101
|
Ranjan S, Choudhary P, Shivalkar S, Dwivedi S, Singh S. Potential of hyaluronic acid and collagen-based scaffolds in promoting stem cell neuronal differentiation for neuroregenerative therapies: A review. Int J Biol Macromol 2025; 309:142981. [PMID: 40216130 DOI: 10.1016/j.ijbiomac.2025.142981] [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: 11/17/2024] [Revised: 03/27/2025] [Accepted: 04/07/2025] [Indexed: 04/18/2025]
Abstract
Stem cell therapy has revolutionized neurodegenerative disease treatment by presenting promising medical applications. Despite their potential, stem cell therapy remains constrained by various limitations, including low differentiation efficiency, difficulties in guiding differentiation, proliferation control, shorter half-life of growth factors, experimental reproducibility, etc. The cellular niche environment is pivotal in effective differentiation of stem cells. Neural regeneration ventures require biomaterial-based 3D scaffolds to simulate in-vivo tissue to solve the niche environment problem. Recent breakthroughs in neural regeneration have led to the development of a biomimetic scaffolds made of Hyaluronic acid (HA) and collagen (COL) that imitate the CNS's extracellular matrix (ECM) for better neural regeneration and repair. HA and COL based scaffold creates a favourable microenvironment for cellular migration, proliferation and survival of the embedded stem cells and promotes neural regeneration. HA regulates cellular activities while COL contributes in healing CNS injuries. Therefore, the utilization of HA-COL based scaffolds is appropriate for regulating cellular responses and behaviour for neural regeneration. This review investigates the synergy between HA and COL in the context of neural-specific applications for repair, regeneration, and recovery as well as augmentation of bioactivity through fabrication techniques such as 3D bioprinting, electrospinning, etc. for neural tissue regeneration.
Collapse
Affiliation(s)
- Sneha Ranjan
- Department of Applied Sciences, Indian Institute of Information Technology Allahabad, Devghat, Jhalwa, Prayagraj 211015, Uttar Pradesh, India.
| | | | - Saurabh Shivalkar
- National Institute of Animal Biotechnology (NIAB), Hyderabad 500032, Telangana, India.
| | - Shrey Dwivedi
- Department of Applied Sciences, Indian Institute of Information Technology Allahabad, Devghat, Jhalwa, Prayagraj 211015, Uttar Pradesh, India.
| | - Sangeeta Singh
- Department of Applied Sciences, Indian Institute of Information Technology Allahabad, Devghat, Jhalwa, Prayagraj 211015, Uttar Pradesh, India.
| |
Collapse
|
102
|
Maartens M, Vlok M, van de Vyver M. Antioxidants improve the viability of diabetic bone marrow MSCs without rescuing their pro-regenerative secretome function. Mol Cell Endocrinol 2025; 601:112519. [PMID: 40057223 DOI: 10.1016/j.mce.2025.112519] [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: 07/22/2024] [Revised: 01/25/2025] [Accepted: 03/05/2025] [Indexed: 04/01/2025]
Abstract
Bone marrow mesenchymal stem cell (BM-MSC) dysfunction and poor viability are prominent in diabetes and limit their therapeutic efficacy. A proteomic investigation was performed to assess disease associated alterations and the efficacy of antioxidants to rescue cellular function. BM-MSCs were isolated from obese diabetic mice (B6.Cg-Lepob/J) cultured in the presence or absence of N-acetylcysteine (NAC) and ascorbic acid-2phosphate (AAP). Label free Liquid Chromatography and Mass Spectrometry (LC-MS) analysis detected 5079 proteins with 251 being differentially expressed between treatment groups. NAC/AAP improved cellular growth/viability post isolation by up-regulating proteins involved in redox status, ATP synthesis, Rho-GTPase signaling and modulated the immunophenotype of BM-MSCs. Despite a single application of the secretome not providing any advantage for wound bed regeneration in full thickness excisional diabetic wounds, the intracellular proteome illustrated the potential mechanisms of action by which NAC/AAP targeted the respiratory chain and modulated the immune phenotype of BM-MSCs. Given these observations, antioxidant supplementation might be more effective as prophylactic strategy to protect MSCs against functional decline instead of using it as a restorative agent and warrants further investigation.
Collapse
Affiliation(s)
- Michelle Maartens
- Experimental Medicine Research Group, Department of Medicine, Faculty of Medicine & Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Mare Vlok
- Proteomics Unit, Central Analytical Facility, Stellenbosch University, South Africa
| | - Mari van de Vyver
- Experimental Medicine Research Group, Department of Medicine, Faculty of Medicine & Health Sciences, Stellenbosch University, Cape Town, South Africa.
| |
Collapse
|
103
|
Sourugeon Y, Boffa A, Perucca Orfei C, de Girolamo L, Magalon J, Sánchez M, Tischer T, Filardo G, Laver L. Cell-based therapies have disease-modifying effects on osteoarthritis in animal models: A systematic review by the ESSKA Orthobiologic Initiative. Part 3: Umbilical cord, placenta, and other sources for cell-based injectable therapies. Knee Surg Sports Traumatol Arthrosc 2025; 33:1695-1708. [PMID: 39302089 PMCID: PMC12022835 DOI: 10.1002/ksa.12472] [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: 07/15/2024] [Revised: 09/02/2024] [Accepted: 09/04/2024] [Indexed: 09/22/2024]
Abstract
PURPOSE This systematic review aimed to investigate in animal models the presence of disease-modifying effects driven by non-bone marrow-derived and non-adipose-derived products, with a particular focus on umbilical cord and placenta-derived cell-based therapies for the intra-articular injective treatment of osteoarthritis (OA). METHODS A systematic review was performed on three electronic databases (PubMed, Web of Science and Embase) according to PRISMA guidelines. The results were synthesised to investigate disease-modifying effects in preclinical animal studies comparing injectable umbilical cord, placenta, and other sources-derived products with OA controls. The risk of bias was assessed using the SYRCLE tool. RESULTS A total of 80 studies were included (2314 animals). Cell therapies were most commonly obtained from the umbilical cord in 33 studies and placenta/amniotic tissue in 18. Cell products were xenogeneic in 61 studies and allogeneic in the remaining 19 studies. Overall, 25/27 (92.6%) of studies on umbilical cord-derived products documented better results compared to OA controls in at least one of the following outcomes: macroscopic, histological and/or immunohistochemical findings, with 19/22 of studies (83.4%) show positive results at the cartilage level and 4/6 of studies (66.7%) at the synovial level. Placenta-derived injectable products documented positive results in 13/16 (81.3%) of the studies, 12/15 (80.0%) at the cartilage level, and 2/4 (50.0%) at the synovial level, but 2/16 studies (12.5%) found overall worse results than OA controls. Other sources (embryonic, synovial, peripheral blood, dental pulp, cartilage, meniscus and muscle-derived products) were investigated in fewer preclinical studies. The risk of bias was low in 42% of items, unclear in 49%, and high in 9% of items. CONCLUSION Interest in cell-based injectable therapies for OA treatment is soaring, particularly for alternatives to bone marrow and adipose tissue. While expanded umbilical cord mesenchymal stem cells reported auspicious disease-modifying effects in preventing OA progression in animal models, placenta/amniotic tissue also reported deleterious effects on OA joints. Lower evidence has been found for other cellular sources such as embryonic, synovial, peripheral blood, dental-pulp, cartilage, meniscus, and muscle-derived products. LEVEL OF EVIDENCE Level II.
Collapse
Affiliation(s)
- Yosef Sourugeon
- Division of Surgery, Orthopaedics DepartmentChaim Sheba Medical CentreRamat GanIsrael
| | - Angelo Boffa
- Applied and Translational Research Center, IRCCS Istituto Ortopedico RizzoliBolognaItaly
- Clinica Ortopedica e Traumatologica 2, IRCCS Istituto Ortopedico RizzoliBolognaItaly
| | - Carlotta Perucca Orfei
- Laboratorio di Biotecnologie Applicate all'Ortopedia, IRCCS Ospedale Galeazzi Sant'AmbrogioMilanItaly
| | - Laura de Girolamo
- Laboratorio di Biotecnologie Applicate all'Ortopedia, IRCCS Ospedale Galeazzi Sant'AmbrogioMilanItaly
| | - Jeremy Magalon
- INSERM, NRA, C2VNAix Marseille UniversityMarseilleFrance
- SAS RemedexMarseilleFrance
- Cell Therapy Laboratory, Hôpital De La Conception, AP‐HMMarseilleFrance
| | - Mikel Sánchez
- Advanced Biological Therapy Unit, Hospital Vithas VitoriaVitoria‑GasteizSpain
- Arthroscopic Surgery Unit, Hospital Vithas VitoriaVitoria‐GasteizSpain
| | - Thomas Tischer
- Department of Orthopaedic and Trauma SurgeryMalteser WaldkrankenhausErlangenGermany
- Department of Orthopaedic SurgeryUniversity of RostockRostockGermany
| | - Giuseppe Filardo
- Applied and Translational Research Center, IRCCS Istituto Ortopedico RizzoliBolognaItaly
- Department of SurgeryService of Orthopaedics and Traumatology, EOCLuganoSwitzerland
- Faculty of Biomedical SciencesUniversità Della Svizzera ItalianaLuganoSwitzerland
| | - Lior Laver
- Arthrosport ClinicTel‑AvivIsrael
- Rappaport Faculty of Medicine, Technion University Hospital (IsraelInstitute of Technology)HaifaIsrael
- Department of OrthopaedicsHillel Yaffe Medical Center (HYMC)HaderaIsrael
| |
Collapse
|
104
|
Liu C, Shao FH, He XC, Du HZ, Liu CM, Zhou B, Teng ZQ. Single-Cell RNA Sequencing Uncovers Molecular Features Underlying the Disrupted Neurogenesis Following Traumatic Brain Injury. Glia 2025; 73:1036-1050. [PMID: 39760225 DOI: 10.1002/glia.24671] [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: 06/15/2024] [Revised: 12/23/2024] [Accepted: 12/26/2024] [Indexed: 01/07/2025]
Abstract
Traumatic brain injury (TBI) is a leading cause of death and disability worldwide, with limited effective treatment strategies. Endogenous neural stem cells (NSCs) give rise to neurons and glial cells throughout life. However, NSCs are more likely to differentiate into glial cells rather than neurons at the lesion site after TBI and the underlying molecular mechanism remains largely unknown. Here, we performed large-scale single-cell transcriptome sequencing of subventricular zone (SVZ) NSCs and NSCs-derived cells in the mouse brain, and provide molecular evidence for previous observations that glial differentiation of NSCs prevails after TBI. In addition, we show that the disrupted neurogenesis following TBI is caused by the reduction of a NSC subcluster (NSC-4) expressing the neuronal gene Tubb3. Finally, we demonstrate that the transcriptional factor Dlx2 is significantly downregulated in NSC-4, and Dlx2 overexpression is sufficient to drive NSCs towards neuronal lineage differentiation at the expense of astrocytic lineage differentiation under pro-inflammatory conditions.
Collapse
Affiliation(s)
- Cong Liu
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute of Mental Health and Drug Discovery, Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Mental Health, Wenzhou Medical University, Zhejiang, China
| | - Fang-Hong Shao
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Xuan-Cheng He
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- Chinese Academy of Sciences, Institute for Stem Cell and Regeneration, Beijing, China
| | - Hong-Zhen Du
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- Chinese Academy of Sciences, Institute for Stem Cell and Regeneration, Beijing, China
| | - Chang-Mei Liu
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- Chinese Academy of Sciences, Institute for Stem Cell and Regeneration, Beijing, China
- Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Bing Zhou
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- Chinese Academy of Sciences, Institute for Stem Cell and Regeneration, Beijing, China
| | - Zhao-Qian Teng
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- Chinese Academy of Sciences, Institute for Stem Cell and Regeneration, Beijing, China
- Medical School, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
105
|
De Simone U, Caloni F, Pignatti P, Gaetano C, Locatelli CA, Coccini T. Human stromal cell-based protocol to generate astrocytes: a straightforward in vitro predictive strategy in neurotoxicology. Toxicol Mech Methods 2025; 35:340-355. [PMID: 39626968 DOI: 10.1080/15376516.2024.2435351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 11/22/2024] [Accepted: 11/23/2024] [Indexed: 05/04/2025]
Abstract
The inherent adaptability of human mesenchymal stromal cells (hMSCs) to differentiate into neural lineages provides a valuable resource for investigating potential neurotoxicity in humans. By harnessing the ability of hMSCs to transform into astrocytes, we can evaluate the effects of various agents on these vital cells. Our protocol employs hMSCs sourced from umbilical cord tissue, ensuring a readily available supply of high-quality cells. The hMSC-to-neural workflow encompasses six essential steps: hMSC culture, followed by the generation of embryoid bodies (EBs) from these cells on specialized surfaces. Next, EBs and cells are expanded in a growth-promoting medium, directing them toward neural lineages. Subsequent differentiation into immature astrocytes is achieved through the use of specific factors. The process continues with the maturation of EBs/cells into astrocyte-like cells (hALCs) under optimized conditions, culminating in the final development of hALCs in a specialized medium. This methodology yields cells that display astrocyte morphology and express characteristic markers such as GFAP and S100β. The protocol is efficient, requiring roughly 6 weeks to generate hALCs from primary hMSCs without genetic manipulation. The application of hMSCs in evaluating cell damage triggered by neurotoxicants like MeHg and MGO underscores their potential as a valuable component within a more extensive battery of neurotoxicity tests.
Collapse
Affiliation(s)
- Uliana De Simone
- Laboratory of Clinical and Experimental Toxicology, and Pavia Poison Center-National Toxicology Information Center, Toxicology Unit, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| | - Francesca Caloni
- Dipartimento di Scienze e Politiche Ambientali (ESP), Università degli Studi di Milano, Milan, Italy
| | - Patrizia Pignatti
- Allergy and Immunology Unit, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| | - Carlo Gaetano
- Laboratory of Epigenetics, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| | - Carlo Alessandro Locatelli
- Laboratory of Clinical and Experimental Toxicology, and Pavia Poison Center-National Toxicology Information Center, Toxicology Unit, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| | - Teresa Coccini
- Laboratory of Clinical and Experimental Toxicology, and Pavia Poison Center-National Toxicology Information Center, Toxicology Unit, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| |
Collapse
|
106
|
Ding M, Lu Y, Lei QK, Zheng YW. Advantages and challenges of ex vivo generation and expansion of human hematopoietic stem cells from pluripotent stem cells. Exp Hematol 2025; 145:104752. [PMID: 40086687 DOI: 10.1016/j.exphem.2025.104752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 01/24/2025] [Accepted: 01/27/2025] [Indexed: 03/16/2025]
Abstract
Hematopoietic stem cell transplantation (HSCT) is an essential and increasing therapeutic approach for treating conditions such as leukemia, lymphoma, and other blood cancers. However, its widespread use faces significant challenges, including limited donor availability, pathogens, and the risk of immune rejection. The emergence of pluripotent stem cells (PSCs) offers a potential solution to these challenges. By enabling the generation of hematopoietic stem cells (HSCs) and blood cells in vitro, PSCs open pathways to address the limitations of traditional HSC sources. Self-induced or gene-edited PSCs from patients may provide an accessible and personalized option for clinical applications. In this review, we examine the current protocols for differentiating PSCs into HSCs and blood cells, highlighting their benefits and shortcomings. Despite advancements in this field, two primary challenges persist: low differentiation efficiency and difficulties in isolating and enriching functional HSCs. These problems make it difficult to obtain HSCs for long-term survival. Thus, we propose innovative strategies and potential improvements including induction scheme optimization, reprogramming, and cell fate tracking. Future research should prioritize the development of efficient and reliable differentiation protocols for PSCs to obtain more functional HSCs. Additionally, establishing effective methods for enriching functional HSCs and blood cells will be critical for optimizing their use in clinical applications. These efforts hold the promise of overcoming current limitations and advancing the therapeutic potential of PSC-derived blood cells.
Collapse
Affiliation(s)
- Min Ding
- Institute of Regenerative Medicine, and Department of Dermatology, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, Jiangsu, China; Haihe Laboratory of Cell Ecosystem, Institute of Hematology, Chinese Academy of Medical Sciences, Tianjin, China; Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, and South China Institute of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen, Guangdong, China
| | - Yu Lu
- Institute of Regenerative Medicine, and Department of Dermatology, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, Jiangsu, China; Haihe Laboratory of Cell Ecosystem, Institute of Hematology, Chinese Academy of Medical Sciences, Tianjin, China; Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, and South China Institute of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen, Guangdong, China
| | - Quan-Kai Lei
- Institute of Regenerative Medicine, and Department of Dermatology, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Yun-Wen Zheng
- Institute of Regenerative Medicine, and Department of Dermatology, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, Jiangsu, China; Haihe Laboratory of Cell Ecosystem, Institute of Hematology, Chinese Academy of Medical Sciences, Tianjin, China; Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, and South China Institute of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen, Guangdong, China; Division of Regenerative Medicine, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, The University of Tokyo, Tokyo, Japan; Department of Medicinal and Life Sciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan.
| |
Collapse
|
107
|
Kazemi N, Hassanzadeh-Tabrizi SA, Koupaei N, Ghomi H, Masaeli E. Incorporation of forsterite nanoparticles in a 3D printed polylactic acid/polyvinylpyrrolidone scaffold for bone tissue regeneration applications. Int J Biol Macromol 2025; 305:141046. [PMID: 39954877 DOI: 10.1016/j.ijbiomac.2025.141046] [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: 05/24/2024] [Revised: 02/05/2025] [Accepted: 02/13/2025] [Indexed: 02/17/2025]
Abstract
Three-dimensional (3D) printing has facilitated the fabrication of customized scaffolds for the repair of complex bone defects. In this study, 3D-printed scaffolds composed of a mixture of polylactic acid-polyvinylpyrrolidone (PLA-PVP) incorporating different amounts of forsterite (F; Mg2SiO4) nanoparticles were fabricated using fused deposition modeling (FDM) technique. The incorporation of PVP and F nanoparticles into the PLA scaffold significantly decreased the water drop contact angle. The mechanical properties of the PLA-PVP scaffold were enhanced with the addition of 10 % F nanoparticles, as the compressive yield strength increased from 10.8 to 16.0 MPa and the elastic modulus from 83.52 to 108.41 MPa. However, the addition of F nanoparticles increased the degradation rate of the PLA-PVP scaffold over 8 weeks. Importantly, the addition of 10 % F nanoparticles into the PLA-PVP scaffold improved bioactivity and formation of apatite deposits on the scaffold after 4 weeks of immersion in simulated body fluid. Moreover, the PLA-PVP/10F scaffold showed strong MG63 cell adhesion and proliferation, as well as promoting osteogenic differentiation of rat bone marrow mesenchymal stem cells. At last, these findings suggest the PLA-PVP/10F scaffold is a promising candidate for application in bone defect repair.
Collapse
Affiliation(s)
- Nafise Kazemi
- Advanced Materials Research Center, Department of Materials Engineering, Najafabad Branch, Islamic Azad University, Najafabad, Iran
| | - S A Hassanzadeh-Tabrizi
- Advanced Materials Research Center, Department of Materials Engineering, Najafabad Branch, Islamic Azad University, Najafabad, Iran.
| | - Narjes Koupaei
- Advanced Materials Research Center, Department of Materials Engineering, Najafabad Branch, Islamic Azad University, Najafabad, Iran
| | - Hamed Ghomi
- Advanced Materials Research Center, Department of Materials Engineering, Najafabad Branch, Islamic Azad University, Najafabad, Iran
| | - Elahe Masaeli
- Department of Cellular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| |
Collapse
|
108
|
Duan G, Zou T, Wu X, Zhang Y, Liu H, Mei C. Neuroprotective role of geniposide-loaded UMSC nanovesicles in depression via P2ry12 downregulation. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 140:156581. [PMID: 40085991 DOI: 10.1016/j.phymed.2025.156581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 02/05/2025] [Accepted: 02/24/2025] [Indexed: 03/16/2025]
Abstract
BACKGROUND Depression is a prevalent mental disorder characterized by persistent low mood, loss of interest, and cognitive impairment. Oxidative stress and inflammation play crucial roles in its pathogenesis. Novel therapeutic strategies targeting these mechanisms are needed to improve treatment outcomes. PURPOSE The purpose of this study is to gauge the therapeutic effectiveness of geniposide (GEN)-loaded umbilical cord-derived mesenchymal stem cell membrane biomimetic nanovesicles (CSPG@UMSC NPs) targeting the P2ry12 factor for depression management, considering its association with oxidative stress and inflammatory pathways. STUDY DESIGN A combination of in vitro neuronal cell culture experiments and an in vivo chronic unpredictable mild stress (CUMS) mouse model was used to assess the effects of CSPG@UMSC NPs. METHODS In vitro investigations involved culture and characterization of CSPG@UMSC NPs and transcriptome sequencing analysis to identify DEGs in neurons. In vivo experiments utilized a depression mouse model treated with CSPG@UMSC NPs, followed by behavioral tests, biomarker analysis, and histological assessments. RESULTS CSPG@UMSC NPs successfully downregulated P2ry12 expression, leading to improved neuronal activity, decreased inflammation, reduced cell apoptosis, and lowered reactive oxygen species levels in both in vitro and in vivo settings. CONCLUSION CSPG@UMSC NPs loaded with GEN inhibit oxidative stress and inflammation by downregulating P2ry12. This research unveils, for the initial instance, the vital role of P2ry12 in depression and proposes a novel nano-therapy strategy based on MSCs and GEN, offering new insights and potential clinical applications for the treatment of depression.
Collapse
Affiliation(s)
- Guoxiang Duan
- Department of Acupuncture, Heilongjiang Academy of Chinese Medical Sciences, Room 602, Zone C, No. 33 Xidazhi Street, Nangang District, Harbin, Heilongjiang Province 150001, China
| | - Tianyu Zou
- Department of Encephalopathy, Shenzhen Luohu District Hospital of Traditional Chinese Medicine, Shenzhen 518000, China
| | - Xue Wu
- Department of Nephrology, Heilongjiang Academy of Chinese Medical Sciences, Harbin 150001, China
| | - Yiming Zhang
- Department of Continuing Education, Heilongjiang Academy of Chinese Medical Sciences, Harbin 150001, China
| | - Hongmei Liu
- Department of Acupuncture, Heilongjiang Academy of Chinese Medical Sciences, Room 602, Zone C, No. 33 Xidazhi Street, Nangang District, Harbin, Heilongjiang Province 150001, China
| | - Cheng Mei
- Department of Acupuncture, Heilongjiang Academy of Chinese Medical Sciences, Room 602, Zone C, No. 33 Xidazhi Street, Nangang District, Harbin, Heilongjiang Province 150001, China.
| |
Collapse
|
109
|
Zheng X, Wang J, Zhou H, Chai Y, Li Z, Chen M, Yang Z, Xu C, Lei C, He Y, Zou D, Ye Q. Dental pulp stem cells alleviate Schwann cell pyroptosis via mitochondrial transfer to enhance facial nerve regeneration. Bioact Mater 2025; 47:313-326. [PMID: 40026822 PMCID: PMC11869962 DOI: 10.1016/j.bioactmat.2025.01.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 01/23/2025] [Accepted: 01/23/2025] [Indexed: 03/05/2025] Open
Abstract
Dental pulp stem cells (DPSCs) have demonstrated remarkable potential in enhancing peripheral nerve regeneration, though the precise mechanisms remain largely unknown. This study investigates how DPSCs alleviate Schwann cell pyroptosis and restore mitochondrial homeostasis through intercellular mitochondrial transfer. In a crab-eating macaque model, we first observed that DPSC-loaded nerve conduits significantly promoted long-term nerve regeneration, facilitating tissue proliferation and myelin recovery. We further established a rat facial nerve injury (FNI) model and found that DPSC treatment reduced pyroptosis and mitochondrial ROS production in Schwann cells. A pivotal mitochondrial protective mechanism, resembling the effects of a ROS-targeted inhibitor, involved the transfer of mitochondria from DPSCs to pyroptosis-induced Schwann cells via tunneling nanotubes, while blocking intercellular junctions or mitochondrial function diminished the therapeutic effects. TNFα secreted by pyroptosis-induced Schwann cells activated the NF-κB pathway in DPSCs, enhancing mitochondrial transfer and adaptive stress responses, thereby promoting mitochondrial protection against pyroptosis in Schwann cells, as reflected in the improved therapeutic efficacy of TNFα-preconditioned DPSCs in the FNI model. These findings unveil a mechanism through which DPSCs foster nerve regeneration via mitochondrial transfer, presenting a promising strategy for enhancing stem cell-based therapies for nerve injuries.
Collapse
Affiliation(s)
- Xiaoyu Zheng
- Centre of Regenerative Medicine, Department of Stomatology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China
| | - Juan Wang
- Centre of Regenerative Medicine, Department of Stomatology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China
| | - Heng Zhou
- Centre of Regenerative Medicine, Department of Stomatology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China
| | - Ying Chai
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, China
| | - Ziwei Li
- Centre of Regenerative Medicine, Department of Stomatology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China
| | - Minjie Chen
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, China
| | - Zihan Yang
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, China
| | - Chun Xu
- Sydney Dental School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, 2006, Australia
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
| | - Chang Lei
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Yan He
- Institute of Regenerative and Translational Medicine, Tianyou Hospital of Wuhan University of Science and Technology, Wuhan, Hubei, 430064, China
| | - Duohong Zou
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, China
| | - Qingsong Ye
- Centre of Regenerative Medicine, Department of Stomatology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China
- Sydney Dental School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, 2006, Australia
| |
Collapse
|
110
|
Xu J, Brown J, Shaik R, Soto-Garcia L, Liao J, Nguyen K, Zhang G, Hong Y. Injectable myocardium-derived hydrogels with SDF-1α releasing for cardiac repair. BIOMATERIALS ADVANCES 2025; 170:214203. [PMID: 39908684 DOI: 10.1016/j.bioadv.2025.214203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 01/23/2025] [Accepted: 01/26/2025] [Indexed: 02/07/2025]
Abstract
Myocardial infarction (MI) is a predominant cause of morbidity and mortality globally. Therapeutic chemokines, such as stromal cell-derived factor 1α (SDF-1α), present a promising opportunity to treat the profibrotic remodeling post-MI if they can be delivered effectively to the injured tissue. However, direct injection of SDF-1α or physical entrapment in a hydrogel has shown limited efficacy. Here, we developed a sustained-release system consisting of SDF-1α loaded poly(lactic-co-glycolic acid) nanoparticles (PLGA NPs) and an injectable porcine cardiac decellularized extracellular matrix (cdECM) hydrogel. This system demonstrated a sustained release of SDF-1α over four weeks while there is one week release for SDF-1α directly encapsulated in the cdECM hydrogel during in vitro testing. The incorporation of PLGA NPs into the cdECM hydrogel significantly enhanced its mechanical properties, increasing the Young's modulus from 561 ± 228 kPa to 1007 ± 2 kPa and the maximum compressive strength from 639 ± 42 kPa to 1014 ± 101 kPa. This nanocomposite hydrogel showed good cell compatibility after 7 days of culture with H9C2 cells, while the released SDF-1α retained its bioactivity, as evidenced by its chemotactic effects in vitro. Furthermore, in vivo studies further highlighted its significant ability to promote angiogenesis in the infarcted area and improve cardiac function after intramyocardial injection. These results demonstrated the therapeutic potential of combining local release of SDF-1α with the cdECM hydrogel for MI treatment.
Collapse
Affiliation(s)
- Jiazhu Xu
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX 76019, United States of America
| | - Jacob Brown
- Department of Biomedical Engineering, The University of Akron, Akron, OH 44325, United States of America
| | - Rubia Shaik
- Department of Biomedical Engineering, The University of Akron, Akron, OH 44325, United States of America
| | - Luis Soto-Garcia
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX 76019, United States of America
| | - Jun Liao
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX 76019, United States of America
| | - Kytai Nguyen
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX 76019, United States of America
| | - Ge Zhang
- Department of Biomedical Engineering, The University of Akron, Akron, OH 44325, United States of America.
| | - Yi Hong
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX 76019, United States of America.
| |
Collapse
|
111
|
Varisli L, Zoumpourlis P, Spandidos DA, Zoumpourlis V, Vlahopoulos S. ALDH1A1 in breast cancer: A prospective target to overcome therapy resistance (Review). Oncol Lett 2025; 29:213. [PMID: 40093866 PMCID: PMC11905208 DOI: 10.3892/ol.2025.14959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Accepted: 02/06/2025] [Indexed: 03/19/2025] Open
Abstract
The expression of cytosolic aldehyde dehydrogenases (ALDHs), which mediate the last step in the pathway of the synthesis of all-trans retinoic acid, is dysregulated in various types of human cancer, and has been associated with the development of cancer stem cells (CSCs) in solid tumors and hematological malignancies. CSCs are considered a minor fraction of cancer cells with the capacity to initiate neoplastic tumors. ALDH1A1 serves a crucial role in the emergence of the CSC phenotype, induces the malignant behavior of cancer cells and promotes treatment resistance. Notably, ALDH1A1-induced therapy resistance is not exclusive to just one group of drugs, but affects diverse types of drugs that use different mechanisms to kill cells. This diversity of drug resistance-inducing effects is associated with the stemness-supporting functions of ALDH1A1. The inhibition of ALDH1A1 activity using chemicals or the depletion of ALDH1A1 via genetic approaches, such as the use of small interfering RNA, can overcome diverse pathways of therapy resistance. In the context of breast cancer, it is critical that only a fraction of malignant cells are expected to manifest stem-like features, which include increased expression of ALDH1A1. From the angle of disease prognosis, the extent of the association of ALDH1A1 with increased malignant behavior and drug resistance remains to be determined through the application of cutting-edge methods that detect the expression of tracked biomarkers within tumors.
Collapse
Affiliation(s)
- Lokman Varisli
- Department of Molecular Biology and Genetics, Science Faculty, Dicle University, Diyarbakir 21280, Turkey
| | - Panagiotis Zoumpourlis
- Biomedical Applications Unit, Institute of Chemical Biology, National Hellenic Research Foundation, 11635 Athens, Greece
| | - Demetrios A. Spandidos
- Laboratory of Clinical Virology, School of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Vassilis Zoumpourlis
- Biomedical Applications Unit, Institute of Chemical Biology, National Hellenic Research Foundation, 11635 Athens, Greece
| | - Spiros Vlahopoulos
- First Department of Pediatrics, National and Kapodistrian University of Athens, 11527 Athens, Greece
| |
Collapse
|
112
|
Ko YC, Chen TC. Hepatic Undifferentiated Carcinoma With Osteoclast Like Giant Cell With Dominant Intraductal Growth: A Case Report With Literature Review. Int J Surg Pathol 2025:10668969251334752. [PMID: 40304634 DOI: 10.1177/10668969251334752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2025]
Abstract
Undifferentiated carcinoma with osteoclast-like giant cells is extremely rare in the liver. Here, we report a tumor with dominant intraductal growth. The unique growth pattern has never been reported before. A 73-year-old female patient presented intermittent right upper quadrant abdominal pain accompanied by fever and general weakness for 1 month. Imaging study revealed an intraductal lesion in the dilated large bile duct. Histologically, the tumor showed predominantly undifferentiated carcinoma with osteoclast-like giant cells, associated with high-grade biliary intraepithelial neoplasia and minor foci of adenocarcinoma. A KRAS codon 61 mutation was detected, supporting the epithelial origin of undifferentiated carcinoma with osteoclast-like giant cells. This tumor not only adds to the limited documented instances of undifferentiated carcinoma with osteoclast-like giant cells in the liver but also demonstrates that the development of an undifferentiated component might occur in a low stage tumor, which is typically considered a late stage in tumor progression.
Collapse
Affiliation(s)
- Yu-Chien Ko
- Department of Pathology, Chang Gung Memorial Hospital, Chang Gung University School of Medicine, Kwei-Shan, Tao-Yuan
| | - Tse-Ching Chen
- Department of Pathology, Chang Gung Memorial Hospital, Chang Gung University School of Medicine, Kwei-Shan, Tao-Yuan
| |
Collapse
|
113
|
Cui J, Luo L, Geng H, Gao Y, Chen Y, Yu Q, Huang X, Wang X, Sun T. Proteomics suggests the role of Cxcl12 secreted by hucMSCs in the treatment of lipopolysaccharide-acute lung injury. Microvasc Res 2025; 160:104815. [PMID: 40311750 DOI: 10.1016/j.mvr.2025.104815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 03/28/2025] [Accepted: 04/25/2025] [Indexed: 05/03/2025]
Abstract
Acute respiratory distress syndrome (ARDS) is a clinical syndrome characterized by a high mortality rate, and its treatment is relatively straightforward. The application of human umbilical cord mesenchymal stem cells (hucMSCs) for the treatment of ARDS has emerged as a novel therapeutic approach and has been the subject of extensive research. In this study, a mouse model of acute lung injury (ALI) was established, and hucMSCs were administered via tail vein injection to investigate the pathogenesis of ARDS and the protein alterations following hucMSC treatment. Data-independent acquisition (DIA) was employed for the proteomic analysis of lung tissue, which included the identification of differentially expressed proteins (DEPs) and their associated pathways. The relevant DEPs identified in the lung tissues of the three groups of mice included Arid5a, Mrpl4, Cxcl12, and Rnf121 (P <0.05). Silencing the expression of Cxcl12 in hucMSCs could significantly inhibit the therapeutic effect of hucMSCs in reducing the permeability of lung tissue and endothelial cells (P < 0.05). Additionally, the signaling pathways associated with the relevant DEPs were analyzed. The DEPs and the enriched pathways discussed herein provide valuable insights into the pathogenesis of ARDS and the potential applications of hucMSCs.
Collapse
Affiliation(s)
- Jinfeng Cui
- Department of Intensive Care Unit, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Liqing Luo
- Department of Hematology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Hongmei Geng
- Medical Insurance Business Center of Bincheng District Medical Security Bureau, Binzhou City, Shandong Province, China
| | - Yunxiu Gao
- Department of Respiratory and Critical Care Medicine, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Yuanyuan Chen
- Department of Intensive Care Unit, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Qilin Yu
- Department of Intensive Care Unit, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Xiao Huang
- Department of Medical Technology, Binzhou Polytechnic, Binzhou, Shandong, China
| | - Xiaozhi Wang
- Department of Intensive Care Unit, Binzhou Medical University Hospital, Binzhou, Shandong, China.
| | - Ting Sun
- Department of Intensive Care Unit, Binzhou Medical University Hospital, Binzhou, Shandong, China.
| |
Collapse
|
114
|
Bashirrohelleh MA, Bavarsad K, Khodadadi A, Shohan M, Asadirad A. Curcumin-enhanced stem cell exosomes: A novel approach to modulating neuroinflammation and improving cognitive function in a rat model of Alzheimer's disease. Eur J Pharmacol 2025; 999:177695. [PMID: 40315951 DOI: 10.1016/j.ejphar.2025.177695] [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: 11/25/2024] [Revised: 04/22/2025] [Accepted: 04/30/2025] [Indexed: 05/04/2025]
Abstract
The effect of Curcumin-enhanced stem cell exosomes on the learning and memory impairment induced by streptozotocin (STZ) and neuro-inflammation in rats was evaluated. An animal model of Alzheimer's disease (AD) was established by intracerebroventricular (ICV) injection of STZ (3 mg/kg) in male Wistar rats (250 ± 50 g). ICV STZ injections chronically reduce cerebral glucose uptake and produce other effects similar to pathological, molecular and behavioral features of AD. Numerous studies confirmed the anti-inflammatory and antioxidant properties of curcumin (a natural polyphenol) against free radicals, as well as its ability to inhibit the aggregation of proteins such as beta-amyloid and alpha-synuclein in disorders such as AD and Parkinson's disease. The use of extracellular vesicles has garnered a lot of interest in research studies because of the important roles that mesenchymal stem cell-derived exosomes play in permeability, retention, and drug delivery as well as their ability to reduce inflammatory cytokines (TNF-α, IL-1β, and IL-6). Furthermore, researches highlighted the positive effect of curcumin on neuronal differentiation of stem cells in vivo and in vitro. Since studies emphasized the ameliorating effect of curcumin-treated macrophage-exosomes on symptoms of Alzheimer's disease by inhibiting tau protein phosphorylation, we proposed that Curcumin-primed MSC exosomes may offer greater efficacy to alleviate AD compared to naïve MSC exosomes. In this study, we investigated the effect of curcumin in stimulating the anti-inflammatory potential of exosome-derived stem cells. We evaluated the effect of MSC-EXO and pre-treated MSC-EXO with curcumin (CUR-MSC-EXO) on inhibiting inflammation and memory and learning impairments. Following four intraperitoneal injections of MSC-EXO and CUR-MSC-EXO at a dosage of 30μg/body over 30 days, we found that MSC-EXO and CUR-MSC-EXO elevated anti-inflammatory cytokines (IL10, TGF-β) and reduced pro-inflammatory cytokines (IL1, TNF-α) in peripheral blood compared to the AD group. The elevated level of M2 anti-inflammatory microglia markers (Arg1, CD206) and decreased level expression of M1 pro-inflammatory markers (iNOS, CD86) indicated that the CUR-MSC-EXO effect was more significant in the polarization of microglia into the M2 phenotype in the rat hippocampus. Both treatment groups demonstrated improvements in memory and learning skills. The results of the passive avoidance learning in the rats with STZ-induced memory impairment, however, were better in the CUR-MSC-EXO. Additionally, after therapy, a decrease in degenerative neurons was seen. Therefore, using curcumin may stimulate the anti-inflammatory and neuroprotective potential of exosome-derived stem cells which could provide hope for Alzheimer's disease treatment in the future.
Collapse
Affiliation(s)
- Mohammad-Ali Bashirrohelleh
- Department of Immunology, Faculty of Medicine, Ahvaz Jundishapur, University of Medical Sciences, Iran; Cellular and Molecular Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Kowsar Bavarsad
- Department of Physiology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran; Persian Gulf Physiology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Ali Khodadadi
- Department of Immunology, Faculty of Medicine, Ahvaz Jundishapur, University of Medical Sciences, Iran; Cancer, Petroleum and Environmental Pollutants Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mojtaba Shohan
- Department of Immunology, Faculty of Medicine, Ahvaz Jundishapur, University of Medical Sciences, Iran
| | - Ali Asadirad
- Department of Immunology, Faculty of Medicine, Ahvaz Jundishapur, University of Medical Sciences, Iran; Cellular and Molecular Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran; Cancer, Petroleum and Environmental Pollutants Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
115
|
Kapoor DU, Pareek A, Sharma S, Prajapati BG, Thanawuth K, Sriamornsak P. Alginate gels: Chemistry, gelation mechanisms, and therapeutic applications with a focus on GERD treatment. Int J Pharm 2025; 675:125570. [PMID: 40199431 DOI: 10.1016/j.ijpharm.2025.125570] [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: 01/07/2025] [Revised: 03/25/2025] [Accepted: 04/05/2025] [Indexed: 04/10/2025]
Abstract
Alginate, a natural polysaccharide derived primarily from marine algae, has become popular in biomedical research due to its versatile gelation properties and biocompatibility. This review explores the chemistry, gelation mechanisms, and therapeutic applications of alginate gels, with a particular focus on their role in gastroesophageal reflux disease (GERD) management. Alginate's structure, comprised of guluronic and mannuronic acid blocks, allows for gel formation by ionic cross-linking with divalent cations like calcium ions, generating a stable "egg-box" structure. The effects of pH, temperature, and ion concentration on gelation are explored, as well as other gel forms such as in situ and heat-sensitive gels. Alginate is widely used in the medical and pharmaceutical areas to promote tissue engineering through cell encapsulation and scaffolding, as well as in drug delivery systems for controlled and targeted release. In GERD therapy, alginate produces a gel raft that inhibits acid reflux, providing an effective alternative to proton pump inhibitors. Alginate-based products have demonstrated clinical success, strengthening alginate's medicinal promise. The review also discusses alginate-related issues, such as source variability and stability, as well as innovative modifications to improve treatment effects. These improvements establish alginate as a potential material for customized medication and tailored delivery systems.
Collapse
Affiliation(s)
- Devesh U Kapoor
- Dr. Dayaram Patel Pharmacy College, Bardoli, Gujarat 394601, India
| | - Anil Pareek
- Department of Pharmaceutics, Lachoo Memorial College of Science and Technology (Autonomous), Jodhpur, Rajasthan 342003, India
| | - Swapnil Sharma
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, Rajasthan 304022, India
| | - Bhupendra G Prajapati
- Shree S. K. Patel College of Pharmaceutical Education and Research, Ganpat University, Kherva, Gujarat 384012, India; Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura 140401 Punjab, India.
| | | | - Pornsak Sriamornsak
- Department of Industrial Pharmacy, Faculty of Pharmacy, Silpakorn University, Nakhon Pathom 73000, Thailand; Academy of Science, The Royal Society of Thailand, Bangkok 10300, Thailand; Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu 602105, India.
| |
Collapse
|
116
|
Wei Z, Li X, Zhou J, Zhou Y, Xiao Z, Yang Q, Liu X, Peng Y, Yang Y, Ding Y, Ru Z, Wang Y, Yang M, Yang X. Inhibition of miRNA-365-2-5p Targeting SIRT1 Regulates Functions of Keratinocytes to Enhance Wound Healing. FASEB J 2025; 39:e70560. [PMID: 40261275 DOI: 10.1096/fj.202401124rrr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 04/01/2025] [Accepted: 04/14/2025] [Indexed: 04/24/2025]
Abstract
The development of drugs to accelerate wound healing is an important area of clinical research. Recent advancements have highlighted the prospects of microRNAs as therapeutic targets for various disorders, although their involvement in mice wound healing remains unclear. Peptides have been proved to be unique and irreplaceable molecules in the elucidation of competing endogenous RNAs mechanisms (ceRNA) involved with skin wound healing. In the present work, CyRL-QN15, a peptide characterized by its minimal length and maximal wound healing efficacy, was applied as a probe to explore the ceRNA mechanism in regard to accelerated wound healing. Results showed that the use of CyRL-QN15 significantly reduced the expression of miRNA-365-2-5p at the wound in mice. In mouse keratinocytes, miRNA-365-2-5p inhibition increased SIRT1 and FOXO1 protein expression and decreased STAT2 protein expression, promoting cell proliferation, migration, and reducing inflammatory factors. Similarly, inhibiting miRNA-365-2-5p at mouse wounds promoted Full-thickness injured skin wounds healing, increased SIRT1 and FOXO1 protein expression, decreased STAT2 protein expression, and reduced inflammatory factors. Overall, these findings demonstrate that miRNA-365-2-5p serves a crucial function in the biological processes underlying cutaneous wound healing in mice, offering a novel target for future therapeutic interventions in wound healing.
Collapse
Affiliation(s)
- Ziqi Wei
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, Yunnan, China
| | - Xingguo Li
- Department of Orthopedics, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Jinyi Zhou
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, Yunnan, China
| | - Yuxuan Zhou
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, Yunnan, China
| | - Zhaoxun Xiao
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, Yunnan, China
| | - Qian Yang
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, Yunnan, China
| | - Xin Liu
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, Yunnan, China
| | - Ying Peng
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, Yunnan, China
| | - Yuliu Yang
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, Yunnan, China
| | - Yujing Ding
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, Yunnan, China
| | - Zeqiong Ru
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, Yunnan, China
| | - Ying Wang
- Key Laboratory of Chemistry in Ethnic Medicinal Resources, State Ethnic Affairs Commission & Ministry of Education, School of Ethnic Medicine, Yunnan Minzu University, Kunming, Yunnan, China
| | - Meifeng Yang
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, Yunnan, China
| | - Xinwang Yang
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, Yunnan, China
| |
Collapse
|
117
|
Ghasemi N, Azizi H, Qorbanee A, Skutella T. From unipotency to pluripotency: deciphering protein networks and signaling pathways in the generation of embryonic stem-like cells from murine spermatogonial stem cells. BMC Genomics 2025; 26:426. [PMID: 40307702 PMCID: PMC12042637 DOI: 10.1186/s12864-025-11612-y] [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: 10/19/2024] [Accepted: 04/17/2025] [Indexed: 05/02/2025] Open
Abstract
With the significant challenges in using human embryonic stem cells (ESCs) for research and clinical applications, there is a growing impetus to seek alternative pluripotent cell sources. Embryonic stem-like (ES-like) cells emerge as a promising avenue in this pursuit. Our research demonstrates the potential for deriving ES-like cells from spermatogonial stem cells (SSCs) in a time-dependent manner under defined culture conditions. To better understand this process, we investigated the gene expression dynamics and underlying pathways associated with ES-like cell generation from SSCs. A deeper understanding of the signaling pathways underlying this biological process can lead us to refine protocols for ES-like cell generation, which could catalyze the development of more efficient and expedited methodologies inspired by the derivation pathway for future research in regenerative medicine. To identify differentially expressed genes (DEGs), we analyzed publicly available microarray data from murine cells obtained from the Gene Expression Omnibus (GEO). This analysis enabled the prediction of protein-protein interactions (PPIs), which were subsequently used for pathway enrichment analysis to identify biologically relevant pathways. Complementing these computational findings, we conducted in vitro experiments, including Fluidigm qPCR and immunostaining. These experiments serve as validation for our microarray data and the DEGs identified, providing reassurance about the reliability of our research. Among the identified enriched pathways in our investigation are the Toll-like receptor (TLR), GDNF/RET, interleukins (ILs), FGF/FGFR, and SMAD signaling pathway, along with the activation of NIMA kinases. Additionally, miR-410-3p, miRNA let-7e, Miat, and Xist are among some of the predicted non-coding RNAs.
Collapse
Affiliation(s)
- Nima Ghasemi
- Department of Applied Biotechnology and System Biology, College of Biotechnology, Amol University of Special Modern Technologies, Amol, Iran
| | - Hossein Azizi
- Department of Stem Cells and Cancer, College of Biotechnology, Amol University of Special Modern Technologies, P.O. Box 49767, Amol, Iran.
| | - Ali Qorbanee
- Department of Surgery, Faculty of General of Medicine, Koya University, Koya, Kurdistan Region FR, KOY45, Iraq
| | - Thomas Skutella
- Institute for Anatomy and Cell Biology, Medical Faculty, University of Heidelberg, Im Neuenheimer Feld 307, Heidelberg, 69120, Germany
| |
Collapse
|
118
|
Ni X, Xu N, Shen J, Wan X, Shen S, Lei C, Wang S, Wang B, Shan X. SVF-gel application for the alleviation of full-thickness skin graft contraction: an experimental study in mice. Sci Rep 2025; 15:15082. [PMID: 40301501 PMCID: PMC12041231 DOI: 10.1038/s41598-025-99649-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 04/22/2025] [Indexed: 05/01/2025] Open
Abstract
Skin grafts often suffer from contracture, complicating recovery. SVF-gel shows promise in addressing scar contracture, but its therapeutic effects and mechanisms are not fully understood. This study evaluates the efficacy of SVF-gel in full-thickness skin grafting. Full-thickness skin grafts were harvested from mice dorsal skin, rotated and sutured. SVF-gel or saline was injected beneath the muscle fascia. Immunohistochemistry assessed SVF-gel's effects on angiogenesis, collagen deposition, fibrosis, and dermal adipocytes. Keloid-related genes from GSE92566 and GSE158395 were analyzed for functional enrichment and protein-protein interactions, with hub genes validated using GSE190626. SVF-gel significantly increased the grafted area, thickness of the epidermal and dermal adipose layers, and hair follicles compared to the control group. SVF-gel enhanced CD31 and perilipin expression, decreased α-SMA expression, and identified HLA+ cells around CD31+ cells in the dermal microvessels and adipose tissue of the graft. Sixty commonly downregulated keloid-related genes were identified, with KEGG pathway analysis indicating enrichment in the PPAR signaling pathway and lipolysis regulation. Five hub genes (ADIPOQ, FABP4, KRT7, LEP, and PIP) were validated. SVF-gel shows promise as a stem cell therapy for skin grafts, improving outcomes by enhancing revascularization, increasing collagen fiber density and regularity, accelerating myofibroblast turnover, promoting adipogenesis, and increasing hair follicles.
Collapse
Affiliation(s)
- Xuejun Ni
- Department of Plastic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
- Department of Plastic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Nating Xu
- Department of Burn and Plastic Surgery, Fujian Provincial Hospital, Fuzhou, 350001, China
| | - Jian Shen
- Department of Burn and Plastic Surgery, Ma'anshan People's Hospital, Ma'anshan, 243000, China
| | - Xiaofen Wan
- Department of Plastic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
| | - Shuyi Shen
- Department of Dermatology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
| | - Chen Lei
- Department of Plastic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
- Department of Plastic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Songyu Wang
- Department of Dermatology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
| | - Biao Wang
- Department of Plastic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China.
- Department of Plastic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China.
| | - Xiuying Shan
- Department of Plastic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China.
- Department of Plastic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China.
| |
Collapse
|
119
|
Rahmawati FN, Takakura N. Development and aging of resident endothelial stem cells in pre-existing blood vessels. Exp Hematol 2025:104795. [PMID: 40311858 DOI: 10.1016/j.exphem.2025.104795] [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: 02/05/2025] [Revised: 04/09/2025] [Accepted: 04/13/2025] [Indexed: 05/03/2025]
Abstract
Organ-specific somatic stem cells play an important role in supporting tissue turnover and facilitating regeneration upon injury. Hematopoietic stem cells are one of the most established organ-specific somatic cells that have been frequently used for transplantation therapy. In recent years, there has been a growing interest in other organ-specific somatic cells, including vascular endothelial stem cells (VESCs). We have previously reported on the use of CD157 and CD200 as markers to isolate VESCs from adult mouse organs, particularly the liver. In this review, we aim to summarize, based on our previous research, how CD157⁺CD200⁺ VESCs in the liver develop from the fetal stage to postnatal life, what transcriptional regulatory mechanisms govern them, and how VESCs change with aging.
Collapse
Affiliation(s)
- Fitriana N Rahmawati
- Department of Signal Transduction, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan.
| | - Nobuyuki Takakura
- Department of Signal Transduction, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan; Immunology Frontier Research Center, Osaka University, Suita, Japan; Center for Infectious Disease Education and Research, Osaka University, Suita, Japan; Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University.
| |
Collapse
|
120
|
Echeverría-Altamar K, Barreto-Gamarra C, Domenech-García M, Resto-Irizarry P. Prediction of cardiac differentiation in human induced pluripotent stem cell-derived cardiomyocyte supernatant using surface-enhanced Raman spectroscopy and machine learning. Biosens Bioelectron 2025; 283:117528. [PMID: 40339557 DOI: 10.1016/j.bios.2025.117528] [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: 02/06/2025] [Revised: 03/24/2025] [Accepted: 04/28/2025] [Indexed: 05/10/2025]
Abstract
The efficient manufacturing of cardiomyocytes from human-induced pluripotent stem cells (hiPSCs) is essential for advancing regenerative therapies for myocardial injuries. However, ensuring cell quality during production is challenging since traditional methods are invasive, destructive, and time-consuming. In this study, we monitored cardiomyocyte differentiation of WTC11 hiPSCs by analyzing conditioned media collected at various stages using Raman spectroscopy, multivariate analysis, and machine learning. Differentiation efficiency was confirmed via flow cytometry and immunostaining. Raman spectra were processed using standard normal variate and second derivative transformations before performing a principal component analysis (PCA) and machine learning (Random Forest, K-Nearest Neighbors, and Deep Neural Networks [DNN]). Results show that PCA was unable to distinguish cells based on differentiation stages, while machine learning could reliably predict cell differentiation early in the cardiac cell manufacturing process. DNN models achieved accuracies exceeding 82 % in predicting differentiation, highlighting their potential as quality control tools. These findings underscore the potential of Raman spectroscopy coupled with machine learning as a tool for real-time monitoring of cardiomyocyte production.
Collapse
Affiliation(s)
- Karla Echeverría-Altamar
- Bioengineering Graduate Program, University of Puerto Rico at Mayagüez, Mayagüez, 00680, Puerto Rico
| | - Carlos Barreto-Gamarra
- Chemical Engineering Department, University of Puerto Rico at Mayagüez, Mayagüez, 00680, Puerto Rico
| | - Maribella Domenech-García
- Bioengineering Graduate Program, University of Puerto Rico at Mayagüez, Mayagüez, 00680, Puerto Rico; Chemical Engineering Department, University of Puerto Rico at Mayagüez, Mayagüez, 00680, Puerto Rico
| | - Pedro Resto-Irizarry
- Bioengineering Graduate Program, University of Puerto Rico at Mayagüez, Mayagüez, 00680, Puerto Rico; Mechanical Engineering Department, University of Puerto Rico at Mayagüez, Mayagüez, 00680, Puerto Rico.
| |
Collapse
|
121
|
Kalındemirtaş FD, Cilasun GE, Kariper A. Enhanced therapeutic efficacy of platinum-doxorubicin nanoparticles on colon and breast cancer cell lines. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04080-4. [PMID: 40299021 DOI: 10.1007/s00210-025-04080-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Accepted: 03/19/2025] [Indexed: 04/30/2025]
Abstract
In this study, platinum nanoparticles (PtNPs) were synthesized and their potential to improve the efficacy of doxorubicin (DOX) in cancer treatment was investigated. H2PtCl6, LiAlH4, and trisodium citrate were used during the synthesis of PtNPs. They were characterized using dynamic light scattering (DLS), Fourier transform infrared spectroscopy (FTIR), and scanning transmission electron microscopy (STEM). The diameter of the PtNPs was measured to be 21.72 nm without DOX loading and approximately 212 nm after DOX loading (DOX-PtNPs). FTIR confirmed the binding of DOX to PtNPs. In addition, MTT assays showed that DOX-PtNPs have a stronger effect on MCF-7 and HCT116 cancer cells than free DOX, even at low doses. The IC50 value for MCF-7 cells treated with DOX was determined to be 4.81 µg/ml, while it was significantly lower for the DOX-PtNP group at 0.64 µg/ml. A similar trend was observed in HCT116 cells, where the IC50 value for DOX was 5.03 µg/ml, while for DOX-PtNPs it was 0.62 µg/ml. In summary, the activity of DOX in these cells was increased approximately eightfold by PtNPs. Moreover, DOX-PtNPs showed no significant cytotoxic effects on normal HUVEC cells at low doses. Moreover, DOX-PtNPs enhanced apoptotic activity in HCT116 cells without inducing drug resistance as demonstrated by Rho123 staining and annexin/PI analyses. The significance of this study lies in the pioneering use of DOX-PtNPs in colon cancer, the synthesis of smaller PtNPs, the eightfold increase in the efficacy of DOX, and the demonstration that DOX-PtNPs do not significantly increase drug resistance.
Collapse
Affiliation(s)
| | - Gökçe Erdemir Cilasun
- Department of Medical Biology, Faculty of Medicine, Biruni University, Istanbul, Turkey
| | - Afşin Kariper
- Department of Science Education, Education Faculty, Erciyes University, Kayseri, Turkey
| |
Collapse
|
122
|
Song L, Gao F, Man J. Ferroptosis: the potential key roles in idiopathic pulmonary fibrosis. Eur J Med Res 2025; 30:341. [PMID: 40296070 PMCID: PMC12036158 DOI: 10.1186/s40001-025-02623-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 04/21/2025] [Indexed: 04/30/2025] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic progressive interstitial lung disease characterized by recurrent injury to alveolar epithelial cells, epithelial-mesenchymal transition, and fibroblast activation, which leads to excessive deposition of extracellular matrix (ECM) proteins. However, effective preventative and therapeutic interventions are currently lacking. Ferroptosis, a unique form of iron-dependent lipid peroxidation-induced cell death, exhibits distinct morphological, physiological, and biochemical features compared to traditional programmed cell death. Recent studies have revealed a close relationship between iron homeostasis and the pathogenesis of pulmonary interstitial fibrosis. Ferroptosis exacerbates tissue damage and plays a crucial role in regulating tissue repair and the pathological processes involved. It leads to recurrent epithelial injury, where dysregulated epithelial cells undergo epithelial-mesenchymal transition via multiple signaling pathways, resulting in the excessive release of cytokines and growth factors. This dysregulated environment promotes the activation of pulmonary fibroblasts, ultimately culminating in pulmonary fibrosis. This review summarizes the latest advancements in ferroptosis research and its role in the pathogenesis and treatment of IPF, highlighting the significant potential of targeting ferroptosis for IPF management. Importantly, despite the rapid developments in this emerging research field, ferroptosis studies continue to face several challenges and issues. This review also aims to propose solutions to these challenges and discusses key concepts and pressing questions for the future exploration of ferroptosis.
Collapse
Affiliation(s)
- Longfei Song
- Department of Rehabilitation Medicine, Affiliated Hospital of Shandong Second Medical University, No. 2428 Yuhe Road, Kuiwen District, Weifang City, 261041, Shandong Province, China
| | - Fusheng Gao
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Shandong Second Medical University, No. 2428, Yuhe Road, Kuiwen District, Weifang City, 261041, Shandong Province, China
| | - Jun Man
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Shandong Second Medical University, No. 2428, Yuhe Road, Kuiwen District, Weifang City, 261041, Shandong Province, China.
- Clinical Research Center, Affiliated Hospital of Shandong Second Medical University, No. 4948, Shengli East Street, Kuiwen District, Weifang City, 261041, Shandong Province, China.
| |
Collapse
|
123
|
Sleep SL, Ranjit E, Gunter J, Hryciw DH, Arany P, George R. Mitochondrial Oxygen Consumption and Immunocytochemistry of Human Dental Pulp Stem Cell Following 808 nm PBM Therapy: A 3D Cell Culture Study. JOURNAL OF BIOPHOTONICS 2025:e70051. [PMID: 40294940 DOI: 10.1002/jbio.70051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 03/27/2025] [Accepted: 04/14/2025] [Indexed: 04/30/2025]
Abstract
This study investigated the impact of 808 nm laser photobiomodulation (PBM) on mitochondrial respiration and osteogenic protein expression (OCN, OPN, ALP, RUNX2, COL-1, BMP-2) in human dental pulp stem cells (hDPSCs) within a 3D hydrogel model. hDPSCs were isolated from third molars and maintained under hypoxic conditions. Cells received PBM at 5 and 15 J/cm2 using an 808 nm diode laser. The study showed that 808 nm PBM can alter mitochondrial respiration, with 5 J/cm2 enhancing osteogenic protein expression (OCN, ALP, OPN, RUNX2) but failing to sustain BMP-2 at 24 h. In contrast, 15 J/cm2 induced stronger upregulation and prolonged BMP-2 expression, suggesting an optimal dose for sustained osteogenic activity. BMP-2 was later downregulated, and COL-1 remained unchanged post-PBM. Importantly, this study indicates the dose-specific PBM modulation of mitochondrial respiration and protein expression, but further research is required to optimize treatment protocols.
Collapse
Affiliation(s)
- Simone L Sleep
- School of Medicine and Dentistry, Griffith University, Southport, Australia
| | - Eliza Ranjit
- School of Medicine and Dentistry, Griffith University, Southport, Australia
| | - Jennifer Gunter
- Australian Prostate Cancer Research Centre, QLD, Centre for Genomics and Personalised Health, School of Biomedical Science, Queensland University of Technology, Translational Research Institute, Brisbane, Australia
| | - Deanne H Hryciw
- School of Environment and Science, Griffith University, Nathan, Australia
- Institute for Biomedicine and Glycomics, Griffith University, Nathan, Australia
| | - Praveen Arany
- Oral Biology, Surgery, and Biomedical Engineering, University at Buffalo, Buffalo, New York, USA
| | - Roy George
- School of Medicine and Dentistry, Griffith University, Southport, Australia
| |
Collapse
|
124
|
Zhang R, Nie P, Zhou Y, He J, Wang L, Xu H, Fu F. Maternal co-exposure to Cd and PS-NPLs induces offspring ovarian inflammatory aging via promoting M1 macrophage polarization. Chem Biol Interact 2025; 416:111535. [PMID: 40306534 DOI: 10.1016/j.cbi.2025.111535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 04/13/2025] [Accepted: 04/27/2025] [Indexed: 05/02/2025]
Abstract
With the growing crisis of plastic pollution, the severe environmental problem threatens human and ecosystem health. And nanoplastics can carry other environmental contaminants, thereby causing severe toxic effects. Cadmium (Cd), as a metal, has been widely concerned because of its long biological half-life, high toxicity and low excretion rate. Additionally, Cd as an endocrine disrupting chemical also has reproductive toxicity and genotoxicity. Studies have found that co-exposed to Cd and PS-NPLs complexes may lead to more severe adverse effects in aquatic and mammals. And the utilization of other non-essential heavy metals such as cadmium are increased due to iron deficiency anemia in women during pregnancy. Therefore, in our study, 8 week-old female C57BL/6 J mice were co-exposed to Cd and NPLs during pregnancy and lactation, and the offspring are raised to four weeks to explore and predict the potential effect on the development of offspring reproductive system by transcriptomics. The results showed that sex hormone levels were interfered in offspring female mice, and gut microbiota disorder and increased LPS levels originated from the mother, which activating TLR4-related signaling pathways, and promoting ovarian M1 macrophage polarization, thereby increasing the risk of ovarian inflammatory aging in female offspring.
Collapse
Affiliation(s)
- Ruiying Zhang
- The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330000, China; State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, 330047, China
| | - Penghui Nie
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, 330047, China
| | - Yuankun Zhou
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, 330047, China
| | - Juanjuan He
- The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330000, China; State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, 330047, China
| | - Lihong Wang
- The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330000, China
| | - Hengyi Xu
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, 330047, China.
| | - Fen Fu
- The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330000, China.
| |
Collapse
|
125
|
Wang J, An H, Tao N. Association of non-insulin-based insulin resistance indices, mean platelet volume and prostate cancer: a cross-sectional study. BMC Cancer 2025; 25:795. [PMID: 40295970 PMCID: PMC12039131 DOI: 10.1186/s12885-025-13839-0] [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: 01/05/2025] [Accepted: 02/28/2025] [Indexed: 04/30/2025] Open
Abstract
PURPOSE Insulin resistance and prostate cancer (PCa) association results remain controversial. However, few studies have compared the role of various non-insulin-based insulin resistance (NI-IR) indices and mean platelet volume (MPV) in PCa. METHODS We conducted a cross-sectional study, the case group included 354 patients with PCa, and the control group included 1,498 non-PCa participants. We performed inverse probability weighting to reduce the impact of differences in baseline information between the case and control groups on results. Weighted logistic regression analysis for assessing the relationship between NI-IR indices and PCa risk. Fitting 4-point restricted cubic spline (RCS) plots to show the trend of NI-IR indices with PCa risk. The interaction between insulin resistance and platelet volume based on generalized additive model (GAM) to reveal the impact of the interaction between insulin resistance and cardiovascular risk on PCa. In the end, we performed three sensitivity analyses to verify the stability of results. RESULTS Weighted logistic regression analysis revealed that all NI-IR indices were associated with PCa. When NI-IR indices were evaluated as continuous variables, in the all variables adjusted model (model 3), the adjusted OR of ZJU index was 1.337 (95%CI: 1.296-1.379), the adjusted OR of TyG index was 5.300 (95%CI:4.208-6.675), the adjusted OR of TG/HDL-c was 1.431 (95%CI:1.335-1.534), and the adjusted OR of METS-IR was 1.129 (95%CI:1.110-1.149). When NI-IR indices were analyzed as categorical variables, also in model 3, using Q1 as reference, the adjusted OR of ZJU index in Q5 was 15.592 (95%CI:10.809-22.492), the adjusted OR of TyG index in Q5 was 7.306 (95%CI:5.182-10.301), the adjusted OR of TG/HDL-c in Q5 was 4.790 (95%CI:3.459-6.632), and the adjusted OR of METS-IR in Q5 was 9.844 (95%CI:6.862-14.121). RCS displayed that PCa risk tended to increase as the ZJU index, TyG index, TG/HDL-c, and METS-IR increased. The interaction test based on the GAM indicated that the value of the interaction between TG/HDL-c and MPV on the PCa risk was χ2 = 6.924(P = 0.009). With the increase in TG/HDL-c and the decrease in MPV, the PCa risk progressively increases. The sensitivity analysis further confirmed the robustness of the results. CONCLUSIONS NI-IR indices were associated with an increased PCa risk. The interaction between MPV and insulin resistance may further contribute to the PCa risk.
Collapse
Affiliation(s)
- Jinru Wang
- College of Public Health, Xinjiang Medical University, Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Hengqing An
- Department of Urology, The First Affiliated Hospital of Xinjiang Medical University, Xinjiang Uygur Autonomous Region, Urumqi, China.
| | - Ning Tao
- College of Public Health, Xinjiang Medical University, Xinjiang Uygur Autonomous Region, Urumqi, China.
| |
Collapse
|
126
|
Cipta NO, Zeng Y, Wong KW, Zheng ZH, Yi Y, Warrier T, Teo JZ, Teo JHJ, Kok YJ, Bi X, Taneja R, Ong DST, Xu J, Ginhoux F, Li H, Liou YC, Loh YH. Rewiring of SINE-MIR enhancer topology and Esrrb modulation in expanded and naive pluripotency. Genome Biol 2025; 26:107. [PMID: 40296153 PMCID: PMC12036290 DOI: 10.1186/s13059-025-03577-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 04/12/2025] [Indexed: 04/30/2025] Open
Abstract
BACKGROUND The interplay between 3D genomic structure and transposable elements (TE) in regulating cell state-specific gene expression program is largely unknown. Here, we explore the utilization of TE-derived enhancers in naïve and expanded pluripotent states by integrative analysis of genome-wide Hi-C-defined enhancer interactions, H3K27ac HiChIP profiling and CRISPR-guided TE proteomics landscape. RESULTS We find that short interspersed nuclear elements (SINEs) are the more involved TEs in the active chromatin and 3D genome architecture. In particular, mammalian-wide interspersed repeat (MIR), a SINE family member, is highly associated with naïve-specific genomic interactions compared to the expanded state. Primarily, in the naïve pluripotent state, MIR enhancer is co-opted by ESRRB for naïve-specific gene expression program. This ESRRB and MIR enhancer interaction is crucial for the formation of loops that build a network of enhancers and super-enhancers regulating pluripotency genes. We demonstrate that loss of a ESRRB-bound MIR enhancer impairs self-renewal. We also find that MIR is co-bound by structural protein complex, ESRRB-YY1, in the naïve pluripotent state. CONCLUSIONS Altogether, our study highlights the topological regulation of ESRRB on MIR in the naïve potency state.
Collapse
Affiliation(s)
- Nadia Omega Cipta
- Epigenetics and Cell Fates Laboratory, Cell Fate Engineering and Therapeutics Laboratory, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive Proteos, Singapore, 138673, Singapore
- Department of Biological Sciences, Faculty of Science, National University of Singapore, 14 Science Drive 4, Singapore, 117543, Singapore
| | - Yingying Zeng
- Epigenetics and Cell Fates Laboratory, Cell Fate Engineering and Therapeutics Laboratory, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive Proteos, Singapore, 138673, Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Ka Wai Wong
- Epigenetics and Cell Fates Laboratory, Cell Fate Engineering and Therapeutics Laboratory, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive Proteos, Singapore, 138673, Singapore
| | - Zi Hao Zheng
- Epigenetics and Cell Fates Laboratory, Cell Fate Engineering and Therapeutics Laboratory, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive Proteos, Singapore, 138673, Singapore
- Department of Physiology, NUS Yong Loo Lin School of Medicine, 2 Medical Drive, MD9, Singapore, Singapore
| | - Yao Yi
- Epigenetics and Cell Fates Laboratory, Cell Fate Engineering and Therapeutics Laboratory, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive Proteos, Singapore, 138673, Singapore
| | - Tushar Warrier
- Epigenetics and Cell Fates Laboratory, Cell Fate Engineering and Therapeutics Laboratory, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive Proteos, Singapore, 138673, Singapore
| | - Jian Zhou Teo
- Epigenetics and Cell Fates Laboratory, Cell Fate Engineering and Therapeutics Laboratory, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive Proteos, Singapore, 138673, Singapore
| | - Jia Hao Jackie Teo
- Epigenetics and Cell Fates Laboratory, Cell Fate Engineering and Therapeutics Laboratory, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive Proteos, Singapore, 138673, Singapore
- Department of Physiology, NUS Yong Loo Lin School of Medicine, 2 Medical Drive, MD9, Singapore, Singapore
| | - Yee Jiun Kok
- Proteomics Group, Agency for Science, Technology and Research (A*STAR), Bioprocessing Technology Institute (BTI), Singapore, 138668, Singapore
| | - Xuezhi Bi
- Proteomics Group, Agency for Science, Technology and Research (A*STAR), Bioprocessing Technology Institute (BTI), Singapore, 138668, Singapore
| | - Reshma Taneja
- Department of Physiology, NUS Yong Loo Lin School of Medicine, 2 Medical Drive, MD9, Singapore, Singapore
| | - Derrick Sek Tong Ong
- Department of Physiology, NUS Yong Loo Lin School of Medicine, 2 Medical Drive, MD9, Singapore, Singapore
- NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Jian Xu
- Department of Pathology, Center of Excellence for Leukemia Studies, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Florent Ginhoux
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- INSERM U1015, Paris Saclay University, Gustave Roussy Cancer Campus, Villejuif, France
- Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Hu Li
- Department of Molecular Pharmacology & Experimental Therapeutics, Center for Individualized Medicine, Mayo Clinic, Rochester, MN, 55905, USA
| | - Yih-Cherng Liou
- Department of Biological Sciences, Faculty of Science, National University of Singapore, 14 Science Drive 4, Singapore, 117543, Singapore.
- NUS Graduate School's Integrative Sciences and Engineering Programme, National University of Singapore, 28 Medical Drive, Singapore, Singapore.
| | - Yuin-Han Loh
- Epigenetics and Cell Fates Laboratory, Cell Fate Engineering and Therapeutics Laboratory, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive Proteos, Singapore, 138673, Singapore.
- Department of Biological Sciences, Faculty of Science, National University of Singapore, 14 Science Drive 4, Singapore, 117543, Singapore.
- Department of Physiology, NUS Yong Loo Lin School of Medicine, 2 Medical Drive, MD9, Singapore, Singapore.
- NUS Graduate School's Integrative Sciences and Engineering Programme, National University of Singapore, 28 Medical Drive, Singapore, Singapore.
| |
Collapse
|
127
|
Ma C, Cheng M, Xiao T, Qi R, Gao X, Chen H, Wu Y, Xu X. Mir-574-3p and Mir-125a-5p in Adipose Derived Mesenchymal Stem Cell Exosomes Synergistically Target TGF-β1/Smad2 Signaling Pathway for the Treatment of Androgenic Alopecia. J Invest Dermatol 2025:S0022-202X(25)00448-8. [PMID: 40306482 DOI: 10.1016/j.jid.2025.03.042] [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: 11/11/2024] [Revised: 03/27/2025] [Accepted: 03/29/2025] [Indexed: 05/02/2025]
Abstract
Androgenetic alopecia (AGA) is the most common hair loss disorder, influenced by distinct genetic factors and intricate environmental factors. The exosomes (Exo) from adipose-derived mesenchymal stem cells (ADMSCs) have diverse effects, including the promotion of cell proliferation, inhibition of apoptosis, analgesia, and enhancement of wound healing. The miRNAs are essential components of the paracrine secretion of ADMSCs-Exo. This study revealed that ADMSCs-Exo could counteract the impairment of dermal papilla cells (DPCs) induced by dihydrotestosterone (DHT) by inhibiting the TGF-β1 signaling pathway`s activation. Two miRNAs, miR-574-3p and miR-125a-5p, were identified as being predominantly expressed and specifically targeting TGF-β1 and SMAD2, respectively. Notably, individually knocking down miR-574-3p or miR-125a-5p did not affect the therapeutic efficacy of ADMSCs-Exo. Yet, when both miR-574-3p and miR-125a-5p were concurrently knocked down, the efficacy of ADMSCs-Exo was markedly reduced. Ultimately, our findings indicate that ADMSCs-Exo target the TGF-β1/SMAD2 signaling pathway via miR-574-3p and miR-125a-5p, which are integral to the therapeutic action of ADMSCs-Exo on DHT-induced DPCs and AGA murine models. This discovery offers significant insights into the pathogenesis of AGA and suggests potential therapeutic approaches. Further investigation into the role and interaction of these miRNAs in ADMSCs-Exo may lead to the development of more precise and efficacious treatments for AGA.
Collapse
Affiliation(s)
- Cong Ma
- Department of Dermatology, The Affiliated Hospital of Inner Mongolia University for the Nationalities, Tongliao, China; Department of Dermatology, The First Hospital of China Medical University, Shenyang, China; National joint Engineering Research Center for Theranostics of Immunological Skin Diseases, The First Hospital of China Medical University and Key Laboratory of Immunodermatology, Ministry of Health and Ministry of Education, Shenyang, China
| | - Ming Cheng
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, China; National joint Engineering Research Center for Theranostics of Immunological Skin Diseases, The First Hospital of China Medical University and Key Laboratory of Immunodermatology, Ministry of Health and Ministry of Education, Shenyang, China
| | - Ting Xiao
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, China; National joint Engineering Research Center for Theranostics of Immunological Skin Diseases, The First Hospital of China Medical University and Key Laboratory of Immunodermatology, Ministry of Health and Ministry of Education, Shenyang, China
| | - Ruiqun Qi
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, China; National joint Engineering Research Center for Theranostics of Immunological Skin Diseases, The First Hospital of China Medical University and Key Laboratory of Immunodermatology, Ministry of Health and Ministry of Education, Shenyang, China
| | - Xinghua Gao
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, China; National joint Engineering Research Center for Theranostics of Immunological Skin Diseases, The First Hospital of China Medical University and Key Laboratory of Immunodermatology, Ministry of Health and Ministry of Education, Shenyang, China
| | - Hongduo Chen
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, China; National joint Engineering Research Center for Theranostics of Immunological Skin Diseases, The First Hospital of China Medical University and Key Laboratory of Immunodermatology, Ministry of Health and Ministry of Education, Shenyang, China
| | - Yan Wu
- Department of Dermatology, Shengjing Hospital of China Medical University, Shenyang, China; National joint Engineering Research Center for Theranostics of Immunological Skin Diseases, The First Hospital of China Medical University and Key Laboratory of Immunodermatology, Ministry of Health and Ministry of Education, Shenyang, China.
| | - Xuegang Xu
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, China; National joint Engineering Research Center for Theranostics of Immunological Skin Diseases, The First Hospital of China Medical University and Key Laboratory of Immunodermatology, Ministry of Health and Ministry of Education, Shenyang, China.
| |
Collapse
|
128
|
Huang TL, Chang YC, Kuo WW, Kao SW, Kuo CH, Hsieh DJY, Lin KH, Ho TJ, Huang CY. Trans-anethole enhances mesenchymal stem cell derived exosomes function to inhibit H 2O 2-induced rheumatoid arthritis-like inflammation in HIG-82 synovial cells. Mol Biol Rep 2025; 52:431. [PMID: 40293555 DOI: 10.1007/s11033-025-10426-1] [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: 01/12/2025] [Accepted: 03/10/2025] [Indexed: 04/30/2025]
Abstract
BACKGROUND Rheumatoid arthritis (RA) is an auto-immune inflammatory disorder for which an effective cure is yet to be found. Trans-anethole (1-methoxy-4-(1E)-1-propen-1-yl-benzene), a key bioactive compound derived from the perennial plant Foeniculum vulgare, exerts multiple medicinal benefits. In this study, we investigated the therapeutic potential of exosomes derived from anethole-preconditioned human Wharton Jelly-derived mesenchymal stem cells (hWJMSCs) against RA-like inflammation in H2O2-treated synoviocyte HIG-82 cells. METHODS The fennel samples were prepared and trans-anethole was purified using LC-ESI-MS/MS analysis. The MTT cell viability assays, hWJMSC derived exosomes, and expression analysis of cellular markers related to proliferation, stemness, apoptosis, and extracellular matrix (ECM)-degrading proteases were performed using Western blotting in HIG-82 cells. RESULTS The results showed that anethole treatment significantly increased cell viability and expression of the MSC marker CD90 in a dose-dependent manner in HIG-82 cells. Cell stemness markers, including proliferation markers cyclin-D, proliferating cell nuclear antigen (PCNA), and minichromosome maintenance complex component 2 (MCM2) were enhanced, whereas p53 and p21 were decreased by anethole. Exosomes derived from anethole-preconditioned hWJMSCs significantly improved the cell viability of H2O2-treated HIG-82 cells. Anethole- preconditioned exosomes decreased ECM-degrading proteases MMP-13, ADAMTS-2, -8, and -17, and AQP-3 expression more significantly than exosomes without preconditioned hWJMSC. Bcl-2 was increased, whereas Bax, Cyto c, and c-caspase 3 were decreased by preconditioned exosomes more prominently than exosomes from without preconditioned hWJMSCs in H2O2-treated HIG-82 cells. CONCLUSION Together, the study showed that exosomes derived from anethole-preconditioned hWJMSC have a greater potential to inhibit RA-like inflammation and apoptosis in H2O2-treated HIG-82 cells.
Collapse
Affiliation(s)
- Tai-Lung Huang
- Department of Orthopedics, Chung-Kang Branch, Cheng Ching General Hospital, Taichung, Taiwan
| | - Yu-Chun Chang
- Department of Biological Science and Technology, College of Life Sciences, China Medical University, Taichung, Taiwan
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Tzu Chi University of Science and Technology, Hualien, Taiwan
| | - Wei-Wen Kuo
- Department of Biological Science and Technology, College of Life Sciences, China Medical University, Taichung, Taiwan
- Program for Biotechnology Industry, China Medical University, Taichung, Taiwan
| | - Shih-Wen Kao
- Graduate Institute of Aging Medicine, China Medical University, Taichung, Taiwan
- Department of Orthopedic Surgery, Chung-Shan Medical University Hospital, Taichung, Taiwan
| | - Chia-Hua Kuo
- Department of Sports Sciences, University of Taipei, Taipei, Taiwan
| | - Dennis Jine-Yuan Hsieh
- School of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung, Taiwan
- Clinical Laboratory, Chung Shan Medical University Hospital, Taichung, 402, Taiwan
| | - Kuan-Ho Lin
- Department of Emergency Medicine, China Medical University Hospital, Taichung, Taiwan
- College of Medicine, China Medical University, Taichung, Taiwan
| | - Tsung-Jung Ho
- School of Post-Baccalaureate Chinese Medicine, College of Medicine, Tzu Chi University, Hualien, Taiwan.
- Department of Chinese Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Tzu Chi University, Hualien, Taiwan.
- Integration Center of Traditional Chinese and Modern Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan.
| | - Chih-Yang Huang
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Tzu Chi University of Science and Technology, Hualien, Taiwan.
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan.
- Department of Medical Laboratory Science and Biotechnology, Asia University, Taichung, Taiwan.
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan.
| |
Collapse
|
129
|
Han D, Ding L, Zheng X, Li S, Yan H, Liu J, Wang H. Proteomics and lipidomics of human umbilical cord mesenchymal stem cells exposed to ionizing radiation. Eur J Med Res 2025; 30:340. [PMID: 40296080 PMCID: PMC12036141 DOI: 10.1186/s40001-025-02578-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2025] [Accepted: 04/10/2025] [Indexed: 04/30/2025] Open
Abstract
OBJECTIVES Mesenchymal stem cell (MSC)-based therapies exhibit beneficial effects on various forms of tissue damage, including ionizing radiation-induced lesions. However, whether ionizing radiation affects the functions of human umbilical cord mesenchymal stem cells (hucMSCs) remains unclear. This study aimed to investigate the effect and possible mechanisms of ionizing radiation on the proliferation and differentiation of hucMSCs. METHODS The hucMSCs were divided into the 1 Gy group (exposure to a single dose (1 Gy) of X-ray radiation (1 Gy/min) for 14 days) and control (without radiation treatment) group. The proliferation, apoptosis, and adipogenic and osteogenic differentiation abilities of hucMSCs in the two groups were evaluated. Moreover, the lipidomics and proteomics analyses were conducted to explore crucial lipids and proteins by which ionizing radiation affected the functions of hucMSCs. In addition, the effects of BYSL on radiation-treated hucMSCs were explore, as well as the involved potential mechanisms. RESULTS X-ray radiation treatment inhibited proliferation, promoted apoptosis, and decreased adipogenic and osteogenic differentiation abilities of hucMSCs. Key lipids, such as triglyceride (TG) and phosphatidylcholine (PC), and hub proteins (BYSL, MRTO4, and RRP9) exhibited significant differences between the 1 Gy group and control group. Moreover, BYSL, MRTO4, and RRP9 were significantly correlated with TG and PC. BYSL overexpression evidently promoted the cell proliferation, adipogenic and osteogenic differentiation abilities of radiation-treated hucMSCs, as well as the protein expression levels of p-GSK-3β/GSK-3β and β-catenin, while suppressed cell apoptosis. However, the GSK-3β inhibitor (1-Az) treatment reversed the protein expression levels of p-GSK-3β/GSK-3β, β-catenin and BYSL, as well as the cell proliferation, apoptosis, adipogenic and osteogenic differentiation abilities of radiation-treated hucMSCs. CONCLUSIONS Our findings reveal that the proliferation and differentiation of hucMSCs are suppressed by radiation, which may be associated with the changes of key lipids (TG and PC) and proteins (BYSL, MRTO4, and RRP9). Furthermore, BYSL promotes adipogenic and osteogenic differentiation abilities of radiation-treated hucMSCs via GSK-3β/β-catenin pathway. These findings help explain the response of hucMSCs to radiation and have clinical implications for improving the outcomes of MSC-based therapies after radiotherapy.
Collapse
Affiliation(s)
- Dongmei Han
- Department of Hematology, Air Force Medical University, Air Force Medical Center, PLA, No.30, Fucheng Road, Beijing, 100142, China
| | - Li Ding
- Department of Hematology, Air Force Medical University, Air Force Medical Center, PLA, No.30, Fucheng Road, Beijing, 100142, China
| | - Xiaoli Zheng
- Department of Hematology, Air Force Medical University, Air Force Medical Center, PLA, No.30, Fucheng Road, Beijing, 100142, China
| | - Sheng Li
- Department of Hematology, Air Force Medical University, Air Force Medical Center, PLA, No.30, Fucheng Road, Beijing, 100142, China
| | - Hongmin Yan
- Department of Hematology, Air Force Medical University, Air Force Medical Center, PLA, No.30, Fucheng Road, Beijing, 100142, China
| | - Jing Liu
- Department of Hematology, Air Force Medical University, Air Force Medical Center, PLA, No.30, Fucheng Road, Beijing, 100142, China
| | - Hengxiang Wang
- Department of Hematology, Air Force Medical University, Air Force Medical Center, PLA, No.30, Fucheng Road, Beijing, 100142, China.
| |
Collapse
|
130
|
Bae BS, Jung JW, Jo GO, Kim SA, Go EJ, Cho ML, Shetty AA, Kim SJ. Treatment of osteoarthritic knee with high tibial osteotomy and allogeneic human umbilical cord blood-derived mesenchymal stem cells combined with hyaluronate hydrogel composite. Stem Cell Res Ther 2025; 16:211. [PMID: 40296133 PMCID: PMC12038988 DOI: 10.1186/s13287-025-04356-9] [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: 12/19/2024] [Accepted: 04/22/2025] [Indexed: 04/30/2025] Open
Abstract
BACKGROUND Delaying total knee arthroplasty is crucial for middle-aged patients with severe osteoarthritis. The long-term outcomes of high tibial osteotomy (HTO) remain uncertain. Recently, mesenchymal stem cells (MSCs) have shown promising potential in enhancing cartilage regeneration. Therefore, this study aimed to assess cartilage regeneration following the implantation of allogeneic human umbilical cord blood-derived mesenchymal stem cells (hUCB-MSCs) with HTO. METHODS In this case series, ten patients underwent hUCB-MSC implantation with HTO. The median age was 58.50 (range: 57.00-60.00) years, and the mean body mass index was 27.81 (range: 24.42-32.24) kg/m2. Clinical outcomes, including the Western Ontario and McMaster Universities Osteoarthritis Index (WOMAC), visual analog scale (VAS), Physical Component Score (PCS) and Mental Component Score (MCS) from the 36-Item Short-Form Health Survey (SF-36), were evaluated 6 months, 1 year, and 2 years postoperatively. Cartilage status of the medial femoral condyle (MFC) was assessed during hardware removal surgery, at least 2 years after the initial procedure, and compared with preoperative MFC cartilage status regarding lesion size and International Cartilage Repair Society (ICRS) grade. Radiological assessments included the Kellgren-Lawrence (KL) grading system for medial compartment osteoarthritis and hip-knee-ankle (HKA) angle. RESULTS Significant improvements were observed in WOMAC scores (preoperative: 57.00 (range: 44.75-63.00), postoperative: 27.50 (range: 22.25-28.75)), VAS scores (preoperative: 66.25 (range: 48.00-74.25), postoperative: 26.25 (range: 14.50-31.13)), SF-36 PCS (preoperative: 27.97 (range: 26.64-31.25), postoperative: 55.31 (range: 51.64-62.50)), and SF-36 MCS (preoperative: 41.04 (range: 29.95-50.96), postoperative: 63.18 (range: 53.83-65.16)) 2 years postoperatively (p = 0.002, 0.002, 0.002, and 0.020, respectively). The MFC chondral lesion demonstrated significant improvement in both lesion size (preoperative: 7.00 cm² (range: 4.38-10.50 cm²), postoperative: 0.16 cm² (range: 0.00-1.75 cm²), p = 0.002) and ICRS grade (preoperative: 4 (range: 4-4), postoperative: 1 (range: 1-2.25), p = 0.002). Additionally, the KL grade significantly decreased from 3 (range: 3-3) preoperatively to 2 (range: 2-2) postoperatively, while the HKA angle was corrected from 7.50° (range: 7.00-10.25°) preoperatively to -1.00° (range: -3.5-0.00°) postoperatively. CONCLUSIONS hUCB-MSC implantation with HTO is an effective treatment for medial compartment osteoarthritis and varus deformities, resulting in significant improvements in cartilage regeneration and overall clinical outcomes. TRIAL REGISTRATION NCT04234412.
Collapse
Affiliation(s)
- Bo Seung Bae
- Department of Orthopaedic Surgery, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jae Woong Jung
- Department of Orthopaedic Surgery, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Gyeong Ok Jo
- Department of Orthopaedic Surgery, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Seon Ae Kim
- Department of Orthopaedic Surgery, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Eun Jeong Go
- Department of Orthopaedic Surgery, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Mi-La Cho
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Asode Ananthram Shetty
- Institute of Medical Sciences, Faculty of Health and Social Care, Canterbury Christ Church University, Canterbury, UK
| | - Seok Jung Kim
- Department of Orthopaedic Surgery, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.
- Department of Orthopedic Surgery, Uijeongbu St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 271, Cheonbo-ro, Uijeongbu-si, Gyeonggi-do, Republic of Korea.
| |
Collapse
|
131
|
Quero FB, Troncoso-Bravo T, Farías MA, Kalergis AM. Cell-Based Therapeutic Strategies for Autoimmune Diseases. Immunotargets Ther 2025; 14:501-514. [PMID: 40322732 PMCID: PMC12047289 DOI: 10.2147/itt.s513629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Accepted: 04/15/2025] [Indexed: 05/08/2025] Open
Abstract
Currently, the management of autoimmune disorders still being a challenge in terms of safety, efficiency, and specificity. Cell-based therapeutic strategies have emerged as a novel approach for autoimmune disease treatment, employing different cell therapy platforms, including tolerogenic dendritic cells, regulatory T cells, conventional and regulatory chimeric antigen receptor-T cells, mesenchymal and hematopoietic stem cells, each with their biological features. Here, we discuss the different cell therapy platforms, their immunological mechanisms of action, their therapeutic potential and benefits in autoimmune diseases, and challenges related to their production, scaling up, risks, and patient safety.
Collapse
Affiliation(s)
- Francisco B Quero
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Tays Troncoso-Bravo
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Mónica A Farías
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alexis M Kalergis
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
132
|
Zou Y, Zhang G, Yang Y, Huang H, Li Z, Chen X, Zheng D, Lu YG, Niu G. Advanced techniques and innovations in peripheral nerve repair: a comprehensive review for clinical and experimental reference. Rev Neurosci 2025; 36:243-265. [PMID: 39566026 DOI: 10.1515/revneuro-2024-0101] [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: 07/27/2024] [Accepted: 09/17/2024] [Indexed: 11/22/2024]
Abstract
Peripheral nerve injury, resulting from various physical and chemical causes, has a high incidence and significant functional impact. This injury, affecting both sensory and motor functions, can severely diminish quality of life and cause mental health issues. Consequently, it is a major focus of current research. Recent advancements in peripheral nerve repair technology, including the application of new techniques and materials, have expanded the options for nerve repair methods. A comprehensive article that combines the pathological process of peripheral nerve repair with these methods is needed to advance research in this field. This review aims to provide a comprehensive overview of various techniques for repairing peripheral nerve injuries. Beginning with the histopathology of nerve injury, it evaluates these techniques in detail to offer clinical guidance. This review summarizes the advantages and disadvantages of various peripheral nerve repair methods, including photobiological modulation therapy, suture repair, nerve graft repair, vein graft catheter repair, muscle graft repair, laser welding repair, nerve catheter repair, nerve sliding repair technology, growth factor-assisted repair, stem cell therapy, and exosome therapy. Additionally, it explores future directions in the treatment of peripheral nerve injuries, providing valuable references for experimental research and clinical treatment.
Collapse
Affiliation(s)
- Yuchun Zou
- Fujian Key Laboratory of Oral Diseases, 74551 School and Hospital of Stomatology, Fujian Biological Materials Engineering and Technology Center of Stomatology, Fujian Medical University , Fuzhou 350004, China
| | - Gonghang Zhang
- Fujian Key Laboratory of Oral Diseases, 74551 School and Hospital of Stomatology, Fujian Biological Materials Engineering and Technology Center of Stomatology, Fujian Medical University , Fuzhou 350004, China
- School of Stomatology, 74551 Fujian Medical University , Fuzhou 350004, China
| | - Yuchen Yang
- Fujian Key Laboratory of Oral Diseases, 74551 School and Hospital of Stomatology, Fujian Biological Materials Engineering and Technology Center of Stomatology, Fujian Medical University , Fuzhou 350004, China
- School of Stomatology, 74551 Fujian Medical University , Fuzhou 350004, China
| | - Hankai Huang
- Fujian Key Laboratory of Oral Diseases, 74551 School and Hospital of Stomatology, Fujian Biological Materials Engineering and Technology Center of Stomatology, Fujian Medical University , Fuzhou 350004, China
- School of Stomatology, 74551 Fujian Medical University , Fuzhou 350004, China
| | - Zongxu Li
- Fujian Key Laboratory of Oral Diseases, 74551 School and Hospital of Stomatology, Fujian Biological Materials Engineering and Technology Center of Stomatology, Fujian Medical University , Fuzhou 350004, China
- School of Stomatology, 74551 Fujian Medical University , Fuzhou 350004, China
| | - Xiaohang Chen
- Fujian Key Laboratory of Oral Diseases, 74551 School and Hospital of Stomatology, Fujian Biological Materials Engineering and Technology Center of Stomatology, Fujian Medical University , Fuzhou 350004, China
- Department of Preventive Dentistry, 74551 School and Hospital of Stomatology, Fujian Medical University , 246 Yangqiao Middle Road, Fuzhou 350001, China
| | - Dali Zheng
- Fujian Key Laboratory of Oral Diseases, 74551 School and Hospital of Stomatology, Fujian Biological Materials Engineering and Technology Center of Stomatology, Fujian Medical University , Fuzhou 350004, China
| | - You-Guang Lu
- Fujian Key Laboratory of Oral Diseases, 74551 School and Hospital of Stomatology, Fujian Biological Materials Engineering and Technology Center of Stomatology, Fujian Medical University , Fuzhou 350004, China
- Department of Preventive Dentistry, 74551 School and Hospital of Stomatology, Fujian Medical University , 246 Yangqiao Middle Road, Fuzhou 350001, China
| | - Gang Niu
- Fujian Key Laboratory of Oral Diseases, 74551 School and Hospital of Stomatology, Fujian Biological Materials Engineering and Technology Center of Stomatology, Fujian Medical University , Fuzhou 350004, China
- Department of Maxillofacial Surgery, School and Hospital of Stomatology, Fujian Medical University, 246 Yangqiao Middle Road, Fuzhou 350002, China
| |
Collapse
|
133
|
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
|
134
|
Rao J, Xia L, Li Q, Ma N, Li X, Li J, Zhu L, Zhao P, Zeng Y, Zhou S, Guo H, Lin S, Dong S, Lou S, Fan F, Wei J, Zhong JF, Gao L, Li SC, Zhang X. A 6-tsRNA signature for early detection, treatment response monitoring, and prognosis prediction in diffuse large B cell lymphoma. Blood Cancer J 2025; 15:79. [PMID: 40295511 PMCID: PMC12037784 DOI: 10.1038/s41408-025-01267-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 03/14/2025] [Accepted: 03/25/2025] [Indexed: 04/30/2025] Open
Abstract
Diffuse large B-cell lymphoma (DLBCL) presents considerable clinical challenges due to its aggressive nature and diverse clinical progression. New molecular biomarkers are urgently needed for outcome prediction. We analyzed blood samples from DLBCL patients and healthy individuals using short, non-coding RNA sequencing. A classifier based on six tsRNAs was developed through random forest and primary component analysis. This classifier, established using Cox proportional hazards modeling with repeated 10-fold cross-validation on an internal cohort of 100 samples analyzed using RT-qPCR, effectively identified high-risk patients with significantly lower overall survival compared to low-risk patients (Hazard ratio: 6.657, 95%CI 2.827-15.68, P = 0.0006). Validation in an external cohort of 160 samples using RT-qPCR confirmed the classifier's robust performance. High-risk status was strongly associated with disease histological subtype, stage, and International Prognostic Index scores. Integration of the classifier into the IPI model enhanced the precision and consistency of prognostic predictions. A dynamic study revealed that patients experiencing a 1.06-fold decrease after one therapy cycle (early molecular response) exhibited better treatment outcomes and prognosis. Furthermore, the 6-tsRNA signature accurately differentiated healthy individuals from DLBCL (AUC 0.882, 95%CI 0.826-0.939). These findings underscore the potential of the identified 6-tsRNA profile as a biomarker for monitoring treatment effectiveness and predicting DLBCL outcomes.
Collapse
MESH Headings
- Humans
- Lymphoma, Large B-Cell, Diffuse/genetics
- Lymphoma, Large B-Cell, Diffuse/diagnosis
- Lymphoma, Large B-Cell, Diffuse/mortality
- Lymphoma, Large B-Cell, Diffuse/therapy
- Lymphoma, Large B-Cell, Diffuse/drug therapy
- Prognosis
- Male
- Female
- Middle Aged
- Aged
- Biomarkers, Tumor/genetics
- Adult
- Aged, 80 and over
- Gene Expression Profiling
- Early Detection of Cancer
Collapse
Affiliation(s)
- Jun Rao
- Medical Center of Hematology, Xinqiao Hospital, Army Medical University, Chongqing, China
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, China
- National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Soochow, China
| | - Lin Xia
- Medical Center of Hematology, Xinqiao Hospital, Army Medical University, Chongqing, China
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, China
- National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Soochow, China
| | - Qiong Li
- Medical Center of Hematology, Xinqiao Hospital, Army Medical University, Chongqing, China
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, China
- National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Soochow, China
| | - NaYa Ma
- Medical Center of Hematology, Xinqiao Hospital, Army Medical University, Chongqing, China
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, China
- National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Soochow, China
| | - Xinlei Li
- Medical Center of Hematology, Xinqiao Hospital, Army Medical University, Chongqing, China
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, China
- National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Soochow, China
| | - Jiali Li
- Medical Center of Hematology, Xinqiao Hospital, Army Medical University, Chongqing, China
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, China
- National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Soochow, China
| | - Lidan Zhu
- Medical Center of Hematology, Xinqiao Hospital, Army Medical University, Chongqing, China
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, China
- National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Soochow, China
| | - Pan Zhao
- Medical Center of Hematology, Xinqiao Hospital, Army Medical University, Chongqing, China
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, China
- National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Soochow, China
| | - Yunjing Zeng
- Medical Center of Hematology, Xinqiao Hospital, Army Medical University, Chongqing, China
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, China
- National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Soochow, China
| | - Sha Zhou
- Medical Center of Hematology, Xinqiao Hospital, Army Medical University, Chongqing, China
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, China
- National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Soochow, China
| | - Huanping Guo
- Medical Center of Hematology, Xinqiao Hospital, Army Medical University, Chongqing, China
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, China
- National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Soochow, China
| | - Shijia Lin
- Medical Center of Hematology, Xinqiao Hospital, Army Medical University, Chongqing, China
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, China
- National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Soochow, China
| | - Song Dong
- Medical Center of Hematology, Xinqiao Hospital, Army Medical University, Chongqing, China
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, China
- National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Soochow, China
| | - Shifeng Lou
- Department of Hematology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Fangyi Fan
- Department of Hematology, General Hospital of Chengdu Military Region, Chengdu, Chongqing, China
| | - Jin Wei
- Department of Hematology, North Sichuan Medical College, Nanchong, China
| | - Jiang F Zhong
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, CA, USA
| | - Li Gao
- Medical Center of Hematology, Xinqiao Hospital, Army Medical University, Chongqing, China
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, China
- National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Soochow, China
| | - Shengwen Calvin Li
- CHOC Children's Research Institute, Children's Hospital of Orange County (CHOC®), part of Rady Children's Heath, Orange, CA, USA.
- Department of Neurology, University of California-Irvine School of Medicine, Orange, CA, USA.
| | - Xi Zhang
- Medical Center of Hematology, Xinqiao Hospital, Army Medical University, Chongqing, China.
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, China.
- National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Soochow, China.
- Jinfeng Laboratory, Chongqing, China.
| |
Collapse
|
135
|
Mante N, Undale V, Sanap A, Bhonde R, Tambe P, Bansode M, Gupta RK. Disease microenvironment preconditioning: An evolving approach to improve therapeutic efficacy of human mesenchymal stromal cells. Int Immunopharmacol 2025; 157:114701. [PMID: 40300358 DOI: 10.1016/j.intimp.2025.114701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 04/10/2025] [Accepted: 04/18/2025] [Indexed: 05/01/2025]
Abstract
Despite the tremendous success in preclinical models, the translation of human mesenchymal stromal cells (hMSCs) as a therapy in the clinic is not up to the expectation. Intrinsic factors (age, sex, health status, life style of the donor, source, cellular senescence, and oxidative stress in hMSCs), extrinsic factors (culture system, batch-to-batch variations, choice of biomaterials, cell processing and preservation protocols), and host microenvironment (inflammatory milieu, oxidative stress, and hypoxia in the recipient) compromise the overall therapeutic efficacy of the transplanted hMSCs. In recent times, the approach of 'Disease Microenvironment Preconditioning (DMP)' has garnered attention to overcome the host-associated attributes involved in compromised hMSCs therapeutic potential. In this review, we discuss various approaches of DMP of hMSCs by employing serum and other body fluids obtained from diseased patients/animals and small molecules, including cytokines such as IFN-γ, IL-6, IL-10, IL- β, TGF-β1, IL-1α, IL-1β, TNF-α, HMGB1, IL-17 A, and IL-8 which are associated with disease conditions. DMP strengthens hMSCs ability to adapt/acclimatize and respond more efficiently to the hostile microenvironment they encounter upon transplantation. DMP modulate hMSCs to withstand inflammation, survive under hypoxic and nutrient-deprived conditions, and resist oxidative stress. Evidence from various disease models ranging from cardiovascular and neurodegenerative disorders to autoimmune diseases and tissue injuries supports the role of DMP in improving hMSC survival, integration, and functional efficacy. While the potential of DMP to revolutionize MSC-based therapies is evident, challenges such as standardizing/optimizing protocols for preconditioning is essential. This review synthesizes current advancements in the approach of DMP aiming to propel the area of regenerative medicine.
Collapse
Affiliation(s)
- Nishant Mante
- Department of Pharmacology, Dr. D. Y. Patil Institute of Pharmaceutical Sciences and Research, Pimpri, Pune 411018, India; Regenerative Medicine Laboratory, Dr. D. Y. Patil Dental College and Hospital, Dr. D. Y. Patil Vidyapeeth, Pune 411018, India; Department of Pharmacology, School of Pharmacy and Research, Dr. D. Y. Patil Dnyan Prasad University, Pimpri, Pune 411018, India
| | - Vaishali Undale
- Department of Pharmacology, Dr. D. Y. Patil Institute of Pharmaceutical Sciences and Research, Pimpri, Pune 411018, India; Department of Pharmacology, School of Pharmacy and Research, Dr. D. Y. Patil Dnyan Prasad University, Pimpri, Pune 411018, India.
| | - Avinash Sanap
- Regenerative Medicine Laboratory, Dr. D. Y. Patil Dental College and Hospital, Dr. D. Y. Patil Vidyapeeth, Pune 411018, India.
| | - Ramesh Bhonde
- Regenerative Medicine Laboratory, Dr. D. Y. Patil Dental College and Hospital, Dr. D. Y. Patil Vidyapeeth, Pune 411018, India
| | - Pratima Tambe
- Department of Pharmacology, Dr. D. Y. Patil Institute of Pharmaceutical Sciences and Research, Pimpri, Pune 411018, India; Department of Pharmacology, School of Pharmacy and Research, Dr. D. Y. Patil Dnyan Prasad University, Pimpri, Pune 411018, India
| | - Manoj Bansode
- Protein Biochemistry Research Centre, Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Tathawade, Pune 411033, Maharashtra, India
| | - Rajesh Kumar Gupta
- Protein Biochemistry Research Centre, Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Tathawade, Pune 411033, Maharashtra, India
| |
Collapse
|
136
|
Tang Q, Lee JM, Li L, Cai C, Jung H, Kwon HJE. p63 co-opts the skin Krt8-to-Krt5 transition for enamel organ development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.11.637463. [PMID: 39990386 PMCID: PMC11844444 DOI: 10.1101/2025.02.11.637463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Tooth enamel, the hardest vertebrate tissue, is crucial for mastication and dental protection. Its formation depends on the enamel organ (EO), a specialized epithelial structure derived from oral epithelium. How uniform oral epithelium differentiates into diverse EO cell types remains unclear. While p63 , an ectodermal development master regulator, is essential for dental placode formation, its specific roles in EO development have been obscured by early arrest in p63 knockout mice. Using single-cell RNA sequencing from mouse incisors, we show p63 expression across all EO cell types with both shared and distinct functions. Through trajectory reconstruction, we identify p63 's role in regulating both amelogenic (AmG) and non-AmG lineage commitment during EO development. Comparative transcriptome analyses reveal that p63 regulates the Krt8-to-Krt5 transition during AmG cell differentiation, paralleling its function in skin development. This parallel is reinforced by comparative motif discovery showing shared transcription factor usage, particularly p63 and AP-2 family members. Chromatin accessibility analyses further illustrate that p63 mediates this transition through chromatin landscape remodeling. These findings demonstrate that p63 co-opts the Krt8-to-Krt5 transition mechanism from skin development for EO formation. Summary statement This study reveals how p63 repurposes a skin keratinization mechanism for tooth enamel formation, providing novel insights into how specialized dental tissues develop and potential therapeutic targets for enamel disorders.
Collapse
|
137
|
Lu JS, Song CY, Cheng WJ, Wang KY. Mechanisms and challenges of mesenchymal stem cells in the treatment of knee osteoarthritis. World J Stem Cells 2025; 17:102923. [PMID: 40308880 PMCID: PMC12038463 DOI: 10.4252/wjsc.v17.i4.102923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 02/22/2025] [Accepted: 03/21/2025] [Indexed: 04/23/2025] Open
Abstract
In this editorial, we comment on the recent article by Xiao et al, focusing on their investigation into whether the therapeutic efficacy of serum-free human umbilical cord mesenchymal stem cells (MSCs) differs from that of serum-containing human umbilical cord MSCs in a mouse model of knee osteoarthritis (OA). The incidence of OA has significantly increased in recent years, primarily owing to sports-related injuries or degenerative diseases. However, the regenerative capacity of articular cartilage remains limited, necessitating artificial joint replacement for patients with advanced OA to enhance the quality of life. MSCs have been widely used in the clinical treatment of OA owing to their multidirectional differentiation potential and immunomodulatory effects. The focus of this paper lies in elucidating the mechanism underlying MSC transplantation for the treatment of knee OA, while also addressing the challenges encountered in MSC therapy and outlining future directions.
Collapse
Affiliation(s)
- Jing-Shun Lu
- Department of Orthopedics, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital, Wuhu 241000, Anhui Province, China
| | - Chen-Yu Song
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Wen-Jing Cheng
- Department of Orthopedics, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital, Wuhu 241000, Anhui Province, China
| | - Kai-Yang Wang
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China.
| |
Collapse
|
138
|
Wang W, Song AR, Liu HW, Li YK. Enhancing the clinical translation of stem cell models by focusing on standardization and international regulatory cooperation. World J Stem Cells 2025; 17:102788. [PMID: 40308886 PMCID: PMC12038456 DOI: 10.4252/wjsc.v17.i4.102788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 02/12/2025] [Accepted: 03/10/2025] [Indexed: 04/23/2025] Open
Abstract
The article by Granjeiro et al provided a thorough review of the role of stem cell models in the development of advanced therapy medicinal products. It emphasized the potential of stem cell models to refine preclinical studies and align with regulatory requirements for clinical applications. This article introduced a new perspective on enhancing the transition of stem cell research into clinical practice, focusing on the importance of international regulatory harmonization and the need for standardization in stem cell-based therapies.
Collapse
Affiliation(s)
- Wei Wang
- Department of Traditional Chinese Medicine, Youyang People's Hospital, Chongqing 409800, China
| | - An-Ran Song
- Department of Traditional Chinese Medicine, Youyang People's Hospital, Chongqing 409800, China
| | - Hong-Wen Liu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| | - Yi-Kai Li
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, Guangdong Province, China.
| |
Collapse
|
139
|
Li ZP, Li H, Ruan YH, Wang P, Zhu MT, Fu WP, Wang RB, Tang XD, Zhang Q, Li SL, Yin H, Li CJ, Tian YG, Han RN, Wang YB, Zhang CJ. Stem cell therapy for intervertebral disc degeneration: Clinical progress with exosomes and gene vectors. World J Stem Cells 2025; 17:102945. [PMID: 40308883 PMCID: PMC12038459 DOI: 10.4252/wjsc.v17.i4.102945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 01/25/2025] [Accepted: 03/10/2025] [Indexed: 04/23/2025] Open
Abstract
Intervertebral disc degeneration is a leading cause of lower back pain and is characterized by pathological processes such as nucleus pulposus cell apoptosis, extracellular matrix imbalance, and annulus fibrosus rupture. These pathological changes result in disc height loss and functional decline, potentially leading to disc herniation. This comprehensive review aimed to address the current challenges in intervertebral disc degeneration treatment by evaluating the regenerative potential of stem cell-based therapies, with a particular focus on emerging technologies such as exosomes and gene vector systems. Through mechanisms such as differentiation, paracrine effects, and immunomodulation, stem cells facilitate extracellular matrix repair and reduce nucleus pulposus cell apoptosis. Despite recent advancements, clinical applications are hindered by challenges such as hypoxic disc environments and immune rejection. By analyzing recent preclinical and clinical findings, this review provided insights into optimizing stem cell therapy to overcome these obstacles and highlighted future directions in the field.
Collapse
Affiliation(s)
- Zhi-Peng Li
- Second Department of Orthopedics, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
- Tianjian Advanced Biomedical Laboratory, Zhengzhou University, Zhengzhou 450001, Henan Province, China
| | - Han Li
- Department of Orthopedics, Affiliated Dongyang Hospital of Wenzhou Medical University, Jinhua 322100, Zhejiang Province, China
| | - Yu-Hua Ruan
- Second Department of Orthopedics, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Peng Wang
- Second Department of Orthopedics, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Meng-Ting Zhu
- Department of Neurology, Union Medical College Hospital of Fujian Medical University, Fuzhou 350001, Fujian Province, China
| | - Wei-Ping Fu
- Second Department of Orthopedics, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Rui-Bo Wang
- Second Department of Orthopedics, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Xiao-Dong Tang
- Second Department of Orthopedics, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Qi Zhang
- Second Department of Orthopedics, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Sen-Li Li
- Second Department of Orthopedics, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - He Yin
- Second Department of Orthopedics, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Cheng-Jin Li
- Second Department of Orthopedics, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Yi-Gong Tian
- Third Department of Orthopedics, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Rui-Ning Han
- Third Department of Orthopedics, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Yao-Bin Wang
- Second Department of Orthopedics, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
- Henan Key Laboratory for Helicobacter pylori and Digestive Tract Microecology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Chang-Jiang Zhang
- Second Department of Orthopedics, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China.
| |
Collapse
|
140
|
Mallis P. Hypoxic endometrial epithelial cell-derived microRNAs effectively regulate the regenerative properties of mesenchymal stromal cells. World J Stem Cells 2025; 17:102482. [PMID: 40308881 PMCID: PMC12038455 DOI: 10.4252/wjsc.v17.i4.102482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 02/06/2025] [Accepted: 03/05/2025] [Indexed: 04/23/2025] Open
Abstract
Endometrial thickness plays an important role in successful embryo implantation and normal pregnancy achievement. However, a thin endometrial layer (≤ 7 mm) may have a significant effect on microenvironment tolerance, which is further related to successful embryo implantation or conception, either naturally or after assisted reproductive technology. Moreover, this microenvironment tolerance shift induces hypoxic damage to endometrial epithelial cells (EECs), which results in altered signaling biomolecule secretion, including exosome content. In the context of endometrium regeneration, mesenchymal stromal cells (MSCs) and umbilical cord (UC)-derived stem cells have been applied in clinical trials with promising results. It has been recently shown that exosomes derived from hypoxic damaged EECs directly contribute to the increased migratory and regenerative abilities of UCs and MSCs. Specifically, microRNAs in exosomes secreted by the hypoxic damaged EECs, such as miR-214-5p and miR-21-5p, play a crucial role in the migratory capacity and differentiation ability of MSCs to EECs mediated through the signal transducer and activator of transcription 3 (STAT3) signaling pathway. Taking into consideration the above information, UC-MSCs may be considered as a modern intervention for endometrial regeneration.
Collapse
Affiliation(s)
- Panagiotis Mallis
- Hellenic Cord Blood Bank, Biomedical Research Foundation Academy of Athens, Athens 11527, Attikí, Greece.
| |
Collapse
|
141
|
Zhang WY, Liu SM, Wang HB, Deng CY. Exosomal miR-137-3p targets UBE3C to activate STAT3, promoting migration and differentiation into endometrial epithelial cell of human umbilical cord mesenchymal stem cells under hypoxia. World J Stem Cells 2025; 17:100359. [PMID: 40308888 PMCID: PMC12038465 DOI: 10.4252/wjsc.v17.i4.100359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 11/29/2024] [Accepted: 03/24/2025] [Indexed: 04/23/2025] Open
Abstract
BACKGROUND Thin endometrium, leading cause of recurrent implantation failure and infertility, has been found to respond to exosomes. AIM To investigate the efficacy of exosomes in addressing the issue of thin endometrium. METHODS RNA sequencing and reverse transcription-quantitative polymerase chain reaction were employed to identify differentially expressed microRNAs (miRNAs) in human umbilical cord mesenchymal stem cell (hucMSC) treated with exosomes enriched with endometrial cell-derived components. Additionally, Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analyses were conducted to highlight significant enrichment in specific biological pathways, molecular functions, and cellular components. Transwell and wound healing assays were performed to assess migratory potential, and western blotting was detected protein level. RESULTS A total of 53 differentially expressed miRNAs were identified in hucMSC treated with exosomes enriched with endometrial cell-derived components, comprising 27 upregulated and 26 downregulated miRNAs, which includes miR-137-3p. Enhanced migratory potential was observed in the Transwell and wound healing assays, and western blotting confirmed the epithelial differentiation of hucMSC and the increased p-signal transducer and activator of transcription 3. These effects were attributed to the upregulation of miR-137-3p. CONCLUSION miR-137-3p in exosomes from hypoxia-affected endometrial epithelial cell stimulates the signal transducer and activator of transcription 3 signaling pathway, enhancing the migration and differentiation of hucMSC into endometrial epithelial cell.
Collapse
Affiliation(s)
- Wan-Yu Zhang
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Si-Miao Liu
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Han-Bi Wang
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Beijing 100730, China
| | - Cheng-Yan Deng
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Beijing 100730, China.
| |
Collapse
|
142
|
Lyu X, Wang J, Su J. Intelligent Manufacturing for Osteoarthritis Organoids. Cell Prolif 2025:e70043. [PMID: 40285592 DOI: 10.1111/cpr.70043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Revised: 03/22/2025] [Accepted: 04/07/2025] [Indexed: 04/29/2025] Open
Abstract
Osteoarthritis (OA) is the most prevalent degenerative joint disease worldwide, imposing a substantial global disease burden. However, its pathogenesis remains incompletely understood, and effective treatment strategies are still lacking. Organoid technology, in which stem cells or progenitor cells self-organise into miniature tissue structures under three-dimensional (3D) culture conditions, provides a promising in vitro platform for simulating the pathological microenvironment of OA. This approach can be employed to investigate disease mechanisms, carry out high-throughput drug screening and facilitate personalised therapies. This review summarises joint structure, OA pathogenesis and pathological manifestations, thereby establishing the disease context for the application of organoid technology. It then examines the components of the arthrosis organoid system, specifically addressing cartilage, subchondral bone, synovium, skeletal muscle and ligament organoids. Furthermore, it details various strategies for constructing OA organoids, including considerations of cell selection, pathological classification and fabrication techniques. Notably, this review introduces the concept of intelligent manufacturing of OA organoids by incorporating emerging engineering technologies such as artificial intelligence (AI) into the organoid fabrication process, thereby forming an innovative software and hardware cluster. Lastly, this review discusses the challenges currently facing intelligent OA organoid manufacturing and highlights future directions for this rapidly evolving field. By offering a comprehensive overview of state-of-the-art methodologies and challenges, this review anticipates that intelligent, automated fabrication of OA organoids will expedite fundamental research, drug discovery and personalised translational applications in the orthopaedic field.
Collapse
Affiliation(s)
- Xukun Lyu
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Trauma Orthopedics Center, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Musculoskeletal Injury and Translational Medicine of Organoids, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Clinical Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jian Wang
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Trauma Orthopedics Center, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Musculoskeletal Injury and Translational Medicine of Organoids, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Translational Medicine, Shanghai University, Shanghai, China
- National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai, China
| | - Jiacan Su
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Trauma Orthopedics Center, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Musculoskeletal Injury and Translational Medicine of Organoids, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Translational Medicine, Shanghai University, Shanghai, China
- National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai, China
| |
Collapse
|
143
|
Rong J, Li YY, Wang X, Wang JN, Song M. Non-coding RNAs in adipose-derived stem cell exosomes: Mechanisms, therapeutic potential, and challenges in wound healing. World J Stem Cells 2025; 17:102917. [PMID: 40308889 PMCID: PMC12038460 DOI: 10.4252/wjsc.v17.i4.102917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/23/2024] [Accepted: 03/21/2025] [Indexed: 04/23/2025] Open
Abstract
The treatment of complex wounds presents a significant clinical challenge due to the limited availability of standardized therapeutic options. Adipose-derived stem cell exosomes (ADSC-Exos) are promising for their capabilities to enhance angiogenesis, mitigate oxidative stress, modulate inflammatory pathways, support skin cell regeneration, and promote epithelialization. These exosomes deliver non-coding RNAs, including microRNAs, long non-coding RNAs, and circular RNAs, which facilitate collagen remodeling, reduce scar formation, and expedite wound healing. This study reviews the mechanisms, therapeutic roles, and challenges of non-coding RNA-loaded ADSC-Exos in wound healing and identifies critical directions for future research. It aims to provide insights for researchers into the potential mechanisms and clinical applications of ADSC-Exos non-coding RNAs in wound healing.
Collapse
Affiliation(s)
- Jian Rong
- Department of Burns and Plastic Surgery, The 940 Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou 730050, Gansu Province, China
- Department of Plateau Medicine, The 940 Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou 730050, Gansu Province, China
| | - Yao-Yao Li
- Department of Burns and Plastic Surgery, The 940 Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou 730050, Gansu Province, China
- Department of Plateau Medicine, The 940 Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou 730050, Gansu Province, China
| | - Xin Wang
- Department of Burns and Plastic Surgery, The 940 Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou 730050, Gansu Province, China
- Department of Plateau Medicine, The 940 Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou 730050, Gansu Province, China
| | - Jia-Ning Wang
- Department of Burns and Plastic Surgery, The 940 Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou 730050, Gansu Province, China
- Department of Plateau Medicine, The 940 Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou 730050, Gansu Province, China
| | - Mei Song
- Department of Burns and Plastic Surgery, The 940 Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou 730050, Gansu Province, China
- Department of Plateau Medicine, The 940 Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou 730050, Gansu Province, China.
| |
Collapse
|
144
|
Habibi MA, Rashidi F, Hajikarimloo B, Karami S, Fathi Tavani S, Zare A, Mirjani MS, Allahdadi A, Didehvar K, Mohammadzadeh I, Kaviari MA. The safety and efficacy of VEGFR tyrosine kinase inhibitors for patients with gliomas: a systematic review, meta-analysis, and a specific analysis on glioblastoma. Neurosurg Rev 2025; 48:390. [PMID: 40285944 DOI: 10.1007/s10143-025-03536-8] [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: 01/10/2025] [Revised: 04/05/2025] [Accepted: 04/13/2025] [Indexed: 04/29/2025]
Abstract
Gliomas account for 24.5% of all primary brain tumors and 80% of all malignant tumors in adults. Vascular endothelial growth factor receptors (VEGFR) tyrosine kinase inhibitors (TKIs) play an important role in disrupting angiogenesis, tumor growth, and invasion. This study evaluates the outcomes of VEGFR TKIs in patients with glioma, with a specific analysis on glioblastoma (GBM). Electronic databases of PubMed/Medline, Embase, Scopus, and Web of Science were conducted until 23 July 2024. Studies that evaluated the survival of patients with glioma treated with VEGFR TKI were included. All statistical analyses were conducted using the R program. A total of 24 studies, including 1,146 glioma patients with a median age range of 5.8 to 62 years were recruited. Regarding progression-free survival (PFS), the six-month PFS rate was reported with a pooled value of 21% [95% CI: 15% to 28%]. The 12-month PFS rate was evaluated in three studies, ranging from 5 to 38% with a pooled rate of 15% [95% CI: 8% to 27%]. Considering the radiological response, the pooled overall response rate (ORR) was 21% [95% CI: 15%-28%]. Evaluation of the subgroups based on drug type at the six-month follow-up showed no significant difference in overall survival (OS) rates among patients (p = 0.06). Our results revealed that VEGFR TKIs in patients with glioma, were associated with limited efficacy. The long-term effectiveness of these treatments remains controversial and requires longer follow-up, which is challenging in cancer cases.
Collapse
Affiliation(s)
- Mohammad Amin Habibi
- Department of Neurosurgery, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran.
| | - Farhang Rashidi
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Bardia Hajikarimloo
- Department of Neurological Surgery, University of Virginia, Charlottesville, VA, USA
| | - Shaghayegh Karami
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Amirhossein Zare
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Ali Allahdadi
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Kimia Didehvar
- School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ibrahim Mohammadzadeh
- Skull Base Research Center, Loghman-Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Amin Kaviari
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Universal Scientific Education and Research Network (USERN) Office, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
145
|
Tian Y, Pan P, Luo X, Sun Y, Yang X, Gao H, Yang Y. Palmitic acid-induced insulin resistance triggers granulosa cell senescence by disruption of the UPR mt/mitophagy/lysosome axis. Chem Biol Interact 2025; 411:111450. [PMID: 40023272 DOI: 10.1016/j.cbi.2025.111450] [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: 11/08/2024] [Revised: 02/18/2025] [Accepted: 02/27/2025] [Indexed: 03/04/2025]
Abstract
Insulin resistance (IR) is the main pathological feature of polycystic ovary syndrome (PCOS), but the adverse impacts of IR on ovary and granulosa cells (GCs) are unknown. Therefore, the role of palmitic acid (PA) induced IR in GCs, and a mitochondrial proteostasis and mitochondrial homeostasis control system, the mitochondrial unfolded protein response (UPRmt)/mitophagy/lysosome axis were investigated to uncover the side effect and the mechanism of IR on GCs. Our results revealed that IR in GC was successfully constructed by 100 μM PA treatment accompanied with cell senescence. In addition, mitochondrial function was impaired by IR-induced GC senescence accompanied by significantly increased reactive oxygen species (ROS) and decreased mitochondrial membrane potential, and mitochondrial proteostasis was impaired by a dysfunctional UPRmt and increased protein aggregation, leading to more unfolded and misfolded proteins accumulating in mitochondria. Mitochondrial homeostasis was maintained by the mitophagy/lysosome degradation system, although mitophagy was significantly increased, lysosomes were damaged; hence, malfunctional mitochondria were not cleared by the mitophagy/lysosome degradation system, more ROS were produced by malfunctional mitochondria. Therefore, accelerated GC senescence was triggered by excessive ROS, and reversed by the mitophagy inhibitor cyclosporin A (CsA) accompanied with reduced IR. Additionally, the mice were administered with PA, and results revealed that the accelerated ovarian aging was caused by PA, which might be attributed to GC senescence. In conclusion, GC senescence was triggered in PA-induced IR by disruption of the UPRmt/mitophagy/lysosome axis, and IR induced GC senescence was reversed by the CsA.
Collapse
Affiliation(s)
- Yuan Tian
- General Hospital, Key Laboratory of Fertility Preservation and Maintenance, Ministry of Education, School of Basic Medicine, Ningxia Medical University, Yinchuan, Ningxia, PR China
| | - Pengge Pan
- General Hospital, Key Laboratory of Fertility Preservation and Maintenance, Ministry of Education, School of Basic Medicine, Ningxia Medical University, Yinchuan, Ningxia, PR China
| | - Xiaoqiang Luo
- Department of Clinical Laboratory, Ningxia Women and Children's Hospital, Beijing University Hospital, Yinchuan, Ningxia, PR China
| | - Yaqi Sun
- General Hospital, Key Laboratory of Fertility Preservation and Maintenance, Ministry of Education, School of Basic Medicine, Ningxia Medical University, Yinchuan, Ningxia, PR China
| | - Xintong Yang
- General Hospital, Key Laboratory of Fertility Preservation and Maintenance, Ministry of Education, School of Basic Medicine, Ningxia Medical University, Yinchuan, Ningxia, PR China
| | - Hui Gao
- General Hospital, Key Laboratory of Fertility Preservation and Maintenance, Ministry of Education, School of Basic Medicine, Ningxia Medical University, Yinchuan, Ningxia, PR China
| | - Yanzhou Yang
- General Hospital, Key Laboratory of Fertility Preservation and Maintenance, Ministry of Education, School of Basic Medicine, Ningxia Medical University, Yinchuan, Ningxia, PR China; Emergency Department, The First People's Hospital of Yinchuan, The Second Clinical Medical College, Ningxia Medical University, Yinchuan, Ningxia, PR China.
| |
Collapse
|
146
|
Park JS, Sung MJ, Na HJ. Drosophila model systems reveal intestinal stem cells as key players in aging. Ann N Y Acad Sci 2025. [PMID: 40276941 DOI: 10.1111/nyas.15351] [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] [Indexed: 04/26/2025]
Abstract
The intestines play important roles in responding immediately and dynamically to food intake, environmental stress, and metabolic dysfunction, and they are involved in various human diseases and aging. A key part of their function is governed by intestinal stem cells (ISCs); therefore, understanding ISCs is vital. Dysregulation of ISC activity, which is influenced by various cell signaling pathways and environmental signals, can lead to inflammatory responses, tissue damage, and increased cancer susceptibility. Aging exacerbates these dynamics and affects ISC function and tissue elasticity. Additionally, proliferation and differentiation profoundly affect ISC behavior and gut health, highlighting the complex interplay between environmental factors and gut homeostasis. Drosophila models help us understand the complex regulatory networks in the gut, providing valuable insights into disease mechanisms and therapeutic strategies targeting human intestinal diseases.
Collapse
Affiliation(s)
- Joung-Sun Park
- Institute of Nanobio Convergence, Pusan National University, Busan, Republic of Korea
- Department of Molecular Biology, Pusan National University, Busan, Republic of Korea
| | - Mi Jeong Sung
- Aging Research Group, Division of Food Functionality Research, Korea Food Research Institute, Wanju, Republic of Korea
| | - Hyun-Jin Na
- Aging Research Group, Division of Food Functionality Research, Korea Food Research Institute, Wanju, Republic of Korea
| |
Collapse
|
147
|
Karam M, Aqel S, Haider MZ, Fathima A, Charafedine A, Daher MA, Shaito A, El-Sabban M, Saliba J. Beyond the Injury: How Does Smoking Impair Stem Cell-Mediated Repair Mechanisms? A Dual Review of Smoking-Induced Stem Cell Damage and Stem Cell-Based Therapeutic Applications. Stem Cell Rev Rep 2025:10.1007/s12015-025-10886-9. [PMID: 40279029 DOI: 10.1007/s12015-025-10886-9] [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] [Accepted: 04/14/2025] [Indexed: 04/26/2025]
Abstract
While the literature on molecular and clinical effects of smoking on the lungs and other organs has been expansively reviewed, there is no comprehensive compilation of the effects of smoking on stem cell (SC) populations. Recent research has shown that tobacco exposure severely compromises the function of SC populations, particularly those involved in tissue regeneration: mesenchymal SCs (MSCs), neural progenitors, and hematopoietic SCs. SC-based therapies have emerged as a promising approach to counteract smoking-related damage. In particular, MSCs have been extensively studied for their immunomodulatory properties, demonstrating the ability to repair damaged tissues, reduce inflammation, and slow disease progression in conditions such as chronic obstructive pulmonary disease. Combination therapies, which integrate pharmaceuticals with SC treatments, have shown potential in enhancing regenerative outcomes. This review examines the impact of smoking on SC biology, describes the processes impairing SC-mediated repair mechanisms and highlights recent advancements in SC-based therapies in the treatment of smoking-induced diseases. This review has two prongs: (1) it attempts to explain potential smoking-related disease etiology, and (2) it addresses a gap in the literature on SC-mediated repair mechanisms in chronic smokers.
Collapse
Affiliation(s)
- Mario Karam
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- Department of Translational Cancer Medicine, Faculty of Medicine, University of Helsinki, Biomedicum Helsinki, Haartmaninkatu 8, 00290, Helsinki, Helsinki, Finland
| | - Sarah Aqel
- Medical Research Center, Hamad Medical Corporation, Doha, Qatar
| | - Mohammad Z Haider
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, P.O. Box 2713, Doha, Qatar
| | - Aseela Fathima
- Biomedical Research Center and Department of Biomedical Sciences at College of Health Sciences, QU Health, Qatar University, P.O. Box 2713, Doha, Qatar
| | - Adib Charafedine
- College Of Pharmacy, American University of Iraq-Baghdad, Baghdad, Iraq
| | - Mira Abou Daher
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- Department of Public Health, Faculty of Health Sciences, University of Balamand, Sin El Fil, PO Box: 55251, Beirut, Lebanon
| | - Abdullah Shaito
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, P.O. Box 2713, Doha, Qatar
- Biomedical Research Center and Department of Biomedical Sciences at College of Health Sciences, QU Health, Qatar University, P.O. Box 2713, Doha, Qatar
| | - Marwan El-Sabban
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Jessica Saliba
- Department of Public Health, Faculty of Health Sciences, University of Balamand, Sin El Fil, PO Box: 55251, Beirut, Lebanon.
- Department of Biology, Faculty of Science, Lebanese University, Beirut, Lebanon.
| |
Collapse
|
148
|
Ali A, Kuo WW, Kuo CH, Lo JF, Hsieh DJY, Pai P, Ho TJ, Shibu MA, Lin SZ, Huang CY. Chaperone-assisted E3 ligase-engineered mesenchymal stem cells target hyperglycemia-induced p53 for ubiquitination and proteasomal degradation ameliorates self-renewal. Biol Res 2025; 58:20. [PMID: 40270049 PMCID: PMC12020092 DOI: 10.1186/s40659-025-00604-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Accepted: 04/09/2025] [Indexed: 04/25/2025] Open
Abstract
BACKGROUND Stem cell therapies may potentially be used in regenerative and reconstructive medicine due to their ability for self-renewal and differentiation. Stressful conditions, such as hyperglycemia, adversely affect stem cell functions, impairing their function and promoting differentiation by opposing self-renewal. The carboxyl terminus of HSP70 interacting protein (CHIP), which is a cochaperone and E3 ligase, maintains protein homeostasis and performs quality control of the cell via ubiquitylation. However, the role of CHIP in regulating stemness remains unknown. RESULTS Hyperglycemia downregulated CHIP-induced p53, arrested the cell cycle at the gap (G1) phase, and promoted the loss of stemness in WJMSCs. Quantitative real-time polymerase chain reaction (qRT-PCR), Western blotting, immunofluorescence, and cell cycle analysis showed that CHIP-overexpressing WJMSCs downregulated the expression of phosphorylated p53 and shortened its half-life while enhancing self-renewal factors. Additionally, co-IP and Western blotting revealed that CHIP promoted the ubiquitination and proteasomal degradation of hyperglycemia-induced p53 through the chaperone system. CONCLUSIONS CHIP may promote ubiquitin-mediated proteasomal degradation of hyperglycemia-induced p53 rescues self-renewal genes, which can maintain the long-term undifferentiated state of WJMSCs. CHIP may be an alternative therapeutic option in regenerative medicine for hyperglycemic-related complications in diabetes.
Collapse
Affiliation(s)
- Ayaz Ali
- Aberdeen Cardiovascular and Diabetes Centre, Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
- Department of Biological Science and Technology, China Medical University, Taichung, 404, Taiwan
| | - Wei-Wen Kuo
- Department of Biological Science and Technology, China Medical University, Taichung, 404, Taiwan
| | - Chia-Hua Kuo
- Laboratory of Exercise Biochemistry, University of Taipei, Taipei, Taiwan
| | - Jeng-Feng Lo
- Institute of Oral Biology, National Yang-Ming University, Taipei, Taiwan
| | | | - Peiying Pai
- School of Medicine, College of Medicine, China Medical University, Taichung, 40402, Taiwan
- Division of Cardiovascular Medicine, Department of Medicine, China Medical University Hospital, Taichung, 40447, Taiwan
| | - Tsung-Jung Ho
- Department of Chinese Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Tzu Chi University, Hualien, Taiwan
| | | | - Shinn-Zong Lin
- Bioinnovation Center, Buddhist Tzu Chi Medical Foundation, Hualien, 970, Taiwan
- Department of Neurosurgery, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, 970, Taiwan
| | - Chih-Yang Huang
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, 404, Taiwan.
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan.
- Department of Biotechnology, Asia University, Taichung, Taiwan.
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Tzu Chi University of Science and Technology, Hualien, Taiwan.
| |
Collapse
|
149
|
Chen Y, Liu B, Tao S, Liu L, Gao J, Liang Y, Dong W, Zhou D. CITED2 Binding to EP300 Regulates Human Spermatogonial Stem Cell Proliferation and Survival Through HSPA6. Stem Cells Int 2025; 2025:2362489. [PMID: 40313859 PMCID: PMC12045681 DOI: 10.1155/sci/2362489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Accepted: 03/22/2025] [Indexed: 05/03/2025] Open
Abstract
Spermatogonial stem cells (SSCs) are essential for the initiation and continuation of spermatogenesis, a process fundamental to male fertility. Despite extensive studies on mouse SSCs, the mechanisms governing self-renewal and differentiation in human SSCs remain to be elucidated. This study investigated the regulatory mechanisms of SSCs by analyzing single-cell sequencing data from the GEO dataset of human testis. Analysis revealed dominant expression of CITED2 in human SSCs. Reduction of CITED2 levels in hSSC lines significantly inhibited proliferation and increased apoptosis. Protein interaction prediction and immunoprecipitation identified interactions between CITED2 and EP300 in SSC lines. RNA sequencing results indicated that CITED2 knockdown significantly affected the MAPK pathway and the HSPA6 gene. Overexpression of HSPA6 mitigated the proliferative and apoptotic changes provoked by CITED2 downregulation. These findings provide novel insights into the regulatory and functional mechanisms of CITED2-mediated hSSC development.
Collapse
Affiliation(s)
- Yongzhe Chen
- Gynecology and Obstetrics and Reproductive Medical Center, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- MOE Key Lab of Rare Pediatric Diseases, University of South China, Hengyang, Hunan 421001, China
| | - Bang Liu
- Hunan Provincial Key Laboratory of Regional Hereditary Birth Defect Prevention and Control, Changsha Hospital for Maternal and Child Health Care Affiliated to Hunan Normal University, Changsha, Hunan 410000, China
| | - Sisi Tao
- Hunan Provincial Key Laboratory of Regional Hereditary Birth Defect Prevention and Control, Changsha Hospital for Maternal and Child Health Care Affiliated to Hunan Normal University, Changsha, Hunan 410000, China
| | - Lvjun Liu
- Hunan Provincial Key Laboratory of Regional Hereditary Birth Defect Prevention and Control, Changsha Hospital for Maternal and Child Health Care Affiliated to Hunan Normal University, Changsha, Hunan 410000, China
| | - Jianxin Gao
- Hunan Provincial Key Laboratory of Regional Hereditary Birth Defect Prevention and Control, Changsha Hospital for Maternal and Child Health Care Affiliated to Hunan Normal University, Changsha, Hunan 410000, China
| | - Ying Liang
- Hunan Provincial Key Laboratory of Regional Hereditary Birth Defect Prevention and Control, Changsha Hospital for Maternal and Child Health Care Affiliated to Hunan Normal University, Changsha, Hunan 410000, China
| | - Weilei Dong
- Gynecology and Obstetrics and Reproductive Medical Center, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- MOE Key Lab of Rare Pediatric Diseases, University of South China, Hengyang, Hunan 421001, China
| | - Dai Zhou
- Hunan Provincial Key Laboratory of Regional Hereditary Birth Defect Prevention and Control, Changsha Hospital for Maternal and Child Health Care Affiliated to Hunan Normal University, Changsha, Hunan 410000, China
- Institute of Reproduction and Stem Cell Engineering, School of Basic Medicine Science, Central South University, Changsha, Hunan 410000, China
- Research Department, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, Hunan 410000, China
| |
Collapse
|
150
|
Araya-Sapag MJ, Lara-Barba E, García-Guerrero C, Herrera-Luna Y, Flores-Elías Y, Bustamante-Barrientos FA, Albornoz GG, Contreras-Fuentes C, Yantén-Fuentes L, Luque-Campos N, Vega-Letter AM, Toledo J, Luz-Crawford P. New mesenchymal stem/stromal cell-based strategies for osteoarthritis treatment: targeting macrophage-mediated inflammation to restore joint homeostasis. J Mol Med (Berl) 2025:10.1007/s00109-025-02547-8. [PMID: 40272537 DOI: 10.1007/s00109-025-02547-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 04/10/2025] [Accepted: 04/14/2025] [Indexed: 04/25/2025]
Abstract
Macrophages are pivotal in osteoarthritis (OA) pathogenesis, as their dysregulated polarization can contribute to chronic inflammatory processes. This review explores the molecular and metabolic mechanisms that influence macrophage polarization and identifies potential strategies for OA treatment. Currently, non-surgical treatments for OA focus only on symptom management, and their efficacy is limited; thus, mesenchymal stem/stromal cells (MSCs) have gained attention for their anti-inflammatory and immunomodulatory capabilities. Emerging evidence suggests that small extracellular vesicles (sEVs) derived from MSCs can modulate macrophage function, thus offering potential therapeutic benefits in OA. Additionally, the transfer of mitochondria from MSCs to macrophages has shown promise in enhancing mitochondrial functionality and steering macrophages toward an anti-inflammatory M2-like phenotype. While further research is needed to confirm these findings, MSC-based strategies, including the use of sEVs and mitochondrial transfer, hold great promise for the treatment of OA and other chronic inflammatory diseases.
Collapse
Affiliation(s)
- María Jesús Araya-Sapag
- Programa de Doctorado en Biomedicina, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Eliana Lara-Barba
- Programa de Doctorado en Biomedicina, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Cynthia García-Guerrero
- Programa de Doctorado en Biomedicina, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Yeimi Herrera-Luna
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Yesenia Flores-Elías
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Felipe A Bustamante-Barrientos
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Guillermo G Albornoz
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Consuelo Contreras-Fuentes
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Liliana Yantén-Fuentes
- Programa de Doctorado en Biomedicina, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
- Red de Equipamiento Científico Avanzado (REDECA), Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Noymar Luque-Campos
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Ana María Vega-Letter
- Escuela de Ingeniería Bioquímica, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Jorge Toledo
- Red de Equipamiento Científico Avanzado (REDECA), Facultad de Medicina, Universidad de Chile, Santiago, Chile.
- Centro de Investigación Clínica Avanzada (CICA), Hospital Clínico Universidad de Chile, Santiago, Chile.
| | - Patricia Luz-Crawford
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile.
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile.
| |
Collapse
|