1
|
Gu NY, Ryu GS, Park GN, Lee JY, Cho YS, Yang DK, Lee HJ. Enhanced susceptibility of porcine muscle-derived mesenchymal stem cells to Aujeszky's virus compared Vero cells. Anim Biotechnol 2025; 36:2479677. [PMID: 40110865 DOI: 10.1080/10495398.2025.2479677] [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: 01/06/2025] [Accepted: 03/10/2025] [Indexed: 03/22/2025]
Abstract
Mesenchymal stem cells (MSCs) can self-renew and differentiate into several lineages and can be isolated from different tissues such as bone marrow, adipose tissue, umbilical cord blood, and muscle. Herein, we established MSCs derived from miniature pig muscle (MpMu-MSCs) and assessed their response to Aujeszky's virus. We characterized the MpMu-MSCs based on their cellular morphology, proliferation properties, cell surface marker expression, and mesodermal differentiation potential. MpMu-MSCs demonstrated a fibroblast-like spindle shape and formed a homogeneous monolayer. They showed a considerable increase in cell proliferation over 16 passages. The cells expressed surface markers CD29, CD44, CD90, and CD105 and demonstrated mesodermal lineage differentiation capabilities. MpMu-MSCs demonstrated faster cytopathic effects than the Vero cells when infected with Aujeszky's virus. The virus titer in MpMu-MSCs was initiated at 101.4 TCID50/ml at 12 h post-infection (hpi) and increased to 106.6 TCID50/ml at 72 hpi. In Vero cells, it was initiated at 102.3 TCID50/ml at 48 hpi and increased to 103.8 TCID50/ml at 72 hpi. This study showed that the stem cells procured from miniature pig muscles exhibit MSC characteristics and that the established cells demonstrate higher susceptibility and virus titer to Aujeszky's virus than Vero cells, indicating their potential use in virus research.
Collapse
Affiliation(s)
- Na-Yeon Gu
- Viral Disease Division, Animal and Plant Quarantine Agency, Gyeongsangbuk-do, Republic of Korea
| | - Gwang Sik Ryu
- Viral Disease Division, Animal and Plant Quarantine Agency, Gyeongsangbuk-do, Republic of Korea
| | - Gyu-Nam Park
- Viral Disease Division, Animal and Plant Quarantine Agency, Gyeongsangbuk-do, Republic of Korea
| | - Ju-Yeon Lee
- Viral Disease Division, Animal and Plant Quarantine Agency, Gyeongsangbuk-do, Republic of Korea
| | - Yun Sang Cho
- Viral Disease Division, Animal and Plant Quarantine Agency, Gyeongsangbuk-do, Republic of Korea
| | - Dong-Kun Yang
- Viral Disease Division, Animal and Plant Quarantine Agency, Gyeongsangbuk-do, Republic of Korea
| | - Hye Jeong Lee
- Viral Disease Division, Animal and Plant Quarantine Agency, Gyeongsangbuk-do, Republic of Korea
| |
Collapse
|
2
|
Lee G, Kim YH, Kim D, Lee DH, Bhang SH, Lee K. PCL-fibrin-alginate hydrogel based cell co-culture system for improving angiogenesis and immune modulation in limb ischemia. Colloids Surf B Biointerfaces 2025; 250:114553. [PMID: 39921993 DOI: 10.1016/j.colsurfb.2025.114553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 12/10/2024] [Accepted: 02/02/2025] [Indexed: 02/10/2025]
Abstract
Stem cell therapy has demonstrated promise in regenerative medicine due to their ability to differentiate into various cell types and secrete growth factors. However, challenges such as poor survival rate of transplanted cells under ischemic and immune conditions limit its effectiveness. To address these issues, we developed a polycaprolactone (PCL)-fibrin-alginate matrix hydrogel, which combines adipose-derived stem cells and human umbilical vein endothelial cells with a PCL fiber, encapsulated within fibrin and alginate hydrogel to enhance cell survival, proliferation, and immune modulation. This structure offers protection to the encapsulated cells, supports angiogenesis, and modulates the immune response, significantly improving therapeutic outcomes in a mouse model of hindlimb ischemia. Our in vitro and in vivo results demonstrate the scaffold's ability to support cell viability, promote angiogenesis, and modulate inflammatory responses, indicating its potential as a promising platform for ischemic tissue repair and regenerative medicine. This innovative approach to cell-based therapy highlights the importance of scaffold design in enhancing the therapeutic efficacy of stem cell treatments for ischemic diseases.
Collapse
Affiliation(s)
- Gyubok Lee
- Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul 08826, Republic of Korea
| | - Yeong Hwan Kim
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Dongwoo Kim
- Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul 08826, Republic of Korea
| | - Dong-Hyun Lee
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Suk Ho Bhang
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| | - Kangwon Lee
- Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul 08826, Republic of Korea.
| |
Collapse
|
3
|
Inafuku N, Sowa Y, Kishida T, Sawai S, Ntege EH, Numajiri T, Yamamoto K, Shimizu Y, Mazda O. Investigation of the stemness and wound-healing potential of long-term cryopreserved stromal vascular fraction cells. Regen Ther 2025; 29:128-139. [PMID: 40162021 PMCID: PMC11952815 DOI: 10.1016/j.reth.2025.02.004] [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: 11/09/2024] [Revised: 01/26/2025] [Accepted: 02/12/2025] [Indexed: 04/02/2025] Open
Abstract
Introduction Stromal vascular fraction (SVF), a heterogeneous cell population primarily derived from adipose tissue, is widely utilized in regenerative therapies for its wound-healing properties and accessibility. While its immediate availability is advantageous, repeated harvesting can be burdensome, especially for elderly patients, and the regenerative capacity of SVF declines with donor age. Long-term cryopreservation offers a potential solution by allowing the banking of SVF from younger donors for future use; however, the impact of this process on SVF functionality remains elusive. This study investigates the stemness and wound-healing potential of SVF following prolonged cryopreservation. Methods SVF cells were isolated from adipose tissue harvested from twelve patients and cryopreserved for either two months (short-term cryopreserved SVF, S-SVF) or 12-13 years (long-term cryopreserved SVF, L-SVF), with six patients in each group. In vitro assays assessed cell viability and stemness, while in vivo assays evaluated wound-healing ability by administering thawed SVF cells from each group to dorsal wounds in immunodeficient mice, compared with a control group. Non-parametric statistical tests analyzed the differences between groups. Results L-SVF exhibited significantly lower stemness compared to S-SVF. Importantly, the L-SVF group showed significantly improved wound healing compared with the control group, although the wound-healing effect of L-SVF was inferior to that of the S-SVF. Conclusion This study demonstrated that, despite reduced stemness, L-SVF retains partial wound-healing potential after 12-13 years of cryopreservation. These findings highlight the need for optimized cryopreservation protocols to enhance SVF viability and regenerative capacity for clinical applications.
Collapse
Affiliation(s)
- Naoki Inafuku
- Department of Plastic and Reconstructive Surgery, Kyoto Prefectural University of Medicine, Kamigyo, Kyoto, Japan
| | - Yoshihiro Sowa
- Department of Plastic Surgery, Jichi Medical University, Shimotsuke, Tochigi, Japan
| | - Tsunao Kishida
- Department of Immunology, Kyoto Prefectural University of Medicine, Kamigyo, Kyoto, Japan
| | - Seiji Sawai
- Department of Orthopedics, Jyujyo Takeda Rehabilitation Hospital, Kyoto, Japan
| | - Edward Hosea Ntege
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Toshiaki Numajiri
- Department of Plastic and Reconstructive Surgery, Kyoto Prefectural University of Medicine, Kamigyo, Kyoto, Japan
| | - Kenta Yamamoto
- Department of Immunology, Kyoto Prefectural University of Medicine, Kamigyo, Kyoto, Japan
| | - Yusuke Shimizu
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Osam Mazda
- Department of Immunology, Kyoto Prefectural University of Medicine, Kamigyo, Kyoto, Japan
| |
Collapse
|
4
|
Storck K, Ussar S, Kotz S, Altun I, Hu F, Birk A, Veit J, Kovacevic M. Characterization of Fat Used for the Optimization of the Soft Tissue Envelope of the Nose in Rhinoplasty. Facial Plast Surg 2025; 41:266-273. [PMID: 38688299 DOI: 10.1055/s-0044-1786185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2024] Open
Abstract
Septorhinoplasty (SRP) is one of the most common aesthetic procedures worldwide. A thin or scarred soft tissue envelope, especially in the context of secondary SRP, can lead to unpredictable scarring, shrinkage, and discoloration of the skin. Other than the careful preparation of the soft tissue mantle, no gold standard exists to minimize the above-mentioned risks. Our aim was to create a thin "separation layer" between the nasal bridge (osseous and cartilaginous) and the skin envelope by autologous fat transfer with the addition of platelet-rich fibrin (PRF) to conceal small irregularities, to improve the quality of the skin soft tissue mantle, and to optimize the mobility of the skin. We report 21 patients who underwent SRP on a voluntary basis. All patients had either thin skin and/or revision SRP with scarring. Macroscopic fat harvested from the periumbilical or rib region was minced and purified. PRF was obtained by centrifugation of autologous whole blood samples and mixed with the fat to form a graft, which was then transferred to the nasal dorsum. Postoperative monitoring of graft survival included sonography and magnetic resonance imaging (MRI) of the nose. The harvested adipose tissue was also analyzed in vitro. In the postoperative follow-up after 1 year, survival of the adipose tissue was demonstrated in all patients by both sonography and MRI. The in vitro analysis showed interindividual differences in the quantity, size, and quality of the transplanted adipocytes. Camouflage of the nasal bridge by using adipose tissue was beneficial for the quality of the skin soft tissue mantle and hence represents a good alternative to known methods. Future aims include the ability to assess the quality of adipose tissue to be transplanted based on clinical parameters. Level of evidence: N/A.
Collapse
Affiliation(s)
- Katharina Storck
- Department of Otorhinolaryngology, Head and Neck Surgery, Klinikum Rechts der Isar, TU Munich, Munich, Germany
| | - Siegfried Ussar
- Research Unit Adipocytes and Metabolism (ADM), Institute for Diabetes and Obesity at Helmholtz Center Munich, Neuherberg, Germany
| | - Sebastian Kotz
- Department of Otorhinolaryngology, Head and Neck Surgery, Klinikum Rechts der Isar, TU Munich, Munich, Germany
| | - Irem Altun
- Research Unit Adipocytes and Metabolism (ADM), Institute for Diabetes and Obesity at Helmholtz Center Munich, Neuherberg, Germany
| | - Fiona Hu
- Research Unit Adipocytes and Metabolism (ADM), Institute for Diabetes and Obesity at Helmholtz Center Munich, Neuherberg, Germany
| | - Amelie Birk
- Department of Otorhinolaryngology, Head and Neck Surgery, Klinikum Rechts der Isar, TU Munich, Munich, Germany
| | | | | |
Collapse
|
5
|
Wan X, Wang L, Khan MA, Peng L, Sun X, Yi X, Wang Z, Chen K. NAT10-mediated N4-acetylcytidine modification in KLF9 mRNA promotes adipogenesis. Cell Death Differ 2025:10.1038/s41418-025-01483-x. [PMID: 40123006 DOI: 10.1038/s41418-025-01483-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 02/14/2025] [Accepted: 03/14/2025] [Indexed: 03/25/2025] Open
Abstract
Dysfunctional adipogenesis is a major contributor of obesity. N-acetyltransferase 10 (NAT10) plays a crucial role in regulating N4-acetylcysteine (ac4C) modification in tRNA, 18SrRNA, and mRNA. As the sole "writer" in the ac4C modification process, NAT10 enhances mRNA stability and translation efficiency. There are few reports on the relationship between NAT10 and adipogenesis, as well as obesity. Our study revealed a significant upregulation of NAT10 in adipose tissues of obese individuals and high-fat diet-fed mice. Furthermore, our findings revealed that the overexpression of NAT10 promotes adipogenesis, while its silencing inhibits adipogenesis in both human adipose tissue-derived stem cells (hADSCs) and 3T3-L1 cells. These results indicate the intimate relationship between NAT10 and obesity. After silencing mouse NAT10 (mNAT10), we identified 30 genes that exhibited both hypo-ac4C modification and downregulation in their expression, utilizing a combined approach of acRIP-sequencing (acRIP-seq) and RNA-sequencing (RNA-seq). Among these genes, we validated KLF9 as a target of NAT10 through acRIP-PCR. KLF9, a pivotal transcription factor that positively regulates adipogenesis. Our findings showed that NAT10 enhances the stability of KLF9 mRNA and further activates the CEBPA/B-PPARG pathway. Furthermore, a dual-luciferase reporter assay demonstrated that NAT10 can bind to three motifs of mouse KLF9 and one motif of human KLF9. In vivo studies revealed that adipose tissue-targeted mouse AAV-NAT10 (AAV-shRNA-mNAT10) inhibits adipose tissue expansion in mice. Additionally, Remodelin, a specific NAT10 inhibitor, significantly reduced body weight, adipocyte size, and adipose tissue expansion in high-fat diet-fed mice by inhibiting KLF9 mRNA ac4C modification. These findings provide novel insights and experimental evidence of the prevention and treatment of obesity, highlighting NAT10 and its downstream targets as potential therapeutic targets.
Collapse
Affiliation(s)
- Xinxing Wan
- Department of Endocrinology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, PR China
| | - Linghao Wang
- Department of Endocrinology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, PR China
| | - Md Asaduzzaman Khan
- Department of Biochemistry and Microbiology, School of Health & Life Sciences, North South University, Dhaka, Bangladesh
| | - Lin Peng
- Department of Nephrology, The First Hospital of Changsha, Changsha, Hunan, PR China
| | - Xiaoying Sun
- Department of Endocrinology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, PR China
| | - Xuan Yi
- Department of Endocrinology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, PR China
| | - Zhouqi Wang
- Department of Endocrinology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, PR China
| | - Ke Chen
- Department of Endocrinology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, PR China.
| |
Collapse
|
6
|
Braitenbach BPDF, Cerqueira CBSD, Lima SS, Hegouet IDC, Moreno BCL, Medrado ARAP. Effects of photobiomodulation on adipocytic infiltration in sites of skin healing: in vivo experimental study. Lasers Med Sci 2025; 40:153. [PMID: 40111643 DOI: 10.1007/s10103-025-04410-1] [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: 09/18/2024] [Accepted: 03/12/2025] [Indexed: 03/22/2025]
Abstract
Adipocyte infiltration consists of a temporary increase in the number of adipocytes in the microenvironment of tissue injury. There is already evidence in the literature of adipocytes' participation in promoting the inflammatory response, and parallelly laser photobiomodulation can benefit the tissue repair process. This study aimed to chronologically analyze adipocytic infiltration in the repair of photobiomodulated skin wounds experimentally induced in rats through histomorphometric analysis. The sample consisted of 20 rats divided into 2 groups: control group and group subjected to laser photobiomodulation. The skin portions of the back of rats were processed and stained with Hematoxylin-Eosin in 4 μm thick sections including the surgical wound 5 and 10 days after the proposed treatments. Qualitative and quantitative analyses were performed by capturing images of tissue sections, describing the organizational pattern of adipocytes around the surgical wound and counting individual adipocytes in the connective tissue in formation. Adipocytic infiltration was observed in both experimental groups on the 5th day, with a decrease on the 10th day. The group treated with photobiomodulation presented a greater number of adipocytes compared to the control group, in both periods analyzed. The findings of the present study seem to corroborate the literature, which indicates that adipose cells might stimulate inflammation and repair, and photobiomodulation can enhance these effects, since it aids the process of adipocytic infiltration in the injured area. Clinical trial number: Not applicable.
Collapse
|
7
|
Storti G, Foti R, Foti R, Palmesano M, Patacchiola M, Incognito D, Cervelli G, Longo B, Scioli MG, Fiorelli E, Terriaca S, Lisa A, Kim BS, Orlandi A, Cervelli V. A Comprehensive Exploration of the Biological Effects of Adipose-Derived Stem Cells in the Treatment of Systemic Sclerosis. Cells 2025; 14:458. [PMID: 40136706 PMCID: PMC11941144 DOI: 10.3390/cells14060458] [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: 02/08/2025] [Revised: 03/10/2025] [Accepted: 03/17/2025] [Indexed: 03/27/2025] Open
Abstract
Systemic sclerosis (SSc) is a complex autoimmune disease characterized by vasculopathy and tissue fibrosis affecting the skin and internal organs. Genetic and environmental factors influence susceptibility, severity, and onset. Current treatments are limited and not always effective, leading researchers to investigate new approaches, such as the use of adipose-derived mesenchymal stem cells (ADSCs) through fat grafting. This review seeks to understand how ADSCs may impact the development and progression of SSc, with a particular focus on how these cells could alter immune responses and reduce fibrosis. ADSCs have been found to affect various immune cells, including T cells, B cells, macrophages, and dendritic cells, by releasing cytokines, chemokines, and growth factors. These interactions generally suppress inflammation and promote a regulatory immune environment. Additionally, ADSCs can influence the extracellular matrix, helping to prevent fibrosis through signaling molecules like exosomes. ADSCs show promise as a treatment for SSc due to their ability to modulate the immune system and reduce fibrosis. Early clinical studies are encouraging, but more research is needed to fully understand how they work and to develop effective treatment protocols.
Collapse
Affiliation(s)
- Gabriele Storti
- Plastic Surgery, Department of Surgical Sciences, University of Rome “Tor Vergata”, 00133 Rome, Italy; (G.S.); (M.P.); (M.P.); (B.L.); (V.C.)
| | - Riccardo Foti
- Plastic Surgery, Department of Surgical Sciences, University of Rome “Tor Vergata”, 00133 Rome, Italy; (G.S.); (M.P.); (M.P.); (B.L.); (V.C.)
| | - Roberta Foti
- Division of Rheumatology, A.O.U. “Policlinico-San Marco”, 95123 Catania, Italy;
- Department of Medicine and Surgery, University of Enna “Kore”, 94100 Enna, Italy
| | - Marco Palmesano
- Plastic Surgery, Department of Surgical Sciences, University of Rome “Tor Vergata”, 00133 Rome, Italy; (G.S.); (M.P.); (M.P.); (B.L.); (V.C.)
- PhD Program in Applied Medical Surgical Sciences, Department of Surgical Sciences, University of Rome “Tor Vergata”, Viale Oxford 81, 00133 Rome, Italy;
| | - Martina Patacchiola
- Plastic Surgery, Department of Surgical Sciences, University of Rome “Tor Vergata”, 00133 Rome, Italy; (G.S.); (M.P.); (M.P.); (B.L.); (V.C.)
| | - Dalila Incognito
- Medical Oncology Unit, Department of Human Pathology “G. Barresi”, University of Messina, 98122 Messina, Italy;
| | - Giulio Cervelli
- Department of Experimental Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy;
| | - Benedetto Longo
- Plastic Surgery, Department of Surgical Sciences, University of Rome “Tor Vergata”, 00133 Rome, Italy; (G.S.); (M.P.); (M.P.); (B.L.); (V.C.)
| | - Maria Giovanna Scioli
- Anatomy Pathology Institute, Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, 00133 Rome, Italy; (M.G.S.); (E.F.); (S.T.); (A.O.)
| | - Elena Fiorelli
- Anatomy Pathology Institute, Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, 00133 Rome, Italy; (M.G.S.); (E.F.); (S.T.); (A.O.)
| | - Sonia Terriaca
- Anatomy Pathology Institute, Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, 00133 Rome, Italy; (M.G.S.); (E.F.); (S.T.); (A.O.)
| | - Andrea Lisa
- PhD Program in Applied Medical Surgical Sciences, Department of Surgical Sciences, University of Rome “Tor Vergata”, Viale Oxford 81, 00133 Rome, Italy;
- Department of Plastic and Reconstructive Surgery, European Institute of Oncology, IRCCS, 20139 Milan, Italy
- Department of Biomedical Sciences, Humanitas University, 20072 Milan, Italy
| | - Bong Sung Kim
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, 8006 Zurich, Switzerland;
| | - Augusto Orlandi
- Anatomy Pathology Institute, Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, 00133 Rome, Italy; (M.G.S.); (E.F.); (S.T.); (A.O.)
| | - Valerio Cervelli
- Plastic Surgery, Department of Surgical Sciences, University of Rome “Tor Vergata”, 00133 Rome, Italy; (G.S.); (M.P.); (M.P.); (B.L.); (V.C.)
| |
Collapse
|
8
|
Goulian AJ, Goldstein B, Saad MA. Advancements in Regenerative Therapies for Orthopedics: A Comprehensive Review of Platelet-Rich Plasma, Mesenchymal Stem Cells, Peptide Therapies, and Biomimetic Applications. J Clin Med 2025; 14:2061. [PMID: 40142869 PMCID: PMC11943164 DOI: 10.3390/jcm14062061] [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: 01/27/2025] [Revised: 03/09/2025] [Accepted: 03/12/2025] [Indexed: 03/28/2025] Open
Abstract
Background/Objectives: Regenerative therapies have gained interest in orthopedic applications for their potential to enhance tissue regeneration, functional recovery, and pain modification. This review evaluates the clinical efficacy of platelet-rich plasma (PRP), mesenchymal stem cells (MSCs), peptide-based treatments, and biomimetic materials in orthopedic care, with a focus on pain reduction and functional outcomes. Methods: A structured literature search in PubMed (January 2009-January 2025) identified 160 studies. After applying inclusion criteria prioritizing randomized controlled trials (RCTs) and clinical trials, 59 studies were included: 20 on PRP, 20 on MSCs, 10 on peptide therapies, and 7 on biomimetics. Data extraction focused on pain reduction and functional recovery, with risk of bias assessed using the Cochrane Risk of Bias (RoB) tool and ROBINS-I tool. A random-effects meta-regression analysis was conducted to evaluate the impact of therapy type, sample size, and risk of bias on reported pain reduction outcomes. Results: Meta-regression analysis identified MSC therapy as the most effective intervention for pain reduction (β = 8.45, p < 0.05), with PRP and peptide-based therapies showing moderate improvements, and biomimetic therapies demonstrating the lowest effect. PRP provided short-term pain relief, particularly in acute injuries and tendon repair, though inconsistencies in preparation methods limited success in chronic conditions. MSC therapies demonstrated cartilage regeneration and early osteoarthritis improvement, but high costs and ethical concerns remain barriers to widespread adoption. Peptide-based therapies and biomimetic materials, including engineered scaffolds and autologous protein solutions, showed promise for infection control and wound healing, though further research is needed to optimize dosing, delivery methods, and long-term safety. Conclusions: Regenerative therapies offer significant potential in orthopedic care, with MSC therapies demonstrating the most reliable regenerative effects, PRP providing short-term symptomatic relief, and peptide-based and biomimetic treatments emerging as promising adjuncts. However, standardized protocols and large-scale clinical trials are needed to establish long-term efficacy and improve clinical translation for broader adoption.
Collapse
Affiliation(s)
- Andrew J. Goulian
- College of Medicine, California Northstate University, Elk Grove, CA 95757, USA; (A.J.G.); (B.G.)
| | - Brielle Goldstein
- College of Medicine, California Northstate University, Elk Grove, CA 95757, USA; (A.J.G.); (B.G.)
| | - Maarouf A. Saad
- Department of Orthopaedic Surgery, University of California, Sacramento, CA 95817, USA
| |
Collapse
|
9
|
Truchan K, Zagrajczuk B, Cholewa-Kowalska K, Osyczka AM. Rapid osteoinduction of human adipose-derived stem cells grown on bioactive surfaces and stimulated by chemically modified media flow. J Biol Eng 2025; 19:23. [PMID: 40087792 PMCID: PMC11908086 DOI: 10.1186/s13036-025-00491-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 02/26/2025] [Indexed: 03/17/2025] Open
Abstract
Adipose-derived stem cells (ASCs) provide an ample, easily accessible source of multipotent cells, an alternative to bone marrow-derived stromal cells (BMSCs), capable of differentiating into osteoblasts. However, the osteogenic potential of ASCs is reportedly lower than that of BMSCs and protocols to effectively differentiate ASCs into osteoblasts are in high demand. Here, we present novel strategies for effective osteogenic differentiation of human ASCs by combining their culture on bioactive growth surfaces with their treatment with specific supplements in osteogenic medium and application of fluid shear stress. Human ASCs were cultured on PLGA-based composites containing 50 wt% sol-gel bioactive glasses (SBGs) from the SiO2-CaO±P2O5 system, either unmodified or modified with 5 wt% ZnO or SrO. The osteogenic medium was supplemented with recombinant human bone morphogenetic protein 2 (BMP-2), MEK1/2 kinase inhibitor (PD98059) and indirect Smurf1 inhibitor (Phenamil). Fluid shear stress was applied with a standard horizontal rocker. ASC culture on SBG-PLGA composites along with the osteogenic medium supplements enhanced the expression of both early and late osteogenic markers. Modification of SBG with either SrO or ZnO further enhanced osteogenic gene expression compared to ASCs cultured on composites containing unmodified SBGs. Notably, the application of fluid shear stress synergistically strengthened the osteogenic effects of bioactive composites and medium supplements. We also show that the presented culture strategies can drive ASCs toward osteoblastic cells in a 3-day culture period and provide mineralizing osteoblasts through a short, 7-day ASC preculture on bioactive composites. Our results also indicate that the applied osteogenic treatment leads to the phosphorylation of β-catenin and CREB or the COX-2 expression. We believe the presented strategies are feasible for rapid ASC differentiation to early osteoblasts or mineralizing osteoblastic cells for various potential cell-based bone regeneration therapies.
Collapse
Affiliation(s)
- Karolina Truchan
- Department of Cell Biology and Imaging, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Gronostajowa St. 9, Krakow, 30-387, Poland
| | - Barbara Zagrajczuk
- Department of Glass Technology and Amorphous Coatings, Faculty of Materials Science and Ceramics, AGH University of Science and Technology, Mickiewicza Ave. 30, Krakow, 30-059, Poland
| | - Katarzyna Cholewa-Kowalska
- Department of Glass Technology and Amorphous Coatings, Faculty of Materials Science and Ceramics, AGH University of Science and Technology, Mickiewicza Ave. 30, Krakow, 30-059, Poland
| | - Anna Maria Osyczka
- Department of Cell Biology and Imaging, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Gronostajowa St. 9, Krakow, 30-387, Poland.
| |
Collapse
|
10
|
Chuang HN, Pei W, Kuo TF, Liu YH, Wang CY, Chang YW, Chuang C, Yang CH, Chuang MH. Effect of injecting adipose stem cells combined with platelet-rich fibrin releasate at Shenshu acupoint (BL23) on acute kidney injury in rabbits. Front Pharmacol 2025; 16:1409056. [PMID: 40144656 PMCID: PMC11936987 DOI: 10.3389/fphar.2025.1409056] [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: 09/24/2024] [Accepted: 02/19/2025] [Indexed: 03/28/2025] Open
Abstract
Introduction Acute kidney injury (AKI) is a major and unmet medical need, characterized by a sudden onset of kidney dysfunction that often occurs within 7 days. Adipose-derived stem cells (ADSCs) are known for their regenerative, differentiative, and repair abilities, making them a promising therapeutic option for kidney injury. Platelet-rich fibrin releasate (PRFr), derived from platelet-rich fibrin after static incubation, contains numerous growth factors that may promote the differentiation and proliferation of stem cells. Additionally, acupoints such as Shenshu (BL23) have been used in clinical practice and experimental settings, particularly in renal failure treatments. Methods This study aimed to evaluate the synergistic effects of ADSCs and PRFr, administered separately or in combination, at the Shenshu acupoint (BL23) in New Zealand white rabbits with acute kidney injury. The treatment groups were injected with ADSCs, PRFr, or a combination of both. Serum creatinine (CRE) and blood urea nitrogen (BUN) levels were measured to assess kidney function. Additionally, histological examination of kidney tissue was performed to observe morphological changes and tissue repair. Results The PRFr + ADSCs treatment group exhibited a significant reduction in CRE and BUN levels during the second week following transplantation. After 7 weeks of treatment, the PRFr + ADSCs group showed the most favorable kidney repair outcomes, with intact glomeruli, no edema or vacuole-like changes in the renal tubular epithelial cells, and no significant infiltration of inflammatory cells in the surrounding tissues. Discussion The administration of PRFr, ADSCs, and their combination at the Shenshu acupoint (BL23) demonstrated a potential therapeutic effect in repairing damaged renal cells, improving kidney function, and reducing serum CRE and BUN levels. These findings suggest that injection of PRFr, ADSCs, and their combination at the Shenshu acupoint (BL23) can effectively repair damaged renal cells and improve kidney function in AKI. The observed synergistic effect indicates that this approach holds potential as a novel therapeutic strategy for kidney injury. Further research is needed to optimize treatment protocols and elucidate the underlying mechanisms.
Collapse
Affiliation(s)
- Hsin-Ni Chuang
- Ph.D. Program of Management, Chung Hua University, Hsinchu, Taiwan
| | - Wen Pei
- College of Management, Chung Hua University, Hsinchu, Taiwan
| | - Tzong-Fu Kuo
- School of Veterinary Medicine, National Taiwan University, Taipei, Taiwan
| | - Yu-Hao Liu
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chia-Yih Wang
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yen-Wei Chang
- Department of Physical Education, Asia University, Taichung, Taiwan
| | | | - Chang-Huan Yang
- Gwo Xi Stem Cell Applied Technology Co., Ltd., Hsinchu, Taiwan
| | - Ming-Hsi Chuang
- College of Management, Chung Hua University, Hsinchu, Taiwan
- Gwo Xi Stem Cell Applied Technology Co., Ltd., Hsinchu, Taiwan
- Institute of Biopharmaceutical Science, National Yang Ming Chiao Tung University, Taipei, Taiwan
- School of Public Health, National Defense Medical Center, Taipei City, Taiwan
| |
Collapse
|
11
|
Cheng Z, Liu D, Park JY, Meng X, Yang Y, Dang M, Dai X, Yang J, Yuan M, Li M, Wang L, Huang Y, Wang J, Liang Y, Fei W. Evaluation of the management of rotator cuff injuries utilising superparamagnetic iron oxide tracking stem cells. Tissue Cell 2025; 95:102836. [PMID: 40081255 DOI: 10.1016/j.tice.2025.102836] [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: 12/30/2024] [Revised: 02/11/2025] [Accepted: 03/01/2025] [Indexed: 03/15/2025]
Abstract
BACKGROUND The ultrastructure of the tendon-bone interface (TBI) is inherently complex. After arthroscopic reconstruction, it is often replaced by disorganized scar tissue, which increases the risk of re-tearing.Stem cell therapies offer a promising approach to regenerate the original tissue structure and enhance the healing environment. The effectiveness of these therapies depends on understanding the localization, proliferation, and overall behavior of the implanted stem cells. This study aimed to track the distribution of stem cells in a rat model of rotator cuff injury using Magnetic Resonance Imaging (MRI) and superparamagnetic iron oxide nanoparticles (SPIO) and to evaluate the mechanisms and therapeutic effects of stem cell therapy. METHODS Adipose-derived mesenchymal stem cells (ADSCs) were isolated and expanded, then labeled with SPIO at an optimized concentration. The visibility of these labeled cells was assessed via MRI, along with evaluations of their viability, potential toxicity, and migration capacity in vitro.For the in vivo study, rats with rotator cuff tears were divided into two groups: a control group that received a PBS injection, and a treatment group that received SPIO-labeled ADSCs (designated as S-A). MRI scans were conducted at 1, 2, and 4 weeks post-surgery, followed by histological analysis after the rats were euthanized. At 8 weeks post-surgery, rats were sacrificed, and their shoulder joints were analyzed biomechanically and histologically to assess the overall treatment efficacy. RESULTS SPIO nanoparticles were successfully incorporated into ADSCs, and MRI imaging demonstrated that these SPIO-labeled cells significantly enhanced MRI contrast without affecting cell viability, proliferation, or migration ability. Both MRI and histological analyses confirmed that the implanted stem cells survived and remained localized for at least two weeks. Further histological and biomechanical evaluations indicated that the stem cells facilitated the repair of the TBI. This repair process appeared to be mediated by an increase in M2 macrophage activity within the injured tissue, promoting improved local healing conditions. CONCLUSION This study confirms that labeling ADSCs with SPIO nanoparticles is an effective method for tracking these cells in vivo using MRI, providing a non-invasive approach to monitor the repair of injured TBI. Moreover, the localized survival of transplanted stem cells supports their role in enhancing TBI repair by modulating the local inflammatory response.
Collapse
Affiliation(s)
- Ziang Cheng
- Department of Orthopedics and Sports Medicine, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, P. R. China
| | - Dianwei Liu
- The Yangzhou school of clinical medicine of Dalian Medical University, Dalian 116000, P. R. China
| | - Jin-Young Park
- Center for Shoulder, Elbow and Sports, Neon Orthopaedic Clinic, Seoul, Republic of Korea
| | - Xiangji Meng
- The Yangzhou school of clinical medicine of Dalian Medical University, Dalian 116000, P. R. China
| | - Yuxia Yang
- Department of Orthopedics and Sports Medicine, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, P. R. China
| | - Mengbo Dang
- The Yangzhou school of clinical medicine of Dalian Medical University, Dalian 116000, P. R. China
| | - Xiaomei Dai
- Department of Orthopedics and Sports Medicine, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, P. R. China
| | - Jian Yang
- Department of Orthopedics and Sports Medicine, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, P. R. China
| | - Meijuan Yuan
- Department of Orthopedics and Sports Medicine, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, P. R. China
| | - Mingjun Li
- Department of Orthopedics and Sports Medicine, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, P. R. China
| | - Liang Wang
- Department of Orthopedics and Sports Medicine, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, P. R. China
| | - Yao Huang
- Department of Orthopedics and Sports Medicine, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, P. R. China
| | - Jingcheng Wang
- Department of Orthopedics and Sports Medicine, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, P. R. China.
| | - Yuan Liang
- Department of Orthopedics and Sports Medicine, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, P. R. China.
| | - Wenyong Fei
- Department of Sports Medicine, Northern Jiangsu People's Hospital, Yangzhou 225001, P. R. China; Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, P. R. China.
| |
Collapse
|
12
|
Farag A, Hendawy H, Emam MH, Hasegawa M, Mandour AS, Tanaka R. Stem Cell Therapies in Canine Cardiology: Comparative Efficacy, Emerging Trends, and Clinical Integration. Biomolecules 2025; 15:371. [PMID: 40149907 PMCID: PMC11940628 DOI: 10.3390/biom15030371] [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: 01/30/2025] [Revised: 02/26/2025] [Accepted: 02/27/2025] [Indexed: 03/29/2025] Open
Abstract
Cardiovascular diseases are a leading cause of morbidity and mortality in dogs, with limited options available for reversing myocardial damage. Stem cell therapies have shown significant potential for cardiac repair, owing to their immunomodulatory, antifibrotic, and regenerative properties. This review evaluates the therapeutic applications of mesenchymal stem cells (MSCs) derived from bone marrow, adipose tissue, and Wharton's jelly with a focus on their role in canine cardiology and their immunoregulatory properties. Preclinical studies have highlighted their efficacy in enhancing cardiac function, reducing fibrosis, and promoting angiogenesis. Various delivery methods, including intracoronary and intramyocardial injections, are assessed for their safety and efficacy. Challenges such as low cell retention, differentiation efficiency, and variability in therapeutic responses are also discussed. Emerging strategies, including genetic modifications and combination therapies, aim to enhance the efficacy of MSCs. Additionally, advances in delivery systems and regulatory frameworks are reviewed to support clinical translation. This comprehensive evaluation underscores the potential of stem cell therapies to revolutionize canine cardiovascular disease management while identifying critical areas for future research and clinical integration.
Collapse
Affiliation(s)
- Ahmed Farag
- Faculty of Agriculture, Veterinary Teaching Hospital, Tokyo University of Agriculture and Technology, Tokyo 183-8509, Japan
- Department of Surgery, Anesthesiology, and Radiology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44519, Egypt
| | - Hanan Hendawy
- Department of Veterinary Surgery, Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522, Egypt
| | - Mahmoud H. Emam
- Animal Medicine Department, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44519, Egypt
| | - Mizuki Hasegawa
- Faculty of Agriculture, Veterinary Teaching Hospital, Tokyo University of Agriculture and Technology, Tokyo 183-8509, Japan
| | - Ahmed S. Mandour
- Department of Animal Medicine (Internal Medicine), Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522, Egypt
| | - Ryou Tanaka
- Faculty of Agriculture, Veterinary Teaching Hospital, Tokyo University of Agriculture and Technology, Tokyo 183-8509, Japan
| |
Collapse
|
13
|
Lemmer P, Sowa JP, Bulut Y, Strnad P, Canbay A. Mechanisms and aetiology-dependent treatment of acute liver failure. Liver Int 2025; 45:e15739. [PMID: 37752801 DOI: 10.1111/liv.15739] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 09/01/2023] [Accepted: 09/05/2023] [Indexed: 09/28/2023]
Abstract
This review compiles the mechanisms of acute liver failure (ALF) as well as the current and potential therapeutic approaches, including aetiology-specific treatment, and the issues encountered with such approaches. On a cellular level, ALF is characterized by massive hepatocyte death due to different types of cellular demise. Compensatory hyperplasia and functional recovery are possible when the regenerative capacity is sufficient to sustain hepatic function. ALF has a high mortality of about 30% and can lead to death in a very short time despite maximum therapeutic intervention. Besides aetiology-specific therapy and intensive care, the therapeutic option of emergency liver transplantation has significantly improved the prognosis of patients with ALF. However, due to limiting factors such as organ shortage, many patients die on the waiting list. In addition to graft assessment, machine perfusion may have the potential to recondition marginal organs and thus expand the organ donor pool.
Collapse
Affiliation(s)
- Peter Lemmer
- Department of Gastroenterology, Hepatology, and Infectious Diseases, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Jan-Peter Sowa
- Department of Medicine, Universitätsklinikum Knappschaftskrankenhaus Bochum, Ruhr University Bochum, Bochum, Germany
| | - Yesim Bulut
- Department of Medicine, Universitätsklinikum Knappschaftskrankenhaus Bochum, Ruhr University Bochum, Bochum, Germany
| | - Pavel Strnad
- Department of Internal Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Ali Canbay
- Department of Medicine, Universitätsklinikum Knappschaftskrankenhaus Bochum, Ruhr University Bochum, Bochum, Germany
| |
Collapse
|
14
|
Yuan X, Long Q, Li W, Yan Q, Zhang P. Characteristics of the Dynamic Evolutionary Pathway of ADSCs Induced Differentiation into Astrocytes Based on scRNA-Seq Analysis. Mol Neurobiol 2025; 62:2926-2944. [PMID: 39190264 DOI: 10.1007/s12035-024-04414-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 07/30/2024] [Indexed: 08/28/2024]
Abstract
We employed single-cell transcriptome sequencing to reveal the dynamic gene expression changes during the differentiation of adipose-derived stromal cells (ADSCs) into astrocytes. Single-cell RNA sequencing was conducted on cells from the ADSCs group and the induced groups at 2, 7, 14, and 21 days using the 10 × Chromium platform. Data underwent quality control and dimensionality reduction. Cell differentiation trajectories were constructed using Monocle2, and differentially expressed genes (DEGs) in each cell cluster were identified using differential selection algorithms. DEGs at each time point were annotated using Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG), and regulatory intensities of transcription factors were analyzed using SCENIC. Integrating all groups, a total of five samples were divided into 13 cell clusters (0-12 clusters). DEGs between clusters and those compared with ADSCs at various induced time points showed distinct specificities. Monocle2 constructed cell differentiation trajectories; ADSCs can differentiate into mature astrocytes not only through the direct pathway from the 1 branch to the 3 branch but also through an indirect pathway, involving the 1 branch to the 2 branch before progressing to the 3 branch. SCENIC analysis highlighted the critical regulatory roles of STAT1, MYEF2, and SOX6 transcription factors during the differentiation of ADSCs into astrocytes. ADSCs can differentiate into mature astrocytes through two distinct pathways: direct and indirect. By the 14th day of induction, mature astrocytes have formed, characterized by a cell cycle arrest in mitosis. Further induction leads to degenerative senescence changes in differentiated cells.
Collapse
Affiliation(s)
- Xiaodong Yuan
- Department of Neurology of Kailuan General Hospital Affiliated North China University of Science and Technology, 57 Xinhua East Road, Lubei District, Tangshan City, 063000, Hebei Province, China
- Hebei Provincial Key Laboratory of Neurobiological Function, 57 Xinhua East Road, Lubei District, Tangshan City, 063000, Hebei Province, China
| | - Qingxi Long
- Department of Neurology of Kailuan General Hospital Affiliated North China University of Science and Technology, 57 Xinhua East Road, Lubei District, Tangshan City, 063000, Hebei Province, China
| | - Wen Li
- Department of Neurology of Kailuan General Hospital Affiliated North China University of Science and Technology, 57 Xinhua East Road, Lubei District, Tangshan City, 063000, Hebei Province, China
| | - Qi Yan
- Department of Neurology of Kailuan General Hospital Affiliated North China University of Science and Technology, 57 Xinhua East Road, Lubei District, Tangshan City, 063000, Hebei Province, China
| | - Pingshu Zhang
- Department of Neurology of Kailuan General Hospital Affiliated North China University of Science and Technology, 57 Xinhua East Road, Lubei District, Tangshan City, 063000, Hebei Province, China.
- Hebei Provincial Key Laboratory of Neurobiological Function, 57 Xinhua East Road, Lubei District, Tangshan City, 063000, Hebei Province, China.
| |
Collapse
|
15
|
Fukutome A, Sakamoto T, Asawa Y, Riu D, Kawakami H, Hoshi K, Hikita A. Establishment of a mouse organ culture model of fetal cleft lip for the evaluation of adipose-derived stem cell therapy. Regen Ther 2025; 28:41-50. [PMID: 39687332 PMCID: PMC11647479 DOI: 10.1016/j.reth.2024.11.012] [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: 10/10/2024] [Revised: 11/12/2024] [Accepted: 11/20/2024] [Indexed: 12/18/2024] Open
Abstract
Introduction Cleft lip and cleft palate are congenital disorders resulting from abnormal facial development. Current treatments require multiple surgeries, which have risks of scar formation and facial deformities. Recently, fetal treatments utilizing "scarless healing" have gained attention, as early intervention shows potential to suppress scarring. In the field of regenerative medicine, mesenchymal stem cell therapies using cell sheets have advanced, by which promotion of tissue repair is expected. However, researches for fetal treatment using small animal models of cleft lip are challenging due to the high fetal mortality caused by surgical invasiveness. Although organ culture methods may offer an alternative approach, no organ culture system for fetal cleft lip research has been reported. Methods In this study, a cleft lip was surgically created on the upper left side lip of E15.5 mouse fetuses. These fetuses were cultured for four days using an organ culture system. Histological evaluation was performed to evaluate cell density, tissue morphology, and epithelialization. Additionally, adipose-derived stem cell (ADSC) sheets were transplanted two days after cleft lip creation to evaluate their effect on tissue repair. Results The histological analysis showed that cell density and tissue morphology were stably maintained in the four-day culture period. Epithelialization of the incision site was observed two days after surgery, confirming the completion of cleft formation. In the ADSC-transplanted group, epithelialization of the cleft site was observed, which indicates that the stem cell sheets contributed to tissue repair. Conclusion This research demonstrates the successful development of a cleft lip organ culture model and highlights the potential of ADSC sheets in promoting tissue repair. These findings provide a foundation for future regenerative medicine strategies in fetal cleft lip therapy.
Collapse
Affiliation(s)
- Ayane Fukutome
- Department of Oral and Maxillofacial Surgery, Dentistry and Orthodontics, The University of Tokyo Graduate School of Medicine, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Tomoaki Sakamoto
- Department of Tissue Engineering, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Yukiyo Asawa
- Department of Tissue Engineering, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Dan Riu
- Department of Tissue Engineering, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Hiroshi Kawakami
- Division of Dentistry and Oral Surgery, Mitsui Memorial Hospital, 1 Kanda Izumicho, Chiyoda-ku, Tokyo 101-8643, Japan
| | - Kazuto Hoshi
- Department of Oral and Maxillofacial Surgery, Dentistry and Orthodontics, The University of Tokyo Graduate School of Medicine, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
- Department of Tissue Engineering, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Atsuhiko Hikita
- Department of Tissue Engineering, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| |
Collapse
|
16
|
Fu H, Wang C. Micro-fragmented adipose tissue-An innovative therapeutic approach: A narrative review. Medicine (Baltimore) 2025; 104:e41724. [PMID: 40020111 PMCID: PMC11875617 DOI: 10.1097/md.0000000000041724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Accepted: 02/12/2025] [Indexed: 03/05/2025] Open
Abstract
Subcutaneous adipose tissue provides distinct advantages as a source of mesenchymal stem cells due to its accessibility and the ease of isolating stem cells. Human adipose stem cells, located in the stromal-vascular fraction, can be harvested using mechanical methods to produce microfragmented adipose tissue (MFAT). Local injections of MFAT have shown potential in promoting natural tissue regeneration. This review introduces the concept of MFAT, highlights its clinical applications, and explores its potential in regenerative medicine, offering insights into its role as an innovative therapeutic approach.
Collapse
Affiliation(s)
- Hongjuan Fu
- Department of Anesthesiology, Yangguangronghe Hospital, Weifang, Shandong, China
| | - Congcong Wang
- Department of Joint Surgery, Weifang People’s Hospital, Weifang, Shandong, China
| |
Collapse
|
17
|
Yan K, Ma F, Song X, Wang H, Liu P, Zhang J, Jin X, Han P, Zuo X, Kang YJ. Unveiling distinctions between mesenchymal stromal cells and stem cells by single-cell transcriptomic analysis. Heliyon 2025; 11:e42311. [PMID: 40034318 PMCID: PMC11872483 DOI: 10.1016/j.heliyon.2025.e42311] [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: 09/08/2024] [Revised: 01/14/2025] [Accepted: 01/27/2025] [Indexed: 03/05/2025] Open
Abstract
Mesenchymal stromal cells (MSCs) and stem cells are distinct types of cells, but they are practically undistinguishable by currently commonly-used identification markers. A single-cell transcriptomic analysis was used to solve this problem. There are eight critical genes involved in self-renewal and differentiation, SOX2, NANOG, POU5F1, SFRP2, DPPA4, SALL4, ZFP42 and MYCN expressed in ESCs, iPSCs and adult stem cells (ASCs), but not in MSCs. There are five functional genes of MSCs, TMEM119, FBLN5, KCNK2, CLDN11 and DKK1, which are not expressed in stem cells. Trajectory analysis displayed clear developmental cliffs from ESCs/iPSCs to ASCs and to MSCs. Adipose-derived MSCs, relative to other types of MSCs, exhibit a more consistent and broader spectrum of gene expression for regulatory and excrete function. This study identifies distinction markers between MSCs and stem cells, providing an alternative approach for quality control of MSCs in their propagation and further mechanistic insights into their action.
Collapse
Affiliation(s)
- Kaijing Yan
- Regenerative Medicine Research Center, Sichuan University West China Hospital, Chengdu, Sichuan, 610044, China
- Tasly Stem Cell Biology Laboratory, Tasly Group, Tianjin, 300410, China
| | - Fei Ma
- Tasly Stem Cell Biology Laboratory, Tasly Group, Tianjin, 300410, China
| | - Xiaoxi Song
- Tasly Stem Cell Biology Laboratory, Tasly Group, Tianjin, 300410, China
| | - Huizhen Wang
- Tasly Stem Cell Biology Laboratory, Tasly Group, Tianjin, 300410, China
| | - Pengchong Liu
- Tasly Stem Cell Biology Laboratory, Tasly Group, Tianjin, 300410, China
| | - Jinlai Zhang
- Tasly Stem Cell Biology Laboratory, Tasly Group, Tianjin, 300410, China
| | - Xin Jin
- Tasly Stem Cell Biology Laboratory, Tasly Group, Tianjin, 300410, China
| | - Pengfei Han
- Tasly Stem Cell Biology Laboratory, Tasly Group, Tianjin, 300410, China
| | - Xiao Zuo
- Tasly Stem Cell Biology Laboratory, Tasly Group, Tianjin, 300410, China
| | - Y. James Kang
- Regenerative Medicine Research Center, Sichuan University West China Hospital, Chengdu, Sichuan, 610044, China
- Tasly Stem Cell Biology Laboratory, Tasly Group, Tianjin, 300410, China
| |
Collapse
|
18
|
Che Y, Shimizu Y, Murohara T. Therapeutic Potential of Adipose-Derived Regenerative Cells for Ischemic Diseases. Cells 2025; 14:343. [PMID: 40072072 PMCID: PMC11898683 DOI: 10.3390/cells14050343] [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: 01/22/2025] [Revised: 02/21/2025] [Accepted: 02/26/2025] [Indexed: 03/15/2025] Open
Abstract
Adipose-derived regenerative cells (ADRCs) are one of the most promising cell sources that possess significant therapeutic effects. They have now become a main source of cell therapy for the treatment of ischemic diseases due to their easy accessibility, expansion, and differentiation. Additionally, ADRCs can release multiple paracrine factors and extracellular vesicles that contribute to tissue regeneration by promoting angiogenesis, regulating inflammation, alleviating apoptosis, and inhibiting fibrosis. However, ADRCs still have some limitations to realize their full therapeutic potential. To address these issues, protective mechanistic studies and bioengineering studies have been carried out. This review focused on the recently studied mechanisms, such as paracrine factors, cell fusion, and mitochondrial transfer, involving the therapeutic potential of ADRCs in ischemic diseases and discussed some modification techniques of ADRCs.
Collapse
Affiliation(s)
| | - Yuuki Shimizu
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | | |
Collapse
|
19
|
Stadelmann N, Horch RE, Schmid R, Ostendorf D, Peddi A, Promny T, Boos AM, Kengelbach-Weigand A. Growth factors IGF-1 and KGF and adipose-derived stem cells promote migration and viability of primary human keratinocytes in an in vitro wound model. Front Med (Lausanne) 2025; 12:1516116. [PMID: 39981084 PMCID: PMC11839819 DOI: 10.3389/fmed.2025.1516116] [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: 10/23/2024] [Accepted: 01/20/2025] [Indexed: 02/22/2025] Open
Abstract
Introduction In the field of plastic surgery, epidermal transplantation is a potential treatment for chronic wounds that results in only minor donor site morbidity. Improving the regenerative capacities of epidermal grafts or single-cell suspensions and therefore accelerating healing processes would be of significant interest. Methods In the present study, we analyzed the effects of growth factors and adipose-derived stem cells (ADSCs) on keratinocyte properties. For optimum translation into the clinical setting, primary human keratinocytes and patient-matched ADSCs were isolated and used in an in vitro wound model. Results The keratinocyte migration and viability increased after treatment with the growth factors insulin-like growth factor 1 (IGF-1) and keratinocyte growth factor (KGF). A similar effect was observed with the use of a concentrated ADSC-conditioned medium (ADSC-CM). It was further possible to isolate the keratinocytes in a xenogen-free medium, which is essential for clinical translation. Importantly, a patient-dependent influence on the effects of the growth factors and ADSC-CM was observed. Discussion This study provides potential for the improvement of epidermal transplantation in the treatment of chronic wounds using xenogen-free isolated and cultivated keratinocytes, growth factors, and ADSC. Translating these results into clinical application may help accelerate wound healing and shorten the time until patients can return to everyday life.
Collapse
Affiliation(s)
| | - Raymund E. Horch
- Department of Plastic and Hand Surgery and Laboratory for Tissue Engineering and Regenerative Medicine, University Hospital of Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | | | | | | | | | | | | |
Collapse
|
20
|
Vembuli H, Rajasingh S, Nabholz P, Guenther J, Morrow BR, Taylor MM, Aghazadeh M, Sigamani V, Rajasingh J. Induced mesenchymal stem cells generated from periodontal ligament fibroblast for regenerative therapy. Exp Biol Med (Maywood) 2025; 250:10342. [PMID: 39963344 PMCID: PMC11830513 DOI: 10.3389/ebm.2025.10342] [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: 08/13/2024] [Accepted: 01/16/2025] [Indexed: 02/20/2025] Open
Abstract
Bone fractures and bone loss represent significant global health challenges, with their incidence rising due to an aging population. Despite autologous bone grafts remain the gold standard for treatment, challenges such as limited bone availability, immune reactions, and the risk of infectious disease transmission have driven the search for alternative cell-based therapies for bone regeneration. Stem cells derived from oral tissues and umbilical cord mesenchymal stem cells (MSCs) have shown potential in both preclinical and clinical studies for bone tissue regeneration. However, their limited differentiation capacity and wound healing abilities necessitate the exploration of alternative cell sources. In this study, we generated induced pluripotent stem cells (iPSCs) using a safe, nonviral and mRNA-based approach from human periodontal ligament fibroblasts (PDLF), an easily accessible cell source. These iPSCs were subsequently differentiated into MSCs, referred to as induced MSCs (iMSCs). The resulting iMSCs were homogeneous, highly proliferative, and possessed anti-inflammatory properties, suggesting their potential as a superior alternative to traditional MSCs for regenerative therapy. These iMSCs demonstrated trilineage differentiation potential, giving rise to osteocytes, chondrocytes, and adipocytes. The iMSC-derived osteocytes (iOSTs) were homogeneous, patient-specific and showed excellent attachment and growth on commercial collagen-based membranes, highlighting their suitability for bone tissue regeneration applications. Given their promising characteristics compared to traditional MSCs, PDLF-derived iMSCs are strong candidates for future clinical studies in bone regeneration and other regenerative dental therapies.
Collapse
Affiliation(s)
- Hemanathan Vembuli
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Sheeja Rajasingh
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Patrick Nabholz
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Jefferson Guenther
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Brian R. Morrow
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Margaret M. Taylor
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Marziyeh Aghazadeh
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Vinoth Sigamani
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Johnson Rajasingh
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN, United States
- Department of Medicine-Cardiology, University of Tennessee Health Science Center, Memphis, TN, United States
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, United States
| |
Collapse
|
21
|
Schultz IC, Dos Santos Pereira Andrade AC, Dubuc I, Laroche A, Allaeys I, Doré E, Bertrand N, Vallières L, Fradette J, Flamand L, Wink MR, Boilard E. Targeting Cytokines: Evaluating the Potential of Mesenchymal Stem Cell Derived Extracellular Vesicles in the Management of COVID-19. Stem Cell Rev Rep 2025; 21:564-580. [PMID: 39340739 DOI: 10.1007/s12015-024-10794-4] [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] [Accepted: 09/24/2024] [Indexed: 09/30/2024]
Abstract
The Coronavirus Disease 2019 (COVID-19), caused by virus SARS-CoV-2, is characterized by massive inflammation and immune system imbalance. Despite the implementation of vaccination protocols, the accessibility of treatment remains uneven. Furthermore, the persistent threat of new variants underscores the urgent need for expanded research into therapeutic options for SARS-CoV-2. Mesenchymal stem cells (MSCs) are known for their immunomodulatory potential through the release of molecules into the extracellular space, either as soluble elements or carried by extracellular vesicles (EVs). The aim of this study was to evaluate the anti-inflammatory potential of EVs obtained from human adipose tissue (ASC-EVs) against SARS-CoV-2 infection. ASC-EVs were purified by size-exclusion chromatography, and co-culture assays confirmed that ASC-EVs were internalized by human lung cells and could colocalize with SARS-CoV-2 into early and late endosomes. To determine the functionality of ASC-EVs, lung cells were infected with SARS-CoV-2 in the presence of increasing concentrations of ASC-EVs, and the release of cytokines, chemokines and viruses were measured. While SARS-CoV-2 replication was significantly reduced only at the highest concentrations tested, multiplex analysis highlighted that lower concentrations of ASC-EV sufficed to prevent the production of immune modulators. Importantly, ASC-EVs did not contain detectable inflammatory cytokines, nor did they trigger inflammatory mediators, nor affect cellular viability. In conclusion, this work suggests that ASC-EVs have the potential to attenuate inflammation by decreasing the production of pro-inflammatory cytokines in lung cells following SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Iago Carvalho Schultz
- Axe Maladies Infectieuses et Immunitaires, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec, QC, Canada
- Departamento de Ciências Básicas da Saúde, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil
| | - Ana Claudia Dos Santos Pereira Andrade
- Axe Maladies Infectieuses et Immunitaires, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec, QC, Canada
- Département de Microbiologie-Infectiologie et d'Immunologie, Faculté de Médecine de l'Université Laval, Québec, QC, Canada
| | - Isabelle Dubuc
- Axe Maladies Infectieuses et Immunitaires, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec, QC, Canada
- Département de Microbiologie-Infectiologie et d'Immunologie, Faculté de Médecine de l'Université Laval, Québec, QC, Canada
| | - Audrée Laroche
- Axe Maladies Infectieuses et Immunitaires, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec, QC, Canada
- Département de Microbiologie-Infectiologie et d'Immunologie, Faculté de Médecine de l'Université Laval, Québec, QC, Canada
| | - Isabelle Allaeys
- Axe Maladies Infectieuses et Immunitaires, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec, QC, Canada
- Département de Microbiologie-Infectiologie et d'Immunologie, Faculté de Médecine de l'Université Laval, Québec, QC, Canada
| | - Etienne Doré
- Axe Maladies Infectieuses et Immunitaires, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec, QC, Canada
- Département de Microbiologie-Infectiologie et d'Immunologie, Faculté de Médecine de l'Université Laval, Québec, QC, Canada
| | - Nicolas Bertrand
- Axe Endocrinologie et Néphrologie, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec, QC, Canada
- Faculté de Pharmacie, Université Laval, Québec, QC, Canada
| | - Luc Vallières
- Axe Neurosciences, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec, QC, Canada
| | - Julie Fradette
- Centre de recherche en organogénèse expérimentale de l'Université Laval/LOEX, Québec, QC, Canada
- Département de Chirurgie, Faculté de Médecine de l'Université Laval, Québec, QC, Canada
- Division of Regenerative Medicine, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec, QC, Canada
| | - Louis Flamand
- Axe Maladies Infectieuses et Immunitaires, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec, QC, Canada
- Département de Microbiologie-Infectiologie et d'Immunologie, Faculté de Médecine de l'Université Laval, Québec, QC, Canada
| | - Marcia Rosangela Wink
- Departamento de Ciências Básicas da Saúde, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil
| | - Eric Boilard
- Axe Maladies Infectieuses et Immunitaires, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec, QC, Canada.
- Département de Microbiologie-Infectiologie et d'Immunologie, Faculté de Médecine de l'Université Laval, Québec, QC, Canada.
| |
Collapse
|
22
|
Sun Y, Liu Q, Zhu J, Hu H, Lu L, Ying J, Guo R, Ye X, Zhu L, Jiang H. Optimal Strategies for Autologous Fat Grafting in Breast Augmentation and Reconstruction: A Systematic Review and Network Meta-Analysis. Plast Reconstr Surg 2025; 155:243e-255e. [PMID: 39874946 DOI: 10.1097/prs.0000000000011653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
BACKGROUND Cell-assisted lipotransfer (CAL) and platelet-rich plasma (PRP)-assisted lipotransfer have been used to overcome the low survival rate of conventional lipotransfer. However, there is still insufficient evidence to determine which technique is the best strategy for autologous fat grafting in breast cosmetic and reconstructive surgery. The present study aimed to compare the efficacy of traditional fat transplantation, CAL, and PRP-assisted lipotransfer. METHODS A systematic search was conducted in several databases, including PubMed, Web of Science, Cochrane, ClinicalTrials.gov, and Embase, concluding on January 21, 2024, to identify studies that met the inclusion criteria. Twelve studies were included after a rigorous selection process based on predefined criteria. Statistical analyses were conducted using R version 4.0.5 software with the netmeta and dmetar packages, utilizing a frequentist approach with a random-effects model. A network meta-analysis was performed to compare different fat graft procedures with regard to fat survival rate and complication events. RESULTS CAL and PRP-assisted lipotransfer were better than traditional fat grafting in terms of fat survival rate. In addition, there was no significant difference in the incidence of postoperative complications among the CAL, PRP, and traditional groups. CONCLUSIONS Given the results of network meta-analysis, it appears that both CAL and PRP-assisted lipotransfer have a higher fat survival rate for autologous fat grafting in breast augmentation and reconstruction. However, the transplantation strategy still needs to be analyzed based on actual conditions in clinical applications.
Collapse
Affiliation(s)
- Yulin Sun
- From the Department of Plastic Surgery, Shanghai East Hospital, Tongji University School of Medicine
| | - Qian Liu
- Shanghai Song Jiang District Sijing Hospital
| | - Jie Zhu
- From the Department of Plastic Surgery, Shanghai East Hospital, Tongji University School of Medicine
| | - Hao Hu
- From the Department of Plastic Surgery, Shanghai East Hospital, Tongji University School of Medicine
| | - Lu Lu
- From the Department of Plastic Surgery, Shanghai East Hospital, Tongji University School of Medicine
| | - Jianghui Ying
- From the Department of Plastic Surgery, Shanghai East Hospital, Tongji University School of Medicine
| | - Rong Guo
- From the Department of Plastic Surgery, Shanghai East Hospital, Tongji University School of Medicine
| | - Xiuyu Ye
- From the Department of Plastic Surgery, Shanghai East Hospital, Tongji University School of Medicine
| | - Lie Zhu
- Department of Plastic and Reconstructive Surgery, Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital)
| | - Hua Jiang
- From the Department of Plastic Surgery, Shanghai East Hospital, Tongji University School of Medicine
| |
Collapse
|
23
|
Cremona M, Gallazzi M, Rusconi G, Mariotta L, Gola M, Soldati G. State of the Art in the Standardization of Stromal Vascular Fraction Processing. Biomolecules 2025; 15:199. [PMID: 40001502 PMCID: PMC11852902 DOI: 10.3390/biom15020199] [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: 12/24/2024] [Revised: 01/22/2025] [Accepted: 01/26/2025] [Indexed: 02/27/2025] Open
Abstract
Stromal Vascular Fraction (SVF) has gained significant attention in clinical applications due to its regenerative and anti-inflammatory properties. Initially identified decades ago, SVF is derived from adipose tissue and has been increasingly utilized in a variety of therapeutic settings. The isolation and processing protocols for SVF have evolved substantially, particularly after its classification as an Advanced Therapy Medicinal Product (ATMP), which mandates adherence to Good Manufacturing Practices to ensure sterility and product quality. Despite the progress, few studies over the last decade have focused on the standardization of SVF processing. Recent advances, driven by the potential of SVF and its derived products such as Adipose-derived Stem Cells, have prompted the development of improved isolation strategies aimed at enhancing their therapeutic and regenerative efficacy. Notable progress includes the advent of automated processing systems, which reduce technical errors, minimize variability, and improve reproducibility across laboratories. These developments, along with the establishment of more precise protocols and guidelines, have enhanced the consistency and clinical applicability of SVF-based therapies. This review discusses the key aspects of SVF isolation and processing, highlighting the efforts to standardize the procedure and ensure the reliability of SVF products for clinical use.
Collapse
Affiliation(s)
- Martina Cremona
- Swiss Stem Cell Foundation, 6900 Lugano, Switzerland; (M.C.)
| | - Matteo Gallazzi
- Swiss Stem Cell Foundation, 6900 Lugano, Switzerland; (M.C.)
| | - Giulio Rusconi
- Swiss Stem Cell Foundation, 6900 Lugano, Switzerland; (M.C.)
| | - Luca Mariotta
- Swiss Stem Cell Foundation, 6900 Lugano, Switzerland; (M.C.)
- Swiss Stem Cells Biotech AG, 8008 Zürich, Switzerland
| | - Mauro Gola
- Swiss Stem Cell Foundation, 6900 Lugano, Switzerland; (M.C.)
| | - Gianni Soldati
- Swiss Stem Cell Foundation, 6900 Lugano, Switzerland; (M.C.)
| |
Collapse
|
24
|
Gallo MC, Elias A, Reynolds J, Ball JR, Lieberman JR. Regional Gene Therapy for Bone Tissue Engineering: A Current Concepts Review. Bioengineering (Basel) 2025; 12:120. [PMID: 40001640 PMCID: PMC11852166 DOI: 10.3390/bioengineering12020120] [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: 12/24/2024] [Revised: 01/20/2025] [Accepted: 01/24/2025] [Indexed: 02/27/2025] Open
Abstract
The management of segmental bone defects presents a complex reconstruction challenge for orthopedic surgeons. Current treatment options are limited by efficacy across the spectrum of injury, morbidity, and cost. Regional gene therapy is a promising tissue engineering strategy for bone repair, as it allows for local implantation of nucleic acids or genetically modified cells to direct specific protein expression. In cell-based gene therapy approaches, a variety of different cell types have been described including mesenchymal stem cells (MSCs) derived from multiple sources-bone marrow, adipose, skeletal muscle, and umbilical cord tissue, among others. MSCs, in particular, have been well studied, as they serve as a source of osteoprogenitor cells in addition to providing a vehicle for transgene delivery. Furthermore, MSCs possess immunomodulatory properties, which may support the development of an allogeneic "off-the-shelf" gene therapy product. Identifying an optimal cell type is paramount to the successful clinical translation of cell-based gene therapy approaches. Here, we review current strategies for the management of segmental bone loss in orthopedic surgery, including bone grafting, bone graft substitutes, and operative techniques. We also highlight regional gene therapy as a tissue engineering strategy for bone repair, with a focus on cell types and cell sources suitable for this application.
Collapse
Affiliation(s)
- Matthew C. Gallo
- Department of Orthopaedic Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033, USA; (M.C.G.); (A.E.); (J.R.); (J.R.B.)
| | - Aura Elias
- Department of Orthopaedic Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033, USA; (M.C.G.); (A.E.); (J.R.); (J.R.B.)
| | - Julius Reynolds
- Department of Orthopaedic Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033, USA; (M.C.G.); (A.E.); (J.R.); (J.R.B.)
| | - Jacob R. Ball
- Department of Orthopaedic Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033, USA; (M.C.G.); (A.E.); (J.R.); (J.R.B.)
| | - Jay R. Lieberman
- Department of Orthopaedic Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033, USA; (M.C.G.); (A.E.); (J.R.); (J.R.B.)
- Alfred E. Mann Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, USA
| |
Collapse
|
25
|
Hajivalili M, Nikkhoo N, Salahi S, Hosseini M. Traumatic brain Injury: Comprehensive overview from pathophysiology to Mesenchymal stem Cell-Based therapies. Int Immunopharmacol 2025; 146:113816. [PMID: 39708488 DOI: 10.1016/j.intimp.2024.113816] [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: 09/23/2024] [Revised: 11/16/2024] [Accepted: 12/05/2024] [Indexed: 12/23/2024]
Abstract
Traumatic brain injury (TBI) is a disastrous phenomenon which is considered to cause high mortality and morbidity rate. Regarding the importance of TBI due to its prevalence and its effects on the brain and other organs, finding new therapeutic methods and improvement of conventional therapies seems to be vital. TBI involves a complex physiological mechanism, with inflammation being a key component among various contributing factors. After incidence of TBI, inflammation can act as a double-edged sword in the process. Inflammation actually plays its role both as initiator and progressive index during TBI which can accumulate myeloid and lymphoid immune cells and trigger cell death pathways. Through this study we made this concept bold that that besides conventional therapies that could be used for traumatic brain injury, treatments based on mesenchyme stem cells (MSCs) and their derivatives including secretomes and exosomes demonstrate more efficacies particularly in preventing secondary injuries caused by TBI. Of note, we highlighted the valuable features of MSC-based therapies such as self-direction toward inflamed tissues and amplifying neuro-regenerative aspects. We listed possible challenges in the way of reaching this therapy to clinic to provide a clear and updated of the field.
Collapse
Affiliation(s)
- Mahsa Hajivalili
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| | - Nima Nikkhoo
- Student research committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sarvenaz Salahi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Maryam Hosseini
- Trauma Research Center, Shahid Rajaee (Emtiaz) Trauma Hospital, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
26
|
Funke S, Wiggenhauser PS, Grundmeier A, Fuchs B, Koban K, Demmer W, Giunta RE, Kuhlmann C. Aspirin Inhibits the In Vitro Adipogenic Differentiation of Human Adipose Tissue-Derived Stem Cells in a Dose-Dependent Manner. Int J Mol Sci 2025; 26:853. [PMID: 39859567 PMCID: PMC11766433 DOI: 10.3390/ijms26020853] [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: 12/15/2024] [Revised: 01/11/2025] [Accepted: 01/15/2025] [Indexed: 01/27/2025] Open
Abstract
Aspirin (ASA) is one of the most used medications worldwide and has shown various effects on cellular processes, including stem cell differentiation. However, the effect of ASA on adipogenesis of adipose tissue-derived stem cells (ASCs) remains largely unknown. Considering the potential application of ASCs in regenerative medicine and cell-based therapies, this study investigates the effects of ASA on adipogenic differentiation in human ASCs. ASCs were exposed to varying concentrations of ASA (0 µM, 400 µM, and 1000 µM) and evaluated for changes in morphology, migration, and adipogenic differentiation. While ASA exposure did not affect self-renewal potential, migration ability, or cell morphology, it significantly reduced lipid vacuole formation at 1000 µM after 21 days of adipogenic differentiation (p = 0.0025). This visible inhibition correlated with decreased expression of adipogenic markers (PPARG, ADIPOQ, and FABP4) and the proliferation marker MKi67 under ASA exposure in comparison to the control (ns). Overall, the findings demonstrate that ASA inhibits adipogenic differentiation of human ASCs in a dose-dependent manner in vitro, contrasting its known role in promoting osteogenic differentiation. This research highlights ASA's complex effects on ASCs and emphasizes the need for further investigation into its mechanisms and potential therapeutic applications in obesity and metabolic diseases. The inhibitory effects of ASA on adipogenesis should be considered in cell-based therapies using ASCs.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Constanze Kuhlmann
- Division of Hand Surgery, Plastic Surgery and Aesthetic Surgery, University Hospital, LMU Munich, Ziemssenstraße 5, 80336 Munich, Germany; (S.F.); (P.S.W.); (A.G.); (B.F.); (K.K.); (W.D.); (R.E.G.)
| |
Collapse
|
27
|
Li P, Cao L, Liu T, Lu X, Ma Y, Wang H. The Effect of Adipose-Derived Stem Cell (ADSC)-Exos on the Healing of Autologous Skin Grafts in Miniature Pigs. Int J Mol Sci 2025; 26:479. [PMID: 39859193 PMCID: PMC11764972 DOI: 10.3390/ijms26020479] [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: 10/16/2024] [Revised: 12/06/2024] [Accepted: 01/06/2025] [Indexed: 01/27/2025] Open
Abstract
The skin functions as the body's primary defense barrier; when compromised, it can lead to dehydration, infection, shock, or potentially life-threatening conditions. Miniature pigs exhibit skin characteristics and healing processes highly analogous to humans. Mesenchymal stem cells contribute to skin injury repair through a paracrine mechanism involving exosomes. This research examines whether adipose-derived MSC exosomes effectively enhance healing following autologous skin grafting in miniature pigs. It also compares the roles and distinctions of ADSCs and ADSC-Exos in inflammatory responses and tissue regeneration. This study found significantly reduced levels of oxidative stress products and pro-inflammatory factors, while antioxidant factors, anti-inflammatory factors, and pro-regenerative factors were elevated, and anti-regenerative factor levels decreased. Moreover, the expression levels of key markers-namely, PI3K, Akt, and mTOR-in the regeneration-associated signaling pathway were increased. The alterations in these indicators indicate that ADSC-Exos can regulate inflammatory responses and promote regeneration. This study provides a novel theoretical foundation for the implementation of acellular therapy in clinical settings.
Collapse
Affiliation(s)
- Pujun Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150300, China; (P.L.); (L.C.); (T.L.); (X.L.); (Y.M.)
- Heilongjiang Provincial Key Laboratory of Pathogenic Mechanism for Animal Disease and Comparative Medicine, Harbin 150300, China
| | - Lei Cao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150300, China; (P.L.); (L.C.); (T.L.); (X.L.); (Y.M.)
- Heilongjiang Provincial Key Laboratory of Pathogenic Mechanism for Animal Disease and Comparative Medicine, Harbin 150300, China
| | - Tao Liu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150300, China; (P.L.); (L.C.); (T.L.); (X.L.); (Y.M.)
- Heilongjiang Provincial Key Laboratory of Pathogenic Mechanism for Animal Disease and Comparative Medicine, Harbin 150300, China
| | - Xiangyu Lu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150300, China; (P.L.); (L.C.); (T.L.); (X.L.); (Y.M.)
- Heilongjiang Provincial Key Laboratory of Pathogenic Mechanism for Animal Disease and Comparative Medicine, Harbin 150300, China
| | - Yajun Ma
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150300, China; (P.L.); (L.C.); (T.L.); (X.L.); (Y.M.)
- Heilongjiang Provincial Key Laboratory of Pathogenic Mechanism for Animal Disease and Comparative Medicine, Harbin 150300, China
| | - Hongbin Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150300, China; (P.L.); (L.C.); (T.L.); (X.L.); (Y.M.)
- Heilongjiang Provincial Key Laboratory of Pathogenic Mechanism for Animal Disease and Comparative Medicine, Harbin 150300, China
| |
Collapse
|
28
|
Chen M, Wang J, Liu Y, Cui X, Liang J, Lei N, Xiao Y, Wang Q, Fan Y, Zhang X. Injectable biomimetic microcarriers harness synergistic effects of paracrine factors and cellular membranes to alleviate osteoarthritis. CHEMICAL ENGINEERING JOURNAL 2025; 503:158451. [DOI: 10.1016/j.cej.2024.158451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
29
|
Long Q, Yuan Y, Ou Y, Li W, Yan Q, Zhang P, Yuan X. Integrative single-cell RNA-seq and ATAC-seq analysis of the evolutionary trajectory features of adipose-derived stem cells induced into astrocytes. J Neurochem 2025; 169:e16269. [PMID: 39700048 DOI: 10.1111/jnc.16269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 10/24/2024] [Accepted: 10/30/2024] [Indexed: 12/21/2024]
Abstract
This study employs single-cell RNA sequencing (scRNA-seq) and assay for transposase-accessible chromatin with high-throughput sequencing technologies (scATAC-seq) to perform joint sequencing on cells at various time points during the induction of adipose-derived stem cells (ADSCs) into astrocytes. We applied bioinformatics approaches to investigate the differentiation trajectories of ADSCs during their induced differentiation into astrocytes. Pseudotemporal analysis was used to infer differentiation trajectories. Additionally, we assessed chromatin accessibility patterns during the differentiation process. Key transcription factors driving the differentiation of ADSCs into astrocytes were identified using motif and footprint methods. Our analysis revealed significant shifts in gene expression during the induction process, with astrocyte-related genes upregulated and stem cell-related genes downregulated. ADSCs first differentiated into neural stem cell-like cells with high plasticity, which further matured into astrocytes via two distinct pathways. Marked changes in chromatin accessibility were observed during ADSC-induced differentiation, affecting transcription regulation and cell function. Transcription factors analysis identified NFIA/B/C/X and CEBPA/B/D as key regulators in ADSCs differentiation into astrocytes. We observed a correlation between chromatin accessibility and gene expression, with ADSCs exhibiting broad chromatin accessibility prior to lineage commitment, where chromatin opening precedes transcription initiation. In summary, we found that ADSCs first enter a neural stem cell-like state before differentiating into astrocytes. ADSCs also display extensive chromatin accessibility prior to astrocyte differentiation, although transcription has not yet been initiated. These findings offer a theoretical framework for understanding the molecular mechanisms underlying this process.
Collapse
Affiliation(s)
- Qingxi Long
- Department of Neurology, Kailuan General Hospital, Affiliated North China University of Science and Technology, Tangshan, China
| | - Yi Yuan
- Department of Pediatric Othopedic, Children's Hospital of Capital Institute of Pediatrics, Beijing, China
| | - Ya Ou
- Department of Neurology, Kailuan General Hospital, Affiliated North China University of Science and Technology, Tangshan, China
- Hebei Provincial Key Laboratory of Neurobiological Function, Tangshan, China
| | - Wen Li
- Department of Neurology, Kailuan General Hospital, Affiliated North China University of Science and Technology, Tangshan, China
| | - Qi Yan
- Department of Neurology, Kailuan General Hospital, Affiliated North China University of Science and Technology, Tangshan, China
| | - Pingshu Zhang
- Department of Neurology, Kailuan General Hospital, Affiliated North China University of Science and Technology, Tangshan, China
- Hebei Provincial Key Laboratory of Neurobiological Function, Tangshan, China
| | - Xiaodong Yuan
- Department of Neurology, Kailuan General Hospital, Affiliated North China University of Science and Technology, Tangshan, China
- Hebei Provincial Key Laboratory of Neurobiological Function, Tangshan, China
| |
Collapse
|
30
|
Sueters J, van Heiningen R, de Vries R, Guler Z, Huirne J, Smit T. Advances in tissue engineering of peripheral nerve and tissue innervation - a systematic review. J Tissue Eng 2025; 16:20417314251316918. [PMID: 39911939 PMCID: PMC11795627 DOI: 10.1177/20417314251316918] [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: 09/10/2024] [Accepted: 01/15/2025] [Indexed: 02/07/2025] Open
Abstract
Although various options are available to treat injured organs and peripheral nerves, none is without limitations. Auto- and allografts are the first choice of treatment, but tissue survival or functionality is not guaranteed due to often limited vascular and neural networks. In response, tissue-engineered solutions have been developed, yet clinical translations is rare. In this study, a systematic review was performed on tissue-engineered advancements for peripheral nerves and tissues, to aid future developments in bridging the gap toward the clinic by identifying high-potential solutions and unexplored areas. A systematic search was performed in PubMed, Embase, Web of Science, and Scopus until November 9, 2023. Search terms involved "tissue engineering," "guided," "tissue scaffold," and "tissue graft," together with "innervation" and "reinnervation." Original in vivo or in vitro studies meeting the inclusion criteria (tissue-engineered peripheral nerve/innervation of tissue) and no exclusion criteria (no full text available; written in foreign language; nonoriginal article; tissue-engineering of central nervous system; publication before 2012; insufficient study quality or reproducibility) were assessed. A total of 68 out of 3626 original studies were included. Data extraction was based on disease model, cell origin and host species, biomaterial nature and composition, and external stimuli of biological, chemical or physical origin. Although tissue engineering is still in its infancy, explored innervation strategies of today were highlighted with respect to biomaterials, cell types, and external stimuli. The findings emphasize that natural biomaterials, pre-seeding with autologous cell sources, and solutions for reproductive organs are beneficial for future research. Natural biomaterials possess important cues required for cell-material interaction and closely resemble native tissue in terms of biomechanical, geometrical and chemical composition. Autologous cells induce biomaterial functionalization. As these solutions pose no risk of immunorejection and have demonstrated good outcomes, they are most likely to fulfill the clinical demands.
Collapse
Affiliation(s)
- Jayson Sueters
- Department of Gynaecology, Amsterdam UMC – location VUmc, Amsterdam, The Netherlands
- Amsterdam Reproduction and Development Research Institute, Amsterdam UMC, Amsterdam, the Netherlands
| | - Rowan van Heiningen
- Department of Gynaecology, Amsterdam UMC – location VUmc, Amsterdam, The Netherlands
- Amsterdam Reproduction and Development Research Institute, Amsterdam UMC, Amsterdam, the Netherlands
- Angiogenesis Laboratory, Cancer Center Amsterdam, Department of Medical Oncology, Amsterdam UMC – location VUmc, Amsterdam, The Netherlands
| | - Ralph de Vries
- Medical Library, Vrije Universiteit, Amsterdam, The Netherlands
| | - Zeliha Guler
- Amsterdam Reproduction and Development Research Institute, Amsterdam UMC, Amsterdam, the Netherlands
- Department of Obstetrics and Gynecology, Amsterdam UMC – location AMC, Amsterdam, The Netherlands
| | - Judith Huirne
- Department of Gynaecology, Amsterdam UMC – location VUmc, Amsterdam, The Netherlands
- Amsterdam Reproduction and Development Research Institute, Amsterdam UMC, Amsterdam, the Netherlands
| | - Theo Smit
- Department of Gynaecology, Amsterdam UMC – location VUmc, Amsterdam, The Netherlands
- Amsterdam Reproduction and Development Research Institute, Amsterdam UMC, Amsterdam, the Netherlands
- Department of Medical Biology, Amsterdam UMC – location AMC, Amsterdam, The Netherlands
| |
Collapse
|
31
|
Ahn SW, Kim EJ, Kim MK, Shin SH, Kwon JJ. Membrane-free stem cell components suppress osteoclast differentiation: Implications for oral regenerative treatment. J Dent Sci 2025; 20:212-219. [PMID: 39873034 PMCID: PMC11762959 DOI: 10.1016/j.jds.2024.08.005] [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: 07/22/2024] [Revised: 08/07/2024] [Indexed: 01/30/2025] Open
Abstract
Background/purpose Membrane-free stem cell components (MFSCCs) have been developed by removing cell membranes with antigens to overcome the limitations associated with cell-based therapies and isolate effective peptides. MFSCCs have been reported to have effects on oral infection sites. Chronic inflammatory diseases cause excessive bone resorption. This study investigated the effects of MFSCCs on osteoclast differentiation in the context of the high prevalence of inflammatory bone resorption. Materials and methods Bone marrow macrophages (BMMs) were treated with macrophage colony-stimulating factor and receptor activator of nuclear factor kappa-B ligand. Osteoclast differentiation was assessed based on the MFSCC concentrations. Tartrate-resistant acid phosphatase (TRAP)-stained mature osteoclasts and multinucleated cells derived from BMMs were analyzed using light microscopy. The messenger RNA (mRNA) expression levels of genes related to osteoclast differentiation were measured using real-time polymerase chain reaction (RT-PCR). The relative expression levels of the key transcription factors c-fos and nuclear factor of activated T cells (NFATc1) were determined using quantitative RT-PCR and western blotting. Results After treatment with MFSCCs, the cell viability was similar, depending on the level of BMMs. As the MFSCC concentration increased, the number of TRAP-positive cells decreased. The mRNA and protein expression of cathepsin K, TRAP, dendritic cell-specific transmembrane protein, c-fos, and NFATc1 decreased as the MFSCC concentration increased. Conclusion Our findings demonstrate that MFSCCs suppress osteoclast differentiation by downregulating transcription factors, particularly, c-fos and NFATc1. Therefore, MFSCCs may serve as a conservative treatment option for chronic inflammatory bone resorption diseases of the oral cavity by suppressing excessive bone resorption.
Collapse
Affiliation(s)
- Sang-Wook Ahn
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Pusan National University, Dental and Life Science Institute, Yangsan, Republic of Korea
| | - Eun-Jung Kim
- Department of Dental Anesthesia and Pain Medicine, School of Dentistry, Pusan National University, Dental Research Institute, Yangsan, Republic of Korea
| | - Mi Kyoung Kim
- Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
| | - Sang-Hun Shin
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Pusan National University, Dental and Life Science Institute, Yangsan, Republic of Korea
| | - Jin-Ju Kwon
- Department of Dentistry, Yeungnam University College of Medicine, Daegu, Republic of Korea
| |
Collapse
|
32
|
Zhang P, Pei H, Zhou G, Fu Q, Bai R, Lin P, Wu Q, Xu X, Chen M. Effectiveness and Safety of Micro-Plasma Radiofrequency Treatment Combined With Autologous Chyle Fat Grafting Treatment for Hypertrophic Scars: A Retrospective Study. J Cosmet Dermatol 2025; 24:e16728. [PMID: 39731280 DOI: 10.1111/jocd.16728] [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: 08/23/2024] [Revised: 11/23/2024] [Accepted: 12/02/2024] [Indexed: 12/29/2024]
Abstract
BACKGROUND Hypertrophic scar (HS) is a fibroproliferative disorder resulting from abnormal healing of skin tissue after injury. Although various therapies are currently employed in clinical to treat HSs, there is no widely accepted standard therapy. Micro-plasma radiofrequency (MPR) and autologous chyle fat grafting are emerging treatments for this condition, and they have demonstrated promising therapeutic outcomes in clinical applications. The aim of this study is to investigate the effectiveness and safety of combining MPR with autologous chyle fat grafting for the treatment of HSs. METHODS We performed a retrospective study on patients diagnosed with HS in a single center between January 2020 and December 2023. According to the treatments, patients were divided into three groups, with 6 months follow-up. The single therapy group received MPR alone for two times. The combined therapy Group 1 first received the MPR treatment followed by the combined treatment. The combined therapy Group 2 first received the combined treatment and then received the MPR treatment. The effectiveness of treatment was evaluated using the Vancouver Scar Scale (VSS) and the Patient Scar Assessment Scale (PSAS). The Visual Analog Scale (VAS) was used to assess the patients' pain on the day of treatment and 1 day after treatment. Adverse events and complications were recorded to assess the safety of treatment. RESULTS A total of 73 patients diagnosed with HS were enrolled in this study, including 35 patients in the single therapy group, 18 patients in the combined therapy Group 1, and 20 patients in the combined therapy Group 2. After the treatments were completed, all three groups exhibited significant effectiveness. The two combined therapy groups scored lower after treatments in the VSS, which includes height, vascularity, pliability, and total scores, as well as in the PSAS, which includes color, stiffness, thickness, and total scores, compared to the single therapy group, with a statistically significant difference. Regarding pain response to treatment, there was no statistical difference in VAS among the three groups. No statistical difference in the overall incidence of adverse events was observed among the three groups, and no severe complications were recorded. CONCLUSIONS This study revealed the combination of MPR and autologous chyle fat grafting showed superior effectiveness compared to MPR alone in treating HSs, without any observed increase in overall adverse event frequency. For patients diagnosed with HS, this combination therapy stands as a promising and effective clinical intervention.
Collapse
Affiliation(s)
- Peixuan Zhang
- Department of Plastic and Reconstructive Surgery, Senior Department of Burns and Plastic Surgery, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Haina Pei
- Department of Burn and Plastic Surgery, Hainan Hospital of Chinese PLA General Hospital, Sanya, Hainan, China
| | - Guiwen Zhou
- Department of Plastic and Reconstructive Surgery, Senior Department of Burns and Plastic Surgery, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Qiang Fu
- Department of Plastic and Reconstructive Surgery, Senior Department of Burns and Plastic Surgery, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Ruiqi Bai
- Department of Plastic and Reconstructive Surgery, Senior Department of Burns and Plastic Surgery, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Pianpian Lin
- Department of Plastic and Reconstructive Surgery, Senior Department of Burns and Plastic Surgery, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Qian Wu
- Department of Plastic and Reconstructive Surgery, Senior Department of Burns and Plastic Surgery, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Xiao Xu
- Department of Ophthalmology, The Third Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Minliang Chen
- Department of Plastic and Reconstructive Surgery, Senior Department of Burns and Plastic Surgery, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
33
|
Wolint P, Hofmann S, von Atzigen J, Böni R, Miescher I, Giovanoli P, Calcagni M, Emmert MY, Buschmann J. Standardization to Characterize the Complexity of Vessel Network Using the Aortic Ring Model. Int J Mol Sci 2024; 26:291. [PMID: 39796147 PMCID: PMC11719671 DOI: 10.3390/ijms26010291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 12/05/2024] [Accepted: 12/30/2024] [Indexed: 01/13/2025] Open
Abstract
Regeneration after ischemia requires to be promoted by (re)perfusion of the affected tissue, and, to date, there is no therapy that covers all needs. In treatment with mesenchymal stem cells (MSC), the secretome acts via paracrine mechanisms and has a positive influence on vascular regeneration via proangiogenic factors. A lack of standardization and the high complexity of vascular structures make it difficult to compare angiogenic readouts from different studies. This emphasizes the need for improved approaches and the introduction of an index in the preclinical setting. A characterization of human MSC secretomes obtained from one of the three formats-single cells, small, and large spheroids-was performed using the chicken aortic ring assay in combination with a modified angiogenic activity index (AAI) and an angiogenic profile. While the secretome of the small spheroid group showed an inhibitory effect on angiogenesis, the large spheroid group impressed with a fully pro-angiogenic response, and a higher AAI compared to the single cell group, underlying the suitability of these three-stem cell-derived secretomes with their distinct angiogenic properties to validate the AAI and the novel angiogenic profile established here.
Collapse
Affiliation(s)
- Petra Wolint
- Division of Surgical Research, University Hospital of Zurich, 8091 Zurich, Switzerland
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, 8091 Zurich, Switzerland; (S.H.); (J.v.A.); (I.M.); (P.G.); (M.C.)
| | - Silvan Hofmann
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, 8091 Zurich, Switzerland; (S.H.); (J.v.A.); (I.M.); (P.G.); (M.C.)
| | - Julia von Atzigen
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, 8091 Zurich, Switzerland; (S.H.); (J.v.A.); (I.M.); (P.G.); (M.C.)
| | - Roland Böni
- White House Center for Liposuction, 8044 Zurich, Switzerland;
| | - Iris Miescher
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, 8091 Zurich, Switzerland; (S.H.); (J.v.A.); (I.M.); (P.G.); (M.C.)
| | - Pietro Giovanoli
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, 8091 Zurich, Switzerland; (S.H.); (J.v.A.); (I.M.); (P.G.); (M.C.)
| | - Maurizio Calcagni
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, 8091 Zurich, Switzerland; (S.H.); (J.v.A.); (I.M.); (P.G.); (M.C.)
| | - Maximilian Y. Emmert
- Institute for Regenerative Medicine (IREM), University of Zurich, 8952 Zurich, Switzerland;
- Deutsches Herzzentrum der Charité (DHZC), Department of Cardiothoracic and Vascular Surgery, 13353 Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 13353 Berlin, Germany
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Johanna Buschmann
- Division of Surgical Research, University Hospital of Zurich, 8091 Zurich, Switzerland
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, 8091 Zurich, Switzerland; (S.H.); (J.v.A.); (I.M.); (P.G.); (M.C.)
| |
Collapse
|
34
|
Ljubić A, Dinić M, Švraka D, Vujović S. Dual-Double Stem Cell Ovarian Therapy: A Comprehensive Approach in Regenerative Medicine. Int J Mol Sci 2024; 26:69. [PMID: 39795929 PMCID: PMC11719681 DOI: 10.3390/ijms26010069] [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: 09/23/2024] [Revised: 11/10/2024] [Accepted: 11/27/2024] [Indexed: 01/13/2025] Open
Abstract
Dual-double stem cell therapy, which integrates mesenchymal stem cells (MSCs) and hematopoietic stem cells (HSCs), represents a cutting-edge approach in regenerative medicine, particularly for conditions such as ovarian decline, premature ovarian insufficiency (POI), and induced ovarian failure. This therapy leverages the unique properties of MSCs and HSCs, enhancing tissue repair, immune modulation, and overall regenerative outcomes. MSCs, known for their ability to differentiate into various cell types, provide a supportive microenvironment and secrete bioactive molecules that promote angiogenesis and reduce inflammation. HSCs, crucial for hematopoiesis and immune function, further enhance this environment by supporting hematopoietic processes and immune regulation. Clinical evidence increasingly supports the effectiveness of stem cell therapy in ovarian regeneration. Studies have demonstrated improved folliculogenesis, normalization of hormone profiles, and successful pregnancies in patients with POI. Furthermore, recent clinical trials in various medical fields underline the superior potential of dual-double therapy compared to monotherapies involving MSCs or HSCs alone, enhancing tissue repair and functional outcomes. However, despite these benefits, the therapy presents risks that require careful consideration. For autologous MSC therapy involving expanded cell populations, risks include tumorigenic potential, with evidence of sarcoma formation in certain cases of cultured MSCs. In contrast, autologous non-expanded MSC and HSC therapies may be limited by low cell yields, potentially compromising therapeutic efficacy. Additionally, non-expanded HSC therapy poses risks of insufficient cell numbers for successful engraftment and delayed immune reconstitution. These considerations underscore the importance of quality control and rigorous screening to optimize safety and efficacy. This article explores the mechanisms of action, clinical applications, and potential complications of dual-double stem cell therapy, underscoring the need for continued research and optimized protocols to enhance safety and outcomes in ovarian insufficiency and related conditions, offering new hope for affected women.
Collapse
Affiliation(s)
- Aleksandar Ljubić
- Pronatal Hospital, 11000 Belgrade, Serbia;
- Academy of Sciences and Arts of Bosnia and Herzegovina, 71000 Sarajevo, Bosnia and Herzegovina
- Medigroup Health System, Dubrovnik International University, 20000 Dubrovnik, Croatia
| | - Marija Dinić
- Department of Therapeutic Apheresis, University Clinical Center of Serbia, 11000 Belgrade, Serbia;
| | | | - Svetlana Vujović
- Clinic of Endocrinology, Diabetes and Diseases of National Center for Infertility and Endocrinology of Gender, 11000 Belgrade, Serbia;
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| |
Collapse
|
35
|
Ouzin M, Wesselborg S, Fritz G, Kogler G. Evaluation of Genotoxic Effects of N-Methyl-N-Nitroso-Urea and Etoposide on the Differentiation Potential of MSCs from Umbilical Cord Blood and Bone Marrow. Cells 2024; 13:2134. [PMID: 39768222 PMCID: PMC11675027 DOI: 10.3390/cells13242134] [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: 12/02/2024] [Revised: 12/19/2024] [Accepted: 12/20/2024] [Indexed: 01/11/2025] Open
Abstract
The present study investigates the influence of nitrosamines and etoposide on mesenchymal stromal cells (MSCs) in a differentiation state- and biological age-dependent manner. The genotoxic effects of the agents on both neonatal and adult stem cell populations after treatment, before, or during the course of differentiation, and the sensitivity of the different MSC types to different concentrations of MNU or etoposide were assessed. Hereby, the multipotent differentiation capacity of MSCs into osteoblasts, adipocytes, and chondrocytes was analyzed. Our findings reveal that while all cell types exhibit DNA damage upon exposure, neonatal CB-USSCs demonstrate enhanced resistance to genotoxic damage compared with their adult counterparts. Moreover, the osteogenic differentiation of MSCs was more susceptible to genotoxic damage, whereas the adipogenic and chondrogenic differentiation potentials did not show any significant changes upon treatment with genotoxin. Furthermore, we emphasize the cell-specific variability in responses to genotoxic damage and the differences in sensitivity and reaction across different cell types, thus advocating the consideration of these variabilities during drug testing and developmental biological research.
Collapse
Affiliation(s)
- Meryem Ouzin
- Institute for Transplantation Diagnostics and Cell Therapeutics, University Hospital, Heinrich Heine University Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany;
| | - Sebastian Wesselborg
- Institute for Molecular Medicine I, University Hospital, Heinrich Heine University Düsseldorf, Universitätsstraße 1, 40225 Düsseldorf, Germany;
| | - Gerhard Fritz
- Institute of Toxicology, University Hospital, Heinrich Heine University Düsseldorf, Universitätsstraße 1, 40225 Düsseldorf, Germany;
| | - Gesine Kogler
- Institute for Transplantation Diagnostics and Cell Therapeutics, University Hospital, Heinrich Heine University Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany;
| |
Collapse
|
36
|
Yuan X, Li W, Liu Q, Ou Y, Li J, Yan Q, Zhang P. Single-Cell RNA-Seq Reveals the Pseudo-temporal Dynamic Evolution Characteristics of ADSCs to Neuronal Differentiation. Cell Mol Neurobiol 2024; 45:5. [PMID: 39661257 PMCID: PMC11634962 DOI: 10.1007/s10571-024-01524-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 12/05/2024] [Indexed: 12/12/2024]
Abstract
Adipose-derived stromal cells (ADSCs) are commonly used in regenerative medicine, but the genetic features of their development into neuronal cells are unknown. This study used single-cell RNA sequencing (scRNA-seq) to reveal gene expression changes during ADSCs to neuronal differentiation. Sequencing of the ADSCs group, the prei-1d group, and the induction 1 h, 3 h, 5 h, 6 h, and 8 h groups was performed using the BD Rhapsody platform. Sequence data were analyzed using t-SNE, Monocle2, GO, and KEGG algorithms. Results showed that a total of 38,453 cells were collected, which were divided into 0-13 clusters. Monocle2 structured analysis revealed that ADSCs were located at the beginning of the trajectory, and the cells after 5 h of induction were mainly distributed at the end of the trajectory in branches 1 and 2. Up-regulated differentially expressed genes (DEGs) at 5 h after induction enriched GO items including cellular protein metabolism, cell adhesion, endocytosis, and cell migration. KEGG analysis showed that induced 6 h and 8 h groups mainly enriched pathways were oxidative phosphorylation, glutathione metabolism, and expression of Parkinson's disease-related genes. In conclusion, two distinct cell state mechanisms stimulate ADSCs to develop into mature neurons. ADSCs induced for 5 h had developed into mature neurons. Later, the differentiated cells undergo degenerative changes associated with senescence.
Collapse
Affiliation(s)
- Xiaodong Yuan
- Department of Neurology, Kailuan General Hospital Affiliated North China University of Science and Technology, 57 Xinhua East Road, Lubei District, Tangshan, 063000, Hebei, China.
- Department of Neurology, Hebei Provincial Key Laboratory of Neurobiological Function, Tangshan, 063000, Hebei, China.
| | - Wen Li
- Department of Neurology, Kailuan General Hospital Affiliated North China University of Science and Technology, 57 Xinhua East Road, Lubei District, Tangshan, 063000, Hebei, China
- Department of Neurology, Hebei Provincial Key Laboratory of Neurobiological Function, Tangshan, 063000, Hebei, China
| | - Qing Liu
- Department of Neurology, Kailuan General Hospital Affiliated North China University of Science and Technology, 57 Xinhua East Road, Lubei District, Tangshan, 063000, Hebei, China
- Department of Neurology, Hebei Provincial Key Laboratory of Neurobiological Function, Tangshan, 063000, Hebei, China
| | - Ya Ou
- Department of Neurology, Kailuan General Hospital Affiliated North China University of Science and Technology, 57 Xinhua East Road, Lubei District, Tangshan, 063000, Hebei, China
| | - Jing Li
- Department of Radiology, Tangshan Maternal and Child Health Hospital, Tangshan, 063000, Hebei, China
| | - Qi Yan
- Department of Neurology, Kailuan General Hospital Affiliated North China University of Science and Technology, 57 Xinhua East Road, Lubei District, Tangshan, 063000, Hebei, China
- Department of Neurology, Hebei Provincial Key Laboratory of Neurobiological Function, Tangshan, 063000, Hebei, China
| | - Pingshu Zhang
- Department of Neurology, Kailuan General Hospital Affiliated North China University of Science and Technology, 57 Xinhua East Road, Lubei District, Tangshan, 063000, Hebei, China.
- Department of Neurology, Hebei Provincial Key Laboratory of Neurobiological Function, Tangshan, 063000, Hebei, China.
| |
Collapse
|
37
|
Mackay G, Allen J. Platelet-rich plasma applications in the larynx: snake oil or stupendous? Curr Opin Otolaryngol Head Neck Surg 2024; 32:374-382. [PMID: 39436958 DOI: 10.1097/moo.0000000000001019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
PURPOSE OF REVIEW To explore and summarize recent work examining the use of platelet-rich plasma (PRP) applications for vocal fold disorder. RECENT FINDINGS Intrachordal PRP injections have recently emerged as a treatment option for patients with damage to the lamina propria of the vocal fold (scar, atrophy, sulcus and inflammatory lesions). Studies support significant improvements in patient-reported and clinician-reported outcomes, and objective phonatory parameters. PRP demonstrates a good safety profile, absence of immune reactions, affordable cost model and improvement in mucosal wave features with resultant improved voice quality up to 12 months following treatment. SUMMARY PRP injection of the vocal folds is a novel repurposing of a previously validated technique, and emerging data shows consistent results across multiple centres, increasing confidence in use of this freely available material. Current published studies confirm improved voice outcomes compared with pretreatment measures and high patient satisfaction. Further research is required to properly assign the role of and ideal candidate for PRP applications, and to delineate durability of treatment.
Collapse
Affiliation(s)
| | - Jacqui Allen
- Department of Surgery, University of Auckland
- Department of Otolaryngology, Te Whatu Ora Waitemata, Auckland, New Zealand
| |
Collapse
|
38
|
You X, Gao J, Yao Y. Advanced methods to mechanically isolate stromal vascular fraction: A concise review. Regen Ther 2024; 27:120-125. [PMID: 38571891 PMCID: PMC10987671 DOI: 10.1016/j.reth.2024.03.020] [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: 11/06/2023] [Revised: 02/29/2024] [Accepted: 03/17/2024] [Indexed: 04/05/2024] Open
Abstract
Adipose tissue is a highly attractive reservoir of stem cells due to its accessibility and abundance, and the SVF within it holds great promise for stem cell-based therapies. The use of mechanical methods for SVF isolation from adipose tissue is preferred over enzymatic methods, as it can be readily applied in clinical settings without additional processing steps. However, there is a lack of consensus on the optimal approach for mechanically isolating SVF. This comprehensive review aims to present and compare the latest mechanical isolation methods for SVF from adipose tissue, including centrifugation, filtration/washing, emulsification, vibration, and mincing/adiponizing. Each of these methods possesses unique advantages and limitations, and yet, no conclusive evidence has emerged demonstrating the superiority of one approach over the others, primarily due to the dearth of well-controlled prospective studies in this field.
Collapse
Affiliation(s)
- Xin You
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong 510515, PR China
| | - JianHua Gao
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong 510515, PR China
| | - Yao Yao
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong 510515, PR China
| |
Collapse
|
39
|
Yuan X, Li W, Yuan Y, Zhu X, Meng Y, Wu Q, Yan Q, Zhang P. Characterization of neuronal differentiation in human adipose-derived stromal cells: morphological, molecular, and ultrastructural insights. J Neurosci Methods 2024; 412:110296. [PMID: 39357604 DOI: 10.1016/j.jneumeth.2024.110296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 09/19/2024] [Accepted: 09/28/2024] [Indexed: 10/04/2024]
Abstract
OBJECTIVE Adipose-derived stromal cells (ADSCs) have shown promise as a potential source of neural differentiation. In this study, we investigated the morphological, molecular and ultrastructural features of ADSCs during neuronal differentiation. METHODS ADSCs were induced in vitro and their differentiation was examined at different time points. Immunocytochemical staining was performed to detect the expression of neuron-specific markers NSE and MAP-2. Immunofluorescence double labeling and Western blot detected the co-expression of presynaptic markers (CaMKII, SynCAM1, SYN) and postsynaptic markers (PSD-95, Synapsin I). Scanning electron microscopy (SEM) was performed to detect the synaptic structural features of differentiated neurons. RESULTS ADSCs showed diverse morphological features during differentiation, gradually acquiring a neuron-like spindle shape and organized arrangement. The expression of neuron-specific markers and synaptic markers peaked at 5 h of induction. Scanning electron microscopy showed polygonal protrusions of ADSC-derived neurons, and transmission electron microscopy showed characteristic ultrastructures such as nidus, synaptic vesicle-like structures, and tight junctions. CONCLUSION Our findings suggest that ADSCs differentiated for 5 h have neuronal features, including morphological, molecular, and ultrastructural resemblance to neurons, as well as the formation of synaptic structures. These insights contribute to a better understanding of ADSC-based neuronal differentiation and pave the way for future applications in regenerative medicine and neurodegenerative diseases.
Collapse
Affiliation(s)
- Xiaodong Yuan
- Department of Neurology, Kailuan General Hospital affiliated to North China University of Science and Technology, Tangshan, Hebei Province 063000, China; Hebei Provincial Key Laboratory of Neurobiological Function, Tangshan, Hebei Province 063000, China.
| | - Wen Li
- Department of Neurology, Kailuan General Hospital affiliated to North China University of Science and Technology, Tangshan, Hebei Province 063000, China; Hebei Provincial Key Laboratory of Neurobiological Function, Tangshan, Hebei Province 063000, China
| | - Yi Yuan
- Children's hospital of Capital institute of pediatrics, department of pediatric othopedic, Beijing 100000, China
| | - Xuhong Zhu
- Department of Neurology, Kailuan General Hospital affiliated to North China University of Science and Technology, Tangshan, Hebei Province 063000, China; Hebei Provincial Key Laboratory of Neurobiological Function, Tangshan, Hebei Province 063000, China
| | - Yan Meng
- Department of Neurology, Kailuan General Hospital affiliated to North China University of Science and Technology, Tangshan, Hebei Province 063000, China; Hebei Provincial Key Laboratory of Neurobiological Function, Tangshan, Hebei Province 063000, China
| | - Qi Wu
- Department of Neurology, Kailuan General Hospital affiliated to North China University of Science and Technology, Tangshan, Hebei Province 063000, China; Hebei Provincial Key Laboratory of Neurobiological Function, Tangshan, Hebei Province 063000, China
| | - Qi Yan
- Department of Neurology, Kailuan General Hospital affiliated to North China University of Science and Technology, Tangshan, Hebei Province 063000, China; Hebei Provincial Key Laboratory of Neurobiological Function, Tangshan, Hebei Province 063000, China
| | - Pingshu Zhang
- Department of Neurology, Kailuan General Hospital affiliated to North China University of Science and Technology, Tangshan, Hebei Province 063000, China; Hebei Provincial Key Laboratory of Neurobiological Function, Tangshan, Hebei Province 063000, China.
| |
Collapse
|
40
|
Gao P, Kajiya M, Motoike S, Ikeya M, Yang J. Application of mesenchymal stem/stromal cells in periodontal regeneration: Opportunities and challenges. JAPANESE DENTAL SCIENCE REVIEW 2024; 60:95-108. [PMID: 38314143 PMCID: PMC10837070 DOI: 10.1016/j.jdsr.2024.01.001] [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: 08/08/2023] [Revised: 12/06/2023] [Accepted: 01/15/2024] [Indexed: 02/06/2024] Open
Abstract
Guided tissue regeneration (GTR) has been widely used in the periodontal treatment of intrabony and furcation defects for nearly four decades. The treatment outcomes have shown effectiveness in reducing pocket depth, improving attachment gain and bone filling in periodontal tissue. Although applying GTR could reconstruct the periodontal tissue, the surgical indications are relatively narrow, and some complications and race ethic problems bring new challenges. Therefore, it is challenging to achieve a consensus concerning the clinical benefits of GTR. With the appearance of stem cell-based regenerative medicine, mesenchymal stem/stromal cells (MSCs) have been considered a promising cell resource for periodontal regeneration. In this review, we highlight preclinical and clinical periodontal regeneration using MSCs derived from distinct origins, including non-odontogenic and odontogenic tissues and induced pluripotent stem cells, and discuss the transplantation procedures, therapeutic mechanisms, and concerns to evaluate the effectiveness of MSCs.
Collapse
Affiliation(s)
- Pan Gao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of General Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Mikihito Kajiya
- Department of Periodontal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8553, Japan
| | - Souta Motoike
- Department of Periodontal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8553, Japan
| | - Makoto Ikeya
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan
| | - Jingmei Yang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| |
Collapse
|
41
|
Manabe N, Hoshino Y, Himaki T, Sakaguchi K, Matsumoto S, Yamamoto T, Murase T. Lysate of bovine adipose-derived stem cells accelerates in-vitro development and increases cryotolerance through reduced content of lipid in the in vitro fertilized embryos. Biochem Biophys Res Commun 2024; 735:150834. [PMID: 39427378 DOI: 10.1016/j.bbrc.2024.150834] [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: 07/23/2024] [Revised: 09/19/2024] [Accepted: 10/12/2024] [Indexed: 10/22/2024]
Abstract
Mesenchymal stem cells such as adipose-derived stem cells (ADSCs) are known to secrete factors that stimulate cell division and promote regeneration in neighboring cells. While conditioned medium from stem cells has been used in blastocyst production, no studies have examined the use of cell lysates. In this study we investigated the effects of adding ADSC lysate to in vitro culture (IVC) medium. ADSCs and fibroblasts were isolated from bovine adipose tissue and auricular tissue, respectively, and their lysates were prepared by freeze-thaw disruption. ADSC lysate was added to synthetic oviductal fluid medium. The effects on cleavage, blastocyst development rates, cell numbers, cryotolerance, gene expression (POU5F1, BAX, IGF1R, IGF2R, SOD2), lipid content, and membrane integrity were evaluated according to the experimental design. In Expt. 1, the comparison involved adding ADSC or fibroblast lysate alongside the control group. The total blastocyst rate increased when ADSC lysate was introduced (ADSCs: 40.1 %, fibroblasts: 33.1 %, control: 27.3 %). However, there were no significant differences in the number of trophoblast cells or in the inner cell mass. Experiment 2 confirmed that this increase in blastocyst development was not due to the solvent, PBS(-). In Expt. 3, addition of 10 % fetal calf serum (FCS) or ADSC lysate increased the total blastocyst rate compared to the control (control, 26.2 %; 10 % FCS, 43.4 %; 1 % ADSC lysate, 34.2 %; 10 % ADSC lysate, 48.1 %). After freezing and thawing, the survival and hatching rates of embryos with FCS were significantly lower than those of the control as well as those with added ADSC lysate. In Expt. 4, the addition of ADSC lysate or FCS had no significant effect on gene expression in blastocysts compared to control. However, the addition of FCS significantly increased the gray intensity, indicating higher lipid content compared to the control, with a significant increase in the number of dead cells in the blastocyst. These results indicate that the addition of ADSC lysate to the IVC medium accelerates bovine blastocyst development and that its 10 % addition, corresponding to 1 × 105 cells/mL, is as effective as 10 % FCS without a decrease in cryotolerance due to the increased lipid content.
Collapse
Affiliation(s)
- Noriyoshi Manabe
- Laboratory of Veterinary Theriogenology, Joint Graduate School of Veterinary Sciences, Gifu University, Gifu, 501-1193, Japan; Department of Dairy Research Center, Gifu Prefectural Livestock Research Institute, Ena, Gifu, 509-7601, Japan
| | - Yoichiro Hoshino
- Kyoto University Livestock Farm, Graduate School of Agriculture, Kyoto University, Funai, Kyoto, 622-0203, Japan
| | - Takehiro Himaki
- Laboratory of Animal Developmental Engineering, Faculty of Applied Biological Sciences, Gifu University, Gifu, 501-1193, Japan
| | - Kenichiro Sakaguchi
- Laboratory of Veterinary Theriogenology, Joint Graduate School of Veterinary Sciences, Gifu University, Gifu, 501-1193, Japan; Laboratory of Veterinary Theriogenology, Joint Department of Veterinary Medicine, Faculty of Applied Biological Sciences, Gifu University, Gifu, 501-1193, Japan; Center for One Medicine Innovative Translational Research (COMIT), Gifu University, Gifu, 501-1193, Japan
| | - Seiji Matsumoto
- Headquarters for Research Promotion, Asahikawa Medical University, Asahikawa, Hokkaido, 078-8510, Japan; Clinical Research Support Center, Asahikawa Medical University Hospital, Asahikawa Medical University, Asahikawa, Hokkaido, 078-8510, Japan
| | - Tokunori Yamamoto
- Clinical Research Support Center, Asahikawa Medical University Hospital, Asahikawa Medical University, Asahikawa, Hokkaido, 078-8510, Japan; Department of Urology, Graduate School of Medicine, Nagoya University, Nagoya, Aichi, 466-8550, Japan
| | - Tetsuma Murase
- Laboratory of Veterinary Theriogenology, Joint Graduate School of Veterinary Sciences, Gifu University, Gifu, 501-1193, Japan; Laboratory of Veterinary Theriogenology, Joint Department of Veterinary Medicine, Faculty of Applied Biological Sciences, Gifu University, Gifu, 501-1193, Japan.
| |
Collapse
|
42
|
Kurimori KT, Bastos EO, Camara PRP, Mascarenhas Dias B, Uyeda GL, Alonso N. Alveolar Bone Graft Supplemented With Stromal Vascular Fraction in Patients With Permanent Dentition: A Randomized Study. Cleft Palate Craniofac J 2024:10556656241296711. [PMID: 39584372 DOI: 10.1177/10556656241296711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2024] Open
Abstract
OBJECTIVE To evaluate bone development in patients with alveolar clefts and permanent dentition treated with alveolar bone graft supplemented with stromal vascular fraction (SVF). DESIGN Clinical, prospective, randomized. LOCATION Single tertiary care institution. PATIENTS Eighteen participants with unilateral alveolar cleft and permanent dentition were included. Patients with other comorbidities were excluded. INTERVENTION The control group underwent alveolar bone grafting (ABG) using iliac crest bone, while the experimental group underwent the same treatment, supplemented with SVF. Adipose tissue was collected by abdominal liposuction, and the SVF was processed using mechanical methods (decantation, microfragmentation, and filtration). MAIN OUTCOME MEASUREMENT Bone formation and bone graft integration rate in alveolar cleft at 6 months postsurgery using 3-dimensional tomographic methods and density measurements. RESULTS The amount of bone graft correlated with an improvement in the relationship between the cleft and noncleft sides (R = 0.78, P < .001) and an improvement in alveolar cleft density (R = 0.69, P = .005), but did not correlate with the graft integration rate. The experimental group showed larger cleft sizes (0.83 × 1.74 cm3, P = .021) and older patients (17.35 × 27.6 years, P = .002), and did not differ in terms of bone development variables when compared to the control group. CONCLUSION ABG supplemented with SVF showed statistically similar bone development results, but with a better trend than conventional ABG. Additionally, the studied groups had asymmetric pre-existing characteristics, with greater severity in the experimental group. A larger study will be necessary to mitigate preoperative characteristic differences and to more accurately compare the results between the methods.
Collapse
Affiliation(s)
| | | | | | | | | | - Nivaldo Alonso
- Burn and Plastic Surgery Department, São Paulo University, Sao Paulo, Brazil
| |
Collapse
|
43
|
Del Vecchio D, Vranis N, Alevrogianni K, Theodorou S. "SAVE"-Shock-Assisted Viable Extraction: A Minimally Manipulative Method of Processing Regenerative Cells for Clinical Use. Aesthet Surg J Open Forum 2024; 7:ojae112. [PMID: 39790718 PMCID: PMC11712265 DOI: 10.1093/asjof/ojae112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2025] Open
Abstract
Background Extraction of adipose-derived stem cells (ASCs) from the stromal vascular fraction (SVF) has gained significant attention lately in the realm of regenerative medicine. However, finding highly efficient methods of extraction that also comply with the US regulations has prevented widespread clinical use. Objectives The objective of this study was to evaluate a novel ASC extraction device to quantify viable ASC extraction and processing efficiency. Methods SVF extracted from abdominal fat samples and processed using a novel shock-assisted viable extraction (SAVE) device was tested for stem-cell count and viability. Additionally, time required for processing was recorded. Results Twelve adipose samples were utilized for this study. After a mean time of 3 min, cell count yield ranged of 47,400 to 189,400 of viable regenerative cells per cc, with an average of 122,464 viable regenerative cells per cc. Conclusions SAVE is a novel fat-processing technique with high stem-cell extraction that shows promise from a regulatory, yield, time efficiency, and cost perspective. Level of Evidence 5 Therapeutic
Collapse
Affiliation(s)
| | - Neil Vranis
- Corresponding Author: Dr Neil Vranis, 433 N Camden Dr #780, Beverly Hills, CA 90210, USA. E-mail:
| | | | | |
Collapse
|
44
|
Zhang C, Cai L, Ma M, Xie X, Wang J, Zhang Y. Hypoxia-Treated Adipose Mesenchymal Stem Cells Derived Exosomes Enhance the Therapeutic Effects on Unilateral Ureteral Obstruction Mice. Pharmacology 2024:1-13. [PMID: 39561719 DOI: 10.1159/000542609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 11/11/2024] [Indexed: 11/21/2024]
Abstract
INTRODUCTION The exosomes from adipose-derived mesenchymal stem cells (AMSCs) had therapeutic effects. However, whether the exosomes derived from hypoxia-treated AMSCs could improve renal functions in unilateral ureteral obstruction (UUO) mice remains unclear. METHODS The exosomes were characterized using a transmission electron microscope and Western blot. Its size distribution was determined using the Zetasizer Nano ZS analysis system. The differentiation ability was assessed by alkaline phosphatase and oil red staining. Consequently, the function of exosomes in inhibiting inflammatory factors was evaluated using an enzyme-linked immunosorbent assay, and apoptosis inhibition was evaluated by Western blot. Finally, the function of exosomes to ameliorate kidney fibrosis was evaluated using quantitative reverse transcription polymerase chain reaction, Western blot, hematoxylin-eosin staining, and Masson staining. RESULTS The cultured AMSCs could differentiate into osteoblast and adipocyte. Meanwhile, the cultured AMSCs could effectively secrete the exosomes, which were characterized by around 110 nm diameter and surface marker expression. Exosomes derived from hypoxia-treated AMSCs improved renal functions in UUO mice. The mechanism exploration revealed that exosomes could decrease the TNF-α and IL-6 and inhibit cell apoptosis. Finally, the fibrosis-associated protein was reversed, and the renal dysfunctions were ameliorated in UUO mice. CONCLUSION The exosomes derived from the hypoxia-treated AMSCs have a better effect than those from normal AMSCs in ameliorating renal dysfunctions in UUO mice.
Collapse
Affiliation(s)
- Chi Zhang
- Department of Nephrology, The Affiliated Suqian Hospital of Xuzhou Medical University, Suqian, China
- Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, China
| | - Longjun Cai
- Department of Urology, The Affiliated Suqian Hospital of Xuzhou Medical University, Suqian, China
| | - Meimei Ma
- Department of Pathology, The Affiliated Suqian Hospital of Xuzhou Medical University, Suqian, China
| | - Xiaohui Xie
- Department of Nephrology, The Affiliated Suqian Hospital of Xuzhou Medical University, Suqian, China
| | - Junsheng Wang
- Department of Nephrology, The Affiliated Suqian Hospital of Xuzhou Medical University, Suqian, China
| | - Yuanyuan Zhang
- Department of Nephrology, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou, China
| |
Collapse
|
45
|
Scatena A, Apicella M, Mantuano M, Ragghianti B, Silverii A, Miranda C, Monge L, Uccioli L, Scevola G, Stabile E, Gargiulo M, Vermigli C, Monami M. Autologous cell therapy for ischemic diabetic foot: a meta-analysis of randomized controlled trials for the development of the Italian guidelines for the treatment of diabetic foot syndrome. Acta Diabetol 2024:10.1007/s00592-024-02393-z. [PMID: 39545964 DOI: 10.1007/s00592-024-02393-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Accepted: 10/13/2024] [Indexed: 11/17/2024]
Abstract
AIM To assess the efficacy and safety of autologous cell therapy (ACT) in patients with ischemic diabetic foot ulcers (DFU). The present meta-analysis was designed to support the development of the Italian Guidelines for the Treatment of Diabetic Foot Syndrome (DFS). METHODS A Medline and Embase search were performed up to Feb 1st, 2024 collecting all RCTs including diabetic patients or reporting subgroup analyses on diabetic patients with ischemic foot ulcers comparing ACT with placebo/no therapy/standard of care (SoC), with a duration of at least 26 weeks. Prespecified endpoints were: major amputation (principal) and minor amputation, ulcer healing, time-to-healing, transcutaneous oxygen pressure (TcPO2), ankle-brachial index (ABI), pain, and all-cause mortality (secondary). Any ACT was allowed, irrespective of cell product type and route of administration (intra-arterial and intramuscular). RESULTS Seven studies fulfilled all inclusion criteria, all using intramuscular transplantation as route of administration, but only 2 had a follow-up greater than 26 weeks. Participants treated with ACT had a significantly lower risk of major amputations in comparison with SoC/placebo (MH-OR 0.47 [0.24, 0.92], p = 0.03). ACT was also associated with a significantly higher rate of ulcer healing (MH-OR: 10.1 [3.5, 29.6], p < 0.001), greater increase of TcPO2 and ABI values (WMD: 17.57 [13.02, 22.12], p < 0.001), and reduction of pain (WMD: -1.83 [-2.32, -1.34], p = 0.003). CONCLUSIONS ACT must be considered as a potential therapy for patients with ischemic diabetic foot ulcers. Further studies are needed to better clarify their role in the treatment and management of DFS.
Collapse
Affiliation(s)
- Alessia Scatena
- Diabetology Unit, Health Authorities South East Tuscany, San Donato Hospital, Via Pietro Nenni 20, 52100, Arezzo, Italy.
| | - Matteo Apicella
- Diabetology Unit, Health Authorities South East Tuscany, San Donato Hospital, Via Pietro Nenni 20, 52100, Arezzo, Italy
| | - Michele Mantuano
- Diabetology Unit, Health Authorities South East Tuscany, San Donato Hospital, Via Pietro Nenni 20, 52100, Arezzo, Italy
| | - Benedetta Ragghianti
- Azienda Ospedaliero Universitaria Careggi and University of Florence, Florence, Italy
| | - Antonio Silverii
- Azienda Ospedaliero Universitaria Careggi and University of Florence, Florence, Italy
| | | | - Luca Monge
- AMD - Italian Association of Clinical Diabetologists, Rome, Italy
| | - Luigi Uccioli
- Diabetes Section CTO Hospital and dept of Biomedicine and prevention Tor Vergata University of Rome, Rome, Italy
| | | | | | - Mauro Gargiulo
- Vascular Surgery, University of Bologna- DIMEC, Bologna, Italy
| | | | - Matteo Monami
- Azienda Ospedaliero Universitaria Careggi and University of Florence, Florence, Italy
| |
Collapse
|
46
|
Trotzier C, Bellanger C, Abdessadeq H, Delannoy P, Mojallal A, Auxenfans C. Deciphering influence of donor age on adipose-derived stem cells: in vitro paracrine function and angiogenic potential. Sci Rep 2024; 14:27589. [PMID: 39528480 PMCID: PMC11555058 DOI: 10.1038/s41598-024-73875-x] [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: 01/17/2024] [Accepted: 09/23/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND As fat grafting is commonly used as a filler, Adipose-derived stem/stromal cells (ASC) have been reported to be key player in retention rate. Paracrine and differentiation potential of those cells confer them strong pro-angiogenic capacities. However, a full characterization of the influence of aging on ASC has not been reported yet. Here we've investigated the effect of age on paracrine function, stemness and angiogenic potential of ASC. METHODS ASC were extracted from young and old adult donors. We assessed stromal vascular fraction cell populations repartition, ASC stemness potential, capability to differentiate into mesenchymal lineages as well as their secretome. Angiogenic potential was assessed using a sprouting assay, an indirect co-culture of ASC and dermal microvascular endothelial cells (EC). Total vascular sprout length was measured, and co-culture soluble factors were quantified. Pro-angiogenic factors alone or in combination as well as ASC-conditioned medium (CM) were added to EC to assess sprouting induction. RESULTS Decrease of endothelial cells yield and percentage is observed in cells extracted from adipose tissue of older patients, whereas ASC percentage increased with age. Clonogenic potential of ASC is stable with age. ASC can differentiate into adipocytes, chondrocytes and osteoblasts, and aging does not alter this potential. Among the 25 analytes quantified, high levels of pro-angiogenic factors were found, but none is significantly modulated with age. ASC induce a significantly longer vascular sprouts compared to fibroblasts, and no difference was found between young and old ASC donors on that parameter. Higher concentrations of FGF-2, G-CSF, HGF and IL-8, and lower concentrations of VEGF-C were quantified in EC/ASC co-cultures compared to EC/fibroblasts co-cultures. EC/ASC from young donors secrete higher levels of VEGF-A compared to old ones. Neither soluble factor nor CM without cells are able to induce organized sprouts, highlighting the requirement of cell communication for sprouting. CM produced by ASC supporting development of long vascular sprouts promote sprouting in co-cultures that establish shorter sprouts. CONCLUSION Our results show cells from young and old donors exhibit no difference in all assessed parameters, suggesting all patients could be included in clinical applications. We emphasized the leading role of ASC in angiogenesis, without impairment with age, where secretome is a key but not sufficient actor.
Collapse
Affiliation(s)
- Chloe Trotzier
- Advanced Research, L'Oréal Research and Innovation, 1, Av. Eugene Schueller, 93600, Aulnay sous Bois, France.
| | - Clement Bellanger
- Advanced Research, L'Oréal Research and Innovation, 1, Av. Eugene Schueller, 93600, Aulnay sous Bois, France
| | - Hakima Abdessadeq
- Advanced Research, L'Oréal Research and Innovation, 1, Av. Eugene Schueller, 93600, Aulnay sous Bois, France
| | - Philippe Delannoy
- Advanced Research, L'Oréal Research and Innovation, 1, Av. Eugene Schueller, 93600, Aulnay sous Bois, France
| | - Ali Mojallal
- Department of Plastic, Reconstructive and Aesthetic Surgery, La Croix Rousse Hospital, Bernard Lyon 1 University, Lyon, France
| | - Celine Auxenfans
- Banque de Tissus et de Cellules des Hospices Civils de Lyon, Edouard Herriot Hospital, Lyon, France
| |
Collapse
|
47
|
Muntión S, Sánchez-Luis E, Díez-Campelo M, Blanco JF, Sánchez-Guijo F, De Las Rivas J. Novel Gene Biomarkers Specific to Human Mesenchymal Stem Cells Isolated from Bone Marrow. Int J Mol Sci 2024; 25:11906. [PMID: 39595975 PMCID: PMC11593895 DOI: 10.3390/ijms252211906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 10/23/2024] [Accepted: 10/25/2024] [Indexed: 11/28/2024] Open
Abstract
In this paper, we present a comparative analysis of the transcriptomic profile of three different human cell types: hematopoietic stem cells (HSCs), bone marrow-derived mesenchymal stem cells (MSCs) and fibroblasts (FIBs). The work aims to identify unique genes that are differentially expressed as specific markers of bone marrow-derived MSCs, and to achieve this undertakes a detailed analysis of three independent datasets that include quantification of the global gene expression profiles of three primary cell types: HSCs, MSCs and FIBs. A robust bioinformatics method, called GlobalTest, is used to assess the specific association between one or more genes expressed in a sample and the outcome variable, that is, the 'cell type' provided as a single univariate response. This outcome variable is predicted for each sample tested, based on the expression profile of the specific genes that are used as input to the test. The precision of the tests is calculated along with the statistical sensitivity and specificity for each gene in each dataset, yielding four genes that mark MSCs with high accuracy. Among these, the best performer is the protein-coding gene Transgelin (TAGLN, Gene ID: 6876) (with a Positive Predictive Value > 0.96 and FDR < 0.001), which identifies MSCs better than any of the currently used standard markers: ENG (CD105), THY1 (CD90) or NT5E (CD73). The results are validated by RT-qPCR, providing novel gene biomarkers specific for human MSCs.
Collapse
Affiliation(s)
- Sandra Muntión
- Cell Therapy Area, Department of Hematology, Institute of Biomedical Research of Salamanca-Hospital Universitario de Salamanca (IBSAL-HUS), 37007 Salamanca, Spain; (S.M.); (F.S.-G.)
- Centro en Red de Medicina Regenerativa y Terapia Celular de Castilla y León, Institute of Biomedical Research of Salamanca-Hospital Universitario de Salamanca (IBSAL-HUS), 37007 Salamanca, Spain
| | - Elena Sánchez-Luis
- Bioinformatics and Functional Genomics Group, Cancer Research Center (CiC-IBMCC), Consejo Superior de Investigaciones Científicas (CSIC) and University of Salamanca (USAL), 37007 Salamanca, Spain;
- Bioinformatics Functional Genomics CANC-14 Group, Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
| | - María Díez-Campelo
- Department of Hematology, Center for Biomedical Research in Network of Cancer (CIBERONC), Institute of Biomedical Research of Salamanca-Hospital Universitario de Salamanca (IBSAL-HUS), 37007 Salamanca, Spain;
- Department of Medicine, Faculty of Medicine, University of Salamanca (USAL), 37007 Salamanca, Spain;
| | - Juan F. Blanco
- Department of Medicine, Faculty of Medicine, University of Salamanca (USAL), 37007 Salamanca, Spain;
- Department of Trauma and Orthopedic Surgery, University Hospital of Salamanca (IBSAL-HUS), 37007 Salamanca, Spain
| | - Fermín Sánchez-Guijo
- Cell Therapy Area, Department of Hematology, Institute of Biomedical Research of Salamanca-Hospital Universitario de Salamanca (IBSAL-HUS), 37007 Salamanca, Spain; (S.M.); (F.S.-G.)
- Centro en Red de Medicina Regenerativa y Terapia Celular de Castilla y León, Institute of Biomedical Research of Salamanca-Hospital Universitario de Salamanca (IBSAL-HUS), 37007 Salamanca, Spain
- Department of Hematology, Center for Biomedical Research in Network of Cancer (CIBERONC), Institute of Biomedical Research of Salamanca-Hospital Universitario de Salamanca (IBSAL-HUS), 37007 Salamanca, Spain;
- Department of Medicine, Faculty of Medicine, University of Salamanca (USAL), 37007 Salamanca, Spain;
| | - Javier De Las Rivas
- Bioinformatics and Functional Genomics Group, Cancer Research Center (CiC-IBMCC), Consejo Superior de Investigaciones Científicas (CSIC) and University of Salamanca (USAL), 37007 Salamanca, Spain;
- Bioinformatics Functional Genomics CANC-14 Group, Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
| |
Collapse
|
48
|
Bang S, Qamar AY, Yun SH, Gu NY, Kim H, Han A, Kang H, Park HS, Kim SI, Saadeldin IM, Lee S, Cho J. Embryotrophic effect of exogenous protein contained adipose-derived stem cell extracellular vesicles. J Anim Sci Biotechnol 2024; 15:145. [PMID: 39488683 PMCID: PMC11531693 DOI: 10.1186/s40104-024-01106-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 09/12/2024] [Indexed: 11/04/2024] Open
Abstract
BACKGROUND Extracellular vesicles (EVs) regulate cell metabolism and various biological processes by delivering specific proteins and nucleic acids to surrounding cells. We aimed to investigate the effects of the cargo contained in EVs derived from adipose-derived stem cells (ASCs) on the porcine embryonic development. METHODS ASCs were isolated from porcine adipose tissue and characterized using ASC-specific markers via flow cytometry. EVs were subsequently extracted from the conditioned media of the established ASCs. These EVs were added to the in vitro culture environment of porcine embryos to observe qualitative improvements in embryonic development. Furthermore, the proteins within the EVs were analyzed to investigate the underlying mechanisms. RESULTS We observed a higher blastocyst development rate and increased mitochondrial activity in early stage embryos in the ASC-EVs-supplemented group than in the controls (24.8% ± 0.8% vs. 28.6% ± 1.1%, respectively). The terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay of blastocysts also revealed significantly reduced apoptotic cells in the ASC-EVs-supplemented group. Furthermore, through proteomics, we detected the proteins in ASC-EVs and blastocysts from each treatment group. This analysis revealed a higher fraction of proteins in the ASC-EVs-supplemented group than in the controls (1,547 vs. 1,495, respectively). Gene analysis confirmed that ASC-EVs showed a high expression of tyrosine-protein kinase (SRC), whereas ASC-EVs supplemented blastocysts showed a higher expression of Cyclin-dependent kinase 1 (CDK1). SRC is postulated to activate protein kinase B (AKT), which inhibits the forkhead box O signaling pathway and activates CDK1. Subsequently, CDK1 activation influences the cell cycle, thereby affecting in vitro embryonic development. CONCLUSION ASC-EVs promote mitochondrial activity, which is crucial for the early development of blastocysts and vital in the downregulation of apoptosis. Additionally, ASC-EVs supply SRC to porcine blastocysts, thereby elongating the cell cycle.
Collapse
Affiliation(s)
- Seonggyu Bang
- College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Republic of Korea
- College of Veterinary Medicine, Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Ahmad Yar Qamar
- College of Veterinary and Animal Sciences, Jhang Sub-campus of University of Veterinary and Animal Sciences, Lahore, 54000, Pakistan
| | - Sung Ho Yun
- Korea Basic Science Institute (KBSI), Ochang, Chungcheongbuk-Do, 28119, Republic of Korea
| | - Na-Yeon Gu
- Viral Disease Research Division, Animal and Plant Quarantine Agency, Gimcheon, Gyeongsangbuk-Do, 39660, Republic of Korea
| | - Heyyoung Kim
- College of Veterinary Medicine, Chungnam National University, Daejeon, 34134, Republic of Korea
- Department of Plastic and Reconstructive Surgery, Vascularized Composite Allotransplantation (VCA) Laboratory, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Ayeong Han
- College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Republic of Korea
- College of Veterinary Medicine, Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Heejae Kang
- College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Republic of Korea
- College of Veterinary Medicine, Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Hye Sun Park
- Korea Basic Science Institute (KBSI), Ochang, Chungcheongbuk-Do, 28119, Republic of Korea
| | - Seung Ii Kim
- Korea Basic Science Institute (KBSI), Ochang, Chungcheongbuk-Do, 28119, Republic of Korea
| | - Islam M Saadeldin
- College of Veterinary Medicine, Chungnam National University, Daejeon, 34134, Republic of Korea
- Comparative Medicine Department, King Faisal Specialist Hospital & Research Centre, Riyadh, 11211, Saudi Arabia
| | - Sanghoon Lee
- College of Veterinary Medicine, Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Jongki Cho
- College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
49
|
Lee GB, Park SM, Jung UJ, Kim SR. The Potential of Mesenchymal Stem Cells in Treating Spinocerebellar Ataxia: Advances and Future Directions. Biomedicines 2024; 12:2507. [PMID: 39595073 PMCID: PMC11591855 DOI: 10.3390/biomedicines12112507] [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: 10/01/2024] [Revised: 10/28/2024] [Accepted: 10/31/2024] [Indexed: 11/28/2024] Open
Abstract
Spinocerebellar ataxia (SCA) is a heterogeneous disorder characterized by impaired balance and coordination caused by cerebellar dysfunction. The absence of treatments approved by the U.S. Food and Drug Administration for SCA has driven the investigation of alternative therapeutic strategies, including stem cell therapy. Mesenchymal stem cells (MSCs), known for their multipotent capabilities, have demonstrated significant potential in treating SCA. This review examines how MSCs may promote neuronal growth, enhance synaptic connectivity, and modulate brain inflammation. Recent findings from preclinical and clinical studies are also reviewed, emphasizing the promise of MSC therapy in addressing the unmet needs of SCA patients. Furthermore, ongoing clinical trials and future directions are proposed to address the limitations of the current approaches.
Collapse
Affiliation(s)
- Gi Beom Lee
- School of Life Science and Biotechnology, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Republic of Korea; (G.B.L.); (S.M.P.)
| | - Se Min Park
- School of Life Science and Biotechnology, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Republic of Korea; (G.B.L.); (S.M.P.)
| | - Un Ju Jung
- Department of Food Science and Nutrition, Pukyong National University, Busan 48513, Republic of Korea;
| | - Sang Ryong Kim
- School of Life Science and Biotechnology, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Republic of Korea; (G.B.L.); (S.M.P.)
- Brain Science and Engineering Institute, Kyungpook National University, Daegu 41404, Republic of Korea
| |
Collapse
|
50
|
Gürünlüoğlu K, Satilmiş B, Gül M, Dündar M, Göktürk N, Akbulut S, Koç A, Gürünlüoğlu S, Aslan M, Karaaslan E, Türköz MA, Toplu ÇG, Ateş H, Üremiş MM, Menevşe İN, Kuştepe EK, Sari Ünal S, Altundaş E, Yildiz T, Şahin TT, Yilmaz S, Demircan M. The impact of subdermal adipose derived stem cell injections and early excision on systemic oxidative stress and wound healing in rats with severe scald burns. Burns 2024; 50:2056-2069. [PMID: 39127577 DOI: 10.1016/j.burns.2024.07.037] [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: 02/19/2024] [Revised: 07/08/2024] [Accepted: 07/28/2024] [Indexed: 08/12/2024]
Abstract
AIM This study aims to develop an experimental treatment model effective against oxidative stress in the acute period of severe burns and to analyze the mechanisms of healing large wound defects. METHODS Five rats, including 2 females and 3 males, were used as donors to obtain adipose-derived stem cells (ADSC) from the inguinal fat pad. The stem cells were labeled with green fluorescent protein. The study included four groups of 17 rats, each with grade 3 scalding burns on 30 % of their body surface, and a control group of 10 rats with an equal number of males and females. After early excision, 106 ADSC-derived stem cells were administered subdermally to the burned wound and autografted to the stem cell group (n = 17). The early excision group (n = 17) received early excision and autograft, with 2 ml of normal saline injected subdermally into the burn wound edge. The PLM group (n = 17) was treated with a polylactic membrane (PLM) dressing after the burn. No treatment was given to the burn group (n = 17). Ten rats from all groups were sacrificed on the 4th day post-burn for oxidative stress evaluation. The control group (n = 10) was sacrificed on day 4. Blood and tissue samples were collected post-sacrifice. Oxidative stress and inflammation in the blood, as well as cell damage in the skin, liver, kidneys, and lungs, were investigated histopathologically and biochemically on the 4th day post-burn. On the 70th day after burn, wound healing was examined macroscopically and histopathologically. RESULTS On the 4th day, oxidative stress results showed that the levels of Total Oxidative Capacity (TOC) in the blood were lowest in the stem cell (7.4 [6-8.8]), control (6.7 [5.9-7.6]), and early excision (7.5 [6.6-8.5]) groups, with no significant difference between them. The burn group (14.7 [12.5-16.9]) had the highest TOC levels. The PLM group (9.7 [8.6-10.7]) had lower TOC levels than the burn group but higher levels than the other groups. Histopathological examination on the 4th day revealed low liver caspase-3 immunoreactivity in the stem cell and early excision groups among the burn groups. Caspase-3 immunoreactivity levels were as follows: stem cell group (20 [10-30]), early excision group (25 [15-50]), PLM group (70 [50-100]), control group (0), and burn group (80 [60-120]). Other oxidative stress and end-organ damage outcomes were consistent with these results. All rats in the stem cell group had burn wounds that healed completely by the 70th day. Examination of the skin and its appendages from the stem cell group with an immunofluorescence microscope demonstrated green coloration, indicating incorporation of stem cells. CONCLUSION Stem cells may have the potential to form new skin and its appendages, providing better healing for large skin defects. Early excision treatment, by removing local necrotic tissues after extensive and deep burns, can prevent end-organ damage due to systemic oxidative stress and inflammation. We also believe that when these two treatments are used together, they can achieve the best results.
Collapse
Affiliation(s)
- Kubilay Gürünlüoğlu
- Department of Pediatric Surgery, İnönü University, Faculty of Medicine, Malatya, Türkiye.
| | - Basri Satilmiş
- Department of Liver Transplantation Institute, İnönü University, Faculty of Medicine, Malatya, Türkiye
| | - Mehmet Gül
- Department of Histology and Embryology, İnönü University, Faculty of Medicine, Malatya, Türkiye
| | - Muhammed Dündar
- Department of Medical Genetics, İnönü University, Faculty of Medicine, Malatya, Türkiye
| | - Nurcan Göktürk
- Department of Medical Biochemistry, İnönü University, Faculty of Medicine, Malatya, Türkiye
| | - Sami Akbulut
- Department of Liver Transplantation Institute, İnönü University, Faculty of Medicine, Malatya, Türkiye
| | - Ahmet Koç
- Department of Medical Genetics, İnönü University, Faculty of Medicine, Malatya, Türkiye
| | - Semra Gürünlüoğlu
- Department of Pathology, İnönü University, Faculty of Medicine, Malatya, Türkiye
| | - Mehmet Aslan
- Department of Pediatrics, Division of Pediatric Emergency Medicine, İnönü University, Faculty of Medicine, Malatya, Türkiye
| | - Ezgi Karaaslan
- Department of Histology and Embryology, İnönü University, Faculty of Medicine, Malatya, Türkiye
| | - Mehmet Akif Türköz
- Department of Radiology, İnönü University, Faculty of Medicine, Malatya, Türkiye
| | - Çağla Güner Toplu
- Department of Pediatric Surgery, İnönü University, Faculty of Medicine, Malatya, Türkiye
| | - Hasan Ateş
- Department of Pediatric Surgery, İnönü University, Faculty of Medicine, Malatya, Türkiye
| | - Muhammed Mehdi Üremiş
- Department of Medical Biochemistry, İnönü University, Faculty of Medicine, Malatya, Türkiye
| | - İrem Nur Menevşe
- Department of Medical Genetics, İnönü University, Faculty of Medicine, Malatya, Türkiye
| | - Elif Kayhan Kuştepe
- Department of Histology and Embryology, İnönü University, Faculty of Medicine, Malatya, Türkiye
| | - Seren Sari Ünal
- Department of Pediatric Surgery, İnönü University, Faculty of Medicine, Malatya, Türkiye
| | - Ebubekir Altundaş
- Department of Pediatric Surgery, İnönü University, Faculty of Medicine, Malatya, Türkiye
| | - Turan Yildiz
- Department of Pediatric Surgery, İnönü University, Faculty of Medicine, Malatya, Türkiye
| | - Tevfik Tolga Şahin
- Department of Liver Transplantation Institute, İnönü University, Faculty of Medicine, Malatya, Türkiye
| | - Sezai Yilmaz
- Department of Liver Transplantation Institute, İnönü University, Faculty of Medicine, Malatya, Türkiye
| | - Mehmet Demircan
- Department of Pediatric Surgery, İnönü University, Faculty of Medicine, Malatya, Türkiye; Pediatric Intensive Burn Care Unit, İnönü University, Faculty of Medicine, Malatya, Türkiye
| |
Collapse
|