioning proteins. Proteasomes have a critical role in controlling protein homeostasis in all cell types, including stem cells. We will discuss the role of UPS enzymes as well as the 26S proteasome complex in stem cell biology from several angles. First, we shall overview common trends of proteasomal activity and gene expression of different proteasomal subunits and UPS enzymes upon passaging and differentiation of stem cells toward various cell lineages. Second, we shall explore the effect of modulating proteasomal activity in stem cells and navigate through the interrelation between proteasomes' activity and various proteasome-related transcription factors. Third, we will shed light on curated microRNAs and long non-coding RNAs using various bioinformatics tools that might have a possible role in regulating UPS in stem cells and possibly, upon manipulation, can enhance the differentiation process into different lineages and/or delay senescence upon cell passaging. This will help to decipher the role played by individual UPS enzymes and subunits as well as various interrelated molecular mediators in stem cells' maintenance and/or differentiation and open new avenues in stem cell research. This can ultimately provide a leap toward developing novel therapeutic interventions related to proteasome dysregulation.
Collapse
Affiliation(s)
- Hind Atta
- School of Life and Medical Sciences, University of Hertfordshire Hosted By Global Academic Foundation, Cairo, Egypt
| | - Dina H Kassem
- Biochemistry Department, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Mohamed M Kamal
- Biochemistry Department, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
- Pharmacology and Biochemistry Department, Faculty of Pharmacy, The British University in Egypt, Cairo, Egypt
- Drug Research and Development Group, Health Research Center of Excellence, The British University in Egypt, Cairo, Egypt
| | - Nadia M Hamdy
- Biochemistry Department, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| |
Collapse
|
35
|
Chakraborty J, Roy C, Kalogeropoulou M, Mota C, Ghosh S, Moroni L. Protocol for developing shape-morphing 4D bioprinted magnetic constructs to promote articular cartilage regeneration using silk fibroin-gelatin bioink. STAR Protoc 2024; 5:103332. [PMID: 39446581 PMCID: PMC11539150 DOI: 10.1016/j.xpro.2024.103332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 08/08/2024] [Accepted: 08/30/2024] [Indexed: 10/26/2024] Open
Abstract
External magnetic fields can regulate cellular responses. Here, we present a protocol to fabricate magnetic constructs by 4D bioprinting with shape-morphing properties using silk fibroin-gelatin bioinks for articular cartilage regeneration. We illustrate the steps for magnetic bioink formulation, bioprinting, and chondrogenic induction of human bone marrow mesenchymal stem/stromal cells. We detail the steps to actuate the constructs using an external magnetic field and then characterize chondrogenesis. Magnetic field actuation may be helpful for mechanically activating constructs for articular cartilage regeneration. For complete details on the use and execution of this protocol, please refer to Chakraborty et al.1.
Collapse
Affiliation(s)
- Juhi Chakraborty
- Department of Textile and Fibre Engineering, Indian Institute of Technology Delhi, New Delhi 110016, India
| | - Chandrashish Roy
- Department of Textile and Fibre Engineering, Indian Institute of Technology Delhi, New Delhi 110016, India
| | - Maria Kalogeropoulou
- Department of Complex Tissue Regeneration, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6211 LK Maastricht, the Netherlands
| | - Carlos Mota
- Department of Complex Tissue Regeneration, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6211 LK Maastricht, the Netherlands
| | - Sourabh Ghosh
- Department of Textile and Fibre Engineering, Indian Institute of Technology Delhi, New Delhi 110016, India.
| | - Lorenzo Moroni
- Department of Complex Tissue Regeneration, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6211 LK Maastricht, the Netherlands.
| |
Collapse
|
36
|
Oh MS, Hong H, Lee OJ, Yi SH, Park HS, Lee JJ, Park CH, Im SW. Transcriptomic Changes in Human Tonsil-Derived Mesenchymal Stem Cells Across Culture Passages. Genes (Basel) 2024; 15:1626. [PMID: 39766894 PMCID: PMC11675251 DOI: 10.3390/genes15121626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/11/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND/OBJECTIVES Tonsil-derived mesenchymal stem cells (TMSCs) are in the limelight in regenerative medicine due to their high proliferation and differentiation potential. It is important to conduct studies to determine the optimal conditions for achieving the maximum yield while maintaining the optimal differentiation capacity of TMSCs. METHODS This study explores the impact of serial subculture on TMSCs by analyzing gene expression at passages 2, 4, 6, and 8. For each culture passage, genes with significant differences in RNA expression from previous passages were selected and their characteristics were observed performing enrichment analysis including KEGG (Kyoto Encyclopedia of Genes and Genomes) and Reactome pathway. RESULTS At each passage, a "cell cycle" term was ranked high with statistical significance in the KEGG and Reactome pathway. Cell cycle gene expression, including Cyclin-dependent kinases (CDKs) and cyclins, increased until passage 6, then decreased by passage 8. The cell cycle is known to be important not only for proliferation but also for determining whether stem cells maintain pluripotency or differentiate into various lineages. CONCLUSIONS The results suggest that cell cycle gene expression can guide the timing for differentiation induction, with passage 6 potentially being a critical point for initiating differentiation.
Collapse
Affiliation(s)
- Moon Sik Oh
- Nano-Bio Regenerative Medical Institute, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea; (M.S.O.); (H.H.); (O.J.L.); (H.S.P.)
| | - Heesun Hong
- Nano-Bio Regenerative Medical Institute, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea; (M.S.O.); (H.H.); (O.J.L.); (H.S.P.)
| | - Ok Joo Lee
- Nano-Bio Regenerative Medical Institute, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea; (M.S.O.); (H.H.); (O.J.L.); (H.S.P.)
| | - Su Hyeon Yi
- Department of Biochemistry and Molecular Biology, Kangwon National University School of Medicine, Chuncheon 24341, Republic of Korea;
| | - Hae Sang Park
- Nano-Bio Regenerative Medical Institute, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea; (M.S.O.); (H.H.); (O.J.L.); (H.S.P.)
- Department of Otorhinolaryngology-Head and Neck Surgery, Chuncheon Sacred Heart Hospital, Hallym University College of Medicine, Chuncheon 24253, Republic of Korea
- Institute of New Frontier Research, Hallym University College of Medicine, Chuncheon 24252, Republic of Korea;
| | - Jae-Jun Lee
- Institute of New Frontier Research, Hallym University College of Medicine, Chuncheon 24252, Republic of Korea;
- Department of Anesthesiology and Pain Medicine, Hallym University College of Medicine, Chuncheon 24252, Republic of Korea
| | - Chan Hum Park
- Nano-Bio Regenerative Medical Institute, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea; (M.S.O.); (H.H.); (O.J.L.); (H.S.P.)
- Department of Otorhinolaryngology-Head and Neck Surgery, Chuncheon Sacred Heart Hospital, Hallym University College of Medicine, Chuncheon 24253, Republic of Korea
| | - Sun-Wha Im
- Department of Biochemistry and Molecular Biology, Kangwon National University School of Medicine, Chuncheon 24341, Republic of Korea;
| |
Collapse
|
37
|
Zhang Z, Zhou C, Yu L. LEP O-GlcNAcylation inactivates NF-κB pathway by suppressing LEP protein level and thus mediates cellular senescence and osteogenic differentiation in mouse mesenchymal stem cells. BMC Mol Cell Biol 2024; 25:26. [PMID: 39695926 DOI: 10.1186/s12860-024-00523-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 11/29/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Cellular senescence is a key driver of decreased bone formation and osteoporosis. Leptin (LEP) has been implicated in cellular senescence and osteogenic differentiation. The aim of this study was to investigate the mechanisms by which LEP mediates cellular senescence and osteogenic differentiation. METHODS C3H10T1/2 cells were treated with etoposide to induce cellular senescence, which was assessed by β-galactosidase staining. Quantitative real-time PCR and western blotting were used to measure the levels of senescence markers p21 and p16, as well as osteogenic differentiation-related genes ALP, COL1A1, and RUNX2. Alkaline phosphatase (ALP) staining and alizarin red S staining were performed to evaluate osteogenic differentiation. The NF-κB pathway and O-GlcNAcylation were assessed by western blotting. RESULTS Etoposide treatment increased the number of senescent cells and the levels of p21 and p16, along with elevated LEP expression. These effects were reversed by LEP knockdown. Additionally, LEP knockdown increased ALP staining density and osteoblast mineralization nodules, as well as the mRNA and protein levels of ALP, COL1A1, and RUNX2, indicating that LEP knockdown promoted osteogenic differentiation in C3H10T1/2 cells. Mechanistically, LEP knockdown inactivated the NF-κB pathway by inhibiting the nuclear translocation of p65. Furthermore, OGT was found to promote O-GlcNAcylation of LEP at the S50 site. CONCLUSION Our findings demonstrated that O-GlcNAcylation of LEP inactivated the NF-κB pathway by reducing LEP protein levels, thereby inhibiting cellular senescence and promoting osteogenic differentiation in C3H10T1/2 cells. This study may provide a novel therapeutic target for the treatment of osteoporosis.
Collapse
Affiliation(s)
- Zhuang Zhang
- Macau University of Science and Technology, Faculty of Chinese Medicine, E205, Avenida Wai Long, Taipa, Macau, 999078, China
- The 2nd People's Hospital of Zhuhai, Zhuhai, China
| | - Chaoqing Zhou
- Department of Traumatology, The 2nd People's Hospital of Zhuhai, No.208 Yuehua Road, Zhuhai, Guangdong, 519020, China.
| | - Lili Yu
- Macau University of Science and Technology, Faculty of Chinese Medicine, E205, Avenida Wai Long, Taipa, Macau, 999078, China.
| |
Collapse
|
38
|
Di Nubila A, Doulgkeroglou MN, Gurdal M, Korntner SH, Zeugolis DI. In vitro and in vivo assessment of a non-animal sourced chitosan scaffold loaded with xeno-free umbilical cord mesenchymal stromal cells cultured under macromolecular crowding conditions. BIOMATERIALS AND BIOSYSTEMS 2024; 16:100102. [PMID: 40225717 PMCID: PMC11993840 DOI: 10.1016/j.bbiosy.2024.100102] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 06/06/2024] [Accepted: 10/08/2024] [Indexed: 04/15/2025] Open
Abstract
There is an increasing demand to not only accelerate the development of advanced therapy tissue engineered medicines, but to also eliminate xenogeneic materials from their development cycle. With these in mind, herein we first assessed the influence of carrageenan as macromolecular crowding agent to enhance and accelerate extracellular matrix deposition in xeno-free human umbilical cord mesenchymal stromal cell cultures and we developed and characterised a non-animal sourced chitosan scaffold. Following appropriate in vitro experimentation, a splinted nude mouse wound healing model was used to assess wound closure and scar size of non-treated control, non-animal sourced chitosan scaffold, non-animal sourced chitosan scaffold loaded with xeno-free human umbilical cord mesenchymal stromal cells and non-animal sourced chitosan scaffold loaded with xeno-free human umbilical cord mesenchymal stromal cells cultured under macromolecular crowding conditions groups. Across all three donors, carrageenan supplementation significantly increased collagen deposition at day 5, day 8 and day 11 without affecting cell morphology, viability, DNA concentration and metabolic activity. Through freeze drying, a non-animal sourced chitosan sponge was developed with appropriate structural and mechanical properties for wound healing applications. In vitro biological analysis made apparent that neither the scaffold nor macromolecular crowding negatively impacted xeno-free human umbilical cord mesenchymal stromal cell metabolic activity and proliferation. In vivo biological analysis revealed no significant differences between the groups in wound closure and scar size, raising question about the suitability of the model. In any case, this work sets the foundations for the development of completely xeno-free tissue engineered medicines.
Collapse
Affiliation(s)
- Alessia Di Nubila
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL) and Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, University of Galway, Galway, Ireland
| | - Meletios-Nikolaos Doulgkeroglou
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL) and Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, University of Galway, Galway, Ireland
| | - Mehmet Gurdal
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Charles Institute of Dermatology, Conway Institute of Biomolecular & Biomedical Research and School of Mechanical & Materials Engineering, University College Dublin (UCD), Dublin, Ireland
| | - Stefanie H. Korntner
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL) and Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, University of Galway, Galway, Ireland
| | - Dimitrios I. Zeugolis
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL) and Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, University of Galway, Galway, Ireland
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Charles Institute of Dermatology, Conway Institute of Biomolecular & Biomedical Research and School of Mechanical & Materials Engineering, University College Dublin (UCD), Dublin, Ireland
| |
Collapse
|
39
|
Jung H, Jung Y, Seo J, Bae Y, Kim HS, Jeong W. Roles of extracellular vesicles from mesenchymal stem cells in regeneration. Mol Cells 2024; 47:100151. [PMID: 39547584 DOI: 10.1016/j.mocell.2024.100151] [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: 09/30/2024] [Revised: 11/09/2024] [Accepted: 11/10/2024] [Indexed: 11/17/2024] Open
Abstract
Mesenchymal stem cells (MSCs) are highly valued in regenerative medicine due to their ability to self-renew and differentiate into various cell types. Their therapeutic benefits are primarily due to their paracrine effects, in particular through extracellular vesicles (EVs), which are related to intercellular communication. Recent advances in EV production and extraction technologies highlight the potential of MSC-derived EVs (MSC-EVs) in tissue engineering and regenerative medicine. MSC-EVs offer several advantages over traditional cell therapies, including reduced toxicity and immunogenicity compared with whole MSCs. EVs carrying functional molecules such as growth factors, cytokines, and miRNAs play beneficial roles in tissue repair, fibrosis treatment, and scar prevention by promoting angiogenesis, skin cell migration, proliferation, extracellular matrix remodeling, and reducing inflammation. Despite the potential of MSC-EVs, there are several limitations to their use, including variability in quality, the need for standardized methods, low yield, and concerns about the composition of EVs and the potential risks. Overall, MSC-EVs are a promising alternative to cell-based therapies, and ongoing studies aim to understand their actions and optimize their use for better clinical outcomes in wound healing and skin regeneration.
Collapse
Affiliation(s)
- Hyeseong Jung
- Department of Biomedical Science, Catholic Kwandong University, Gangneung 25601, Republic of Korea
| | - Yuyeon Jung
- Department of Dental Hygiene, Catholic Kwandong University, Gangneung 25601, Republic of Korea
| | - Junsik Seo
- Department of Biomedical Science, Catholic Kwandong University, Gangneung 25601, Republic of Korea
| | - Yeongju Bae
- Department of Biomedical Science, Catholic Kwandong University, Gangneung 25601, Republic of Korea; Research Center for Marine Bio-Food and Medicine, Catholic Kwandong University, Gangneung 25601, Republic of Korea
| | - Han-Soo Kim
- Department of Biomedical Science, Catholic Kwandong University, Gangneung 25601, Republic of Korea
| | - Wooyoung Jeong
- Department of Biomedical Science, Catholic Kwandong University, Gangneung 25601, Republic of Korea; Research Center for Marine Bio-Food and Medicine, Catholic Kwandong University, Gangneung 25601, Republic of Korea.
| |
Collapse
|
40
|
Cheng G, Wang X, Zhang F, Wang K, Li Y, Guo T, Xu N, Wei W, Yan S. Reparative homing of bone mesenchymal stem cells induced by iMSCs via the SDF-1/CXCR4 axis for articular cartilage defect restoration. Biomed Pharmacother 2024; 181:117649. [PMID: 39536539 DOI: 10.1016/j.biopha.2024.117649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 10/17/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND The intrinsic healing ability of articular cartilage is poor after injury or illness, and untreated injury could lead to cartilage degeneration and ultimately osteoarthritis. iMSCs are derived from embryonic induced pluripotent stem cells and have strong therapeutic capabilities in the repair of cartilage defects, while the mechanism of action is unclear. The aim of this study is to clarify the repair mode of iMSCs on cartilage defects in rat knee joints, elucidate the chemotactic effect of iMSCs on autologous BMSCs in rats, and provide a basis for the treatment of cartilage defects and endogenous regeneration with iMSCs. METHODS Based on the establishment of the rat cartilage defect model, the reparative effect of iMSCs on the rat cartilage defect was evaluated. The cartilage repair was evaluated by quantitative score, H&E staining, Masson staining and Safranin-O staining, and the metabolic changes of iMSCs in the joint cavity were detected in vivo. The expression of SOX9, CD29, CD90, ColⅠ, ColⅡ, PCNA, SDF-1, and CXCR4 was detected by immunohistochemistry (IHC), IF, flow cytometry, respectively. After co-culturing iMSCs with BMSCs in vitro, the expression of CXCR4/SDF-1 on the cell membrane surface of BMSCs was detected by western blotting.; The level of p-Akt and p-Erk1/2 in total protein of BMSCs were detected by western blotting. SIGNIFICANCE Our research results provide experimental evidence for the treatment of cartilage defects and endogenous regeneration with iMSCs; This also provides new ideas for the clinical treatment of cartilage defects using iMSCs.
Collapse
Affiliation(s)
- Gang Cheng
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China; Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei 230032, China
| | - Xulei Wang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China; Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei 230032, China; Laboratory Animal Center, Anhui Medical University, Hefei 230032, China
| | - Feng Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China; Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei 230032, China
| | - Kang Wang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China; Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei 230032, China
| | - Ying Li
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China; Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei 230032, China
| | - Tingting Guo
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China; Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei 230032, China
| | - Nuo Xu
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China; Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei 230032, China
| | - Wei Wei
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China; Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei 230032, China.
| | - Shangxue Yan
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China; Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei 230032, China; Laboratory Animal Center, Anhui Medical University, Hefei 230032, China.
| |
Collapse
|
41
|
Heinrichs DP, Maldonado VV, Ardana IKK, Porter RM, Samsonraj RM. Assessing the Effects of Dasatinib on Mesenchymal Stem/Stromal Cells. Cell Mol Bioeng 2024; 17:609-618. [PMID: 39926377 PMCID: PMC11799476 DOI: 10.1007/s12195-024-00830-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 10/11/2024] [Indexed: 02/11/2025] Open
Abstract
Introduction Progressive aging, or senescence, of mesenchymal stem/stromal cells (MSCs) is a major obstacle faced when trying to culture potent stem cells for use in therapy. Senescent cells are irreversibly nondividing cells that cease performing critical functional effects. Elimination of senescent cells using biochemical means, such as the use of senolytic drugs like dasatinib, may be useful in retaining the viable and proliferating populations of the cells. Methods An in vitro approach was used to investigate the effect of dasatinib on phenotypic, genotypic, and immunomodulatory functionality of osteogenic and adipogenic differentiated MSCs. Replicative senescence was achieved through multiple sub-culturing in vitro, then senescent and non-senescent cultures were treated with a standard dosage of dasatinib. MSCs were then differentiated into osteogenic, adipogenic or chondrogenic cultures using conditioned media to be tested for the three criteria being investigated. Results Significant changes were observed in these criteria, indicated by evidence gathered from proliferation and indoleamine 2,3 dioxygenase activity assays. Phenotypic results of dasatinib were shown to reduce the population of senescent MSCs while allowing non-senescent MSCs to continue differentiating and proliferating without interference from senescent cells. Genotypic results showed no change to upregulation in markers associated with osteogenic and adipogenic cells when exposed to dasatinib. Indoleamine Dioxygenase activity showed insignificant differences in cells exposed to dasatinib versus control groups, providing evidence against compromised cellular immune function. Conclusion This investigation provides insight into how dasatinib effects MSCs functional ability and provides a better understanding of the function of senolytic agents.
Collapse
Affiliation(s)
- David P. Heinrichs
- Department of Biomedical Engineering, Engineering Research Center, College of Engineering, University of Arkansas, 700 W Research Center Boulevard, Fayetteville, AR 72701 USA
| | - Vitali V. Maldonado
- Department of Biomedical Engineering, Engineering Research Center, College of Engineering, University of Arkansas, 700 W Research Center Boulevard, Fayetteville, AR 72701 USA
| | - I. Kade K. Ardana
- Cell and Molecular Biology Interdisciplinary Program, University of Arkansas, Fayetteville, AR 72701 USA
| | - Ryan M. Porter
- Department of Orthopedic Surgery, University of Arkansas for Medical Sciences, Little Rock, AR 72205 USA
| | - Rebekah M. Samsonraj
- Department of Biomedical Engineering, Engineering Research Center, College of Engineering, University of Arkansas, 700 W Research Center Boulevard, Fayetteville, AR 72701 USA
- Cell and Molecular Biology Interdisciplinary Program, University of Arkansas, Fayetteville, AR 72701 USA
- Department of Orthopedic Surgery, University of Arkansas for Medical Sciences, Little Rock, AR 72205 USA
| |
Collapse
|
42
|
Kim D, Lee C, Kim M, Park JH. Gold Kiwi-Derived Nanovesicles Mitigate Ultraviolet-Induced Photoaging and Enhance Osteogenic Differentiation in Bone Marrow Mesenchymal Stem Cells. Antioxidants (Basel) 2024; 13:1474. [PMID: 39765803 PMCID: PMC11673108 DOI: 10.3390/antiox13121474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 11/25/2024] [Accepted: 11/28/2024] [Indexed: 01/11/2025] Open
Abstract
Bone marrow mesenchymal stem cells (BM-MSCs) play a crucial role in bone formation through their ability to differentiate into osteoblasts. Aging, however, detrimentally affects the differentiation and proliferation capacities of BM-MSCs, consequently impairing bone regeneration. Thus, mitigating the aging effects on BM-MSCs is vital for addressing bone-related pathologies. In this study, we demonstrate that extracellular nanovesicles isolated from gold kiwi (GK-NVs) protect human BM-MSCs from ultraviolet (UV)-induced photoaging, thereby alleviating aging-related impairments in cellular functions that are crucial for bone homeostasis. Notably, GK-NVs were efficiently taken up by BM-MSCs without causing cytotoxicity. GK-NVs reduced intracellular reactive oxygen species (ROS) levels upon UV irradiation, restoring impaired proliferation and migration capabilities. Furthermore, GK-NVs corrected the skewed differentiation capacities of UV-irradiated BM-MSCs by enhancing osteoblast differentiation, as evidenced by the increased expression in osteoblast-specific genes and the calcium deposition, and by reducing adipocyte differentiation, as indicated by the decreased lipid droplet formation. These findings position GK-NVs as a promising biomaterial for the treatment of bone-related diseases such as osteoporosis.
Collapse
Affiliation(s)
- Doyeon Kim
- Department of Biomedical Science, Kangwon National University, Chuncheon-si 24341, Gangwon-do, Republic of Korea; (D.K.); (C.L.); (M.K.)
| | - Chanho Lee
- Department of Biomedical Science, Kangwon National University, Chuncheon-si 24341, Gangwon-do, Republic of Korea; (D.K.); (C.L.); (M.K.)
| | - Manho Kim
- Department of Biomedical Science, Kangwon National University, Chuncheon-si 24341, Gangwon-do, Republic of Korea; (D.K.); (C.L.); (M.K.)
| | - Ju Hyun Park
- Department of Biomedical Science, Kangwon National University, Chuncheon-si 24341, Gangwon-do, Republic of Korea; (D.K.); (C.L.); (M.K.)
- Institute of Molecular Science and Fusion Technology, Kangwon National University, Chuncheon-si 24341, Gangwon-do, Republic of Korea
| |
Collapse
|
43
|
Wang X, Wang Q, Xia Z, Yang Y, Dai X, Zhang C, Wang J, Xu Y. Mesenchymal stromal cell therapies for traumatic neurological injuries. J Transl Med 2024; 22:1055. [PMID: 39578845 PMCID: PMC11583761 DOI: 10.1186/s12967-024-05725-3] [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: 07/09/2024] [Accepted: 10/01/2024] [Indexed: 11/24/2024] Open
Abstract
Improved treatment options are urgently needed for neurological injuries resulting from trauma or iatrogenic events causing long-term disabilities that severely impact patients' quality of life. In vitro and animal studies have provided promising proof-of-concept examples of regenerative therapies using mesenchymal stromal cells (MSC) for a wide range of pathological conditions. Over the previous decade, various MSC-based therapies have been investigated in clinical trials to treat traumatic neurological injuries. However, while the safety and feasibility of MSC treatments has been established, the patient outcomes in these studies have not demonstrated significant success in the translation of MSC regenerative therapy for the treatment of human brain and spinal cord injuries. Herein, we have reviewed the literature and ongoing registered trials on the application of MSC for the treatment of traumatic brain injury, traumatic spinal cord injury, and peripheral nerve injury. We have focused on the shortcomings and technological hurdles that must be overcome to further advance clinical research to phase 3 trials, and we discuss recent advancements that represent potential solutions to these obstacles to progress.
Collapse
Affiliation(s)
- Xiujuan Wang
- Technology Department, Tianjin Everunion Biotechnology Co., Ltd, SOHO Nexus Center, No. 19A East 3rd Ring North Road, Chaoyang District, Beijing, 100020, China
| | - Qian Wang
- HELP Therapeutics Co., Ltd, No. 568 Longmian Avenue, Jiangning District, Nanjing, 211166, Jiangsu Province, China
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, E12 Avenida da Universidade, Macau, 519000, SAR, China
| | - Ziyao Xia
- Department of Ophthalmology, Peking University Third Hospital, No. 49 North Garden Road, Haidian District, Beijing, 100191, China
- Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, No. 49 North Garden Road, Haidian District, Beijing, 100191, China
| | - Ying Yang
- Technology Department, Tianjin Everunion Biotechnology Co., Ltd, SOHO Nexus Center, No. 19A East 3rd Ring North Road, Chaoyang District, Beijing, 100020, China
| | - Xunan Dai
- Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, No. 49 North Garden Road, Haidian District, Beijing, 100191, China
| | - Chun Zhang
- Department of Ophthalmology, Peking University Third Hospital, No. 49 North Garden Road, Haidian District, Beijing, 100191, China.
- Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, No. 49 North Garden Road, Haidian District, Beijing, 100191, China.
| | - Jiaxian Wang
- HELP Therapeutics Co., Ltd, No. 568 Longmian Avenue, Jiangning District, Nanjing, 211166, Jiangsu Province, China.
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, E12 Avenida da Universidade, Macau, 519000, SAR, China.
| | - Yongsheng Xu
- Technology Department, Tianjin Everunion Biotechnology Co., Ltd, SOHO Nexus Center, No. 19A East 3rd Ring North Road, Chaoyang District, Beijing, 100020, China.
- Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, No. 49 North Garden Road, Haidian District, Beijing, 100191, China.
| |
Collapse
|
44
|
Setiawan AM, Kamarudin TA. Differentiation of Human Mesenchymal Stem Cells into Corneal Epithelial Cells: Current Progress. Curr Issues Mol Biol 2024; 46:13281-13295. [PMID: 39727920 DOI: 10.3390/cimb46120792] [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: 09/17/2024] [Revised: 11/13/2024] [Accepted: 11/17/2024] [Indexed: 12/28/2024] Open
Abstract
The limited availability of corneal tissue grafts poses significant challenges in the treatment of corneal blindness. Novel treatment utilizes stem cell grafts transplanted from the healthy side of the cornea to the damaged side. However, this procedure is only possible for those who have one-sided corneal blindness. Human stem cells offer promising potential for corneal tissue engineering, providing an alternative solution. Among the different types of stem cells, mesenchymal stem cells (MSCs) stand out due to their abundance and ease of isolation. Human MSCs can be derived from bone marrow, adipose, and umbilical cord tissues. Differentiating MSC toward corneal tissue can be achieved through several methods including chemical induction and co-culture with adult corneal cells such as human limbal epithelial stem cells (LESCs) and human corneal epithelial cells (hTCEpi). Adipose-derived stem cells (ADSCs) are the most common type of MSC that has been studied for corneal differentiation. Corneal epithelial cells are the most common corneal cell type targeted by researchers for corneal differentiation. Chemical induction with small molecules, especially bone morphogenetic protein 4 (BMP4), all-trans retinoic acid (ATRA), and epidermal growth factor (EGF), has gained more popularity in corneal epithelial cell differentiation. This review highlights the current progress in utilizing MSCs for corneal differentiation studies, showcasing their potential to revolutionize treatments for corneal blindness.
Collapse
Affiliation(s)
- Abdul Malik Setiawan
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia
- Department of Anatomy, Maulana Malik Ibrahim State Islamic University, Malang 65144, Indonesia
| | - Taty Anna Kamarudin
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia
| |
Collapse
|
45
|
Jin W, Jiang S, Liu X, He Y, Li T, Ma J, Chen Z, Lu X, Liu X, Shou W, Jin G, Ding J, Zhou Z. Disorganized chromatin hierarchy and stem cell aging in a male patient of atypical laminopathy-based progeria mandibuloacral dysplasia type A. Nat Commun 2024; 15:10046. [PMID: 39567511 PMCID: PMC11579472 DOI: 10.1038/s41467-024-54338-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 11/01/2024] [Indexed: 11/22/2024] Open
Abstract
Studies of laminopathy-based progeria offer insights into aging-associated diseases and highlight the role of LMNA in chromatin organization. Mandibuloacral dysplasia type A (MAD) is a largely unexplored form of atypical progeria that lacks lamin A post-translational processing defects. Using iPSCs derived from a male MAD patient carrying homozygous LMNA p.R527C, premature aging phenotypes are recapitulated in multiple mesenchymal lineages, including mesenchymal stem cells (MSCs). Comparison with 26 human aging MSC expression datasets reveals that MAD-MSCs exhibit the highest similarity to senescent primary human MSCs. Lamina-chromatin interaction analysis reveals reorganization of lamina-associating domains (LADs) and repositioning of non-LAD binding peaks may contribute to the observed accelerated senescence. Additionally, 3D genome organization further supports hierarchical chromatin disorganization in MAD stem cells, alongside dysregulation of genes involved in epigenetic modification, stem cell fate maintenance, senescence, and geroprotection. Together, these findings suggest LMNA missense mutation is linked to chromatin alterations in an atypical progeroid syndrome.
Collapse
Affiliation(s)
- Wei Jin
- Guangdong Cardiovascular Institute, Medical Research Institute, Guangdong Key Laboratory for Immune and Genetic Research of Chronic Nephropathy, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong, Hong Kong SAR
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR
| | - Shaoshuai Jiang
- RNA Biomedical Institute, Sun Yat-Sen Memorial Hospital, Key Laboratory for Stem Cells and Tissue Engineering of Ministry of Education, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Xinyi Liu
- RNA Biomedical Institute, Sun Yat-Sen Memorial Hospital, Key Laboratory for Stem Cells and Tissue Engineering of Ministry of Education, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Yi He
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR
| | - Tuo Li
- Department of Endocrinology, Changzheng Hospital, Shanghai, China
| | - Jingchun Ma
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR
| | - Zhihong Chen
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR
- Department of Andrology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xiaomei Lu
- Dongguan Institute of Pediatrics, Dongguan Children's Hospital, Dongguan, China
| | - Xinguang Liu
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Institute of Aging Research, Institute of Biochemistry & Molecular Biology, Guangdong Medical University, Dongguan, China
| | - Weinian Shou
- Indiana University School of Medicine, Indianapolis, IN, USA
| | - Guoxiang Jin
- Guangdong Cardiovascular Institute, Medical Research Institute, Guangdong Key Laboratory for Immune and Genetic Research of Chronic Nephropathy, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
| | - Junjun Ding
- RNA Biomedical Institute, Sun Yat-Sen Memorial Hospital, Key Laboratory for Stem Cells and Tissue Engineering of Ministry of Education, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China.
| | - Zhongjun Zhou
- Guangdong Cardiovascular Institute, Medical Research Institute, Guangdong Key Laboratory for Immune and Genetic Research of Chronic Nephropathy, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China.
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR.
- University of Hong Kong-Shenzhen Hospital, Shenzhen, China.
| |
Collapse
|
46
|
Melo WGGD, Bezerra DDO, Silva ERDDFS, Campêlo CB, Carvalho MAMD, Argôlo Neto NM. Behavioral dynamics of medicinal signaling cells from porcine bone marrow in long-term culture. Can J Physiol Pharmacol 2024; 102:672-679. [PMID: 39189463 DOI: 10.1139/cjpp-2023-0458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/28/2024]
Abstract
Medicinal signaling cells (MSC) hold promise for regenerative medicine due to their ability to repair damaged tissues. However, their effectiveness can be affected by how long they are cultured in the lab. This study investigated how passage number influences key properties for regenerative medicine of pig bone marrow MSC. The medicinal signiling cells derived from pig bone marrow (BM-MSC) were cultured in D-MEM High Glucose supplemented with 15% foetal bovine serum until the 25th passage and assessed their growth, viability, ability to differentiate into different cell types (plasticity), and cell cycle activity. Our findings showed that while the cells remained viable until the 25th passage, their ability to grow and differentiate declined after the 5th passage. Additionally, cells in later passages spent more time in a resting phase, suggesting reduced activity. In conclusion, the number of passages is a critical factor for maintaining ideal MSC characteristics. From the 9th passage BM-MSC exhibit decline in proliferation, differentiation potential, and cell cycle activity. Given this, it is possible to suggest that the use of 5th passage cells is the most suitable for therapeutic applications.
Collapse
Affiliation(s)
- Wanderson Gabriel Gomes de Melo
- Núcleo Integrado de Morfologia e Pesquisa com Células-Tronco, Programa de Pós Graduação em Tecnologias Aplicadas a Animais de Interesse Regional, Universidade Federal do Piauí, Teresina, Piauí, Brazil
| | - Dayseanny de Oliveira Bezerra
- Núcleo Integrado de Morfologia e Pesquisa com Células-Tronco, Programa de Pós Graduação em Tecnologias Aplicadas a Animais de Interesse Regional, Universidade Federal do Piauí, Teresina, Piauí, Brazil
| | | | - Camile Benício Campêlo
- Núcleo Integrado de Morfologia e Pesquisa com Células-Tronco, Programa de Pós Graduação em Tecnologias Aplicadas a Animais de Interesse Regional, Universidade Federal do Piauí, Teresina, Piauí, Brazil
| | - Maria Acelina Martins de Carvalho
- Núcleo Integrado de Morfologia e Pesquisa com Células-Tronco, Programa de Pós Graduação em Tecnologias Aplicadas a Animais de Interesse Regional, Universidade Federal do Piauí, Teresina, Piauí, Brazil
| | - Napoleão Martins Argôlo Neto
- Núcleo Integrado de Morfologia e Pesquisa com Células-Tronco, Programa de Pós Graduação em Tecnologias Aplicadas a Animais de Interesse Regional, Universidade Federal do Piauí, Teresina, Piauí, Brazil
| |
Collapse
|
47
|
Kord-Parijaee E, Ferdosi-Shahandashti E, Bakhshandeh B, Pournajaf A. Enhancing Gingival-Derived Mesenchymal Stem Cell Potential in Tissue Engineering and Regenerative Medicine Through Paraprobiotics. Tissue Eng Part C Methods 2024; 30:512-521. [PMID: 39165236 DOI: 10.1089/ten.tec.2024.0169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/22/2024] Open
Abstract
Gingival-derived mesenchymal stem cells (GMSCs) stand for a unique source of mesenchymal stem cells (MSCs) isolated from a neural crest origin with potential application in regenerative medicine. However, there are some limitations to the usage of these cells in clinical cell therapy such as reduced cell number and undesirable differentiation of the cell throughout frequent passages. Nowadays, studies have applied manipulation strategies to improve MSCs' effectiveness in clinical therapy. Among all of the materials used for this purpose, there is a growing trend for the use of biomaterials such as probiotic extracts or their conditioned media due to their lower toxicity. In the present study, we utilized extracts from Lactobacillus reuteri and Lactobacillus rhamnosus to assess their potential to enhance the function of GMSCs. We compared the effectiveness of these bacterial extracts to determine their relative efficacy. Bacterial extracts of two lactic acid bacteria were prepared using an ultrasonic homogenizing device. The impact of these bacterial extracts on GMSCs was evaluated through Alizarin Red and Oil Red O staining, cell counting by Trypan Blue staining, and real-time polymerase chain reaction. The findings of our study indicate that the administration of 50 μg/mL L. rhamnosus extract resulted in a greater enhancement of stemness marker expression, osteogenic differentiation, and proliferation of GMSCs compared with an equivalent concentration of L. reuteri extract. Neither of these bacterial extracts revealed any effect on the differentiation of the GMSCs into the adipogenic lineage. These findings suggest that L. rhamnosus extract could be more effective at promoting GMSCs' efficacy in tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Ensiyeh Kord-Parijaee
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
- Biomedical and Microbial Advanced Technologies (BMAT) Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Elaheh Ferdosi-Shahandashti
- Biomedical and Microbial Advanced Technologies (BMAT) Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Behnaz Bakhshandeh
- Department of Biotechnology, College of Science, University of Tehran, Tehran, Iran
| | - Abazar Pournajaf
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| |
Collapse
|
48
|
Hu M, Liu R, Chen X, Yan S, Gao J, Zhang Y, Wu D, Sun L, Jia Z, Sun G, Liu D. Metabolomics Dysfunction in Replicative Senescence of Periodontal Ligament Stem Cells Regulated by AMPK Signaling Pathway. Stem Cells Dev 2024; 33:607-615. [PMID: 39302052 DOI: 10.1089/scd.2024.0112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/22/2024] Open
Abstract
Periodontal ligament mesenchymal stem cells (PDLSCs) are a promising cell resource for stem cell-based regenerative medicine in dentistry, but they inevitably acquire a senescent phenotype after prolonged in vitro expansion. The key regulators of PDLSCs during replicative senescence remain unclear. Here, we sought to elucidate the role of metabolomic changes in determining the cellular senescence of PDLSCs. PDLSCs were cultured to passages 4, 10, and 20. The senescent phenotypes of PDLSCs were detected, and metabolomics analysis was performed. We found that PDLSCs manifested senescence phenotype during passaging. Metabolomics analysis showed that the metabolism of replicative senescence in PDLSCs varied significantly. The AMP-activated protein kinase (AMPK) signaling pathway was closely related to adenosine monophosphate (AMP) levels. The AMP:ATP ratio increased in senescent PDLSCs; however, the levels of p-AMPK, FOXO1 and FOXO3a decreased with senescence. We treated PDLSCs with an activator of the AMPK pathway (AICAR) and observed that the phosphorylated AMPK level at P20 PDLSCs was partially restored. These data delineate that the metabolic process of PDLSCs is active in the early stage of senescence and attenuated in the later stages of senescence; however, the sensitivity of AMPK phosphorylation sites is impaired, causing senescent PDLSCs to fail to respond to changes in energy metabolism.
Collapse
Affiliation(s)
- Meilin Hu
- Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, Tianjin Medical University School of Stomatology, Tianjin Medical University, Heping, China
| | - Ruiqi Liu
- Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, Tianjin Medical University School of Stomatology, Tianjin Medical University, Heping, China
| | - Xiaoyu Chen
- Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, Tianjin Medical University School of Stomatology, Tianjin Medical University, Heping, China
| | - Shen Yan
- Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, Tianjin Medical University School of Stomatology, Tianjin Medical University, Heping, China
| | - Jian Gao
- Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, Tianjin Medical University School of Stomatology, Tianjin Medical University, Heping, China
| | - Yao Zhang
- Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, Tianjin Medical University School of Stomatology, Tianjin Medical University, Heping, China
| | - Di Wu
- Division of Oral and Craniofacial Health Sciences, Adams School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Lu Sun
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, Michigan, USA
| | - Zhi Jia
- Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, Tianjin Medical University School of Stomatology, Tianjin Medical University, Heping, China
| | - Guangyunhao Sun
- Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, Tianjin Medical University School of Stomatology, Tianjin Medical University, Heping, China
| | - Dayong Liu
- Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, Tianjin Medical University School of Stomatology, Tianjin Medical University, Heping, China
| |
Collapse
|
49
|
Košuthová H, Fecskeová LK, Matejová J, Slovinská L, Morávek M, Bártová Z, Harvanová D. Effects of Replicative Senescence of Human Chorionic MSCs on their EV-miRNA Profile. Stem Cell Rev Rep 2024; 20:2318-2335. [PMID: 39305404 PMCID: PMC11554840 DOI: 10.1007/s12015-024-10790-8] [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: 09/15/2024] [Indexed: 11/12/2024]
Abstract
Chorionic mesenchymal stromal cells (CHO-MSCs) and their extracellular vesicles (EVs) are becoming increasingly popular, since chorion is ethically harmless and an easily accessible source of MSCs. However, until now there is only a limited number of studies with a thorough characterization of CHO-MSCs derived EVs and their miRNA profile. In this study, we monitored changes in the EV-miRNA profile between early and late passage of human CHO-MSCs. First, senescence of CHO-MSCs was induced by serial passaging and confirmed by morphological changes, shortened telomeres and changes in the expression of selected genes. The expression of MSCs-specific surface markers CD73, CD90, CD105 did not change with increasing passages. Next, EVs and their miRNA profiles were compared between early vs late passage cells. Number of EVs and their size were not significantly changed. Seven of the top 10 most expressed EV-miRNAs were common to both early and late passages. A differential expression study between early and late passages identified 37 significantly differentially expressed EV-miRNAs, out of which 23 were found to be associated with pathways of cellular senescence based on KEGG pathway analysis. A set of 9 miRNAs were identified as the most frequently associated with senescence and/or with the most altered expression between early and late passages, out of which miR-145-5p, miR-335-5p and miR-199b-3p were the most significant downregulated miRNAs in late passages. The most upregulated EV-miRNAs were miR-1307-3p, miR-3615 and miR320b. Targeting these miRNAs in future experiments may prolong the therapeutic potential of CHO-MSCs and their EVs.
Collapse
Affiliation(s)
- Hedviga Košuthová
- Associated Tissue Bank, Faculty of Medicine, Pavol Jozef Safarik University and Louis Pasteur University Hospital, Trieda SNP 1, 04011, Kosice, Slovakia
| | - Lívia K Fecskeová
- Associated Tissue Bank, Faculty of Medicine, Pavol Jozef Safarik University and Louis Pasteur University Hospital, Trieda SNP 1, 04011, Kosice, Slovakia.
| | - Jana Matejová
- Associated Tissue Bank, Faculty of Medicine, Pavol Jozef Safarik University and Louis Pasteur University Hospital, Trieda SNP 1, 04011, Kosice, Slovakia
| | - Lucia Slovinská
- Associated Tissue Bank, Faculty of Medicine, Pavol Jozef Safarik University and Louis Pasteur University Hospital, Trieda SNP 1, 04011, Kosice, Slovakia
| | - Marko Morávek
- Associated Tissue Bank, Faculty of Medicine, Pavol Jozef Safarik University and Louis Pasteur University Hospital, Trieda SNP 1, 04011, Kosice, Slovakia
| | - Zuzana Bártová
- Institute of Geotechnics of the Slovak Academy of Sciences, Watsonova 45, 040 01, Kosice, Slovakia
| | - Denisa Harvanová
- Associated Tissue Bank, Faculty of Medicine, Pavol Jozef Safarik University and Louis Pasteur University Hospital, Trieda SNP 1, 04011, Kosice, Slovakia
| |
Collapse
|
50
|
Liu C, Li Q, Ma JX, Lu B, Criswell T, Zhang Y. Exosome-mediated renal protection: Halting the progression of fibrosis. Genes Dis 2024; 11:101117. [PMID: 39263535 PMCID: PMC11388648 DOI: 10.1016/j.gendis.2023.101117] [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: 02/27/2023] [Revised: 08/09/2023] [Accepted: 08/13/2023] [Indexed: 09/13/2024] Open
Abstract
Renal fibrosis is a complex and multifactorial process that involves inflammation, cell proliferation, collagen, and fibronectin deposition in the kidney, ultimately leading to chronic kidney disease and even end-stage renal disease. The main goal of treatment is to slow down or halt the progression of fibrosis and to improve or preserve kidney function. Despite significant progress made in understanding the underlying mechanisms of renal fibrosis, current therapies have limited renal protection as the disease progresses. Exosomes derived from stem cells are a newer area of research for the treatment of renal fibrosis. Exosomes as nano-sized extracellular vesicles carry proteins, lipids, and nucleic acids, which can be taken up by local or distant cells, serving as mediators of intercellular communication and as drug delivery vehicles. Exosomes deliver molecules that reduce inflammation, renal fibrosis and extracellular matrix protein production, and promote tissue regeneration in animal models of kidney disease. Additionally, they have several advantages over stem cells, such as being non-immunogenic, having low risk of tumor formation, and being easier to produce and store. This review describes the use of natural and engineered exosomes containing therapeutic agents capable of mediating anti-inflammatory and anti-fibrotic processes during both acute kidney injury and chronic kidney disease. Exosome-based therapies will be compared with stem cell-based treatments for tissue regeneration, with a focus on renal protection. Finally, future directions and strategies for improving the therapeutic efficacy of exosomes are discussed.
Collapse
Affiliation(s)
- Chuanqi Liu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Qingfeng Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Jian-Xing Ma
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC 27101, United States
| | - Baisong Lu
- Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, United States
| | - Tracy Criswell
- Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, United States
| | - Yuanyuan Zhang
- Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, United States
| |
Collapse
|