1
|
Tumentemur G, Aygun EG, Yurtsever B, Cakirsoy D, Ovali E. Effect of amniotic fluid on hair follicle growth. J DERMATOL TREAT 2025; 36:2451389. [PMID: 39827901 DOI: 10.1080/09546634.2025.2451389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 01/02/2025] [Indexed: 01/22/2025]
Abstract
Purpose: Human amniotic fluid stem cells (hAFSCs) have shown significant regenerative potential in treating hair loss, wound healing, and tissue repair. This study aims to evaluate the effects of human amniotic fluid (hAF) on hair follicle (HF) regeneration and immune system modulation. Materials and Methods: The hAF used was pooled, acellular, and gamma-irradiated to standardize its contents and enhance its stability. Both irradiated (FAFI) and non-irradiated (FAF) hAF were assessed for their efficacy and safety in promoting hair growth and modulating immune responses in a rat model of hair loss. The study examined HF regeneration, transition to the anagen phase, and macrophage polarization from the pro-inflammatory M1 phenotype to the anti-inflammatory M2 phenotype. Results: Both FAF and FAFI treatments significantly increased HF density, with FAFI exhibiting enhanced effects. Histological analysis demonstrated improved HF regeneration, increased M2 macrophages, and reduced collagen fiber deposition in treated areas. Gamma irradiation likely improved the efficacy of FAFI by stabilizing active components and inhibiting protease activity. Conclusions: Irradiated hAF is a safe and effective therapeutic candidate for alopecia and HF growth disorders. These findings support further evaluation of hAF in clinical trials to validate its potential for hair regeneration therapies.
Collapse
Affiliation(s)
- Gamze Tumentemur
- Vocational School of Health Services, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Elif Ganime Aygun
- Department of Obstetrics and Gynecology, Acibadem Mehmet Ali Aydinlar University Atakent Hospital, Istanbul, Turkey
| | - Bulut Yurtsever
- Acibadem Labcell Cellular Therapy Laboratory, Istanbul, Turkey
| | - Didem Cakirsoy
- Acibadem Labcell Cellular Therapy Laboratory, Istanbul, Turkey
| | - Ercument Ovali
- Acibadem Labcell Cellular Therapy Laboratory, Istanbul, Turkey
| |
Collapse
|
2
|
Ozkan S, Isildar B, Neccar D, Koyuturk M. Dynamic analysis of metabolic and ultrastructural changes in mesenchymal stem cells at hypoxic preconditioning and post-preconditioning stages: Cobalt chloride on the spotlight. Tissue Cell 2025; 95:102923. [PMID: 40267849 DOI: 10.1016/j.tice.2025.102923] [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: 01/16/2025] [Revised: 03/21/2025] [Accepted: 04/11/2025] [Indexed: 04/25/2025]
Abstract
Mesenchymal stem cells (MSCs) have come up as a potential remedy for treatment of various diseases thanks to their regenerative abilities. However, MSC-based therapies face challenges like reduced cell survival and functionality after transplantation. Preconditioning, particularly with hypoxia-mimicking agents like cobalt chloride (CoCl2), has been explored to enhance MSCs' effectiveness. This study aims to evaluate MSC survival, migration, and therapeutic outcomes at the CoCI2-preconditioning and post-preconditioning stages. Human umbilical cord-MSCs were treated with 100 µM CoCI2 with/out serum for 24-hours, and then passaged and planted in corresponding culture conditions without CoCI2, these two consecutive passages were named as the preconditioning and post-preconditioning stages, respectively. In each stage, total protein concentrations, total antioxidant and total oxidant status (TAS/TOS) of the conditioned media derived from the cells were investigated with bicinchoninic acid assay and TAS/TOS kits, respectively. The proliferation rates, migratory capacities, cellular senescence, expression levels of hypoxia-inducible factor1-α (HIF1-α), Ki-67, active caspase-3 and beclin-1 proteins and ultrastructures of the cells were evaluated by 3-(4,5-Dimethylthiazol-2-yl)-2,5-Diphenyltetrazolium Bromide test, wound healing assay, β-galactosidase-activity assessment, immunocytochemistry and transmission electron microscopy, respectively. Our results indicated that preconditioning MSCs with CoCl2 did not significantly enhance their proliferation, migration, or secretory abilities. However, it increased antioxidant capacity and along with normalization of senescence-status post-preconditioning, possibly by shifting energy metabolism from oxidative-phosphorylation to glycolysis through the upregulation of the HIF1-α signalling pathway. These findings indicate that CoCl2 preconditioning could be an effective approach to boost the therapeutic potential of MSCs, especially in enhancing their survival and functionality after transplantation.
Collapse
Affiliation(s)
- Serbay Ozkan
- Faculty of Medicine, Histology and Embryology Department, Izmir Katip Çelebi University, Çiğli, Izmir, 35620, Turkiye.
| | - Basak Isildar
- Faculty of Medicine, Histology and Embryology Department, Balıkesir University, Balıkesir 10145, Turkiye.
| | - Duygu Neccar
- Cerrahpasa Faculty of Medicine, Histology and Embryology Department, Istanbul University-Cerrahpasa, Fatih, Istanbul 34098, Turkiye.
| | - Meral Koyuturk
- Cerrahpasa Faculty of Medicine, Histology and Embryology Department, Istanbul University-Cerrahpasa, Fatih, Istanbul 34098, Turkiye.
| |
Collapse
|
3
|
Deng XH, Wu ZC, Sun Q, Huang LX, Xie YC, Lou DX, Li CG, Liu XQ, Zhou ZR, Tian T, Lian CL, Fu QL. The effects of Klotho delivering mesenchymal stem cell-derived small extracellular vesicles on acute kidney injury. J Nanobiotechnology 2025; 23:427. [PMID: 40481485 PMCID: PMC12144696 DOI: 10.1186/s12951-025-03499-4] [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: 02/05/2025] [Accepted: 05/27/2025] [Indexed: 06/11/2025] Open
Abstract
Acute kidney injury (AKI) is a life-threating syndrome characterized by sudden loss of kidney function, and its management is challenging and often suboptimal. Mesenchymal stem cells (MSCs) have shown promise in AKI therapy in pre-clinical and clinical trials; however, their clinical application still faces many challenges. MSC-derived small extracellular vesicles (sEV) may help overcome these challenges. In the current study, we overexpressed Klotho in MSCs and then isolated Klotho-loaded sEV (Klotho-sEV) using anion-exchange chromatography. Klotho-sEV displayed characteristics comparable to those of sEV in terms of size, morphology, conventional markers, and biosafety, as well as a higher abundance of Klotho protein. In rhabdomyolysis-induced AKI, sEV showed preferential tropism in injured kidneys. We found significantly and stably accelerated renal recovery, mitigated functional and histological abnormalities, stimulated tubular cell proliferation, reduced injury and inflammatory marker expression, and restored endogenous Klotho loss in mice after the administration of Klotho-sEV. In addition, Klotho-sEV treatment activated the mTOR and MEK1/2 signaling pathways. Proteomics and small RNA sequencing analyses of sEV and Klotho-sEV revealed abundant proteins and miRNAs involved in anti-inflammation and reno-protection, and Klotho-sEV showed characteristics that were different from those of sEV. In conclusion, Klotho-sEV may be a promising cell-free strategy for the treatment of AKI.
Collapse
Affiliation(s)
- Xiao-Hui Deng
- Department of Otorhinolaryngology, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou, 510080, Guangdong, PR China
- Extracellular Vesicle Research and Clinical Translational Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, PR China
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, PR China
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, 510080, Guangdong, PR China
| | - Zi-Cong Wu
- Department of Otorhinolaryngology, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou, 510080, Guangdong, PR China
- Extracellular Vesicle Research and Clinical Translational Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, PR China
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, 510080, Guangdong, PR China
| | - Qi Sun
- Department of Otorhinolaryngology, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou, 510080, Guangdong, PR China
| | - Long-Xin Huang
- Department of Otorhinolaryngology, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou, 510080, Guangdong, PR China
| | - Ying-Chun Xie
- Department of Otorhinolaryngology, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou, 510080, Guangdong, PR China
| | - Dong-Xiao Lou
- Department of Otorhinolaryngology, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou, 510080, Guangdong, PR China
| | - Chan-Gu Li
- Department of Otorhinolaryngology, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou, 510080, Guangdong, PR China
| | - Xiao-Qing Liu
- Department of Otorhinolaryngology, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou, 510080, Guangdong, PR China
| | - Zhi-Rou Zhou
- Department of Otorhinolaryngology, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou, 510080, Guangdong, PR China
| | - Tian Tian
- Department of Otorhinolaryngology, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou, 510080, Guangdong, PR China
| | - Chang-Lin Lian
- Extracellular Vesicle Research and Clinical Translational Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, PR China
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, 510080, Guangdong, PR China
| | - Qing-Ling Fu
- Department of Otorhinolaryngology, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou, 510080, Guangdong, PR China.
- Extracellular Vesicle Research and Clinical Translational Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, PR China.
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, 510080, Guangdong, PR China.
| |
Collapse
|
4
|
Gao Y, Chen C, Liu R, Zhang Z, Zhao X, Ma H. Research progress of connexin 43 mediated gap junction communication regulating bone metabolism in glucocorticoid-induced osteonecrosis of the femoral head. Exp Cell Res 2025; 449:114598. [PMID: 40339781 DOI: 10.1016/j.yexcr.2025.114598] [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: 11/13/2024] [Revised: 05/05/2025] [Accepted: 05/05/2025] [Indexed: 05/10/2025]
Abstract
Osteonecrosis of the femoral head (ONFH) is a refractory orthopedic disease that commonly affects young and middle-aged individuals. Long-term and high-dose use of glucocorticoids (GCs) is one of the main causes. Currently, the pathological mechanism of GCs-induced ONFH remains unclear, which poses difficulties for clinical prevention and treatment. This article focuses on reviewing the roles of gap junctions (GJs) and connexin 43 (Cx43) in GCs-induced ONFH. Under normal circumstances, cells in bone tissue form a network structure through GJs to maintain bone metabolic balance. However, GCs can obstruct the normal connections and signal transmission between bone tissue cells, leading to bone metabolic imbalance and triggering ONFH. As a key component of GJs in bone tissue, Cx43 is of great significance in bone metabolism. It not only participates in the construction of the osteocyte network but also regulates osteocyte activity, osteoblast differentiation, and osteogenic activities. Meanwhile, in vascular endothelial cells, Cx43 plays an important role in angiogenesis and maintaining vascular homeostasis, and is closely related to the vascularization of bone tissue. In addition, Cx43 is associated with the release of prostaglandin E2 (PGE2). GCs can inhibit the activity of Cx43, reduce the release of PGE2, and disrupt the balance of bone metabolism. Studies have shown that measuring changes in the expression level of Cx43 is expected to become an early diagnostic biomarker for GCs-induced ONFH. Enhancing its expression through small - molecule drugs, biological agents, and gene therapy may be potential treatment approaches for ONFH. This article proposes the PI3K/Akt/GSK3β/β-catenin pathway and conducts research on the regulatory mechanism of Cx43-mediated GJ-based intercellular communication, aiming to provide new ideas for the treatment of ONFH and bone metabolism-related diseases.
Collapse
Affiliation(s)
- Yang Gao
- Department of Orthopaedics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, People's Republic of China
| | - Changjun Chen
- Department of Orthopedics, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, People's Republic of China
| | - Rongxing Liu
- Department of Orthopaedics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, People's Republic of China
| | - Zhongkai Zhang
- Department of Orthopaedics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, People's Republic of China
| | - Xin Zhao
- Department of Orthopaedics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, People's Republic of China.
| | - Huanzhi Ma
- Department of Orthopaedics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, People's Republic of China.
| |
Collapse
|
5
|
Kharat A, Bhate K, Sanap A, Kheur S, Contractor M, Suryawanshi P, Bhonde R. Exploring the Differentiation Abilities of Hair Follicle and Dental Pulp Stem Cells Into Islet Like Cells. Cell Biol Int 2025; 49:634-644. [PMID: 40014298 DOI: 10.1002/cbin.70010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 01/03/2025] [Accepted: 02/16/2025] [Indexed: 02/28/2025]
Abstract
This study aimed to compare the differentiation potential of dental pulp-derived mesenchymal stem cells (DP-MSCs) and hair follicle-derived mesenchymal stem cells (HF-MSCs), which originated from the ectoderm. Dental pulps were separated from the extracted wisdom teeth during dental surgery, and Hair follicles were extracted from the scalp of patients undergoing hair transplantation. We cultivated the cell in cell culture media, supplemented with additional nutrients. After the fourth passage, the homogeneous population of DP-MSCs and HF-MSCs was analyzed for the surface markers (CD73, CD90, and CD105) by fluorescence-activated cell sorting. In vitro, the multi-lineage differentiation potential for both the MSCs was tested with respective induction media such as osteogenic, chondrogenic, adipogenic, and insulin-producing cells. Following the fourth passage, identical fibroblast-like cells were noted in each culture plate. Mesenchymal stem cell marker was expressed in both DP-MSCs and HF-MSCs. Both the DP-MSCs and HF-MSCs exhibited similar differentiation potential toward osteogenic, chondrogenic, and adipogenic differentiation. However, there was a difference in the differentiation potential into IPCs. HF-MSCs showed higher C-peptide and insulin secretion response to glucose, PDX1, and Insulin gene expression compared to DP-MSCs. These findings suggest that although DP-MSCs and HF-MSCs showed similar stemness properties, they differ in their differentiation potential towards insulin-producing cells (IPCs). This is the first report showing the potential of HF-MSCs to generate IPCs, revealing hair follicles as a novel and promising source for autologous stem cell therapy in diabetes. The generated islet organoids can be used for diabetic drug toxicity testing and screening.
Collapse
Affiliation(s)
- Avinash Kharat
- Regenerative Medicine Laboratory, Dr. D. Y. Patil Dental College and Hospital, Dr. D. Y. Patil Vidyapeeth, Pimpri, Pune, Maharashtra, India
| | - Kalyani Bhate
- Department of Oral and Maxillofacial Surgery, Dr. D. Y. Patil Dental College and Hospital, Dr. D. Y. Patil Vidyapeeth, Pimpri, Pune, Maharashtra, India
| | - Avinash Sanap
- Regenerative Medicine Laboratory, Dr. D. Y. Patil Dental College and Hospital, Dr. D. Y. Patil Vidyapeeth, Pimpri, Pune, Maharashtra, India
| | - Supriya Kheur
- Regenerative Medicine Laboratory, Dr. D. Y. Patil Dental College and Hospital, Dr. D. Y. Patil Vidyapeeth, Pimpri, Pune, Maharashtra, India
| | - Murtuza Contractor
- Department of Oral and Maxillofacial Surgery, Dr. D. Y. Patil Dental College and Hospital, Dr. D. Y. Patil Vidyapeeth, Pimpri, Pune, Maharashtra, India
| | - Poonam Suryawanshi
- The Central Research Facility, Dr. D. Y. Patil Medical College, Hospital and Research Centre, Dr. D. Y. Patil Vidyapeeth, Pimpri, Pune, Maharashtra, India
| | - Ramesh Bhonde
- Regenerative Medicine Laboratory, Dr. D. Y. Patil Dental College and Hospital, Dr. D. Y. Patil Vidyapeeth, Pimpri, Pune, Maharashtra, India
| |
Collapse
|
6
|
Mojtahedi A, Ghaderi S, Ghiasi M, Halabian R, Dehghan H, Padash A, Eftekhari E, Salimi A. Investigating the enhancement of neural differentiation of adipose-derived mesenchymal stem cell with Foeniculum vulgare nanoemulsions: An in vitro research. Tissue Cell 2025; 94:102806. [PMID: 40022910 DOI: 10.1016/j.tice.2025.102806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 02/15/2025] [Accepted: 02/15/2025] [Indexed: 03/04/2025]
Abstract
BACKGROUND Neurons, distributed throughout the body, regulate various bodily functions. The recovery of the nervous system is often slow and can be irreversible. Currently, the approach of using mesenchymal stem cells (MSCs) in conjunction with conventional treatments for nervous system injuries is being explored. Nanoemulsions are systems designed for the nanoscale delivery of drug cargoes. Foeniculum vulgare (F. vulgare), a medicinal plant long utilized in complementary medicine, is the focus of this study. The aim is to utilize nanoemulsions of fennel to induce the differentiation of MSCs into neural-like cells in vitro. MATERIALS AND METHODS Human adipose-derived mesenchymal stem cells (hADSCs) were commercially purchased. These cells were cultured in DMEM medium containing 10 % fetal bovine serum and 1 % penicillin-streptomycin antibiotic. Based on a sequential extraction method, n-hexane (Hex), ethyl acetate (EtAc), and ethanolic extracts were obtained from the seeds of F. vulgare. To prepare the F. vulgare extract nanoemulsion, the aqueous phase (distilled water), the oily part (F. vulgare extract), Span 80 and Tween 20 were used. The optimal dose of F. vulgare nanoemulsion was determined using the MTT assay and acridine orange/ethidium bromide (AO/EB) staining. Neural differentiation was induced using a specialized differentiation medium on the MSCs, with the prepared nanoemulsions acting as inducers. The neural differentiation of the human differentiated hADSCs was studied and evaluated through Real-time PCR and immunocytochemistry (ICC) techniques on days 7 and 14. RESULTS The results obtained from the MTT and AO/EB tests indicated that the optimal dose of F. vulgare nanoemulsions is 1 μg/ml. Analysis of neural differentiation index gene expression revealed a significant (P ≤ 0.05) upregulation of MAP-2, β-tubulin III, and NSE genes on days 7 and 14 following treatment with the nanoemulsions. It is noteworthy that the nanoemulsion prepared from the hexane extract of the plant showed a significant increase in the expression of marker genes in the process of neural differentiation. Protein expression analysis demonstrated an increase in MAP-2, β-tubulin III, and NSE (gamma enolase) proteins in response to the nanoemulsion inducers compared to the control group (TCPS). DISCUSSION Overall, our findings indicate that F. vulgare nanoemulsions have a positive effect on the expression of genes and proteins related to neural differentiation in hADSCs. The proposed protocol may serve as a potential therapeutic strategy in complementary medicine for patients seeking to improve injuries to the nervous system. However, further studies and performance measurements are necessary in future research to confirm these results.
Collapse
Affiliation(s)
- Arya Mojtahedi
- Department of Biology, Borujerd Branch, Islamic Azad University, Borujerd, Iran
| | - Shima Ghaderi
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Mohsen Ghiasi
- Cardiovascular Research Center, Rajaie Cardiovascular Institute, Tehran, Iran
| | - Raheleh Halabian
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Hossein Dehghan
- Department of Basic Sciences, Medicinal Plants Research Center, Shahed University, Tehran, Iran
| | - Arash Padash
- Department of Medical Nanotechnology, Faculty of Advanced Sciences and Technology, Pharmaceutical Sciences Branch, Islamic Azad University, Tehran, Iran
| | - Elahe Eftekhari
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Ali Salimi
- Tissue Engineering and Regenerative Medicine Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
7
|
Tu GC, Abedi F, Chang AY, Shen X, Soleimani M, Araujo I, Jung R, Kwon J, Anwar KN, Arabpour Z, Mahmud N, Tu EY, Dana R, Hematti P, Joslin CE, Djalilian AR. Safety of subconjunctival injection of mesenchymal stromal cells in persistent corneal epithelial disease - A phase 1b clinical trial. Ocul Surf 2025; 38:8-13. [PMID: 40414287 DOI: 10.1016/j.jtos.2025.05.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2025] [Revised: 04/19/2025] [Accepted: 05/20/2025] [Indexed: 05/27/2025]
Abstract
PURPOSE To investigate the safety and tolerability of subconjunctival injection of three escalating doses of allogeneic bone marrow-derived mesenchymal stromal cells (MSCs) in patients with persistent corneal epithelial defect/disease (PCED). DESIGN Prospective single-center open label phase 1b clinical trial. PARTICIPANTS Patients with PCED in the setting of neurotrophic keratitis and/or limbal stem cell deficiency. METHODS A dose escalation study design was used. The first three patients received a subconjunctival injection of 1 × 106 MSCs/50 μL suspension; subsequently, three participants were treated with 1 subconjunctival injection of 3 × 106 MSCs/150 μL; and two participants received 2 subconjunctival injections of 3 × 106 MSCs/150 μl in 2 conjunctival sites. MAIN OUTCOME MEASURES The primary outcome was the safety of the treatment determined on day 28 post-injection. Ocular surface toxicity and other ocular or systemic treatment emergent adverse events (TEAEs) were assessed at 1, 7, 14, 28 and 90 days. Demonstration of safety on day 28 was required before escalating to the next higher dose. Changes in the PCED were also monitored. RESULTS Eight participants completed the 90-day study. All 3 doses of subconjunctival MSCs were well tolerated. No participant developed ocular surface toxicity or other ocular or systemic TEAEs. The size of the PCED improved in 5 (63 %) patients; it increased in 2 (25 %) patients; and no progressive improvement was observed with dose escalation. CONCLUSION Subconjunctival administration of MSCs was safe and well tolerated with no systemic or ocular toxicity in patients with PCED. Improvement in epithelial defect size was observed in 63 % of the participants.
Collapse
Affiliation(s)
- Grace C Tu
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, University of Illinois at Chicago, Chicago, IL, USA
| | - Farshad Abedi
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, University of Illinois at Chicago, Chicago, IL, USA
| | - Arthur Y Chang
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, University of Illinois at Chicago, Chicago, IL, USA
| | - Xiang Shen
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, University of Illinois at Chicago, Chicago, IL, USA
| | - Mohammad Soleimani
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, University of Illinois at Chicago, Chicago, IL, USA
| | - Iskra Araujo
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, University of Illinois at Chicago, Chicago, IL, USA
| | - Rebecca Jung
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, University of Illinois at Chicago, Chicago, IL, USA
| | - Jeonghyun Kwon
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, University of Illinois at Chicago, Chicago, IL, USA
| | - Khandeker N Anwar
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, University of Illinois at Chicago, Chicago, IL, USA
| | - Zohreh Arabpour
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, University of Illinois at Chicago, Chicago, IL, USA
| | - Nadim Mahmud
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, University of Illinois at Chicago, Chicago, IL, USA
| | - Elmer Y Tu
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, University of Illinois at Chicago, Chicago, IL, USA
| | - Reza Dana
- Massachusetts Eye and Ear, Harvard Medical School Department of Ophthalmology, Boston, MA, USA
| | - Peiman Hematti
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Charlotte E Joslin
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, University of Illinois at Chicago, Chicago, IL, USA; Division of Epidemiology and Biostatistics, University of Illinois at Chicago, Chicago, IL, USA
| | - Ali R Djalilian
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, University of Illinois at Chicago, Chicago, IL, USA.
| |
Collapse
|
8
|
Gao F, Pan L, Liu W, Chen J, Wang Y, Li Y, Liu Y, Hua Y, Li R, Zhang T, Zhu T, Jin F, Gao Y. Idiopathic pulmonary fibrosis microenvironment: Novel mechanisms and research directions. Int Immunopharmacol 2025; 155:114653. [PMID: 40222273 DOI: 10.1016/j.intimp.2025.114653] [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: 01/23/2025] [Revised: 04/08/2025] [Accepted: 04/08/2025] [Indexed: 04/15/2025]
Abstract
Idiopathic Pulmonary Fibrosis (IPF) is a progressive interstitial lung disease marked by increasing dyspnea and respiratory failure. The underlying mechanisms remain poorly understood, given the complexity of its pathogenesis. This review investigates the microenvironment of IPF to identify novel mechanisms and therapeutic avenues. Studies have revealed that various cell types, including alveolar epithelial cells, fibroblasts, myofibroblasts, and immune cells, are integral to disease progression, engaging in cellular stress responses and inflammatory regulation via signaling pathways such as TGF-β, Wnt, mTOR, and ROS. Non-coding RNAs, particularly miRNAs, are critical in IPF and may serve as diagnostic and prognostic biomarkers. Regarding treatment, mesenchymal stem cells (MSCs) and their extracellular vesicles (EVs) or non-vesicular derivatives offer promise by modulating immune responses, enhancing tissue repair, and inhibiting fibrosis. Additionally, alterations in the lung microbiota are increasingly recognized as a contributing factor to IPF progression, offering fresh insights into potential treatments. Despite the encouraging results of MSC-based therapies, the precise mechanisms and clinical applications remain subjects of ongoing research. This review emphasizes the significance of the IPF microenvironment and highlights the need for further exploration to develop effective therapies that could enhance patient outcomes.
Collapse
Affiliation(s)
- Fuguo Gao
- Department of Pulmonary and Critical Care Medicine, Tangdu hospital, Air Force Medical University, Xi'an, 710038, China
| | - Lei Pan
- Department of Pulmonary and Critical Care Medicine, Tangdu hospital, Air Force Medical University, Xi'an, 710038, China
| | - Wei Liu
- Department of Pulmonary and Critical Care Medicine, Tangdu hospital, Air Force Medical University, Xi'an, 710038, China
| | - Jian Chen
- Department of Pulmonary and Critical Care Medicine, Tangdu hospital, Air Force Medical University, Xi'an, 710038, China
| | - Yifeng Wang
- Department of Pulmonary and Critical Care Medicine, Tangdu hospital, Air Force Medical University, Xi'an, 710038, China
| | - Yan Li
- Department of Pulmonary and Critical Care Medicine, Tangdu hospital, Air Force Medical University, Xi'an, 710038, China; Department of Pulmonary and Critical Care Medicine, Shaanxi provincal people's hospital, Xi'an, 710068, China
| | - Yurou Liu
- Department of Pulmonary and Critical Care Medicine, Tangdu hospital, Air Force Medical University, Xi'an, 710038, China
| | - Yiying Hua
- Department of Pulmonary and Critical Care Medicine, Tangdu hospital, Air Force Medical University, Xi'an, 710038, China
| | - Ruiqi Li
- Department of Pulmonary and Critical Care Medicine, Tangdu hospital, Air Force Medical University, Xi'an, 710038, China
| | - Tongtong Zhang
- Department of Pulmonary and Critical Care Medicine, Tangdu hospital, Air Force Medical University, Xi'an, 710038, China
| | - Ting Zhu
- Department of Pulmonary and Critical Care Medicine, Tangdu hospital, Air Force Medical University, Xi'an, 710038, China
| | - Faguang Jin
- Department of Pulmonary and Critical Care Medicine, Tangdu hospital, Air Force Medical University, Xi'an, 710038, China.
| | - Yongheng Gao
- Department of Pulmonary and Critical Care Medicine, Tangdu hospital, Air Force Medical University, Xi'an, 710038, China.
| |
Collapse
|
9
|
Liu J, Chen Y, Li Z, Li Z, Lyu F, Wang F, Wang A, Liu Z, Liao X, Wu J. Human dental pulp stem cells attenuate airway inflammation in mice with PM 2.5-induced asthma exacerbation by inhibiting the pyroptosis pathway. Stem Cell Res Ther 2025; 16:240. [PMID: 40361181 PMCID: PMC12076901 DOI: 10.1186/s13287-025-04368-5] [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: 11/05/2024] [Accepted: 04/25/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND Fine particulate matter (PM2.5) exposure significantly exacerbates respiratory morbidity, particularly in asthmatic individuals, highlighting an urgent need for effective therapeutic interventions. In this study, we evaluated the therapeutic potential and underlying mechanisms of human dental pulp stem cells (hDPSCs), a promising mesenchymal stem cell population, in mitigating airway inflammation in mice with PM2.5-induced asthma exacerbation. METHODS In a PM2.5-exacerbated ovalbumin (OVA)-asthma murine model, hDPSCs were intravenously administered with MCC950 (NLRP3 inhibitor) as positive control, systematically evaluating their therapeutic effects on airway inflammation and pyroptosis through pulmonary function tests, histopathological examination, cytological and molecular analyses. RESULTS The administration of hDPSCs ameliorated airway inflammation. Moreover, hDPSCs further alleviated Th2 inflammation and decreased serum IgE concentrations, along with a decrease in eosinophils in BALF. At the same time, interleukin-1β (IL-1β) and IL-18 levels in BALF and caspase-1 activity in lung tissues were reduced. In addition, immunohistochemistry showed that the expression levels of NLRP3, caspase-1, GSDMD, cleaved capsase-1 and IL-1β were reduced. The western blot results also showed that the expression level of NLRP3/caspase-1/GSDMD/cleaved capsase-1 in the classical pathway of pyroptosis decreased after hDPSCs intervention. CONCLUSIONS These findings provided the first evidence that hDPSCs transplantation attenuated allergic airway inflammation and mucus secretion in mice with PM2.5-induced asthma exacerbation. Thus, hDPSCs exert these protective effects through suppression of the NLRP3/caspase-1/GSDMD-mediated pyroptosis pathway, suggesting their potential as a novel cell-based therapy for PM2.5 inhalation-mediated asthma.
Collapse
Affiliation(s)
- Jianling Liu
- Second Department of Elderly Respiratory, Guangdong Provincial People's Hospital, Southern Medical University, Guangzhou, 510080, China
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Yuehua Chen
- Second Department of Elderly Respiratory, Guangdong Provincial People's Hospital, Southern Medical University, Guangzhou, 510080, China
| | - Zhongpeng Li
- Critical Care Medicine Department, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Zhangwen Li
- Second Department of Elderly Respiratory, Guangdong Provincial People's Hospital, Southern Medical University, Guangzhou, 510080, China
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Fengjuan Lyu
- Joint Center for Regenerative Medicine Research of South China University of Technology and the University of Western Australia, School of Medicine, South China University of Technology, Guangzhou, 515000, P.R. China
- The Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, 510000, P.R. China
| | - Fang Wang
- Second Department of Elderly Respiratory, Guangdong Provincial People's Hospital, Southern Medical University, Guangzhou, 510080, China
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Aili Wang
- Second Department of Elderly Respiratory, Guangdong Provincial People's Hospital, Southern Medical University, Guangzhou, 510080, China
| | - Zhangquan Liu
- Second Department of Elderly Respiratory, Guangdong Provincial People's Hospital, Southern Medical University, Guangzhou, 510080, China
| | - Xiaoyang Liao
- Second Department of Elderly Respiratory, Guangdong Provincial People's Hospital, Southern Medical University, Guangzhou, 510080, China
| | - Jian Wu
- Second Department of Elderly Respiratory, Guangdong Provincial People's Hospital, Southern Medical University, Guangzhou, 510080, China.
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, China.
| |
Collapse
|
10
|
Zheng Y, Yang G, Li P, Tian B. Bioelectric and physicochemical foundations of bioelectronics in tissue regeneration. Biomaterials 2025; 322:123385. [PMID: 40367812 DOI: 10.1016/j.biomaterials.2025.123385] [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: 12/21/2024] [Revised: 04/15/2025] [Accepted: 05/01/2025] [Indexed: 05/16/2025]
Abstract
Understanding and exploiting bioelectric signaling pathways and physicochemical properties of materials that interface with living tissues is central to advancing tissue regeneration. In particular, the emerging field of bioelectronics leverages these principles to develop personalized, minimally invasive therapeutic strategies tailored to the dynamic demands of individual patients. By integrating sensing and actuation modules into flexible, biocompatible devices, clinicians can continuously monitor and modulate local electrical microenvironments, thereby guiding regenerative processes without extensive surgical interventions. This review provides a critical examination of how fundamental bioelectric cues and physicochemical considerations drive the design and engineering of next-generation bioelectronic platforms. These platforms not only promote the formation and maturation of new tissues across neural, cardiac, musculoskeletal, skin, and gastrointestinal systems but also precisely align therapies with the unique structural, functional, and electrophysiological characteristics of each tissue type. Collectively, these insights and innovations represent a convergence of biology, electronics, and materials science that holds tremendous promise for enhancing the efficacy, specificity, and long-term stability of regenerative treatments, ushering in a new era of advanced tissue engineering and patient-centered regenerative medicine.
Collapse
Affiliation(s)
- Yuze Zheng
- Department of Chemistry, The University of Chicago, Chicago, IL, 60637, USA
| | - Guangqing Yang
- Department of Chemistry, The University of Chicago, Chicago, IL, 60637, USA
| | - Pengju Li
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL, 60637, USA
| | - Bozhi Tian
- Department of Chemistry, The University of Chicago, Chicago, IL, 60637, USA; The James Franck Institute, The University of Chicago, Chicago, IL, 60637, USA; The Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL, 60637, USA.
| |
Collapse
|
11
|
Surico PL, Barone V, Singh RB, Coassin M, Blanco T, Dohlman TH, Basu S, Chauhan SK, Dana R, Di Zazzo A. Potential applications of mesenchymal stem cells in ocular surface immune-mediated disorders. Surv Ophthalmol 2025; 70:467-479. [PMID: 39097173 DOI: 10.1016/j.survophthal.2024.07.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 07/29/2024] [Accepted: 07/29/2024] [Indexed: 08/05/2024]
Abstract
We explore the interaction between corneal immunity and mesenchymal stem/stromal cells (MSCs) and their potential in treating corneal and ocular surface disorders. We outline the cornea's immune privilege mechanisms and the immunomodulatory substances involved. In this realm, MSCs are characterized by their immunomodulatory properties and regenerative potential, making them promising for therapeutic application. Therefore, we focus on the role of MSCs in immune-mediated corneal diseases such as dry eye disease, corneal transplantation rejection, limbal stem cell deficiency, and ocular graft-versus-host disease. Preclinical and clinical studies demonstrate MSCs' efficacy in promoting corneal healing and reducing inflammation in these conditions. Overall, we emphasize the potential of MSCs as innovative therapies in ophthalmology, offering promising solutions for managing various ocular surface pathologies.
Collapse
Affiliation(s)
- Pier Luigi Surico
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA; Department of Ophthalmology, Campus Bio-Medico University Hospital, Rome 00128, Italy; Cornea Rare Diseases Center, Fondazione Policlinico Campus Bio-Medico, Rome 00128, Italy
| | - Vincenzo Barone
- Department of Ophthalmology, Campus Bio-Medico University Hospital, Rome 00128, Italy; Cornea Rare Diseases Center, Fondazione Policlinico Campus Bio-Medico, Rome 00128, Italy
| | - Rohan Bir Singh
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Marco Coassin
- Department of Ophthalmology, Campus Bio-Medico University Hospital, Rome 00128, Italy; Cornea Rare Diseases Center, Fondazione Policlinico Campus Bio-Medico, Rome 00128, Italy
| | - Tomas Blanco
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Thomas H Dohlman
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Sayan Basu
- Brien Holden Eye Research Centre (BHERC), L. V. Prasad Eye Institute, Hyderabad, Telangana, India
| | - Sunil K Chauhan
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Reza Dana
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Antonio Di Zazzo
- Department of Ophthalmology, Campus Bio-Medico University Hospital, Rome 00128, Italy; Cornea Rare Diseases Center, Fondazione Policlinico Campus Bio-Medico, Rome 00128, Italy.
| |
Collapse
|
12
|
Zhao DZ, Wei HX, Yang YB, Yang K, Chen F, Zhang Q, Zhang T. Advances in the Research of Mesenchymal Stromal Cells in the Treatment of Maxillofacial Neurological Disorders and the Promotion of Facial Nerve Regeneration. Mol Neurobiol 2025:10.1007/s12035-025-04981-8. [PMID: 40295362 DOI: 10.1007/s12035-025-04981-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Accepted: 04/18/2025] [Indexed: 04/30/2025]
Abstract
Maxillofacial neurological disorders include a range of disorders affecting the cranial nerves, which can be caused by a variety of reasons, including infection, trauma, tumor, and surgical complications, resulting in severe dysfunction, and the study of new approaches for the treatment of these disorders is crucial for the restoration of sensory and motor functions of the face. In recent years, due to the excellent tissue regenerative ability of mesenchymal stromal cells (MSCs), research on MSCs and MSC-derived exosomes has been progressively deepened, bringing many new perspectives to the therapeutic strategies for many diseases. Facial nerve regeneration is a complex process involving various pathophysiological mechanisms and therapeutic strategies to restore nerve function after injury. And the rapid development of stem cell tissue engineering has greatly facilitated the research process of facial nerve regeneration. In this paper, we review the characteristics of MSCs and neural stem cells (NSCs), the roles they play in the neural microenvironment and the mechanisms that promote nerve regeneration, summarize the research progress of MSCs in the treatment of maxillofacial neurological disorders, and highlight the promising directions for future development.
Collapse
Affiliation(s)
- De-Zhi Zhao
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
- Department of Prosthetics, Affiliated Stomatology Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Han-Xiao Wei
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Yi-Bing Yang
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Kang Yang
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Fang Chen
- Department of Prosthetics, Affiliated Stomatology Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Qian Zhang
- Department of Human Anatomy, Zunyi Medical University, Zunyi, Guizhou, China.
| | - Tao Zhang
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China.
| |
Collapse
|
13
|
Wang C, Ge F, Ge F, Xu Z, Jiang J. Harnessing stem cell therapeutics in LPS-induced animal models: mechanisms, efficacies, and future directions. Stem Cell Res Ther 2025; 16:176. [PMID: 40221751 PMCID: PMC11993993 DOI: 10.1186/s13287-025-04290-w] [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: 02/11/2025] [Accepted: 03/19/2025] [Indexed: 04/14/2025] Open
Abstract
The severity and threat posed by inflammation are well documented, and lipopolysaccharides (LPS), as important inducers of inflammatory responses, are widely recognized for studying host immunity and the resulting tissue and organ damage. The LPS-induced disease model, triggers a remarkable release of inflammatory factors, immune and coagulation dysfunction, and damage to vital organs such as the brain, lungs, heart, liver, and kidneys. Recently, the role of mesenchymal stem cells (MSCs) in various clinical diseases has garnered significant attention due to their immunomodulatory, anti-inflammatory, tissue healing, anti-apoptotic, and antibacterial properties. Despite the common use of LPS models to induce disease models and simulate acute inflammation, the integration of stem cell therapy within these models remains underexplored. This article integrates the LPS induced animal model and reviews the current evidence regarding the therapeutic mechanisms of stem cells in LPS-induced disease models across various human body systems. Furthermore, this review predicts and hypothesizes the feasibility and potential of using stem cells in disease models that have not yet been extensively studied, based on existing animal inflammation models.
Collapse
Affiliation(s)
- Chengran Wang
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, 130033, Jilin Province, People's Republic of China
| | - Fanghong Ge
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, 130033, Jilin Province, People's Republic of China
| | - Fangjun Ge
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, Guangdong Province, People's Republic of China
| | - Zhonghang Xu
- Department of Breast Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, Jilin Province, People's Republic of China.
| | - Jinlan Jiang
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, 130033, Jilin Province, People's Republic of China.
| |
Collapse
|
14
|
Hoseini SM, Montazeri F. Cell origin and microenvironment: The players of differentiation capacity in human mesenchymal stem cells. Tissue Cell 2025; 93:102709. [PMID: 39765135 DOI: 10.1016/j.tice.2024.102709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 12/12/2024] [Accepted: 12/26/2024] [Indexed: 03/05/2025]
Abstract
Mesenchymal stem cells (MSCs) have several important properties that make them desirable for regenerative medicine. These properties include immunomodulatory ability, growth factor production, and differentiation into various cell types. Despite extensive research and promising results in clinical trials, our understanding of MSC biology, their mechanism of action, and their targeted and routine use in clinics is limited. Differentiation of human MSCs (hMSCs) is a complex process influenced by various elements such as growth factors, pharmaceutical compounds, microRNAs, 3D scaffolds, and mechanical and electrical stimulation. Research has shown that different culture conditions can affect the differentiation potential of hMSCs obtained from multiple fetal and adult sources. Additionally, it seems that what affects the differentiation capacities of these cells is their secretory characteristics, which are influenced by the origin of the cells and the local microenvironment where the cells are located. The review can provide insights into the microenvironment-based mechanisms involved in MSC differentiation, which can be valuable for future therapeutic applications.
Collapse
Affiliation(s)
- Seyed Mehdi Hoseini
- Biotechnology Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran; Hematology and Oncology Research Center, Non-communicable Diseases Research Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Fateme Montazeri
- Abortion Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran.
| |
Collapse
|
15
|
Li W, He S, Lin C, Yang S, Zhang W. Mesenchymal stem cell-derived exosomes carry miR-125a-5p to improve diabetic keratopathy by regulating endoplasmic reticulum stress. Tissue Cell 2025; 93:102669. [PMID: 39674096 DOI: 10.1016/j.tice.2024.102669] [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: 08/18/2024] [Revised: 12/03/2024] [Accepted: 12/04/2024] [Indexed: 12/16/2024]
Abstract
BACKGROUND Diabetic keratopathy is a prevalent but sometimes ignored visual condition in diabetic patients, which significantly affects patients with diabetes mellitus (DM) in terms of their visual acuity. Exosomes regulate diabetes-related conditions like diabetic keratopathy (DK) by secreting their components into the body. OBJECTIVE Aim to investigate the effect and mechanism of mesenchymal stem cell (MSC)-derived exosome miR-125a-5p on DK. METHODS Transmission electron microscopy, along with nanoparticle tracking analysis, was used to determine the morphology and size of exosomes. To evaluate cell viability, proliferation, and migration, Western blotting and RT-qPCR methods were used. CCK-8, cell cloning, and scratch assays were used to measure protein levels and mRNA expression. RESULTS High glucose treatment of corneal epithelial cells weakened cell viability, proliferation and migration, and the level of miR-125a-5p was significantly reduced. It has been proposed that elevated levels of miR-125a-5p could enhance cell viability, proliferation, and migration, can inhibit endoplasmic reticulum stress induced by high glucose, which is the same as the effect of endoplasmic reticulum stress inhibitors. CONCLUSION Mouse bone marrow MSC-derived exosome miR-125a-5p repairs corneal epithelial cell viability and proliferation as well as migration ability to improve DK by inhibiting high glucose-induced endoplasmic reticulum stress.
Collapse
Affiliation(s)
- Weina Li
- Ophthalmology Department, Nanxishan Hospital of Guangxi Zhuang Autonomous Region (The second Hospital of Guangxi Zhuang Autonomous Region), Guilin, Guangxi 541000, China; Ophthalmology Department, The Second Affiliated Hospital of Guangxi University of Science and Technology, Liuzhou, Guangxi 545005, China.
| | - Shiping He
- Glaucoma and Cataract Department, Liuzhou Aier Ophthalmology Hospital, Liuzhou, Guangxi 545005, China
| | - Chaoqun Lin
- Neurosurgery Department, University of Chinese Academy of Sciences-Shenzhen Hospital (Guangming District), Shenzhen, Guangdong 518106, China
| | - Sheng Yang
- Glaucoma and Cataract Department, Liuzhou Aier Ophthalmology Hospital, Liuzhou, Guangxi 545005, China
| | - Wenbin Zhang
- Ophthalmology Department, Nanxishan Hospital of Guangxi Zhuang Autonomous Region (The second Hospital of Guangxi Zhuang Autonomous Region), Guilin, Guangxi 541000, China
| |
Collapse
|
16
|
Xiong J, Ma R, Xie K, Shan C, Chen H, Wang Y, Liao Y, Deng Y, Ye G, Wang Y, Zhu Q, Zhang Y, Cai H, Guo W, Yin Y, Li Z. Recapitulation of endochondral ossification by hPSC-derived SOX9 + sclerotomal progenitors. Nat Commun 2025; 16:2781. [PMID: 40118845 PMCID: PMC11928506 DOI: 10.1038/s41467-025-58122-9] [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: 03/27/2024] [Accepted: 03/11/2025] [Indexed: 03/24/2025] Open
Abstract
Endochondral ossification generates most of the load-bearing bones, recapitulating it in human cells remains a challenge. Here, we report generation of SOX9+ sclerotomal progenitors (scl-progenitors), a mesenchymal precursor at the pre-condensation stage, from human pluripotent stem cells and development of osteochondral induction methods for these cells. Upon lineage-specific induction, SOX9+ scl-progenitors have not only generated articular cartilage but have also undergone spontaneous condensation, cartilaginous anlagen formation, chondrocyte hypertrophy, vascular invasion, and finally bone formation with stroma, thereby recapitulating key stages during endochondral ossification. Moreover, self-organized growth plate-like structures have also been induced using SOX9+ scl-progenitor-derived fusion constructs with chondro- and osteo-spheroids, exhibiting molecular and cellular similarities to the primary growth plates. Furthermore, we have identified ITGA9 as a specific surface marker for reporter-independent isolation of SOX9+ scl-progenitors and established a culture system to support their expansion. Our work highlights SOX9+ scl-progenitors as a promising tool for modeling human skeletal development and bone/cartilage bioengineering.
Collapse
Affiliation(s)
- Jingfei Xiong
- Center of Growth Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu, China
| | - Runxin Ma
- Center of Growth Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu, China
| | - Kun Xie
- Center of Growth Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu, China
| | - Ce Shan
- Center of Growth Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu, China
| | - Hanyi Chen
- Center of Growth Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu, China
| | - Yuqing Wang
- Center of Growth Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu, China
| | - Yuansong Liao
- Center of Growth Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu, China
| | - Yanhui Deng
- Center of Growth Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu, China
| | - Guogen Ye
- Center of Growth Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu, China
| | - Yifu Wang
- Center of Growth Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu, China
| | - Qing Zhu
- Center of Growth Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu, China
- Department of Anesthesiology, West China Second University Hospital, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, Sichuan University, Chengdu, China
| | - Yunqiu Zhang
- Center of Growth Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu, China
| | - Haoyang Cai
- Center of Growth Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu, China
| | - Weihua Guo
- Yunnan Key Laboratory of Stomatology, Department of Pediatric Dentistry, The Affiliated Stomatology Hospital of Kunming Medical University, Kunming Medical University, Kunming, China
| | - Yike Yin
- Center of Growth Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu, China.
| | - Zhonghan Li
- Center of Growth Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu, China.
- Yunnan Key Laboratory of Stomatology, Department of Pediatric Dentistry, The Affiliated Stomatology Hospital of Kunming Medical University, Kunming Medical University, Kunming, China.
| |
Collapse
|
17
|
Gharandouq MH, Ismail MA, Saleh T, Zihlif M, Ababneh NA. Metformin Protects Human Induced Pluripotent Stem Cell (hiPSC)-Derived Neurons from Oxidative Damage Through Antioxidant Mechanisms. Neurotox Res 2025; 43:15. [PMID: 40100475 DOI: 10.1007/s12640-025-00734-6] [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: 07/28/2024] [Revised: 02/18/2025] [Accepted: 03/08/2025] [Indexed: 03/20/2025]
Abstract
The antidiabetic drug metformin possesses antioxidant and cell protective effects including in neuronal cells, suggesting its potential use for treating neurodegenerative diseases. This study aimed to assess metformin's effects on viability and antioxidant activity in human-induced pluripotent stem cell (hiPSC)-derived neurons under varying concentrations and stress conditions. Six lines of hiPSC-derived neuronal progenitors derived from healthy human iPSCs were treated with metformin (1-500 µM) on day 18 of differentiation. For mature neurons (day 30), three concentrations (10 µM, 50 µM, and 100 µM) were used to assess cytotoxicity. MG132 proteasomal inhibitor and sodium arsenite (NaArs) were used to investigate oxidative stress, and 50 µM of metformin was tested for its protective effects against oxidative stress in hiPSC-derived neurons. Metformin treatment did not affect cell viability, neuronal differentiation, or trigger reactive oxygen species (ROS) generation in healthy hiPSC-derived motor neurons. Additionally, mitochondrial membrane potential (MMP) loss was not observed at 50 µM metformin. Metformin effectively protected neurons from stress agents and elevated the expression of antioxidant genes when treated with MG132. However, an interplay between MG132 and metformin resulted in lower expression of Nrf2 and NQO1 compared to the MG132 group alone, indicating reduced JC-1 aggregate levels due to MG132 proteasomal inhibition. Metformin upregulated antioxidant genes in hiPSC-derived neurons under stress conditions and protected the cells from oxidative damage.
Collapse
Affiliation(s)
- Mohammad H Gharandouq
- Faculty of Biological Sciences, The University of Jordan, Amman, Jordan
- Cell Therapy Center, The University of Jordan, Queen Rania Street, Amman, 11942, Jordan
| | - Mohammad A Ismail
- Cell Therapy Center, The University of Jordan, Queen Rania Street, Amman, 11942, Jordan
- South Australian ImmunoGENomics Cancer Institute, Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Tareq Saleh
- Department of Pharmacology and Public Health, Faculty of Medicine, The Hashemite University, Zarqa, 13133, Jordan
- Department of Pharmacology & Therapeutics, College of Medicine & Health Sciences, Arabian Gulf University, Manama, Bahrain
| | - Malik Zihlif
- Department of Pharmaceutical Sciences, School of Medicine, University of Jordan, Amman, Jordan
| | - Nidaa A Ababneh
- Cell Therapy Center, The University of Jordan, Queen Rania Street, Amman, 11942, Jordan.
| |
Collapse
|
18
|
Li L, Dou X, Song X, Wang F. The Current Status and Future Prospects of Intra-articular Injection Therapy for Hip Osteoarthritis: A Review. Curr Pain Headache Rep 2025; 29:64. [PMID: 40100299 PMCID: PMC11919992 DOI: 10.1007/s11916-025-01378-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/05/2025] [Indexed: 03/20/2025]
Abstract
PURPOSE OF REVIEW Hip osteoarthritis constitutes a prevalent condition among individuals aged 55 and above, serving as one of the primary triggers for joint discomfort and impairment, and marking a substantial origin of chronic pain particularly affecting the elderly population. Our article provides an exhaustive summary of the mechanisms of action, therapeutic efficacy, and potential adverse consequences associated with novel therapeutic modalities including glucocorticoids, hyaluronic acid, platelet-rich plasma, mesenchymal stem cells, and stromal vascular fraction. Concurrently, we conducted a comprehensive evaluation of the clinical efficacy and potential applications of various medications. RECENT FINDINGS In comparison to physical therapy, oral analgesics, and other nonsurgical modalities, intra-articular injection therapy is characterized by enhanced safety and greater efficacy. Moreover, when contrasted with surgical intervention, intra-articular injection demonstrates a lower degree of invasiveness and incurs fewer adverse reactions. Intra-articular treatments have shown excellent local efficacy while significantly minimizing adverse reactions in patients. These methods hold significant potential for development but require comprehensive research and thorough discussion within the academic community.
Collapse
Affiliation(s)
- Li Li
- Center for Rehabilitation Medicine, Department of Orthopedics, Zhejiang Provincial People'S Hospital (Affiliated People'S Hospital, Hangzhou Medical College), No. 158 Shangtang Road, Hangzhou, 310014, Zhejiang, China
- Nursing department, Zhejiang Provincial People'S Hospital (Affiliated People'S Hospital, Hangzhou Medical College), No. 158 Shangtang Road, Hangzhou, 310014, Zhejiang, China
| | - Xiaofan Dou
- Center for Rehabilitation Medicine, Department of Orthopedics, Zhejiang Provincial People'S Hospital (Affiliated People'S Hospital, Hangzhou Medical College), No. 158 Shangtang Road, Hangzhou, 310014, Zhejiang, China
- Nursing department, Zhejiang Provincial People'S Hospital (Affiliated People'S Hospital, Hangzhou Medical College), No. 158 Shangtang Road, Hangzhou, 310014, Zhejiang, China
| | - Xueliang Song
- Center for Rehabilitation Medicine, Department of Orthopedics, Zhejiang Provincial People'S Hospital (Affiliated People'S Hospital, Hangzhou Medical College), No. 158 Shangtang Road, Hangzhou, 310014, Zhejiang, China
- Nursing department, Zhejiang Provincial People'S Hospital (Affiliated People'S Hospital, Hangzhou Medical College), No. 158 Shangtang Road, Hangzhou, 310014, Zhejiang, China
| | - Fengxian Wang
- Center for Rehabilitation Medicine, Department of Orthopedics, Zhejiang Provincial People'S Hospital (Affiliated People'S Hospital, Hangzhou Medical College), No. 158 Shangtang Road, Hangzhou, 310014, Zhejiang, China.
- Nursing department, Zhejiang Provincial People'S Hospital (Affiliated People'S Hospital, Hangzhou Medical College), No. 158 Shangtang Road, Hangzhou, 310014, Zhejiang, China.
| |
Collapse
|
19
|
Zhao J, Quan Z, Wang H, Wang J, Xie Y, Li J, Zhang R. Novel strategy for hair regeneration: Exosomes and collagenous sequences of human a1(XVII) chain enhance hair follicle stem cell activity by regulating the hsa-novel-238a-CASP9 axis. Exp Cell Res 2025; 446:114483. [PMID: 40010561 DOI: 10.1016/j.yexcr.2025.114483] [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: 02/09/2025] [Accepted: 02/23/2025] [Indexed: 02/28/2025]
Abstract
The regenerative capacity of hair follicles is fundamentally influenced by the intricate interactions between hair follicle stem cells (HFSCs) and their microenvironment. Our study presents a novel strategy for hair regeneration, highlighting the synergistic relationship between dermal papilla cell-derived exosomes (DPC-Exos) and collagenous sequences of Human a1(XVII) Chain (CS-COL17A1) in modulating HFSC activity via the hsa-novel-238a-CASP9 axis. We characterized DPC-Exos using nanoparticle tracking analysis and transmission electron microscopy and confirmed, their purity with the exosomal markers CD81, CD63, and CD9.A dose-dependent CCK-8 assay showed that both DPC-Exos and CS-COL17A1 significantly improved HFSC viability. Scratch and Transwell assays showed improved HFSC migration after treatment. MiRNA sequencing revealed a significant upregulation of hsa-novel-238a in HFSCs after treatment with DPC-Exos and CS-COL17A1, suggesting its involvement in the regulation of HFSCs activity. A dual-luciferase assay confirmed that hsa-novel-238a directly targets the CASP9 gene, elucidating the underlying molecular mechanisms. The combined application of DPC-Exos and CS-COL17A1 significantly improved HFSC migration and proliferation (p < 0.01), highlighting the importance of the hsa-novel-238a-CASP9 axis. This research provides insights into the regulatory network of exosomes and CS-COL17A1, paving the way for innovative therapeutic approaches to treat hair loss and enhance hair follicle regeneration through modulation of the hsa-novel-238a-CASP9 axis.
Collapse
Affiliation(s)
- Jingyu Zhao
- Department of Dermatology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, China; Department of Dermatology, The Third Affiliated Hospital of Naval Medical University, Shanghai, 200433, China
| | - Zhe Quan
- Department of Dermatology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, China; Department of Dermatology, Shanghai United Family XinCheng Hospital, Shanghai, 200003, China
| | - Huiying Wang
- Department of Dermatology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, China
| | - Jun Wang
- Department of Dermatology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, China
| | - Yong Xie
- Jiangsu Trautec Medical Technology Co.,Ltd.,Changzhou, 213100, China
| | - Jiajia Li
- Jiangsu Trautec Medical Technology Co.,Ltd.,Changzhou, 213100, China
| | - Ruzhi Zhang
- Department of Dermatology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, China; Department of Dermatology, The Second Affiliated Hospital of Wannan Medical College, Wuhu, 241100, China.
| |
Collapse
|
20
|
Su H, Liu L, Yan Z, Guo W, Huang G, Zhuang R, Pan Y. Therapeutic potential of total flavonoids of Rhizoma Drynariae: inhibiting adipogenesis and promoting osteogenesis via MAPK/HIF-1α pathway in primary osteoporosis. J Orthop Surg Res 2025; 20:260. [PMID: 40069718 PMCID: PMC11895304 DOI: 10.1186/s13018-025-05665-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Accepted: 02/27/2025] [Indexed: 03/15/2025] Open
Abstract
AIM This study seeks to confirm the therapeutic effectiveness of TRFD in inhibiting adipogenesis and promoting osteogenesis in primary osteoporosis through the MAPK/HIF-1α signaling pathway. C57BL/6J mice underwent ovariectomy (OVX) to induce osteoporosis. Mice were administered TRFD (Low and high doses)estradiol for a duration of 12 weeks. Bone microarchitecture evaluated using Micro-CT, while serum biomarkers and protein expressions were analyzed through enzyme-linked immunosorbent assay, Western blotting, and immunohistochemistry. Furthermore, BMSC were isolated to show differentiation, Osteogenic and adipogenic induction were performed, including ALP activity and Oil Red O staining. Bioinformatics analysis of RNA sequencing data was conducted to identify differentially expressed genes. RESULTS Total flavonoids of Rhizoma Drynariae treatment significantly improved bone microarchitecture and reversed histopathological damage in OVX mice. It increased serum levels of osteogenesis markers (RUNX2, BMP-2) and enhanced MAPK and HIF-1α signaling pathways, The results also showed a significant dose, TFDR enhanced the osteogenic differentiation of BMSCs while suppressing adipogenic differentiation, as demonstrated by increased ALP activity and mineralization, alongside, the expression of lipid markers (PPAR-γ, C/EBPα) was inhibited. Furthermore, MAPK/HIF-1α pathway was confirmed be crucial in mediating these effects. CONCLUSION TRFD exhibits significant therapeutic potential in treating primary osteoporosis by promoting osteogenesis and inhibiting adipogenesis through the MAPK/HIF-1α pathway. These establish an investigation of TRFD as a natural treatment option for managing osteoporosis. CLINICAL TRIAL NUMBER Not applicable.
Collapse
Affiliation(s)
- Hui Su
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, 310053, China
| | - Luyao Liu
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, 310053, China
| | - Zechen Yan
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, 310053, China
| | - WenXuan Guo
- Orthopedic Department, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, 310053, China
| | - Guangxin Huang
- Department of Pharmacy, Tianjin FirstCentral Hospital, Tianjin, 300000, China
| | - Rujie Zhuang
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, 310053, China.
- Orthopedic Department, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, 310053, China.
- Quzhou TCM Hospital at the Junction of Four Provinces Affiliated to Zhejiang Chinese Medical University, Quzhou, 324000, China.
| | - Yu Pan
- Orthopedic Department, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, 310053, China.
| |
Collapse
|
21
|
Allouh MZ, Rizvi SFA, Alamri A, Jimoh Y, Aouda S, Ouda ZH, Hamad MIK, Perez-Cruet M, Chaudhry GR. Mesenchymal stromal/stem cells from perinatal sources: biological facts, molecular biomarkers, and therapeutic promises. Stem Cell Res Ther 2025; 16:127. [PMID: 40055783 PMCID: PMC11889844 DOI: 10.1186/s13287-025-04254-0] [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: 10/11/2024] [Accepted: 02/25/2025] [Indexed: 05/13/2025] Open
Abstract
The use of mesenchymal stem cells (MSCs) from perinatal tissue sources has gained attention due to their availability and lack of significant ethical or moral concerns. These cells have a higher proliferative capability than adult MSCs and less immunogenic or tumorigenesis risk than fetal and embryonic stem cells. Additionally, they do not require invasive isolation methods like fetal and adult MSCs. We reviewed the main biological and therapeutic aspects of perinatal MSCs in a three-part article. In the first part, we revised the main biological features and characteristics of MSCs and the advantages of perinatal MSCs over other types of SCs. In the second part, we provided a detailed molecular background for the main biomarkers that can be used to identify MSCs. In the final part, we appraised the therapeutic application of perinatal MSCs in four major degenerative disorders: degenerative disc disease, retinal degenerative diseases, ischemic heart disease, and neurodegenerative diseases. In conclusion, there is no single specific molecular marker to identify MSCs. We recommend using at least two positive markers of stemness (CD29, CD73, CD90, or CD105) and two negative markers (CD34, CD45, or CD14) to exclude the hematopoietic origin. Moreover, utilizing perinatal MSCs for managing degenerative diseases presents a promising therapeutic approach. This review emphasizes the significance of employing more specialized progenitor cells that originated from the perinatal MSCs. The review provides scientific evidence from the literature that applying these progenitor cells in therapeutic procedures provides a greater regenerative capacity than the original primitive MSCs. Finally, this review provides a valuable reference for researchers exploring perinatal MSCs and their therapeutic applications.
Collapse
Affiliation(s)
- Mohammed Z Allouh
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, P. O. Box: 15551, Al Ain, UAE.
- OU-WB Institute for Stem Cell and Regenerative Medicine, Oakland University, Rochester, MI, 48309, USA.
| | - Syed Faizan Ali Rizvi
- OU-WB Institute for Stem Cell and Regenerative Medicine, Oakland University, Rochester, MI, 48309, USA
- Department of Biological Sciences, Oakland University, Rochester, MI, 48309, USA
| | - Ali Alamri
- OU-WB Institute for Stem Cell and Regenerative Medicine, Oakland University, Rochester, MI, 48309, USA
- Department of Biological Sciences, Oakland University, Rochester, MI, 48309, USA
| | - Yusuf Jimoh
- OU-WB Institute for Stem Cell and Regenerative Medicine, Oakland University, Rochester, MI, 48309, USA
- Department of Biological Sciences, Oakland University, Rochester, MI, 48309, USA
| | - Salma Aouda
- College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, UAE
| | - Zakaria H Ouda
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, P. O. Box: 15551, Al Ain, UAE
| | - Mohammad I K Hamad
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, P. O. Box: 15551, Al Ain, UAE
| | - Mick Perez-Cruet
- OU-WB Institute for Stem Cell and Regenerative Medicine, Oakland University, Rochester, MI, 48309, USA
- Department of Neurosurgery, Corewell Health, Royal Oak, MI, USA
| | - G Rasul Chaudhry
- OU-WB Institute for Stem Cell and Regenerative Medicine, Oakland University, Rochester, MI, 48309, USA.
- Department of Biological Sciences, Oakland University, Rochester, MI, 48309, USA.
| |
Collapse
|
22
|
Li J, Wu M, He L. Immunomodulatory effects of mesenchymal stem cell therapy in chronic kidney disease: a literature review. BMC Nephrol 2025; 26:107. [PMID: 40033224 PMCID: PMC11874639 DOI: 10.1186/s12882-025-04029-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 02/19/2025] [Indexed: 03/05/2025] Open
Abstract
Chronic kidney disease (CKD) has been a growing public medical concern in recent years which calls for effective interventions. Mesenchymal stem cells (MSCs) have garnered increased interest in past decades due to their potential to repair and regenerate damaged tissues. Many clinical trials have highlighted the safety and effectiveness of kidney disease with this novel cell therapy. MSC infusion can improve renal function indices such as glomerular filtration rate, urine protein, serum creatinine, and blood urea nitrogen, while inhibiting immune response by increasing regulatory T cells. The therapeutic mechanisms may be primarily attributed to a function combined with immunomodulation, anti-inflammation, anti-fibrosis, promoting angiogenesis, anti-oxidation, anti-apoptosis, or tissue healing produced by cell secretsome. However, CKD is a broad concept due to many pathological etiologies including diabetes, hypertension, heart disease, immunological damage, a family history of renal failure, and so on. Furthermore, the therapeutic efficacy of MSCs may be influenced by different cell sources, injection methods, medication dosage, or homing proportion. As a result, it is timely and essential to access recent advancements in the MSC application on CKD.
Collapse
Affiliation(s)
- Jipeng Li
- Department of Nephrology, Xijing Hospital, Air Force Medical University, Xi'an, Shaan Xi, China
| | - Mengting Wu
- Department of Nephrology, Xijing Hospital, Air Force Medical University, Xi'an, Shaan Xi, China
| | - Lijie He
- Department of Nephrology, Xijing Hospital, Air Force Medical University, Xi'an, Shaan Xi, China.
| |
Collapse
|
23
|
Zhu Y, Zhu J, Wang X, Wang P, Liu R. Molecular roles in membrane receptor signaling pathways and cascade reactions in chondrocytes: a review. J Mol Histol 2025; 56:94. [PMID: 39988650 DOI: 10.1007/s10735-025-10368-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 02/03/2025] [Indexed: 02/25/2025]
Abstract
Articular cartilage (AC) is a specialized connective tissue with unique biological and mechanical properties, which depends on the biological effects of each resident chondrocyte and its surrounding extracellular matrix (ECM) to form a unit that operates in a constant and balanced feedback loop. The surface membrane receptors of chondrocytes play a crucial role in the feedback balance of this biological unit. Various biological signals outside chondrocytes, such as water-soluble chemical signal molecules and mechanical signals, are unable to directly enter the cell and must first bind to the plasma membrane receptors to induce changes in the level and activity of intracellular signal transduction molecules. These changes then transmit through signaling cascade pathways into the nucleus, changing the cell phenotype, and producing physiological or pathological changes. Specific chemical and mechanical signals break the feedback balance of cartilage tissue units through membrane receptors. In the ECM environment, the molecular actions of chondrocyte membrane receptors in response to these specific signals, along with associated ion channel receptors, collectively regulate the biological effects of chondrocytes. This leads to decreased chondrocyte survival and an imbalance in ECM regulation, ultimately disrupting the tissue's molecular framework and physiological feedback mechanisms, and resulting in pathological changes in cartilage tissue. To provide insights into addressing the complexities associated with cartilage tissue injury and repair engineering, this review provides a comprehensive overview of the molecular mechanisms and biological implications of chondrocyte membrane receptor-mediated signal transduction, including G protein-coupled receptors (GPCRs), enzyme-linked receptors (tyrosine kinase receptors (TKRs)), and integrin receptors.
Collapse
Affiliation(s)
- Yingkang Zhu
- Department of Orthopedics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Jingjing Zhu
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Qingdao University, Qingdao, 266000, Shandong, China
| | - Xu Wang
- Department of Orthopedics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Pengbo Wang
- Department of Orthopedics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Ruiyu Liu
- Department of Orthopedics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China.
| |
Collapse
|
24
|
Cho YW, Kang MJ, Park JH, Eom YS, Kim TH. Spatially controlled multicellular differentiation of stem cells using triple factor-releasing metal-organic framework-coated nanoline arrays. Nat Commun 2025; 16:1389. [PMID: 39910083 PMCID: PMC11799339 DOI: 10.1038/s41467-025-56373-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Accepted: 01/17/2025] [Indexed: 02/07/2025] Open
Abstract
Improved in vitro models are needed for regenerative therapy and drug screening. Here, we report on functionally aligned nanoparticle-trapped nanopattern arrays for spatially controlled, precise mesenchymal stem cell differentiation on a single substrate. The arrays comprise nanohole and nanoline arrays fabricated through interference lithography and selectively capture of UiO-67 metal-organic frameworks on nanoline arrays with a 99.8% efficiency using an optimised asymmetric spin-coating method. The UiO-67 metal-organic frameworks contain three osteogenic differentiation factors for sustained release over four weeks. The combination of differentiation factors and patterned array allows for generation of adipocytes, osteoblasts, and adipocyte-osteoblast mixtures on nanohole arrays, nanoline arrays, and at the nanohole-nanoline interface, respectively, with mature osteoblasts exhibiting higher marker expression and mineralisation. The sustained release patterned array holds potential for constructing advanced therapeutic and disease state in vitro cellular models.
Collapse
Affiliation(s)
- Yeon-Woo Cho
- Department of Intelligent Precision Healthcare Convergence, Institute for Cross-disciplinary Studies (ICS), Sungkyunkwan University (SKKU), Suwon, Gyeonggi, 16419, Republic of Korea
- School of Integrative Engineering, Chung-Ang University, 84 Heukseuk-ro, Dongjak-gu, Seoul, 06974, Republic of Korea
| | - Min-Ji Kang
- Department of Intelligent Precision Healthcare Convergence, Institute for Cross-disciplinary Studies (ICS), Sungkyunkwan University (SKKU), Suwon, Gyeonggi, 16419, Republic of Korea
| | - Joon-Ha Park
- Department of Intelligent Precision Healthcare Convergence, Institute for Cross-disciplinary Studies (ICS), Sungkyunkwan University (SKKU), Suwon, Gyeonggi, 16419, Republic of Korea
| | - Yun-Sik Eom
- Department of Intelligent Precision Healthcare Convergence, Institute for Cross-disciplinary Studies (ICS), Sungkyunkwan University (SKKU), Suwon, Gyeonggi, 16419, Republic of Korea
- School of Integrative Engineering, Chung-Ang University, 84 Heukseuk-ro, Dongjak-gu, Seoul, 06974, Republic of Korea
| | - Tae-Hyung Kim
- Department of Intelligent Precision Healthcare Convergence, Institute for Cross-disciplinary Studies (ICS), Sungkyunkwan University (SKKU), Suwon, Gyeonggi, 16419, Republic of Korea.
- Department of Biomedical Engineering, ICS, SKKU, Suwon, Gyeonggi, 16419, Republic of Korea.
| |
Collapse
|
25
|
Sheikhi K, Ghaderi S, Firouzi H, Rahimibarghani S, Shabani E, Afkhami H, Yarahmadi A. Recent advances in mesenchymal stem cell therapy for multiple sclerosis: clinical applications and challenges. Front Cell Dev Biol 2025; 13:1517369. [PMID: 39963155 PMCID: PMC11830822 DOI: 10.3389/fcell.2025.1517369] [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: 10/25/2024] [Accepted: 01/09/2025] [Indexed: 02/20/2025] Open
Abstract
Multiple sclerosis (MS), a chronic autoimmune disorder of the central nervous system (CNS), is characterized by inflammation, demyelination, and neurodegeneration, leading to diverse clinical manifestations such as fatigue, sensory impairment, and cognitive dysfunction. Current pharmacological treatments primarily target immune modulation but fail to arrest disease progression or entirely reverse CNS damage. Mesenchymal stem cell (MSC) therapy offers a promising alternative, leveraging its immunomodulatory, neuroprotective, and regenerative capabilities. This review provides an in-depth analysis of MSC mechanisms of action, including immune system regulation, promotion of remyelination, and neuroregeneration. It examines preclinical studies and clinical trials evaluating the efficacy, safety, and limitations of MSC therapy in various MS phenotypes. Special attention is given to challenges such as delivery routes, dosing regimens, and integrating MSCs with conventional therapies. By highlighting advancements and ongoing challenges, this review underscores the potential of MSCs to revolutionize MS treatment, paving the way for personalized and combinatory therapeutic approaches.
Collapse
Affiliation(s)
- Kamran Sheikhi
- Kurdistan University of Medical Sciences, Kurdistan, Iran
| | | | - Hassan Firouzi
- Department of Medical Laboratory, Faculty of Medicine, Sari Branch, Islamic Azad University, Sari, Iran
| | - Sarvenaz Rahimibarghani
- Department of Physical Medicine and Rehabilitation, Tehran University of Medical Sciences, Tehran, Iran
| | - Ehsan Shabani
- Department of Clinical Pharmacy, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamed Afkhami
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran
- Department of Medical Microbiology, Faculty of Medicine, Shahed University, Tehran, Iran
| | - Aref Yarahmadi
- Department of Biology, Khorramabad Branch, Islamic Azad University, Khorramabad, Iran
| |
Collapse
|
26
|
Mohan PP, Deo S, Liu ZJ, Dikici E, Kaneku H, Chang D, Garcia-Buitrago M, Jalaeian H, Zeynaloo E, Ortiz YY, Li Y, Bhatia S, Velazquez O, Daunert S. Liver Regeneration Following Thermal Ablation Using Nanocarrier Mediated Targeted Mesenchymal Stem Cell Therapy. Cardiovasc Intervent Radiol 2025; 48:233-243. [PMID: 39505737 PMCID: PMC11790787 DOI: 10.1007/s00270-024-03862-2] [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: 01/24/2024] [Accepted: 09/07/2024] [Indexed: 11/08/2024]
Abstract
PURPOSE To test the efficacy of nanocarrier (NC) mediated mesenchymal stem cell (MSC) therapy for liver regeneration following thermal ablation of porcine livers. MATERIALS AND METHODS Liver radiofrequency ablation was performed in 18 swines divided into MSC, MSC + NC and control groups. The test groups received infusion of MSC or MSC + NC labeled with enhanced green fluorescent protein (eGFP) via hepatic artery. MSC + NC group had MSCs coated with dendrimer nanocarrier complexed with I-Domain of lymphocyte function-associated antigen-1 (LFA-1). Nanocarriers direct homing of MSCs by binding to its counterpart protein, intercellular adhesion molecule-1 (ICAM-1), which is overexpressed at the periablation margins from inflammation. Ablation cavity reduction by CT volumetry was used as surrogate marker for liver regeneration. Cell proliferation was assessed with Ki67 and HepPar-1 stains. GFP identified MSC derived cells. RESULTS Total number of ablations in control animals were 13 across 4 animals. In the MSC group, there were 23 ablations across 6 animals, and in MSC + NC group there were 21 ablations across 6 animals. Ablation cavity volume reduction from day 0 to 30 were 64.4 ± 15.0%, 61.5 ± 12.9% and 80.3 ± 9.4% for control, MSC and MSC + NC groups, respectively (MSC + NC vs MSC: p < 0.001, MSC + NC vs. control: p = 0.001). GFP+ cell count at margins was 426.8 ± 193.2 for MSC group and 498.6 ± 235.2 for MSC + NC group (p = 0.01). The mean Ki67 and HepPar-1 staining at margins were 9.81 ± 4.5% and 6.12 ± 4.2% for MSC + NC group versus 7.59 ± 3.7% and 5.09 ± 3.7% for MSC group, respectively (P < 0.001 and P = 0.09, respectively). CONCLUSION Nanocarrier-mediated MSC therapy promotes liver regeneration by engrafting MSCs at ablation margins, potentially making liver-directed therapy viable for patients with severe liver dysfunction. This technology may also benefit other solid organs.
Collapse
Affiliation(s)
- Prasoon P Mohan
- Department of Interventional Radiology, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Interventional Radiology, UMHC-SCC, 1475 NW 12th Ave., Miami, FL, 33136, USA
| | - Sapna Deo
- Department of Biochemistry, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Interventional Radiology, UMHC-SCC, 1475 NW 12th Ave., Miami, FL, 33136, USA
| | - Zhao-Jun Liu
- Department of Biochemistry, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Interventional Radiology, UMHC-SCC, 1475 NW 12th Ave., Miami, FL, 33136, USA
| | - Emre Dikici
- Department of Biochemistry, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Interventional Radiology, UMHC-SCC, 1475 NW 12th Ave., Miami, FL, 33136, USA
| | - Hugo Kaneku
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Doyoung Chang
- Department of Interventional Radiology, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Interventional Radiology, UMHC-SCC, 1475 NW 12th Ave., Miami, FL, 33136, USA
| | - Monica Garcia-Buitrago
- Department of Interventional Radiology, UMHC-SCC, 1475 NW 12th Ave., Miami, FL, 33136, USA
| | - Hamed Jalaeian
- Department of Interventional Radiology, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Interventional Radiology, UMHC-SCC, 1475 NW 12th Ave., Miami, FL, 33136, USA
| | - Elnaz Zeynaloo
- Department of Biochemistry, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Interventional Radiology, UMHC-SCC, 1475 NW 12th Ave., Miami, FL, 33136, USA
| | - Yulexi Y Ortiz
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Interventional Radiology, UMHC-SCC, 1475 NW 12th Ave., Miami, FL, 33136, USA
| | - Yan Li
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Interventional Radiology, UMHC-SCC, 1475 NW 12th Ave., Miami, FL, 33136, USA
| | - Shivank Bhatia
- Department of Interventional Radiology, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Interventional Radiology, UMHC-SCC, 1475 NW 12th Ave., Miami, FL, 33136, USA
| | - Omaida Velazquez
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Interventional Radiology, UMHC-SCC, 1475 NW 12th Ave., Miami, FL, 33136, USA
| | - Sylvia Daunert
- Department of Biochemistry, University of Miami Miller School of Medicine, Miami, FL, USA.
- Department of Interventional Radiology, UMHC-SCC, 1475 NW 12th Ave., Miami, FL, 33136, USA.
| |
Collapse
|
27
|
Ozkan S, Isildar B, Koyuturk M. Comparative analysis of the effects of different hypoxia mimetic agents on the secretome contents of conditioned medium obtained from mesenchymal stem/stromal cells cultured in 2 or 3-dimensional cell culture systems. Cytotechnology 2025; 77:11. [PMID: 39654545 PMCID: PMC11625095 DOI: 10.1007/s10616-024-00659-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Accepted: 10/28/2024] [Indexed: 12/12/2024] Open
Abstract
Paracrine factors secreted by mesenchymal stem/stromal cells (MSCs) have been demonstrated to have significant therapeutic potential. The secretome profiles of MSCs variate depending on culture conditions. Generally, the effects of a single preconditioning strategy on secretome profiles of MSCs were investigated. However, until now, there has been no study examining the combinatory effects of different preconditioning strategies in a comparative manner. This study aimed to evaluate the secretome contents of conditioned media obtained from human umbilical cord-derived MSCs cultured in 2- or 3-dimensional (D) culture systems preconditioned with deferoxamine (DFS) or dimethyloxalylglycine (DMOG). Immunocytochemical analysis showed that MSCs preconditioned with DFS or DMOG have increased nuclear hypoxia-inducible factor-1α expression. Transmission electron microscopic analysis showed that cells preconditioned with DFS or DMOG have increased autophagic vesicles, which could be attributed to altered energy metabolism under hypoxic conditions. It was revealed that hypoxia-mimetic agents added to the 2D-, or 3D-culture environment raised total protein concentrations per cell along with vascular endothelial growth factor. The concentrations of glial cell-derived neurotrophic factor (GDNF) and nerve growth factor (NGF) were differentially regulated in 2D-, and 3D-culture system, that the secretions of GDNF and NGF per cell were more prominent in 3D- and 2D-culture systems, respectively. These findings indicate that hypoxic conditions alone significantly elevate total protein concentrations, while the contribution of the 3D environment is more modest than initially anticipated. However, concentrations of secreted growth factors may be affected differently depending on the topography of the culture condition and the types of hypoxia mimetic agents.
Collapse
Affiliation(s)
- Serbay Ozkan
- Faculty of Medicine, Histology and Embryology Department, Izmir Katip Çelebi University, Izmir, Turkey
| | - Basak Isildar
- Faculty of Medicine, Histology and Embryology Department, Balıkesir University, Balikesir, Turkey
| | - Meral Koyuturk
- Cerrahpaşa Faculty of Medicine, Histology and Embryology Department, Istanbul University-Cerrahpaşa, Istanbul, Turkey
| |
Collapse
|
28
|
Veret D, Tejedor G, Perez E, Chomette A, Farno M, Ferreira-Lopez R, Dagneaux L, Pers YM, Jorgsensen C, Gondeau C, Brondello JM. Combination of rapamycin and adipose-derived mesenchymal stromal cells enhances therapeutic potential for osteoarthritis. Stem Cell Res Ther 2025; 16:9. [PMID: 39815291 PMCID: PMC11737215 DOI: 10.1186/s13287-024-04090-8] [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: 07/05/2024] [Accepted: 12/03/2024] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND The regenerative potential of mesenchymal stromal/stem cells (MSCs) has been extensively studied in clinical trials in the past decade. However, despite the promising regenerative properties documented in preclinical studies, for instance in osteoarthritis (OA), the therapeutic translation of these results in patients has not been fully conclusive. One factor contributing to this therapeutic barrier could be the presence of senescent cells in OA joints. METHODS This study evaluated a novel approach to OA treatment by combining adipose tissue-derived MSCs (AD-MSCs) with rapamycin, a clinically approved immunosuppressive drug with anti-senescence properties. First, rapamycin effects on senescence and fibrosis markers were investigated in freshly isolated OA chondrocytes by immunostaining. Next, the in vitro differentiation capacities of AD-MSCs, their regulatory immune functions on activated immune cells and their regenerative effects on OA chondrocyte signature were assessed in the presence of rapamycin. RESULTS In OA chondrocytes, rapamycin reduced the senescence marker p15INK4B and the fibrosis marker COL1A1 without affecting the expression of the master chondrogenic markers SOX9 and COL2. Rapamycin also enhanced AD-MSC differentiation into chondrocytes and reduced their differentiation into adipocytes. In addition, rapamycin improved AD-MSC immunoregulatory functions by promoting the expression of immunosuppressive factors, such as IDO1, PTGS2 and also CD274 (encoding PD-L1). Finally, RNA sequencing analysis showed that in the presence of rapamycin, AD-MSCs displayed improved chondroprotective regenerative effects on co-cultured OA chondrocytes. CONCLUSIONS Our findings suggest that the rapamycin and AD-MSC combination enhances the therapeutic efficacy of these cells in senescence-driven degenerative diseases such as OA, notably by improving their anti-fibrotic and anti-inflammatory properties.
Collapse
Affiliation(s)
- Damien Veret
- IRMB, Univ Montpellier, INSERM, CHU St Eloi, 80 AV A Fliche, 34295-Cedex-05, Montpellier, France
- MedXCell, IRMB, CHU St Eloi, Cyborg, Montpellier, France
| | | | - Esther Perez
- MedXCell, IRMB, CHU St Eloi, Cyborg, Montpellier, France
| | | | - Maylis Farno
- MedXCell, IRMB, CHU St Eloi, Cyborg, Montpellier, France
| | - Rosanna Ferreira-Lopez
- IRMB, Univ Montpellier, INSERM, CHU St Eloi, 80 AV A Fliche, 34295-Cedex-05, Montpellier, France
- Clinical Immunology and Osteoarticular Diseases Therapeutic Unit, Hôpital Lapeyronie, Montpellier, France
- Rheumatology department, Regional Narbonne Hospital, Narbonne, France
| | - Louis Dagneaux
- Hôpital Lapeyronie, Orthopedic Service, Montpellier, France
| | - Yves-Marie Pers
- IRMB, Univ Montpellier, INSERM, CHU St Eloi, 80 AV A Fliche, 34295-Cedex-05, Montpellier, France
- Clinical Immunology and Osteoarticular Diseases Therapeutic Unit, Hôpital Lapeyronie, Montpellier, France
| | - Christian Jorgsensen
- IRMB, Univ Montpellier, INSERM, CHU St Eloi, 80 AV A Fliche, 34295-Cedex-05, Montpellier, France
- Clinical Immunology and Osteoarticular Diseases Therapeutic Unit, Hôpital Lapeyronie, Montpellier, France
| | - Claire Gondeau
- MedXCell, IRMB, CHU St Eloi, Cyborg, Montpellier, France
| | - Jean-Marc Brondello
- IRMB, Univ Montpellier, INSERM, CHU St Eloi, 80 AV A Fliche, 34295-Cedex-05, Montpellier, France.
| |
Collapse
|
29
|
Zhou L, Cai W, Zhang Y, Zhong W, He P, Ren J, Gao X. Therapeutic effect of mesenchymal stem cell-derived exosome therapy for periodontal regeneration: a systematic review and meta-analysis of preclinical trials. J Orthop Surg Res 2025; 20:27. [PMID: 39780243 PMCID: PMC11715287 DOI: 10.1186/s13018-024-05403-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 12/21/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND To assess the therapeutic effects of mesenchymal stem cell (MSC)-derived exosome therapy on periodontal regeneration and identify treatment factors associated with enhanced periodontal regeneration in recent preclinical studies. METHODS Searches were conducted in PubMed, Cochrane Library, EMBASE, and Web of Science databases until October 10, 2024. A risk of bias (ROB) assessment was performed using the SYRCLE tool. Osteogenic-related parameters were used as the primary outcome measures. RESULTS In total, 1360 articles were identified, of which 17 preclinical studies were based on MSC-derived exosome therapy, and they demonstrated a beneficial effect on BV/TV (SMD = 13.99; 95% Cl = 10.50, 17.48; p < 0.00001), CEJ-ABC (SMD = -0.22; 95% Cl = -0.31, -0.13; p < 0.00001), BMD (SMD = 0.29; 95% Cl = 0.14, 0.45; p = 0.0002), and Tp.Sp (SMD = -0.08; 95% Cl= -0.15, -0.02; p = 0.02) compared with the control group. However, no significant differences were observed in Tp.Th (SMD = 0.03; 95% CI = 0.00, 0.07; p = 0.09) between the exosome-treated group and control group. Additionally, subgroup analysis indicated that preconditioned exosomes (p = 0.03) significantly improved BV/TV. In contrast, there were no significant differences in the enhancement of BV/TV with respect to the application method (p = 0.29), application frequency (p = 0.10), treatment duration (p = 0.15), or source of MSCs (p = 0.31). CONCLUSIONS MSC-derived exosomes show great promise for enhancing the quality of periodontal regeneration. However, more standardized and robust trials are needed to reduce heterogeneity and bias across studies and to confirm the therapeutic parameters associated with the enhancement of periodontal regeneration by MSC-derived exosomes. REGISTRATION CRD42024546236.
Collapse
Affiliation(s)
- Liping Zhou
- College of Stomatology, Chongqing Medical University, Chongqing, 401147 , China
- Chongqing Key Laboratory of Oral Diseases , Chongqing, 401147, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147 , China
| | - Wenjia Cai
- College of Stomatology, Chongqing Medical University, Chongqing, 401147 , China
- Chongqing Key Laboratory of Oral Diseases , Chongqing, 401147, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147 , China
| | - Yuhan Zhang
- College of Stomatology, Chongqing Medical University, Chongqing, 401147 , China
- Chongqing Key Laboratory of Oral Diseases , Chongqing, 401147, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147 , China
| | - Wenjie Zhong
- College of Stomatology, Chongqing Medical University, Chongqing, 401147 , China
- Chongqing Key Laboratory of Oral Diseases , Chongqing, 401147, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147 , China
| | - Ping He
- Department of Stomatology, Dazhou Central Hospital, Dazhou, 635000, China.
| | - Jingsong Ren
- Department of Stomatology, Dazhou Central Hospital, Dazhou, 635000, China.
| | - Xiang Gao
- College of Stomatology, Chongqing Medical University, Chongqing, 401147 , China.
- Chongqing Key Laboratory of Oral Diseases , Chongqing, 401147, China.
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147 , China.
| |
Collapse
|
30
|
Lv Y, Hua Z, Lu X. Protective effects and possible mechanisms of mesenchymal stem cells and mesenchymal stem cell-derived extracellular vesicles against kidney fibrosis in animal models: a systematic review and meta-analysis. Front Pharmacol 2025; 15:1511525. [PMID: 39830341 PMCID: PMC11739157 DOI: 10.3389/fphar.2024.1511525] [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: 10/15/2024] [Accepted: 12/05/2024] [Indexed: 01/22/2025] Open
Abstract
Introduction The risk of kidney fibrosis is significantly elevated in individuals with diabetes, chronic nephritis, trauma, and other underlying conditions. Concurrently, human umbilical cord blood-derived mesenchymal stem cells (hUCB-MSCs) and their extracellular vesicles (MSC-Exos) have gained prominence in regenerative medicine. In light of these observations, we are undertaking a meta-analysis to elucidate the influence of hUCB-MSCs and MSC-Exos on kidney fibrosis. Methods To identify eligible trials, we conducted a comprehensive search of the CNKI, PubMed, Web of Science and Wanfang databases from inception to 24 October 2022. Furthermore, the methodological quality of the included studies was evaluated using the Systematic Review Center for Laboratory Animal Experimentation (SYRCLE) risk-of-bias tool. Besides, the weighted standard mean difference (SMD) with a 95% confidence interval (CI) was calculated using the Review Manager 5.4 software. The Stata (12.0) software was employed to assess the impact of factors on outcome heterogeneity and publication bias in the study. A total of 645 related research studies were retrieved, of which 14 that involved 219 experimental animals were included in the study. Results In comparison to the control treatment, treatment with Human UCB MSC and MSC-Exos was observed to significantly enhance renal function in animal models of kidney fibrosis. This was evidenced by a reduction in serum creatinine (Scr) levels (p < 0.00001) and blood urea nitrogen (BUN) levels (p < 0.00001), as well as reduction of CD68+ macrophages (p < 0.00001), TdT-mediated dUTP Nick-End labeling (TUNEL)+ tubular cells(p < 0.00001), α-SMA levels (p = 0.0009) and TGF-β1 (p < 0.00001). P < 0.05 is deemed to indicate a statistically significant difference. Alpha-smooth muscle actin (α-SMA) is a specific protein that is normally expressed in myofibroblasts. The term "CD68+ macrophages" refers to macrophages that express the CD68 protein on their cell surface. Both macrophages and myofibroblasts have been linked to the development of kidney fibrosis. In this study, the quantity of CD68+ macrophages and α-SMA was employed as a means of gauging the extent of renal fibrosis. Additionally, transforming growth factor beta 1 (TGF-β1) is a significant cytokine implicated in the pathogenesis of kidney fibrosis. TUNEL-positive tubular cells represent tubular cells undergoing apoptosis. It is hypothesized that this may result in a reduction of tubular apoptosis and a delay in kidney fibrosis, due to the inhibition of the transformation of macrophages into myofibroblasts (MMT) and the disruption of the kidney fibrogenic niche. Conclusion The principal findings of this preclinical systematic review indicate that hUCB MSC and MSC-Exos have a substantial protective impact against kidney fibrosis. Kidney transfer remains the final option for traditional renal fibrosis treatment. The lack of donors and high cost make it challenging for many patients to access appropriate treatment. Although this study still suffers from three shortcomings: sample size, methodological consistency and translational challenges, the hUCB MSC and MSC-Exos have been demonstrated to reduce tubular apoptosis and inhibit fibrotic progression. The hUCB MSC and MSC-Exos offer a promising alternative due to their lower price and accessibility. Nevertheless, further high-quality studies are required in the future to address the methodological limitations identified in this review. Systematic Review Registration Identifier INPLASY2022100104.
Collapse
Affiliation(s)
- Yuanchen Lv
- 1st Clinical Department, China Medical University, Shenyang, Liaoning, China
| | - Zibo Hua
- 1st Clinical Department, China Medical University, Shenyang, Liaoning, China
| | - Xiaomei Lu
- Department of Pathophysiology, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
31
|
Zhao DZ, Yang RL, Wei HX, Yang K, Yang YB, Wang NX, Zhang Q, Chen F, Zhang T. Advances in the research of immunomodulatory mechanism of mesenchymal stromal/stem cells on periodontal tissue regeneration. Front Immunol 2025; 15:1449411. [PMID: 39830512 PMCID: PMC11739081 DOI: 10.3389/fimmu.2024.1449411] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Accepted: 12/13/2024] [Indexed: 01/22/2025] Open
Abstract
Periodontal disease is a highly prevalent disease worldwide that seriously affects people's oral health, including gingivitis and periodontitis. Although the current treatment of periodontal disease can achieve good control of inflammation, it is difficult to regenerate the periodontal supporting tissues to achieve a satisfactory therapeutic effect. In recent years, due to the good tissue regeneration ability, the research on Mesenchymal stromal/stem cells (MSCs) and MSC-derived exosomes has been gradually deepened, especially its ability to interact with the microenvironment of the body in the complex immunoregulatory network, which has led to many new perspectives on the therapeutic strategies for many diseases. This paper systematically reviews the immunomodulatory (including bone immunomodulation) properties of MSCs and their role in the periodontal inflammatory microenvironment, summarizes the pathways and mechanisms by which MSCs and MSC-EVs have promoted periodontal regeneration in recent years, lists potential areas for future research, and describes the issues that should be considered in future basic research and the direction of development of "cell-free therapies" for periodontal regeneration.
Collapse
Affiliation(s)
- De-Zhi Zhao
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Rui-Lin Yang
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Han-Xiao Wei
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Kang Yang
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Yi-Bing Yang
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Nuo-Xin Wang
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Qian Zhang
- Department of Human Anatomy, Zunyi Medical University, Zunyi, Guizhou, China
| | - Fang Chen
- Department of Prosthetics, Affiliated Stomatology Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Tao Zhang
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| |
Collapse
|
32
|
Li J, Guo Z, Wu Z, Wang Y, Wang Z, Guo M, Zhang P. Highly precise strategy of polygalacturonic acid microcarriers functionalized with zwitterions and specific peptides for MSC screening. Carbohydr Polym 2024; 345:122564. [PMID: 39227103 DOI: 10.1016/j.carbpol.2024.122564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 07/17/2024] [Accepted: 07/30/2024] [Indexed: 09/05/2024]
Abstract
Microcarriers for large-scale cell culture have a broader prospect in cell screening compared with the traditional high cost, low efficiency, and cell damaging methods. However, the equal biological affinity to cells has hindered its application. Therefore, based on the antifouling strategy of zwitterionic polymer, we developed a cell-specific microcarrier (CSMC) for shielding non-target cells and capturing mesenchymal stem cells (MSCs), which has characteristics of high biocompatibility, low background noise and high precision. Briefly, [2-(methacryloyloxy) ethyl] dimethyl-(3-sulfopropyl) ammonium hydroxide and glycidyl methacrylate were grafted onto polygalacturonic acid, respectively. The former built a hydration layer through solvation to provide an excellent antifouling surface, while the latter provided active sites for the click reaction with sulfhydryl-modified cell-specific peptides, resulting in rapid immobilization of peptides. This method is applicable to the vast majority of polysaccharide materials. The accurate capture ratio of MSCs by CSMC in a mixed multicellular environment is >95 % and the proliferation rate of MSCs on microcarriers is satisfactory. In summary, this grafting strategy of bioactive components lays a foundation for the application of polysaccharide materials in the biomedical field, and the specific adhesive microcarriers also open up new ideas for the development of stem cell screening as well.
Collapse
Affiliation(s)
- Jianchao Li
- Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China; School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
| | - Ziyuan Guo
- Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China; School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
| | - Zhenxv Wu
- Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China; School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
| | - Yu Wang
- Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China; School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
| | - Zongliang Wang
- Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China; School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
| | - Min Guo
- Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China; School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
| | - Peibiao Zhang
- Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China; School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China.
| |
Collapse
|
33
|
Moñivas Gallego E, Zurita Castillo M. Mesenchymal stem cell therapy in ischemic stroke trials. A systematic review. Regen Ther 2024; 27:301-306. [PMID: 38633415 PMCID: PMC11021793 DOI: 10.1016/j.reth.2024.03.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 03/11/2024] [Accepted: 03/24/2024] [Indexed: 04/19/2024] Open
Abstract
Cerebrovascular accidents, also known as strokes, are the leading cause of permanent disability in society, presenting significant socioeconomic and healthcare costs. They can be caused by ischemic factors or hemorrhages, with ischemic strokes being the most common among the population. Therapies for patients suffering from this condition are limited and primarily focus on acute-phase treatment. In recent years, there has been an increase in cellular therapies, employing Stem Cells to mitigate or eliminate the consequences arising from this disease. Mesenchymal Stem Cells (MSCs) hold substantial therapeutic potential in Nervous System pathologies due to their low antigenicity and capacity to differentiate into various human tissues, such as adipogenic, chondrogenic, and osteogenic tissues. This study conducts a literature review using the "clinical trials" and "Pubmed" database, summarizing all ongoing clinical trials for ischemic strokes that utilize MSCs as treatment.
Collapse
Affiliation(s)
- Ester Moñivas Gallego
- Fundación para la Investigación Biomédica del Hospital Universitario Puerta de Hierro Majadahonda, Spain
| | | |
Collapse
|
34
|
Song L, Yang C, Ji G, Hu R. The role and potential treatment of macrophages in patients with infertility and endometriosis. J Reprod Immunol 2024; 166:104384. [PMID: 39442472 DOI: 10.1016/j.jri.2024.104384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 10/04/2024] [Accepted: 10/12/2024] [Indexed: 10/25/2024]
Abstract
Endometriosis is characterized as a macrophage-related ailment due to its strong link with immune dysfunction. Understanding the status of macrophage polarization in the context of endometriosis-related infertility is crucial for advancing diagnostic and therapeutic strategies. Our comprehensive review delves into the foundational understanding of macrophages and their profound influence on both endometriosis and infertility. Additionally, we illuminate the complex role of macrophages in infertility and endometriosis specifically. Finally, we focused on four critical dimensions: follicular fluid, the intraperitoneal environment, endometrial receptivity, and strategies for managing endometriosis. It is clear that throughout the progression of endometriosis, the diverse polarization states of macrophages play a pivotal role in the internal reproductive environment of infertile individuals grappling with this condition. Modulating macrophage polarization in the reproductive environment of endometriosis patients could address infertility challenges more effectively, offering a promising pathway for treating infertility associated with endometriosis.
Collapse
Affiliation(s)
- Linlin Song
- Department of Gynecology, The General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, China; Reproductive Medicine Center, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, China; Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, China
| | - Caihong Yang
- Department of Gynecology, The General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, China; Reproductive Medicine Center, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, China
| | - Guiyi Ji
- Reproductive Medicine Center, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, China; Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, China; Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan, Ningxia, 750004, China
| | - Rong Hu
- Reproductive Medicine Center, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, China; Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, China; Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan, Ningxia, 750004, China.
| |
Collapse
|
35
|
Ahmadi Somaghian S, Pajouhi N, Dezfoulian O, Pirnia A, Kaeidi A, Rasoulian B. The protective effects of hyperoxic pre-treatment in human-derived adipose tissue mesenchymal stem cells against in vitro oxidative stress and a rat model of renal ischaemia-reperfusion. Arch Physiol Biochem 2024; 130:606-615. [PMID: 37506037 DOI: 10.1080/13813455.2023.2238918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 06/23/2023] [Accepted: 07/11/2023] [Indexed: 07/30/2023]
Abstract
OBJECTIVE Improvement of cell survival is essential for achieving better clinical outcomes in stem cell therapy. We investigated the effects of hyperoxic pre-treatment (HP) on the viability of human adipose stromal stem cells (ASCs). MATERIALS AND METHODS MTT and Western blot tests were used to assess cell viability and the expression of apoptosis-related proteins, respectively. For the in-vivo trial, the rats were subjected to renal ischaemia-reperfusion (IR). RESULTS The results showed that HP could significantly increase the viability of ASCs and decrease apoptotic markers (Bax/BCL-2 ratio and Caspase-3) compared with control cells. There were some additional effects with regard to the improvement of renal structure and function in the animal model. However, the difference between the treated and non-treated transplanted ASCs failed to reach significance. CONCLUSION These results suggested that HP could increase the survival of ASCs against oxidative stress-induced damages in the in-vitro condition, but this strategy was not highly effective in renal IR.
Collapse
Affiliation(s)
- Shahram Ahmadi Somaghian
- Razi Herbal Medicines Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Naser Pajouhi
- Razi Herbal Medicines Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Omid Dezfoulian
- Department of Pathobiology, School of Veterinary Medicine, Lorestan University, Khorramabad, Iran
| | - Afshin Pirnia
- Razi Herbal Medicines Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Ayat Kaeidi
- Physiology-Pharmacology Research Center, Research Institute of Basic Medical Science, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Bahram Rasoulian
- Nutritional Health Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
| |
Collapse
|
36
|
Kahts M, Mellet J, Durandt C, Moodley K, Summers B, Ebenhan T, Zeevaart JR, Aras O, Pepper MS. A proof-of-concept study to investigate the radiolabelling of human mesenchymal and hematopoietic stem cells with [ 89Zr]Zr-Df-Bz-NCS. EJNMMI Radiopharm Chem 2024; 9:82. [PMID: 39611856 PMCID: PMC11607195 DOI: 10.1186/s41181-024-00311-w] [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: 10/12/2024] [Accepted: 11/15/2024] [Indexed: 11/30/2024] Open
Abstract
BACKGROUND The transplantation of hematopoietic stem and progenitor cells (HSPCs) or mesenchymal stromal/stem cells (MSCs) for the treatment of a wide variety of diseases has been studied extensively. A challenge with cell-based therapies is that migration to and retention at the target site is often difficult to monitor and quantify. Zirconium-89 (89Zr) is a positron-emitting radionuclide with a half-life of 3.3 days, which allows long-term cell tracking. Para-isothiocyanatobenzyl-desferrioxamine B (Df-Bz-NCS) is the chelating agent of choice for 89Zr-cell surface labelling. We utilised a shortened labelling method, thereby avoiding a 30-60-min incubation step for [89Zr]Zr-Df-Bz-NCS chelation, to radiolabel HSPCs and MSCs with zirconium-89. RESULTS Three 89Zr-MSC labelling attempts were performed. High labelling efficiencies (81.30 and 87.30%) and relatively good labelling yields (59.59 and 67.00%) were achieved with the use of a relatively larger number of MSCs (4.425 and 3.855 million, respectively). There was no significant decrease in MSC viability after 89Zr-labeling (p = 0.31). This labelling method was also translatable to prepare 89Zr-HSPC; preliminary data from one preparation indicated high 89Zr-HSPC labelling efficiency (88.20%) and labelling yield (71.06%), as well as good HSPC viability after labelling (68.65%). CONCLUSIONS Successful 89Zr-MSC and 89Zr-HSPC labelling was achieved, which underlines the prospects for in vivo cell tracking studies with positron emission tomography. In vitro investigations with larger sample sizes and preclinical studies are recommended.
Collapse
Affiliation(s)
- Maryke Kahts
- Pharmaceutical Sciences Department, School of Pharmacy, Sefako Makgatho Health Sciences University (SMU), Ga-Rankuwa, 0208, South Africa.
| | - Juanita Mellet
- Department of Medical Immunology, Institute for Cellular and Molecular Medicine, South African Medical Research Council (SAMRC) Extramural Unit for Stem Cell Research and Therapy, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Chrisna Durandt
- Department of Medical Immunology, Institute for Cellular and Molecular Medicine, South African Medical Research Council (SAMRC) Extramural Unit for Stem Cell Research and Therapy, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Kinosha Moodley
- Department of Medical Immunology, Institute for Cellular and Molecular Medicine, South African Medical Research Council (SAMRC) Extramural Unit for Stem Cell Research and Therapy, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Beverley Summers
- Pharmaceutical Sciences Department, School of Pharmacy, Sefako Makgatho Health Sciences University (SMU), Ga-Rankuwa, 0208, South Africa
| | - Thomas Ebenhan
- Radiochemistry, South African Nuclear Energy Corporation, Pelindaba, Hartebeespoort, South Africa
| | - Jan Rijn Zeevaart
- Radiochemistry, South African Nuclear Energy Corporation, Pelindaba, Hartebeespoort, South Africa
- Nuclear Medicine Research Infrastructure (NuMeRI) NPC, Pretoria, South Africa
- DST/NWU, Preclinical Drug Development Platform, North-West University, Potchefstroom, South Africa
| | - Omer Aras
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Radiology Department, AMRIC Health, New York, NY, USA
| | - Michael S Pepper
- Department of Medical Immunology, Institute for Cellular and Molecular Medicine, South African Medical Research Council (SAMRC) Extramural Unit for Stem Cell Research and Therapy, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| |
Collapse
|
37
|
Danev N, Poggi JM, Dewever EA, Bartlett AP, Oliveira L, Huntimer L, Harman RM, Van de Walle GR. Immortalized mammosphere-derived epithelial cells retain a bioactive secretome with antimicrobial, regenerative, and immunomodulatory properties. Stem Cell Res Ther 2024; 15:429. [PMID: 39543714 PMCID: PMC11566417 DOI: 10.1186/s13287-024-04019-1] [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: 08/08/2024] [Accepted: 10/25/2024] [Indexed: 11/17/2024] Open
Abstract
BACKGROUND The secretome of primary bovine mammosphere-derived epithelial cells (MDECs) has been shown to exert antimicrobial, regenerative, and immunomodulatory properties in vitro, which warrants its study as a potential biologic treatment with the potential to be translated to human medicine. Currently, the use of the MDEC secretome as a therapy is constrained by the limited life span of primary cell cultures and the decrease of secretome potency over cell passages. METHODS To address these limitations, early-passage bovine MDECs were immortalized using hTERT, a human telomerase reverse transcriptase. The primary and immortal MDECs were compared morphologically, transcriptomically, and phenotypically. The functional properties and proteomic profiles of the secretome of both cell lines were evaluated and compared. All experiments were performed with both low and high passage cell cultures. RESULTS We confirmed through in vitro experiments that the secretome of immortalized MDECs, unlike that of primary cells, maintained antimicrobial and pro-migratory properties over passages, while pro-angiogenic effects of the secretome from both primary and immortalized MDECs were lost when the cells reached high passage. The secretome from primary and immortalized MDECs, at low and high passages exerted immunomodulatory effects on neutrophils in vitro. CONCLUSIONS High passage immortalized MDECs retain a bioactive secretome with antimicrobial, regenerative, and immunomodulatory properties, suggesting they may serve as a consistent cell source for therapeutic use.
Collapse
Affiliation(s)
- Nikola Danev
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, 235 Hungerford Hill Road, Ithaca, NY, 14853, USA
| | - Julia M Poggi
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, 235 Hungerford Hill Road, Ithaca, NY, 14853, USA
| | - Emilie A Dewever
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, 235 Hungerford Hill Road, Ithaca, NY, 14853, USA
| | - Arianna P Bartlett
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, 235 Hungerford Hill Road, Ithaca, NY, 14853, USA
| | - Leane Oliveira
- Elanco Animal Health, 2500 Innovation Way, Indianapolis, IN, 46241, USA
| | - Lucas Huntimer
- Elanco Animal Health, 2500 Innovation Way, Indianapolis, IN, 46241, USA
| | - Rebecca M Harman
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, 235 Hungerford Hill Road, Ithaca, NY, 14853, USA
| | - Gerlinde R Van de Walle
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, 235 Hungerford Hill Road, Ithaca, NY, 14853, USA.
- Department of Veterinary Pathobiology, Royal (Dick) School of Veterinary Studies and Roslin Institute, University of Edinburgh, Easter Bush, Roslin, Midlothian, UK.
| |
Collapse
|
38
|
Xu Q, Hou W, Zhao B, Fan P, Wang S, Wang L, Gao J. Mesenchymal stem cells lineage and their role in disease development. Mol Med 2024; 30:207. [PMID: 39523306 PMCID: PMC11552129 DOI: 10.1186/s10020-024-00967-9] [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: 04/09/2024] [Accepted: 10/18/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) are widely dispersed in vivo and are isolated from several tissues, including bone marrow, heart, body fluids, skin, and perinatal tissues. Bone marrow MSCs have a multidirectional differentiation potential, which can be induced to differentiate the medium in a specific direction or by adding specific regulatory factors. MSCs repair damaged tissues through lineage differentiation, and the ex vivo transplantation of bone marrow MSCs can heal injured sites. MSCs have different propensities for lineage differentiation and pathological evolution for different diseases, which are crucial in disease progression. In this study, we describe various lineage analysis methods to explore lineage ontology in vitro and in vivo, elucidate the impact of MSC lineage differentiation on diseases, advance our understanding of the role of MSC differentiation in physiological and pathological states, and explore new targets and ideas associated with disease diagnosis and treatment.
Collapse
Affiliation(s)
- Qi Xu
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Wenrun Hou
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Baorui Zhao
- Stem cell Translational laboratory, Shanxi Technological Innovation Center for Clinical Diagnosis and Treatment of Immune and Rheumatic Diseases, Shanxi Bethune Hospital, Tongji Shanxi Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China
| | - Peixin Fan
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Sheng Wang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Lei Wang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Jinfang Gao
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China.
| |
Collapse
|
39
|
Brookshaw T, Fuller B, Erro E, Islam T, Chandel S, Zotova E, Selden C. Cryobiological aspects of upscaling cryopreservation for encapsulated liver cell therapies. Cryobiology 2024; 117:105155. [PMID: 39461406 DOI: 10.1016/j.cryobiol.2024.105155] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 09/26/2024] [Accepted: 10/23/2024] [Indexed: 10/29/2024]
Abstract
For the efficient delivery of a cell therapy a treatment must be provided rapidly, at clinical scale, contain a sufficient active cellular component (biomass), and adhere to a multitude of regulatory requirements. Cryopreservation permits many of these demands to be met more readily. Here we present the cryopreservation and recovery of large volume (2.5L) alginate encapsulated liver cell spheroids (AELS), suitable for use with a novel bioartificial liver device (HepatiCan™) for the treatment of those suffering from acute liver failure (ALF), in regulatory approved cryobags and a cryopreservation process optimised for large volumes. By first assessing the thermal profiles of large scale cryobags with a thermal mimic, the feasibility of cryopreserving a full patient dose simultaneously (3x cryobags containing 833 ml biomass each) was investigated, allowing for small and subsequently large-scale testing of cellular functional recoveries. Work presented here demonstrates that optimised reproducible cooling and warming profiles could be achieved with these large volumes, leading to high biomass recoveries at full clinical scale. The recovered AELS also had high regeneration potential, achieving full pre-freeze viable cell densities within 3 days, indicating that the cell therapy could be delivered rapidly to patients with ALF. This study has presented the feasibility for rapid delivery of large volume cell therapies, whilst further research into improved speed of post-thaw recovery is warranted.
Collapse
Affiliation(s)
- Tom Brookshaw
- UCL Institute for Liver and Digestive Health, UCL Medical School, Royal Free Campus, London, NW3 2PF, UK
| | - Barry Fuller
- UCL Division of Surgery and Interventional Science, UCL Medical School, Royal Free Campus, London, NW3 2QG, UK
| | - Eloy Erro
- UCL Institute for Liver and Digestive Health, UCL Medical School, Royal Free Campus, London, NW3 2PF, UK
| | - Tamnia Islam
- UCL Institute for Liver and Digestive Health, UCL Medical School, Royal Free Campus, London, NW3 2PF, UK
| | - Sweta Chandel
- UCL Institute for Liver and Digestive Health, UCL Medical School, Royal Free Campus, London, NW3 2PF, UK
| | - Elizaveta Zotova
- UCL Institute for Liver and Digestive Health, UCL Medical School, Royal Free Campus, London, NW3 2PF, UK
| | - Clare Selden
- UCL Institute for Liver and Digestive Health, UCL Medical School, Royal Free Campus, London, NW3 2PF, UK.
| |
Collapse
|
40
|
Pandit A, Indurkar A, Locs J, Haugen HJ, Loca D. Calcium Phosphates: A Key to Next-Generation In Vitro Bone Modeling. Adv Healthc Mater 2024; 13:e2401307. [PMID: 39175382 PMCID: PMC11582516 DOI: 10.1002/adhm.202401307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 08/06/2024] [Indexed: 08/24/2024]
Abstract
The replication of bone physiology under laboratory conditions is a prime target behind the development of in vitro bone models. The model should be robust enough to elicit an unbiased response when stimulated experimentally, giving reproducible outcomes. In vitro bone tissue generation majorly requires the availability of cellular components, the presence of factors promoting cellular proliferation and differentiation, efficient nutrient supply, and a supporting matrix for the cells to anchor - gaining predefined topology. Calcium phosphates (CaP) are difficult to ignore while considering the above requirements of a bone model. Therefore, the current review focuses on the role of CaP in developing an in vitro bone model addressing the prerequisites of bone tissue generation. Special emphasis is given to the physico-chemical properties of CaP that promote osteogenesis, angiogenesis and provide sufficient mechanical strength for load-bearing applications. Finally, the future course of action is discussed to ensure efficient utilization of CaP in the in vitro bone model development field.
Collapse
Affiliation(s)
- Ashish Pandit
- Institute of Biomaterials and BioengineeringFaculty of Natural Sciences and TechnologyRiga Technical UniversityPulka Street 3RigaLV‐1007Latvia
- Baltic Biomaterials Centre of ExcellenceHeadquarters at Riga Technical UniversityRigaLV‐1007Latvia
| | - Abhishek Indurkar
- Institute of Biomaterials and BioengineeringFaculty of Natural Sciences and TechnologyRiga Technical UniversityPulka Street 3RigaLV‐1007Latvia
- Baltic Biomaterials Centre of ExcellenceHeadquarters at Riga Technical UniversityRigaLV‐1007Latvia
| | - Janis Locs
- Institute of Biomaterials and BioengineeringFaculty of Natural Sciences and TechnologyRiga Technical UniversityPulka Street 3RigaLV‐1007Latvia
- Baltic Biomaterials Centre of ExcellenceHeadquarters at Riga Technical UniversityRigaLV‐1007Latvia
| | | | - Dagnija Loca
- Institute of Biomaterials and BioengineeringFaculty of Natural Sciences and TechnologyRiga Technical UniversityPulka Street 3RigaLV‐1007Latvia
- Baltic Biomaterials Centre of ExcellenceHeadquarters at Riga Technical UniversityRigaLV‐1007Latvia
| |
Collapse
|
41
|
Merlin JPJ, Abrahamse H. Optimizing CRISPR/Cas9 precision: Mitigating off-target effects for safe integration with photodynamic and stem cell therapies in cancer treatment. Biomed Pharmacother 2024; 180:117516. [PMID: 39332185 DOI: 10.1016/j.biopha.2024.117516] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 09/22/2024] [Accepted: 09/25/2024] [Indexed: 09/29/2024] Open
Abstract
CRISPR/Cas9 precision genome editing has revolutionized cancer treatment by introducing specific alterations to the cancer genome. But the therapeutic potential of CRISPR/Cas9 is limited by off-target effects, which can cause undesired changes to genomic regions and create major safety concerns. The primary emphasis lies in their implications within the realm of cancer photodynamic therapy (PDT), where precision is paramount. PDT is a promising cancer treatment method; nevertheless, its effectiveness is severely limited and readily leads to recurrence due to the therapeutic resistance of cancer stem cells (CSCs). With a focus on targeted genome editing into cancer cells during PDT and stem cell treatment (SCT), the review aims to further the ongoing search for safer and more accurate CRISPR/Cas9-mediated methods. At the core of this exploration are recent advancements and novel techniques that offer promise in mitigating the risks associated with off-target effects. With a focus on cancer PDT and SCT, this review critically assesses the landscape of off-target effects in CRISPR/Cas9 applications, offering a comprehensive knowledge of their nature and prevalence. A key component of the review is the assessment of cutting-edge delivery methods, such as technologies based on nanoparticles (NPs), to optimize the distribution of CRISPR components. Additionally, the study delves into the intricacies of guide RNA design, focusing on advancements that bolster specificity and minimize off-target effects, crucial elements in ensuring the precision required for effective cancer PDT and SCT. By synthesizing insights from various methodologies, including the exploration of innovative genome editing tools and leveraging robust validation methods and bioinformatics tools, the review aspires to chart a course towards more reliable and precise CRISPR-Cas9 applications in cancer PDT and SCT. For safe PDT and SCT integration in cancer therapy, CRISPR/Cas9 precision optimization is essential. Utilizing sophisticated molecular and computational techniques to address off-target effects is crucial to realizing the therapeutic promise of these technologies, which will ultimately lead to the development of individualized and successful cancer treatment strategies. Our long-term goals are to improve precision genome editing for more potent cancer therapy approaches by refining the way CRISPR/Cas9 is integrated with photodynamic and stem cell therapies.
Collapse
Affiliation(s)
- J P Jose Merlin
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, South Africa.
| | - Heidi Abrahamse
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, South Africa
| |
Collapse
|
42
|
Han K, Wang F, Ma X, Wu Y, Zhang H, Zhao Y, Wang H, Ma J, Luan X. Human placental mesenchymal stromal cells alleviate intestinal oxidative damage in mice with graft-versus-host disease via CD73/adenosine/PI3K/Akt/GSK-3β axis. Cell Signal 2024; 123:111372. [PMID: 39209221 DOI: 10.1016/j.cellsig.2024.111372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 08/03/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Intestinal damage is a common and serious complication in patients with graft-versus-host disease (GVHD). Human placental mesenchymal stromal cells (hPMSCs) ameliorate GVHD tissue damage by exerting anti-oxidative effects; however, the underlying mechanisms remain not fully clear. METHODS A GVHD mouse model and tumor necrosis factor-α (TNF-α)-stimulated human colon epithelial cell lines NCM460 and HT-29 cells were used to investigate the mechanisms of hPMSCs alleviating GVHD-induced intestinal oxidative damage. RESULTS hPMSCs reduced TNF-α concentrations and the number of CD3+TNF-α+ T-cells, which were negatively correlated with the expression of claudin-1, occludin, and ZO-1, through CD73 in the colon tissue of GVHD mice. Meanwhile, hPMSCs reduced the mean fluorescence intensity (MFI) of reactive oxygen species (ROS) and the concentration of malondialdehyde (MDA), promoted superoxide dismutase (SOD), glutathione peroxidase (GSH-Px), and catalase (CAT) activities, as well as claudin-1, occludin, and ZO-1 expression, in colonic epithelial cells of GVHD mice and TNF-α-stimulated cells via CD73. Moreover, hPMSCs upregulated adenosine (ADO) concentrations in GVHD mice and TNF-α-stimulated cells and mitigated the loss of tight junction proteins via the CD73/ADO/ADO receptors. Further analysis showed that hPMSCs diminished Fyn expression and enhanced Nrf2, GCLC, and HO-1 expression in both TNF-α-stimulated cells and colonic epithelial cells of GVHD mice by activating PI3K/Akt/GSK-3β pathway. CONCLUSIONS The results suggested that hPMSC-mediated redox metabolism balance and promoted tight junction protein expression were achieved via CD73/ADO/PI3K/Akt/GSK-3β/Fyn/Nrf2 axis, by which alleviating intestinal oxidative injury in GVHD mice.
Collapse
Affiliation(s)
- Kaiyue Han
- Department of Immunology, Binzhou Medical University, Yantai, 264000, Shandong Province, China
| | - Feifei Wang
- Anesthesiology, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, 264000, Shandong Province, China
| | - Xiaolin Ma
- Hematology, the Affiliated Hospital of Qingdao University, Qingdao, 266000, Shandong Province, China
| | - Yunhua Wu
- Department of Immunology, Binzhou Medical University, Yantai, 264000, Shandong Province, China
| | - Hengchao Zhang
- Yantaishan Hospital Affiliated to Binzhou Medical University, Yantai, 264000, Shandong Province, China
| | - Yaxuan Zhao
- Department of Immunology, Binzhou Medical University, Yantai, 264000, Shandong Province, China
| | - Hua Wang
- Hematology, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, 264000, Shandong Province, China
| | - Junjie Ma
- Yuhuangding Hospital Affiliated to Qingdao University, Yantai, 264000, Shandong Province, China.
| | - Xiying Luan
- Department of Immunology, Binzhou Medical University, Yantai, 264000, Shandong Province, China.
| |
Collapse
|
43
|
Hamad-Alrashid H, Muntión S, Sánchez-Guijo F, Borrajo-Sánchez J, Parreño-Manchado F, García-Cenador MB, García-Criado FJ. Bone Regeneration with Dental Pulp Stem Cells in an Experimental Model. J Pers Med 2024; 14:1075. [PMID: 39590567 PMCID: PMC11595977 DOI: 10.3390/jpm14111075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 10/01/2024] [Accepted: 10/16/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES The therapeutic approach to bone mass loss and bone's limited self-regeneration is a major focus of research, emphasizing new biomaterials and cell therapy. Tissue bioengineering emerges as a potential alternative to conventional treatments. In this study, an experimental model of a critical bone lesion in rats was used to investigate bone regeneration by treating the defect with biomaterials Evolution® and Gen-Os® (OsteoBiol®, Turín, Italy), with or without mesenchymal stromal cells from dental pulp (DP-MSCs). METHODS Forty-six adult male Wistar rats were subjected to a 5-mm critical bone defect in the right mandible, which does not regenerate without intervention. The rats were randomly assigned to a Simulated Group, Control Group, or two Study Groups (using Evolution®, Gen-Os®, and DP-MSCs). The specimens were euthanized at three or six months, and radiological, histological, and ELISA tests were conducted to assess bone regeneration. RESULTS The radiological results showed that the DP-MSC group achieved uniform radiopacity and continuity in the bone edge, with near-complete structural defect restitution. Histologically, full bone regeneration was observed, with well-organized, vascularized lamellar bone and no lesion edges. These findings were supported by increases in endoglin, transforming growth factor-beta 1 (TGF-β1), protocollagen, parathormone, and calcitonin, indicating a conducive environment for bone regeneration. CONCLUSIONS The use of DP-MSCs combined with biomaterials with appropriate three-dimensional matrices is a promising therapeutic option for further exploration.
Collapse
Affiliation(s)
- Haifa Hamad-Alrashid
- Doctoral School “Studii Salamantini”, University of Salamanca, 37008 Salamanca, Spain;
| | - Sandra Muntión
- Biomedical Research Institute (IBSAL), 37007 Salamanca, Spain; (S.M.); (F.J.G.-C.)
- Regenerative Medicine and Cellular Therapy Network Center of Castilla y León, 37007 Salamanca, Spain;
| | - Fermín Sánchez-Guijo
- Regenerative Medicine and Cellular Therapy Network Center of Castilla y León, 37007 Salamanca, Spain;
- Hematology Department, University Hospital of Salamanca, 37007 Salamanca, Spain
- Department of Medicine, Faculty of Medicine, University of Salamanca, 37007 Salamanca, Spain
| | - Javier Borrajo-Sánchez
- Department of Biomedical and Diagnostic Sciences, Faculty of Medicine, University of Salamanca, 37007 Salamanca, Spain;
| | - Felipe Parreño-Manchado
- Department of Surgery, Faculty of Medicine, University of Salamanca, 37007 Salamanca, Spain;
- Coordinator of the Esophagogastric Surgery and Obesity Unit, University Hospital of Salamanca, 37007 Salamanca, Spain
| | - M. Begoña García-Cenador
- Biomedical Research Institute (IBSAL), 37007 Salamanca, Spain; (S.M.); (F.J.G.-C.)
- Department of Surgery, Faculty of Medicine, University of Salamanca, 37007 Salamanca, Spain;
| | - F. Javier García-Criado
- Biomedical Research Institute (IBSAL), 37007 Salamanca, Spain; (S.M.); (F.J.G.-C.)
- Department of Surgery, Faculty of Medicine, University of Salamanca, 37007 Salamanca, Spain;
| |
Collapse
|
44
|
Zhai R, Tai F, Ding K, Tan X, Li H, Cao Z, Ge C, Zheng X, Fu H. Comparative Analysis of the Therapeutic Effects of MSCs From Umbilical Cord, Bone Marrow, and Adipose Tissue and Investigating the Impact of Oxidized RNA on Radiation-Induced Lung Injury. Stem Cells Int 2024; 2024:7419270. [PMID: 39483952 PMCID: PMC11527546 DOI: 10.1155/2024/7419270] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 09/16/2024] [Accepted: 09/22/2024] [Indexed: 11/03/2024] Open
Abstract
Radiation-induced lung injury (RILI) is frequently observed in patients undergoing radiotherapy for thoracic malignancies, constituting a significant complication that hampers the effectiveness and utilization of tumor treatments. Ionizing radiation exerts both direct and indirect detrimental effects on cellular macromolecules, including DNA, RNA and proteins, but the impact of oxidized RNA in RILI remains inadequately explored. Mesenchymal stem cells (MSCs) can repair injured tissues, and the reparative potential and molecular mechanism of MSCs in treating RILI remains incompletely understood. This study aimed to investigate the therapeutic effects and mechanisms of action of three distinct sources of MSCs, including human umbilical cord mesenchymal stem cells (UCMSCs), bone marrow mesenchymal stem cells (BMSCs), and adipose-derived stem cells (ADSCs), in thoracically irradiated mice. Comparative analysis revealed that all three types of MSCs exhibited the ability to mitigate radiation-induced inflammatory infiltration, alveolar hemorrhage, and alveolar wall thickening in the lung tissue of the mice. MSCs also attenuated RILI by decreasing inflammatory factors, upregulating anti-inflammatory factor expression, and reducing collagen accumulation. Immunohistochemical results showed that all three MSCs reduced radiation-induced cell apoptosis and promoted the regeneration of lung tissue cells. The analysis of malondialdehyde (MDA) and 8-hydroyguanosine (8-OHG) content indicated that MSCs possess reparative properties against radiation-induced oxidative damage in lung tissue. The study provides evidence that UCMSCs are a more appropriate therapeutic option for RILI compared to BMSCs and ADSCs. Additionally, MSCs effectively reduce the accumulation of oxidized RNA in RILI, thereby, presenting a unique avenue for investigating the underlying mechanism of MSC-based treatment for RILI.
Collapse
Affiliation(s)
- Rui Zhai
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
- Department of Experimental Hematology and Biochemistry, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Fumin Tai
- Department of Experimental Hematology and Biochemistry, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Kexin Ding
- Department of Experimental Hematology and Biochemistry, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Xin Tan
- Department of Experimental Hematology and Biochemistry, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing 100850, China
- Department of Radiation Oncology, Chinese PLA General Hospital, Beijing 100853, China
| | - Hujie Li
- Department of Experimental Hematology and Biochemistry, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Zhengyue Cao
- Department of Experimental Hematology and Biochemistry, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Changhui Ge
- Department of Experimental Hematology and Biochemistry, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Xiaofei Zheng
- Department of Experimental Hematology and Biochemistry, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Hanjiang Fu
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
- Department of Experimental Hematology and Biochemistry, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing 100850, China
| |
Collapse
|
45
|
Wang S, Li X, Wang T, Sun Z, Feng E, Jin Y. Overexpression of USP35 enhances the protective effect of hUC-MSCs and their extracellular vesicles in oxygen-glucose deprivation/reperfusion-induced SH-SY5Y cells via stabilizing FUNDC1. Commun Biol 2024; 7:1330. [PMID: 39406943 PMCID: PMC11480199 DOI: 10.1038/s42003-024-07024-5] [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: 11/21/2023] [Accepted: 10/07/2024] [Indexed: 10/19/2024] Open
Abstract
Ischemia-reperfusion (IR) injury is associated with neurological disorders such as stroke. The therapeutic potential of human umbilical cord mesenchymal stem cells (hUC-MSCs) and their secreted extracellular vesicles (EVs) in alleviating IR injury across various cell types including neuronal cells has been documented. However, the underlying mechanisms through which hUC-MSCs and hUC-MSC-EVs protect neuronal cells from IR-triggered damage are not well understood. In this study, we co-cultured SH-SY5Y neuroblastoma cells with hUC-MSCs or hUC-MSC-EVs and subjected them to oxygen-glucose deprivation/reperfusion (OGD/R) treatment. Our findings indicate that both hUC-MSCs and hUC-MSC-EVs significantly improved viability, reduced apoptosis, promoted autophagy of OGD/R-induced SH-SY5Y cells, and decreased mitochondrial reactive oxygen species levels within them. Furthermore, the neuroprotective effect of hUC-MSCs and hUC-MSC-EVs in OGD/R-induced SH-SY5Y cells was enhanced by overexpressing USP35, a deubiquitinase. Mechanistically, USP35 interacted with and stabilized FUNDC1, a positive regulator of mitochondrial metabolism. Knockdown of FUNDC1 in USP35-overexpressing hUC-MSCs and their secreted EVs eliminated the augmented neuroprotective function induced by excess USP35. In conclusion, these findings underscore the crucial role of USP35 in enhancing the neuroprotective function of hUC-MSCs and their secreted EVs, achieved through the stabilization of FUNDC1 in OGD/R-induced SH-SY5Y cells.
Collapse
Affiliation(s)
- Shuo Wang
- Department of Orthopedics Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, People's Republic of China
| | - Xigong Li
- Department of Orthopedics Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, People's Republic of China
| | - Tianjiao Wang
- Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, Zhejiang, 310051, People's Republic of China
| | - Zeyu Sun
- Department of Orthopedics Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, People's Republic of China
| | - Erwei Feng
- Department of Orthopedics Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, People's Republic of China
| | - Yongming Jin
- Department of Orthopedics Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, People's Republic of China.
| |
Collapse
|
46
|
Rajendran R, Gangadaran P, Oh JM, Hong CM, Ahn BC. Engineering Three-Dimensional Spheroid Culture for Enrichment of Proangiogenic miRNAs in Umbilical Cord Mesenchymal Stem Cells and Promotion of Angiogenesis. ACS OMEGA 2024; 9:40358-40367. [PMID: 39372025 PMCID: PMC11447852 DOI: 10.1021/acsomega.4c02037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 07/30/2024] [Accepted: 08/14/2024] [Indexed: 10/08/2024]
Abstract
In the field of regenerative medicine, umbilical cord-derived mesenchymal stem cells (UC-MSCs) have a plausible potential. However, traditional two-dimensional (2D) culture systems remain limited in replicating the complex in vivo microenvironment. Thus, three-dimensional (3D) cultures offer a more physiologically relevant model. This study explored the impact of 3D culture conditions on the UC-MSC secretome and its ability to promote angiogenesis, both in vitro and in vivo. In this study, using two distinct methods, we successfully cultured UC-MSCs: in a monolayer (2D-UC-MSCs) and as spheroids formed in U-shaped 96-well plates (3D-UC-MSCs). The presence and expression of proangiogenic miRNAs in the conditioned media (CM) of these cultures were investigated, and differential expression patterns were explored. Particularly, the CM of 3D-UC-MSCs revealed significantly higher levels of miR-21-5p, miR-126-5p, and miR-130a-3p compared to 2D-UC-MSCs. Moreover, the CM from 3D-UC-MSCs revealed a higher effect on endothelial cell proliferation, migration, and tube formation than did the CM from 2D-UC-MSCs, indicating their proangiogenic potential. In an in vivo Matrigel plug mouse model, 3D-UC-MSCs (cells) stimulated greater vascular formation compared to 2D-UC-MSCs (cells). 3D culture of UC-MSCs' secretome improves the promotion of angiogenesis.
Collapse
Affiliation(s)
- Ramya
Lakshmi Rajendran
- Department
of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Prakash Gangadaran
- Department
of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea
- BK21
FOUR KNU Convergence Educational Program of Biomedical Sciences for
Creative Future Talents, Department of Biomedical Science, School
of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Ji Min Oh
- Department
of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Chae Moon Hong
- Department
of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea
- Department
of Nuclear Medicine, Kyungpook National
University Hospital, Daegu 41944, Korea
| | - Byeong-Cheol Ahn
- Department
of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea
- BK21
FOUR KNU Convergence Educational Program of Biomedical Sciences for
Creative Future Talents, Department of Biomedical Science, School
of Medicine, Kyungpook National University, Daegu 41944, Korea
- Department
of Nuclear Medicine, Kyungpook National
University Hospital, Daegu 41944, Korea
| |
Collapse
|
47
|
Nakamura K, Kitahashi T, Kogawa R, Yoshino Y, Ogura I. Definition of Synovial Mesenchymal Stem Cells for Meniscus Regeneration by the Mechanism of Action and General Amp1200 Gene Expression. Int J Mol Sci 2024; 25:10510. [PMID: 39408838 PMCID: PMC11476826 DOI: 10.3390/ijms251910510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 09/22/2024] [Accepted: 09/24/2024] [Indexed: 10/20/2024] Open
Abstract
The quality control (QC) of pharmaceutical-grade cell-therapy products, such as mesenchymal stem cells (MSCs), is challenging. Attempts to develop such products have been hampered by difficulties defining cell-type-specific characteristics and therapeutic mechanisms of action (MoAs). Although we have developed a cell therapy product, FF-31501, consisting of human synovial MSCs (SyMSCs), it was difficult to find specific markers for SyMSCs and to define the cells separately from other MSCs. The purpose of this study was to create a method for identifying and defining SyMSCs from other tissue-derived MSCs and to delve deeper into the mechanism of action of SyMSC-induced meniscus regeneration. Specifically, as a cell-type-dependent approach, we constructed a set of 1143 genes (Amp1200) reported to be associated with MSCs and established a method to evaluate them by correlating gene expression patterns. As a result, it was possible to define SyMSCs separately from other tissue-derived MSCs and non-MSCs. In addition, the gene expression analysis also highlighted TNSF-15. The in vivo rat model of meniscus injury found TNSF-15 to be an essential molecule for meniscus regeneration via SyMSC administration. This molecule and previously reported MoA molecules allowed an MoA-dependent approach to define the mechanism of action for SyMSCs. Therefore, SyMSCs for meniscus regeneration were defined by means of two approaches: the method to separate them from other MSCs and the identification of the MoA molecules. These approaches would be useful for the QC of cell therapy products.
Collapse
Affiliation(s)
- Kentaro Nakamura
- Bioscience & Engineering Laboratory, FUJIFILM Corporation, Ashigarakamigun 258-8577, Kanagawa, Japan; (T.K.); (R.K.); (Y.Y.); (I.O.)
| | | | | | | | | |
Collapse
|
48
|
Zhao Y, Chen Z, Wu Y, Zhang J, Zhang H, Han K, Wang H, Li H, Luan X. Human placental mesenchymal stromal cells promote the formation of CD8 +CD122 +PD-1 +Tregs via CD73/Foxo1 to alleviate liver injury in graft-versus-host disease mice. Int Immunopharmacol 2024; 138:112554. [PMID: 38968861 DOI: 10.1016/j.intimp.2024.112554] [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: 04/25/2024] [Revised: 06/19/2024] [Accepted: 06/21/2024] [Indexed: 07/07/2024]
Abstract
BACKGROUND Human placental mesenchymal stromal cells (hPMSCs) are known to limit graft-versus-host disease (GVHD). CD8+CD122+PD-1+Tregs have been shown to improve the survival of GVHD mice. However, the regulatory roles of hPMSCs in this subgroup remain unclear. Here, the regulatory mechanism of hPMSCs in reducing liver fibrosis in GVHD mice by promoting CD8+CD122+PD-1+Tregs formation and controlling the balance of IL-6 and IL-10 were explored. METHODS A GVHD mouse model was constructed using C57BL/6J and BALB/c mice and treated with hPMSCs. LX-2 cells were explored to study the effects of IL-6 and IL-10 on the activation of hepatic stellate cells (HSCs). The percentage of CD8+CD122+PD-1+Tregs and IL-10 secretion were determined using FCM. Changes in hepatic tissue were analysed by HE, Masson, multiple immunohistochemical staining and ELISA, and the effects of IL-6 and IL-10 on LX-2 cells were detected using western blotting. RESULTS hPMSCs enhanced CD8+CD122+PD-1+Treg formation via the CD73/Foxo1 and promoted IL-10, p53, and MMP-8 levels, but inhibited IL-6, HLF, α-SMA, Col1α1, and Fn levels in the liver of GVHD mice through CD73. Positive and negative correlations of IL-6 and IL-10 between HLF were found in liver tissue, respectively. IL-6 upregulated HLF, α-SMA, and Col1α1 expression via JAK2/STAT3 pathway, whereas IL-10 upregulated p53 and inhibited α-SMA and Col1α1 expression in LX-2 cells by activating STAT3. CONCLUSIONS hPMSCs promoted CD8+CD122+PD-1+Treg formation and IL-10 secretion but inhibited HSCs activation and α-SMA and Col1α1 expression by CD73, thus controlling the balance of IL-6 and IL-10, and alleviating liver injury in GVHD mice.
Collapse
Affiliation(s)
- Yaxuan Zhao
- Department of Immunology, Binzhou Medical University, Yantai, Shandong Province 264003, China
| | - Zhenghua Chen
- Department of Surgery, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong Province 264100, China
| | - Yunhua Wu
- Department of Immunology, Binzhou Medical University, Yantai, Shandong Province 264003, China
| | - Jiashen Zhang
- Department of Immunology, Binzhou Medical University, Yantai, Shandong Province 264003, China
| | - Hengchao Zhang
- Department of Immunology, Binzhou Medical University, Yantai, Shandong Province 264003, China
| | - Kaiyue Han
- Department of Immunology, Binzhou Medical University, Yantai, Shandong Province 264003, China
| | - Hua Wang
- Department of Hematology, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong Province 264100, China
| | - Heng Li
- Traditional Chinese Medicine Hospital of Muping District of Yantai City, Yantai, Shandong Province 264003, China.
| | - Xiying Luan
- Department of Immunology, Binzhou Medical University, Yantai, Shandong Province 264003, China.
| |
Collapse
|
49
|
Selestin Raja I, Kim C, Oh N, Park JH, Hong SW, Kang MS, Mao C, Han DW. Tailoring photobiomodulation to enhance tissue regeneration. Biomaterials 2024; 309:122623. [PMID: 38797121 DOI: 10.1016/j.biomaterials.2024.122623] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 04/25/2024] [Accepted: 05/19/2024] [Indexed: 05/29/2024]
Abstract
Photobiomodulation (PBM), the use of biocompatible tissue-penetrating light to interact with intracellular chromophores to modulate the fates of cells and tissues, has emerged as a promising non-invasive approach to enhancing tissue regeneration. Unlike photodynamic or photothermal therapies that require the use of photothermal agents or photosensitizers, PBM treatment does not need external agents. With its non-harmful nature, PBM has demonstrated efficacy in enhancing molecular secretions and cellular functions relevant to tissue regeneration. The utilization of low-level light from various sources in PBM targets cytochrome c oxidase, leading to increased synthesis of adenosine triphosphate, induction of growth factor secretion, activation of signaling pathways, and promotion of direct or indirect gene expression. When integrated with stem cell populations, bioactive molecules or nanoparticles, or biomaterial scaffolds, PBM proves effective in significantly improving tissue regeneration. This review consolidates findings from in vitro, in vivo, and human clinical outcomes of both PBM alone and PBM-combined therapies in tissue regeneration applications. It encompasses the background of PBM invention, optimization of PBM parameters (such as wavelength, irradiation, and exposure time), and understanding of the mechanisms for PBM to enhance tissue regeneration. The comprehensive exploration concludes with insights into future directions and perspectives for the tissue regeneration applications of PBM.
Collapse
Affiliation(s)
| | - Chuntae Kim
- Institute of Nano-Bio Convergence, Pusan National University, Busan, 46241, Republic of Korea; Center for Biomaterials Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Nuri Oh
- Department of Chemistry and Biology, Korea Science Academy of KAIST, Busan, 47162, Republic of Korea
| | - Ji-Ho Park
- Department of Bio and Brain Engineering and KAIST Institute for Health Science and Technology, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
| | - Suck Won Hong
- Department of Cogno-Mechatronics Engineering, College of Nanoscience & Nanotechnology, Pusan National University, Busan, 46241, Republic of Korea
| | - Moon Sung Kang
- Department of Cogno-Mechatronics Engineering, College of Nanoscience & Nanotechnology, Pusan National University, Busan, 46241, Republic of Korea
| | - Chuanbin Mao
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Sha Tin, Hong Kong SAR, China.
| | - Dong-Wook Han
- Institute of Nano-Bio Convergence, Pusan National University, Busan, 46241, Republic of Korea; Department of Cogno-Mechatronics Engineering, College of Nanoscience & Nanotechnology, Pusan National University, Busan, 46241, Republic of Korea.
| |
Collapse
|
50
|
Natsir Kalla DS, Alkaabi SA, Hendra FN, Nasrun NE, Ruslin M, Forouzanfar T, Helder MN. Stem Cell-Based Tissue Engineering for Cleft Defects: Systematic Review and Meta-Analysis. Cleft Palate Craniofac J 2024; 61:1439-1460. [PMID: 37203174 PMCID: PMC11323438 DOI: 10.1177/10556656231175278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/20/2023] Open
Abstract
This study aimed to analyze the efficacy of stem cell-based tissue engineering for the treatment of alveolar cleft (AC) and cleft palate (CP) defects in animal models. Systematic review and meta-analysis. Preclinical studies on alveolar cleft repair in maxillofacial practice. Electronic search was performed using PubMed, Embase, and Cochrane databases. Pre-clinical studies, where stem cell-based tissue engineering was used in the reconstruction of AC and CP in animal models were included. Quality of the selected articles was evaluated using SYRCLE (SYstematic Review Centre for Laboratory animal Experimentation). Review of alveolar cleft bone augmentation interventions in preclinical models. Outcome parameters registered were new bone formation (NBF) and/or bone mineral density (BMD). Thirteen large and twelve small animal studies on AC (21) and CP (4) reconstructions were included. Studies had an unclear-to-high risk of bias. Bone marrow mesenchymal stem cells were the most widely used cell source. Meta-analyses for AC indicated non-significant benefits in favor of: (1) scaffold + cells over scaffold-only (NBF P = .13); and (2) scaffold + cells over empty control (NBF P = .66; BMD P = .31). Interestingly, dog studies using regenerative grafts showed similar to superior bone formation compared to autografts. Meta analysis for the CP group was not possible. AC and CP reconstructions are enhanced by addition of osteogenic cells to biomaterials. Directions and estimates of treatment effect are useful to predict therapeutic efficacy and guide future clinical trials of bone tissue engineering.
Collapse
Affiliation(s)
- Diandra S. Natsir Kalla
- Department of Oral and Maxillofacial Surgery/Oral Pathology, Amsterdam University Medical Centers and Academic Centre for Dentistry Amsterdam (ACTA), Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, The Netherlands
- Department of Biochemistry, Faculty of Medicine, Hasanuddin University, Makassar, Indonesia
| | - Salem A. Alkaabi
- Department of Oral and Maxillofacial Surgery/Oral Pathology, Amsterdam University Medical Centers and Academic Centre for Dentistry Amsterdam (ACTA), Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, The Netherlands
- Department of Oral and Maxillofacial Surgery, Fujairah Hospital, Ministry of Health, Fujairah, UAE
| | - Faqi N. Hendra
- Department of Oral and Maxillofacial Surgery/Oral Pathology, Amsterdam University Medical Centers and Academic Centre for Dentistry Amsterdam (ACTA), Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, The Netherlands
- Department of Anatomy, Faculty of Medicine, Hasanuddin University, Makassar, Indonesia
| | - Nisrina E. Nasrun
- Division of Reconstructive Surgery for Oral and Maxillofacial Region, Department of Human Biology and Pathophysiology, School of Dentistry, Health Sciences University of Hokkaido, Hokkaido, Japan
| | - Muhammad Ruslin
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Hasanuddin University, Makassar, Indonesia
| | - Tymour Forouzanfar
- Department of Oral and Maxillofacial Surgery/Oral Pathology, Amsterdam University Medical Centers and Academic Centre for Dentistry Amsterdam (ACTA), Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, The Netherlands
- Department of Oral and Maxillofacial Surgery, Leiden University Medical Centre (LUMC), Leiden, The Netherlands
| | - Marco N. Helder
- Department of Oral and Maxillofacial Surgery/Oral Pathology, Amsterdam University Medical Centers and Academic Centre for Dentistry Amsterdam (ACTA), Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, The Netherlands
| |
Collapse
|