1
|
Chen L, Wu Y, Lv T, Tuo R, Xiao Y. Mesenchymal stem cells enchanced by salidroside to inhibit ferroptosis and improve premature ovarian insufficiency via Keap1/Nrf2/GPX4 signaling. Redox Rep 2025; 30:2455914. [PMID: 39874130 PMCID: PMC11776066 DOI: 10.1080/13510002.2025.2455914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND Regenerative medicine researches have shown that mesenchymal stem cells (MSCs) may be an effective treatment method for premature ovarian insufficiency (POI). However, the efficacy of MSCs is still limited. PURPOSE This study aims to explain whether salidroside and MSCs combination is a therapeutic strategy to POI and to explore salidroside-enhanced MSCs inhibiting ferroptosis via Keap1/Nrf2/GPX4 signaling. METHODS The effect of salidroside and MSCs on ovarian granular cells (GCs) was analyzed. After treatment, hormone levels and -fertility of rats were measured. Lipid peroxidation levels, iron deposition and mitochondrial morphology were detected. The genes and proteins of Keap1/Nrf2/GPX4 signaling were examined. RESULTS Salidroside and MSCs were found to inhibit cell death of GCs by reducing peroxidation and intracellular ferrous. Salidroside promotes the proliferation of MSCs and supports cell survival in ovary. Salidroside combined with MSCs therapy restored ovarian function, which was better than MSCs monotherapy. Salidroside-enhanced MSCs to inhibit ferroptosis. The results showed activation of the Keap1/Nrf2/GPX4 signaling and an increase in anti-ferroptosis molecule. CONCLUSIONS Salidroside-enhanced MSCs as a ferroptosis inhibitor and provide new therapeutic strategies for POI. The possible mechanisms of MSCs were related to maintaining redox homeostasis via a Keap1/Nrf2/GPX4 signaling.
Collapse
Affiliation(s)
- Lixuan Chen
- Department of Hematology, Shenzhen Qianhai Shekou Pilot Free Trade Zone Hospital, Shenzhen, People’s Republic of China
| | - Yingnan Wu
- Translational Medicine Center, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, People’s Republic of China
| | - Tiying Lv
- Guangzhou University of Chinese Medicine, Guangzhou, People’s Republic of China
| | - Rui Tuo
- Guangzhou University of Chinese Medicine, Guangzhou, People’s Republic of China
| | - Yang Xiao
- Department of Hematology, Shenzhen Qianhai Shekou Pilot Free Trade Zone Hospital, Shenzhen, People’s Republic of China
- Guangzhou University of Chinese Medicine, Guangzhou, People’s Republic of China
| |
Collapse
|
2
|
Yang Y, Ma K, Li S, Xiong T. Multifaceted role of nitric oxide in vascular dementia. Med Gas Res 2025; 15:496-506. [PMID: 40300885 DOI: 10.4103/mgr.medgasres-d-24-00158] [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: 12/30/2024] [Accepted: 02/24/2025] [Indexed: 05/01/2025] Open
Abstract
Vascular dementia is a highly heterogeneous neurodegenerative disorder induced by a variety of factors. Currently, there are no definitive treatments for the cognitive dysfunction associated with vascular dementia. However, early detection and preventive measures have proven effective in reducing the risk of onset and improving patient prognosis. Nitric oxide plays an integral role in various physiological and pathological processes within the central nervous system. In recent years, nitric oxide has been implicated in the regulation of synaptic plasticity and has emerged as a crucial factor in the pathophysiology of vascular dementia. At different stages of vascular dementia, nitric oxide levels and bioavailability undergo dynamic alterations, with a marked reduction in the later stages, which significantly contributes to the cognitive deficits associated with the disease. This review provides a comprehensive review of the emerging role of nitric oxide in the physiological and pathological processes underlying vascular dementia, focusing on its effects on synaptic dysfunction, neuroinflammation, oxidative stress, and blood‒brain barrier integrity. Furthermore, we suggest that targeting the nitric oxide soluble guanylate cyclase-cyclic guanosine monophosphate pathway through specific therapeutic strategies may offer a novel approach for treating vascular dementia, potentially improving both cognitive function and patient prognosis. The review contributes to a better understanding of the multifaceted role of nitric oxide in vascular dementia and to offering insights into future therapeutic interventions.
Collapse
Affiliation(s)
- Yi Yang
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, Jiangsu Province, China
| | - Kangrong Ma
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, Jiangsu Province, China
| | - Shun Li
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Tianqing Xiong
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, Jiangsu Province, China
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Key Laboratory of the Jiangsu Higher Education Institutions for Integrated Traditional Chinese and Western Medicine in Senile Diseases Control (Yangzhou University), Yangzhou, Jiangsu Province, China
| |
Collapse
|
3
|
Poyraz M, Poyraz AK, Dogan Y, Gunes S, Mir HS, Paul JK, Barua PD, Baygin M, Dogan S, Tuncer T, Molinari F, Acharya R. BrainNeXt: novel lightweight CNN model for the automated detection of brain disorders using MRI images. Cogn Neurodyn 2025; 19:53. [PMID: 40124704 PMCID: PMC11929658 DOI: 10.1007/s11571-025-10235-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 12/19/2024] [Accepted: 02/28/2025] [Indexed: 03/25/2025] Open
Abstract
The main aim of this study is to propose a novel convolutional neural network, named BrainNeXt, for the automated brain disorders detection using magnetic resonance images (MRI) images. Furthermore, we aim to investigate the performance of our proposed network on various medical applications. To achieve high/robust image classification performance, we gathered a new MRI dataset belonging to four classes: (1) Alzheimer's disease, (2) chronic ischemia, (3) multiple sclerosis, and (4) control. Inspired by ConvNeXt, we designed BrainNeXt as a lightweight classification model by incorporating the structural elements of the Swin Transformers Tiny model. By training our model on the collected dataset, a pretrained BrainNeXt model was obtained. Additionally, we have suggested a feature engineering (FE) approach based on the pretrained BrainNeXt, which extracted features from fixed-sized patches. To select the most discriminative/informative features, we employed the neighborhood component analysis selector in the feature selection phase. As the classifier for our patch-based FE approach, we utilized the support vector machine classifier. Our recommended BrainNeXt approach achieved an accuracy of 100% and 91.35% for training and validation. The recommended model obtained the test classification accuracy of 94.21%. To further improve the classification performance, we suggested a patch-based DFE approach, which achieved a test accuracy of 99.73%. The obtained results, surpassing 90% accuracy on the test dataset, demonstrate the effectiveness and high classification performance of the proposed models.
Collapse
Affiliation(s)
- Melahat Poyraz
- Department of Radiology, Elazig Fethi Sekin City Hospital, Elazig, Turkey
| | - Ahmet Kursad Poyraz
- Department of Radiology, School of Medicine, Firat University, 23119 Elazig, Turkey
| | - Yusuf Dogan
- Department of Radiology, School of Medicine, Firat University, 23119 Elazig, Turkey
| | - Selva Gunes
- Department of Radiology, School of Medicine, Firat University, 23119 Elazig, Turkey
| | - Hasan S. Mir
- Department of Electrical Engineering, American University of Sharjah, Sharjah, UAE
| | - Jose Kunnel Paul
- Department of Neurology, Government Medical College, Thiruvananthapuram, Kerala India
| | - Prabal Datta Barua
- School of Business (Information System), University of Southern Queensland, Springfield, Australia
| | - Mehmet Baygin
- Department of Computer Engineering, Engineering Faculty, Erzurum Technical University, Erzurum, Turkey
| | - Sengul Dogan
- Department of Digital Forensics Engineering, Technology Faculty, Firat University, Elazig, Turkey
| | - Turker Tuncer
- Department of Digital Forensics Engineering, Technology Faculty, Firat University, Elazig, Turkey
| | - Filippo Molinari
- Department of Electronics and Telecommunications, Politecnico Di Torino, Turin, Italy
| | - Rajendra Acharya
- School of Mathematics, Physics and Computing, University of Southern Queensland, Springfield, Australia
| |
Collapse
|
4
|
Xiang Z, Ma B, Pei X, Wang W, Gong W. Mechanism of action of genistein on breast cancer and differential effects of different age stages. PHARMACEUTICAL BIOLOGY 2025; 63:141-155. [PMID: 39996512 PMCID: PMC11864014 DOI: 10.1080/13880209.2025.2469607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 02/12/2025] [Accepted: 02/14/2025] [Indexed: 02/26/2025]
Abstract
CONTEXT Genistein, a soy-derived isoflavone, exhibits structural similarities with 17β-estradiol and demonstrates antioxidant, anti-inflammatory, and estrogenic properties. Despite its low bioavailability limiting its clinical application, it shows potential for breast cancer prevention and treatment. OBJECTIVE This review aims to summarize the pharmacological effects and molecular mechanisms of genistein in breast cancer, focusing on its therapeutic potential, strategies to overcome bioavailability limitations, and its role in personalized medicine. Differential impacts among population subgroups are also discussed. METHODS A systematic review was conducted using PubMed, ScienceDirect, and Google Scholar databases. Studies were selected based on their focus on genistein's mechanisms of action, strategies to enhance its bioavailability, and interactions with other therapies. RESULTS Genistein exerted anticancer effects by modulating estrogen receptor β (ERβ), inhibiting angiogenesis, arresting the cell cycle, and inducing apoptosis. Its antioxidant properties help mitigate tumor-associated oxidative stress. Bioavailability enhancement strategies, such as nanoparticle and lipid-based formulations, show promise. Age-dependent effects were evident, with distinct responses observed in prepubertal, menopausal, and postmenopausal populations, underscoring its potential for personalized therapies. Furthermore, genistein influences epigenetic modifications, including DNA methylation and miRNA expression, bolstering its anticancer efficacy. CONCLUSION Genistein is a promising candidate for breast cancer therapy, particularly for personalized treatment. Strategies to enhance bioavailability and further clinical research are essential to optimize its therapeutic potential and evaluate its efficacy in combination therapies.
Collapse
Affiliation(s)
- Zhebin Xiang
- Zhejiang Chinese Medical University, Hangzhou, China
| | - Bo Ma
- Zhejiang Hospital, Hangzhou, China
| | - Xiujun Pei
- Shandong Provincial Hospital, Shandong, China
| | - Wenjie Wang
- Zhejiang Chinese Medical University, Hangzhou, China
| | - Weilun Gong
- Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
5
|
Yuan Y, Kuang M, Yu T, Huang S, Jiang F, Lu B, Cai M, Lu X. Adipogenic dedifferentiation enhances survival of human umbilical cord-derived mesenchymal stem cells under oxidative stress. Adipocyte 2025; 14:2467150. [PMID: 39976240 PMCID: PMC11845070 DOI: 10.1080/21623945.2025.2467150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 12/16/2024] [Accepted: 02/05/2025] [Indexed: 02/21/2025] Open
Abstract
Mesenchymal stem cells (MSCs) serve as ideal candidates for a broad range of cell-based therapies. However, cell ageing caused by long-term in vitro expansion and poor survival after in vivo delivery greatly limits their success in preclinical and clinical applications. Dedifferentiation represents a potential strategy for enhancing the retention and function of MSCs in hostile environments. In this study, we evaluated the cell phenotype, proliferation, and differentiation potential, as well as the anti-oxidative stress ability of human umbilical cord-derived MSCs (hMSCs) manipulated with adipogenic priming and subsequent dedifferentiation. After an in vitro differentiation and dedifferentiation procedure, the resultant dedifferentiated hMSCs (De-hMSCs) displayed properties similar to their original counterparts, including immunophenotype and mesodermal potential. Upon re-induction, De-hMSCs exhibited a significantly higher adipogenic differentiation capability than unmanipulated hMSCs. Importantly, De-hMSCs showed a significantly enhanced ability to resist tert-butyl hydroperoxide (t-BHP) induced apoptosis compared to undifferentiated hMSCs. Mechanisms involving bcl-2 family proteins and autophagy may contribute to the demonstrated advantages of dedifferentiation-reprogrammed hMSCs. These results indicate that adipogenic dedifferentiation promotes adipogenesis and cell persistence, as well as preserves the stemness of human umbilical cord-derived MSCs that have been committed to the adipocytic lineage. As a unique stem cell population, dedifferentiated MSCs may represent an attractive and promising candidate for MSC-based therapy.
Collapse
Affiliation(s)
- Yin Yuan
- School of Life Science & Biopharmacology, Guangdong Pharmaceutical University, Guangzhou, China
| | - Meina Kuang
- School of Life Science & Biopharmacology, Guangdong Pharmaceutical University, Guangzhou, China
| | - Tengye Yu
- School of Life Science & Biopharmacology, Guangdong Pharmaceutical University, Guangzhou, China
| | - Sirui Huang
- School of Life Science & Biopharmacology, Guangdong Pharmaceutical University, Guangzhou, China
| | - Fujie Jiang
- School of Life Science & Biopharmacology, Guangdong Pharmaceutical University, Guangzhou, China
| | - Biyi Lu
- School of Life Science & Biopharmacology, Guangdong Pharmaceutical University, Guangzhou, China
| | - Mingen Cai
- School of Life Science & Biopharmacology, Guangdong Pharmaceutical University, Guangzhou, China
| | - Xin Lu
- School of Life Sciences, South China Normal University, Guangzhou, China
| |
Collapse
|
6
|
Vidman S, Ma YHE, Fullenkamp N, Plant GW. Human induced pluripotent stem cell-derived therapies for regeneration after central nervous system injury. Neural Regen Res 2025; 20:3063-3075. [PMID: 39715081 PMCID: PMC11881715 DOI: 10.4103/nrr.nrr-d-24-00901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 09/26/2024] [Accepted: 10/29/2024] [Indexed: 12/25/2024] Open
Abstract
In recent years, the progression of stem cell therapies has shown great promise in advancing the nascent field of regenerative medicine. Considering the non-regenerative nature of the mature central nervous system, the concept that "blank" cells could be reprogrammed and functionally integrated into host neural networks remained intriguing. Previous work has also demonstrated the ability of such cells to stimulate intrinsic growth programs in post-mitotic cells, such as neurons. While embryonic stem cells demonstrated great potential in treating central nervous system pathologies, ethical and technical concerns remained. These barriers, along with the clear necessity for this type of treatment, ultimately prompted the advent of induced pluripotent stem cells. The advantage of pluripotent cells in central nervous system regeneration is multifaceted, permitting differentiation into neural stem cells, neural progenitor cells, glia, and various neuronal subpopulations. The precise spatiotemporal application of extrinsic growth factors in vitro, in addition to microenvironmental signaling in vivo, influences the efficiency of this directed differentiation. While the pluri- or multipotency of these cells is appealing, it also poses the risk of unregulated differentiation and teratoma formation. Cells of the neuroectodermal lineage, such as neuronal subpopulations and glia, have been explored with varying degrees of success. Although the risk of cancer or teratoma formation is greatly reduced, each subpopulation varies in effectiveness and is influenced by a myriad of factors, such as the timing of the transplant, pathology type, and the ratio of accompanying progenitor cells. Furthermore, successful transplantation requires innovative approaches to develop delivery vectors that can mitigate cell death and support integration. Lastly, host immune responses to allogeneic grafts must be thoroughly characterized and further developed to reduce the need for immunosuppression. Translation to a clinical setting will involve careful consideration when assessing both physiologic and functional outcomes. This review will highlight both successes and challenges faced when using human induced pluripotent stem cell-derived cell transplantation therapies to promote endogenous regeneration.
Collapse
Affiliation(s)
- Stephen Vidman
- Department of Neuroscience, Ohio State University, Columbus, OH, USA
| | - Yee Hang Ethan Ma
- Department of Neuroscience, Ohio State University, Columbus, OH, USA
| | - Nolan Fullenkamp
- Department of Neuroscience, Ohio State University, Columbus, OH, USA
| | - Giles W. Plant
- Department of Neuroscience, Ohio State University, Columbus, OH, USA
| |
Collapse
|
7
|
Aldali F, Deng C, Nie M, Chen H. Advances in therapies using mesenchymal stem cells and their exosomes for treatment of peripheral nerve injury: state of the art and future perspectives. Neural Regen Res 2025; 20:3151-3171. [PMID: 39435603 PMCID: PMC11881730 DOI: 10.4103/nrr.nrr-d-24-00235] [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: 02/26/2024] [Revised: 07/26/2024] [Accepted: 08/26/2024] [Indexed: 10/23/2024] Open
Abstract
"Peripheral nerve injury" refers to damage or trauma affecting nerves outside the brain and spinal cord. Peripheral nerve injury results in movements or sensation impairments, and represents a serious public health problem. Although severed peripheral nerves have been effectively joined and various therapies have been offered, recovery of sensory or motor functions remains limited, and efficacious therapies for complete repair of a nerve injury remain elusive. The emerging field of mesenchymal stem cells and their exosome-based therapies hold promise for enhancing nerve regeneration and function. Mesenchymal stem cells, as large living cells responsive to the environment, secrete various factors and exosomes. The latter are nano-sized extracellular vesicles containing bioactive molecules such as proteins, microRNA, and messenger RNA derived from parent mesenchymal stem cells. Exosomes have pivotal roles in cell-to-cell communication and nervous tissue function, offering solutions to changes associated with cell-based therapies. Despite ongoing investigations, mesenchymal stem cells and mesenchymal stem cell-derived exosome-based therapies are in the exploratory stage. A comprehensive review of the latest preclinical experiments and clinical trials is essential for deep understanding of therapeutic strategies and for facilitating clinical translation. This review initially explores current investigations of mesenchymal stem cells and mesenchymal stem cell-derived exosomes in peripheral nerve injury, exploring the underlying mechanisms. Subsequently, it provides an overview of the current status of mesenchymal stem cell and exosome-based therapies in clinical trials, followed by a comparative analysis of therapies utilizing mesenchymal stem cells and exosomes. Finally, the review addresses the limitations and challenges associated with use of mesenchymal stem cell-derived exosomes, offering potential solutions and guiding future directions.
Collapse
Affiliation(s)
- Fatima Aldali
- Department of Rehabilitation Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Chunchu Deng
- Department of Rehabilitation Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Mingbo Nie
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Hong Chen
- Department of Rehabilitation Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| |
Collapse
|
8
|
Yuan M, Yin Z, Wang Z, Xiong Z, Chen P, Yao L, Liu P, Sun M, Shu K, Li L, Jiang Y. Modification of MSCs with aHSCs-targeting peptide pPB for enhanced therapeutic efficacy in liver fibrosis. Biomaterials 2025; 321:123295. [PMID: 40188718 DOI: 10.1016/j.biomaterials.2025.123295] [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: 09/15/2024] [Revised: 02/22/2025] [Accepted: 03/26/2025] [Indexed: 05/04/2025]
Abstract
Mesenchymal stem cells (MSCs) hold significant therapeutic potential for liver fibrosis but face translational challenges due to suboptimal homing efficiency and poor retention at injury sites. Activated hepatic stellate cells (aHSCs), the primary drivers of fibrogenesis, overexpress platelet-derived growth factor receptor-beta (PDGFRB), a validated therapeutic target in liver fibrosis. Here, we engineered pPB peptide-functionalized MSCs (pPB-MSCs) via hydrophobic insertion of DMPE-PEG-pPB (DPP) into the MSC membrane, creating a targeted "MSC-pPB-aHSC" delivery system. Our findings demonstrated that pPB modification preserved MSC viability, differentiation potential, and paracrine functions. pPB-MSCs exhibited higher binding affinity to TGF-β1-activated HSCs in vitro and greater hepatic accumulation in TAA-induced fibrotic mice, as quantified by in vivo imaging. Moreover, pPB-MSCs attenuated collagen deposition, suppressed α-SMA+ HSCs, and restored serum ALT/AST levels to near-normal ranges. Mechanistically, pPB-MSCs promoted hepatocyte regeneration via HGF upregulation, inhibited epithelial-mesenchymal transition through TGF-β/Smad pathway suppression, and polarized macrophages toward an M2 phenotype, reducing pro-inflammatory IL-6/TNF-α while elevating anti-inflammatory IL-10. Overall, our study raised a non-genetic MSC surface engineering strategy that synergizes PDGFRB-targeted homing with multifactorial tissue repair, addressing critical barriers in cell therapy for liver fibrosis. By achieving enhanced spatial delivery without compromising MSC functionality, our approach provides a clinically translatable platform for enhancing regenerative medicine outcomes.
Collapse
Affiliation(s)
- Mengqin Yuan
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, Wuhan 430000, China; Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Clinical Research Center for Medical Imaging in Hubei Province, Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Zhengrong Yin
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Molecular Biological Targeted Therapies of the Ministry of Education, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Zheng Wang
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, Wuhan 430000, China
| | - Zhiyu Xiong
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, Wuhan 430000, China
| | - Ping Chen
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, Wuhan 430000, China
| | - Lichao Yao
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, Wuhan 430000, China
| | - Pingji Liu
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, Wuhan 430000, China
| | - Muhua Sun
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, Wuhan 430000, China
| | - Kan Shu
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, Wuhan 430000, China
| | - Lanjuan Li
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, Wuhan 430000, China; State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, China.
| | - Yingan Jiang
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, Wuhan 430000, China.
| |
Collapse
|
9
|
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] [MESH Headings] [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
|
10
|
Chen TY, Xu J, Tai CH, Wen TK, Hsu SH. Biodegradable, electroconductive self-healing hydrogel based on polydopamine-coated polyurethane nano-crosslinker for Parkinson's disease therapy. Biomaterials 2025; 320:123268. [PMID: 40121830 DOI: 10.1016/j.biomaterials.2025.123268] [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: 12/03/2024] [Revised: 02/19/2025] [Accepted: 03/17/2025] [Indexed: 03/25/2025]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterized by loss of dopaminergic neurons, causing motor and neurological impairments. Current treatments offer only temporary symptom relief without halting progression. Herein, a fully biodegradable, electroconductive self-healing hydrogel (CPUD gel) is developed, incorporating electroconductive polydopamine-coated polyurethane nanoparticles (PUD) as crosslinker. The core-shell PUD nanoparticles have a highly uniform size of ∼36 nm with a polydopamine shell of ∼4.8 nm thick on polyurethane core, revealed by small angle X-ray scattering, and own a conductivity of ∼0.82 mS/cm. As nano-crosslinker, the PUD can react with chitosan to form the dynamic CPUD hydrogel with shear modulus (∼280 Pa) and conductivity (∼4.34 mS/cm), mimicking brain tissue properties. In vitro, CPUD gel supports neural stem cell (NSC) proliferation (∼565 %) and differentiation, with elevated neuronal marker expression at 14 days, while exhibiting strong antioxidative and anti-inflammatory effects, rescuing ∼88 % inflamed NSCs. A therapeutic strategy combining injectable CPUD gel with acupuncture in a PD rat model, aiming to activate the innate regenerative mechanisms of body through mobilized endogenous stem cells, is further established. Using this approach, this hydrogel significantly elevates serum TGF-β1/SDF-1 levels, promotes dopaminergic neuron regeneration (>80 %), modulates neuroinflammation through M1-to M2-microglia transition (∼12.6-fold M2/M1 ratio), and improves motor function (from 8 % to 37 % forelimb contacts) in 14 days. Particularly, the electrophysiological spike rate is recovered from 66 to 19 spikes/s, close to the healthy rate 15 spikes/s. The synergistic immunomodulation and neuroprotection highlight the potential of CPUD gel as an advanced therapeutic tool for PD.
Collapse
Affiliation(s)
- Tsai-Yu Chen
- Institute of Polymer Science and Engineering, National Taiwan University, Taipei, Taiwan, ROC
| | - Junpeng Xu
- Institute of Polymer Science and Engineering, National Taiwan University, Taipei, Taiwan, ROC
| | - Chun-Hwei Tai
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan, ROC
| | - Tsung-Kai Wen
- School of Post-Baccalaureate Chinese Medicine, Tzu Chi University, Hualien, Taiwan, ROC
| | - Shan-Hui Hsu
- Institute of Polymer Science and Engineering, National Taiwan University, Taipei, Taiwan, ROC.
| |
Collapse
|
11
|
Ozkan S, Isildar B, Neccar D, Koyuturk M. Dynamic analysis of metabolic and ultrastructural changes in mesenchymal stem cells at hypoxic preconditioning and post-preconditioning stages: Cobalt chloride on the spotlight. Tissue Cell 2025; 95:102923. [PMID: 40267849 DOI: 10.1016/j.tice.2025.102923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 03/21/2025] [Accepted: 04/11/2025] [Indexed: 04/25/2025]
Abstract
Mesenchymal stem cells (MSCs) have come up as a potential remedy for treatment of various diseases thanks to their regenerative abilities. However, MSC-based therapies face challenges like reduced cell survival and functionality after transplantation. Preconditioning, particularly with hypoxia-mimicking agents like cobalt chloride (CoCl2), has been explored to enhance MSCs' effectiveness. This study aims to evaluate MSC survival, migration, and therapeutic outcomes at the CoCI2-preconditioning and post-preconditioning stages. Human umbilical cord-MSCs were treated with 100 µM CoCI2 with/out serum for 24-hours, and then passaged and planted in corresponding culture conditions without CoCI2, these two consecutive passages were named as the preconditioning and post-preconditioning stages, respectively. In each stage, total protein concentrations, total antioxidant and total oxidant status (TAS/TOS) of the conditioned media derived from the cells were investigated with bicinchoninic acid assay and TAS/TOS kits, respectively. The proliferation rates, migratory capacities, cellular senescence, expression levels of hypoxia-inducible factor1-α (HIF1-α), Ki-67, active caspase-3 and beclin-1 proteins and ultrastructures of the cells were evaluated by 3-(4,5-Dimethylthiazol-2-yl)-2,5-Diphenyltetrazolium Bromide test, wound healing assay, β-galactosidase-activity assessment, immunocytochemistry and transmission electron microscopy, respectively. Our results indicated that preconditioning MSCs with CoCl2 did not significantly enhance their proliferation, migration, or secretory abilities. However, it increased antioxidant capacity and along with normalization of senescence-status post-preconditioning, possibly by shifting energy metabolism from oxidative-phosphorylation to glycolysis through the upregulation of the HIF1-α signalling pathway. These findings indicate that CoCl2 preconditioning could be an effective approach to boost the therapeutic potential of MSCs, especially in enhancing their survival and functionality after transplantation.
Collapse
Affiliation(s)
- Serbay Ozkan
- Faculty of Medicine, Histology and Embryology Department, Izmir Katip Çelebi University, Çiğli, Izmir, 35620, Turkiye.
| | - Basak Isildar
- Faculty of Medicine, Histology and Embryology Department, Balıkesir University, Balıkesir 10145, Turkiye.
| | - Duygu Neccar
- Cerrahpasa Faculty of Medicine, Histology and Embryology Department, Istanbul University-Cerrahpasa, Fatih, Istanbul 34098, Turkiye.
| | - Meral Koyuturk
- Cerrahpasa Faculty of Medicine, Histology and Embryology Department, Istanbul University-Cerrahpasa, Fatih, Istanbul 34098, Turkiye.
| |
Collapse
|
12
|
Ribeiro M. Advances in Cell-based therapies for peripheral arterial disease. Tissue Cell 2025; 95:102909. [PMID: 40250109 DOI: 10.1016/j.tice.2025.102909] [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: 12/05/2024] [Revised: 04/02/2025] [Accepted: 04/07/2025] [Indexed: 04/20/2025]
Abstract
PURPOSE To examine recent advances in cell-based therapies for peripheral arterial disease (PAD), focusing on mechanisms of action, clinical applications, and regulatory considerations. The review aimed to evaluate the therapeutic potential of various cell types and assess their efficacy in addressing the unmet needs of PAD patients,particularly those with critical limb ischemia (CLI). METHODS The review analysed current literature on cell-based therapies for PAD, including preclinical studies using animal models, clinical trials from phase I to III, and regulatory frameworks. Multiple cell types were evaluated, including mesenchymal stem cells (MSCs), endothelial progenitor cells (EPCs), bone marrow mononuclear cells (BMMNCs),and adipose-derived stem cells (ADSCs). RESULTS Preclinical studies demonstrated significant improvements in limb perfusion and neovascularization across various cell types. Clinical trials, particularly those utilizing MSCs and BM-MNCs, showed encouraging outcomes in wound healing and reduced amputation rates. The therapeutic effects were mediated through multiple mechanisms, including direct vessel formation, paracrine signalling, immunomodulation, and tissue repair. The FDA's implementation of a tiered, risk-based system for human cells, tissues, and cellular and tissue-based products (HCT/Ps) has provided a regulatory framework balancing innovation with safety. CONCLUSION Cell-based therapies show promising potential for PAD treatment, particularly for patients with limited conventional treatment options. While clinical trials demonstrate encouraging results, challenges remain in standardizing cell characterization methods and establishing appropriate potency assays. Future research should focus on optimizing cell delivery methods, identifying the most effective cell types, and conducting larger clinical trials to establish definitive efficacy.
Collapse
Affiliation(s)
- Maisa Ribeiro
- Medical College, Health Sciences Academic Unit, University Center of Mineiros, Mineiros, Goias, Brazil.
| |
Collapse
|
13
|
Wang H, Zhang W, Sun Y, Xu X, Chen X, Zhao K, Yang Z, Liu H. Nanotherapeutic strategies exploiting biological traits of cancer stem cells. Bioact Mater 2025; 50:61-94. [PMID: 40242505 PMCID: PMC12002948 DOI: 10.1016/j.bioactmat.2025.03.016] [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: 11/30/2024] [Revised: 03/08/2025] [Accepted: 03/20/2025] [Indexed: 04/18/2025] Open
Abstract
Cancer stem cells (CSCs) represent a distinct subpopulation of cancer cells that orchestrate cancer initiation, progression, metastasis, and therapeutic resistance. Despite advances in conventional therapies, the persistence of CSCs remains a major obstacle to achieving cancer eradication. Nanomedicine-based approaches have emerged for precise CSC targeting and elimination, offering unique advantages in overcoming the limitations of traditional treatments. This review systematically analyzes recent developments in nanomedicine for CSC-targeted therapy, emphasizing innovative nanomaterial designs addressing CSC-specific challenges. We first provide a detailed examination of CSC biology, focusing on their surface markers, signaling networks, microenvironmental interactions, and metabolic signatures. On this basis, we critically evaluate cutting-edge nanomaterial engineering designed to exploit these CSC traits, including stimuli-responsive nanodrugs, nanocarriers for drug delivery, and multifunctional nanoplatforms capable of generating localized hyperthermia or reactive oxygen species. These sophisticated nanotherapeutic approaches enhance selectivity and efficacy in CSC elimination, potentially circumventing drug resistance and cancer recurrence. Finally, we present an in-depth analysis of current challenges in translating nanomedicine-based CSC-targeted therapies from bench to bedside, offering critical insights into future research directions and clinical implementation. This review aims to provide a comprehensive framework for understanding the intersection of nanomedicine and CSC biology, contributing to more effective cancer treatment modalities.
Collapse
Affiliation(s)
- Hongyu Wang
- State Key Laboratory of Organic-Inorganic Composites, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing Key Laboratory of Bioprocess, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Wenjing Zhang
- State Key Laboratory of Green Biomanufacturing, Innovation Center of Molecular Diagnostics, College of Life Science and Technology, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Yun Sun
- State Key Laboratory of Organic-Inorganic Composites, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing Key Laboratory of Bioprocess, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Xican Xu
- State Key Laboratory of Organic-Inorganic Composites, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing Key Laboratory of Bioprocess, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Xiaoyang Chen
- State Key Laboratory of Green Biomanufacturing, Innovation Center of Molecular Diagnostics, College of Life Science and Technology, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Kexu Zhao
- State Key Laboratory of Green Biomanufacturing, Innovation Center of Molecular Diagnostics, College of Life Science and Technology, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Zhao Yang
- State Key Laboratory of Green Biomanufacturing, Innovation Center of Molecular Diagnostics, College of Life Science and Technology, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Huiyu Liu
- State Key Laboratory of Organic-Inorganic Composites, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing Key Laboratory of Bioprocess, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, 100029, Beijing, China
| |
Collapse
|
14
|
Eisa NM, Elshaer SS, Bakry S, Abdelzaher OF, Eldesoky NAR. Placental extract augments mesenchymal stem cells in pancreatic tissue regeneration: A new insight into diabetes treatment. Tissue Cell 2025; 95:102883. [PMID: 40157219 DOI: 10.1016/j.tice.2025.102883] [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: 01/05/2025] [Revised: 03/19/2025] [Accepted: 03/19/2025] [Indexed: 04/01/2025]
Abstract
BACKGROUND Although a wide variety of medicinal interventions and lifestyles have been endeavored so far for the treatment of diabetes mellitus, it is still intractable. The current study aimed to examine the effect of mesenchymal stem cells (MSCs) and/or placental extract (PE) on streptozotocin (STZ) induced diabetic rats. METHODS Fifty male albino rats were used. Ten of them as negative control (group I) and the remaining forty rats were subjected to diabetes induction using 50 mg/kg STZ then divided into; group II (positive controls), group III (MSCs treated), group IV (PE treated), and group V (MSCs/PE combination treated). After 4 weeks of treatment, animals were sacrificed; blood samples were collected for determination of glycated hemoglobin by HPLC, and serum was separated for determination of glucose spectrophotometrically and insulin by ELISA. Pancreatic tissues were harvested for histopathological examination and pancreatic duodenal homeobox 1 (Pdx1) gene expression by PCR. RESULTS The three treated groups showed significant enhancement in glycemic parameters and Pdx1 gene expression compared with positive control group (P < 0.05). Histopathological examination revealed great improvement in the three treated groups where group V showed the best picture and the best glycemic control. CONCLUSIONS This study points to the possible role of PE in DM treatment. The MSCs/PE combination had the ability to return all parameters and Pdx1 gene expression to their normal levels. This action could be attributed to MSCs homing into the pancreas and the pancreatic rejuvenation provided by PE contents of growth factors; EGF, HGF, IGF-1 and IGF-II.
Collapse
Affiliation(s)
- Nehal Mohamed Eisa
- Clinical Research Department at Giza health affairs Directorate, MOHP, Giza, Egypt; Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Girls), Al-Azhar University, Nasr city, Cairo 11823, Egypt.
| | - Shereen Saeid Elshaer
- Biochemistry Department, Faculty of Pharmacy, Heliopolis University, Cairo 11785, Egypt; Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Girls), Al-Azhar University, Nasr city, Cairo 11823, Egypt
| | - Sayed Bakry
- Zoology Department, Faculty of Science, Al-Azhar University, Cairo, Egypt
| | | | - Noha Abdel-Rahman Eldesoky
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Girls), Al-Azhar University, Nasr city, Cairo 11823, Egypt
| |
Collapse
|
15
|
Wu K, Meng Q, Nong X, Chen X, Liu Y, Mok SWF, Huang R, Zhang X. New insights into the cytotoxic mechanism of marine-fungus-derived citrinin in three-dimensional Hepa1-6 cell model. Comp Biochem Physiol C Toxicol Pharmacol 2025; 294:110196. [PMID: 40164369 DOI: 10.1016/j.cbpc.2025.110196] [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: 11/25/2024] [Revised: 02/26/2025] [Accepted: 03/23/2025] [Indexed: 04/02/2025]
Abstract
Although recent studies have demonstrated that Marine-fungus-derived citrinin (MFDC) has a significant cytotoxic effect in traditional two-dimensional (2D) monolayer cell culture and animal models, its precise cytotoxic mechanism, particularly in a three-dimensional (3D) cell culture model remains unclear. In this study, a 3D Hepa1-6 cell model based on Matrigel was used to investigate the potential cytotoxic mechanism of MFDC (0-100 μg/mL). The results revealed that, after treatment of 60-100 μg/mL MFDC, the increases of reactive oxygen species (ROS), lactate dehydrogenase (LDH), aspartate aminotransferase (AST) and alanine aminotransferase (ALT) in 2D cell model were more significant than those in 3D cell model. In addition, the metabolomic results revealed that the significantly altered metabolic pathways were pyrimidine metabolism and vitamin B6 metabolism, which might be related to the interference of MFDC in the pyrimidine synthesis pathway, as well as the upregulation of pyridoxine 5'phosphate oxidase and pyridoxal kinase activities. This study was the first to compare the cytotoxicology of 2D and Matrigel-based 3D cell models after MFDC induction, and to detect differences in cell metabolites after MFDC induction in 3D cell models, providing a new scientific basis for the use of a 3D cell model and a novel research idea for the cellular damage caused by MFDC.
Collapse
Affiliation(s)
- Keyue Wu
- University Joint Laboratory of Guangdong Province, Hong Kong and Macao Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou 510642, China
| | - Qingyue Meng
- Department of Medical Genetics, Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing 211166, China
| | - Xuhua Nong
- Key Laboratory of Tropical Medicinal Plant Chemistry of Hainan Province, Hainan Normal University, Haikou 571158, China
| | - Xinye Chen
- University Joint Laboratory of Guangdong Province, Hong Kong and Macao Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou 510642, China
| | - Yonghong Liu
- Key Laboratory of Tropical Marine Bio-resources and Ecology, South China Sea Institute of Oceanology, Chinese academy of sciences, 164 West Xingang Road, Guangzhou 510301, China
| | - Simon Wing-Fai Mok
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macao
| | - Riming Huang
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou 510642, China.
| | - Xiaoyong Zhang
- University Joint Laboratory of Guangdong Province, Hong Kong and Macao Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou 510642, China.
| |
Collapse
|
16
|
Liang B, Bai R, Wang J, Shi S, Guo Y, Wang Q, Peng H, Tang J, Liu S, Zhu J, Yi C, Hou M, Li H. Innovative applications of acellular adipose matrix derived film in skin soft tissue expansion. BIOMATERIALS ADVANCES 2025; 173:214291. [PMID: 40154149 DOI: 10.1016/j.bioadv.2025.214291] [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: 11/18/2024] [Revised: 03/04/2025] [Accepted: 03/22/2025] [Indexed: 04/01/2025]
Abstract
BACKGROUND Skin dilation generates "extra" skin tissue through mechanical traction, but its effectiveness is limited by the proliferation capacity of keratinocytes, fibroblasts and the level of angiogenesis. Cutaneous application of drug and subcutaneous injection are common interventions to promote skin dilation, but they have defects such as uneven drug distribution, high risk of infection and single targeting. Although Acellular adipose matrix (AAM) has the potential to promote cell proliferation and angiogenesis, its hydrogel/powder dosage forms still need frequent injection, which limits clinical application. RESULTS In this study, Acellular adipose matrix derived film (AAF) was successfully developed, and a flexible film was formed by acellular - lyophilized - enzymolysis - self-assembly process. In vitro experiments confirmed that AAF significantly promoted the activity of Human Immortalized Epidermal Cells (HaCaTs), Normal Skin Fibroblasts (NFbs) and Human Umbilical Endothelial Cells (HUVECs); It was also found that AAF can induce adipose mesenchymal stem cells (ASCs) to differentiate into adipocytes and promote subcutaneous fat regeneration. In vivo, the rat model showed that AAF wrapping expander could effectively improve the skin expansion efficiency, promote the skin thickness increase in the expanded area, and the density of new blood vessels was significantly increased compared with the comparative group, and there was no complication such as infection or skin collapse. It was found for the first time that AAF successfully formed new adipose tissue in the subcutaneous area. CONCLUSION AAF innovatively integrates the bionic structure of extracellular matrix and slow-release function, and solves the uneven drug distribution and associated infection risk of traditional intervention methods by regulating the synergistic regeneration of epidermodermis and vascular units. Its mechanical adaptability (dry toughness/wet plasticity) and the ability of inducing adipose regeneration provide a new strategy of both structural strengthening and metabolic support for skin dilation, also laying a mechanism and empirical foundation for clinical transformation of tissue engineering materials.
Collapse
Affiliation(s)
- Baoyan Liang
- Department of Plastic and Reconstructive Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Ruoxue Bai
- Department of Plastic and Reconstructive Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Jiayang Wang
- Department of Plastic and Reconstructive Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Shuyang Shi
- Department of Plastic and Reconstructive Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Yajie Guo
- Department of Digestive Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Qi Wang
- Department of Digestive Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Han Peng
- Department of Plastic and Reconstructive Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China; Innovation Research Institute, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Jiezhang Tang
- Department of Plastic and Burn Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an 710038, China
| | - Shuai Liu
- Department of Digestive Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Jun Zhu
- Department of Digestive Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Chenggang Yi
- Department of Plastic Surgery, The Second Affiliated Hospital of Zhejiang University College of Medicine, Hangzhou 310000, China
| | - Mengmeng Hou
- Department of Plastic and Reconstructive Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China; Innovation Research Institute, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Huichen Li
- Department of Plastic and Reconstructive Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China; Innovation Research Institute, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China.
| |
Collapse
|
17
|
Cristallini C, Rossin D, Vanni R, Barbani N, Bulgheresi C, Labardi M, Perveen S, Burchielli S, Terlizzi D, Kusmic C, Del Ry S, Cabiati M, Trouki C, Rossino D, Sergi F, Villano A, Aquaro GD, Scarpellino G, Ruffinatti FA, Amorim S, Pires RA, Reis RL, Rastaldo R, Giachino C. A biodegradable, microstructured, electroconductive and nano-integrated drug eluting patch (MENDEP) for myocardial tissue engineering. Bioact Mater 2025; 50:246-272. [PMID: 40270551 PMCID: PMC12017858 DOI: 10.1016/j.bioactmat.2025.04.008] [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: 12/04/2024] [Revised: 03/24/2025] [Accepted: 04/05/2025] [Indexed: 04/25/2025] Open
Abstract
We produced a microstructured, electroconductive and nano-functionalized drug eluting cardiac patch (MENDEP) designed to attract endogenous precursor cells, favor their differentiation and counteract adverse ventricular remodeling in situ. MENDEP showed mechanical anisotropy and biaxial strength comparable to porcine myocardium, reduced impedance, controlled biodegradability, molecular recognition ability and controlled drug release activity. In vitro, cytocompatibility and cardioinductivity were demonstrated. Migration tests showed the chemoattractive capacity of the patches and conductivity assays showed unaltered cell-cell interactions and cell beating synchronicity. MENDEP was then epicardially implanted in a rat model of ischemia/reperfusion (I/R). Histological, immunofluorescence and biomarker analysis indicated that implantation did not cause damage to the healthy myocardium. After I/R, MENDEP recruited precursor cells into the damaged myocardium and triggered their differentiation towards the vascular lineage. Under the patch, the myocardial tissue appeared well preserved and cardiac gap junctions were correctly distributed at the level of the intercalated discs. The fibrotic area measured in the I/R group was partially reduced in the patch group. Overall, these results demonstrate that MENDEP was fully retained on the epicardial surface of the left ventricle over 4-week implantation period, underwent progressive vascularization, did not perturb the healthy myocardium and showed great potential in repairing the infarcted area.
Collapse
Affiliation(s)
- Caterina Cristallini
- Institute for Chemical and Physical Processes, CNR-IPCF, Via Giuseppe Moruzzi 1, 56124, Pisa, Italy
- Department of Civil and Industrial Engineering, DICI, University of Pisa, Largo Lucio Lazzarino, 56126, Pisa, Italy
| | - Daniela Rossin
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, 10043, Orbassano, Italy
| | - Roberto Vanni
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, 10043, Orbassano, Italy
| | - Niccoletta Barbani
- Institute for Chemical and Physical Processes, CNR-IPCF, Via Giuseppe Moruzzi 1, 56124, Pisa, Italy
- Department of Civil and Industrial Engineering, DICI, University of Pisa, Largo Lucio Lazzarino, 56126, Pisa, Italy
| | - Chiara Bulgheresi
- Department of Civil and Industrial Engineering, DICI, University of Pisa, Largo Lucio Lazzarino, 56126, Pisa, Italy
| | - Massimiliano Labardi
- Institute for Chemical and Physical Processes, CNR-IPCF, Via Giuseppe Moruzzi 1, 56124, Pisa, Italy
| | - Sadia Perveen
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, 10043, Orbassano, Italy
| | | | | | - Claudia Kusmic
- Clinical Physiology Institute, CNR-IFC, Via Giuseppe Moruzzi 1, 56124, Pisa, Italy
| | - Silvia Del Ry
- Clinical Physiology Institute, CNR-IFC, Via Giuseppe Moruzzi 1, 56124, Pisa, Italy
| | - Manuela Cabiati
- Clinical Physiology Institute, CNR-IFC, Via Giuseppe Moruzzi 1, 56124, Pisa, Italy
| | - Cheherazade Trouki
- Institute for Chemical and Physical Processes, CNR-IPCF, Via Giuseppe Moruzzi 1, 56124, Pisa, Italy
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126, Pisa, Italy
| | - Dawid Rossino
- Institute for Chemical and Physical Processes, CNR-IPCF, Via Giuseppe Moruzzi 1, 56124, Pisa, Italy
- Department of Civil and Industrial Engineering, DICI, University of Pisa, Largo Lucio Lazzarino, 56126, Pisa, Italy
| | - Francesca Sergi
- Department of Civil and Industrial Engineering, DICI, University of Pisa, Largo Lucio Lazzarino, 56126, Pisa, Italy
| | - Anthea Villano
- Institute for Chemical and Physical Processes, CNR-IPCF, Via Giuseppe Moruzzi 1, 56124, Pisa, Italy
| | - Giovanni D. Aquaro
- Academic Radiology Unit, Department of Surgical, Medical and Molecular Pathology and of the Critical Area, University of Pisa, Via Paradisa 2, 56124, Pisa, Italy
| | - Giorgia Scarpellino
- Department of Life Sciences and Systems Biology, University of Turin, Via Accademia Albertina 13, 10123, Torino, Italy
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, Via Adolfo Ferrata 9, 27100, Pavia, Italy
| | - Federico A. Ruffinatti
- Department of Life Sciences and Systems Biology, University of Turin, Via Accademia Albertina 13, 10123, Torino, Italy
| | - Sara Amorim
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017, Barco, Guimarães, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - Ricardo A. Pires
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017, Barco, Guimarães, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - Rui L. Reis
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017, Barco, Guimarães, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - Raffaella Rastaldo
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, 10043, Orbassano, Italy
| | - Claudia Giachino
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, 10043, Orbassano, Italy
| |
Collapse
|
18
|
Severo NC, Inês de Assumpção T, Silva Peixer MA, da Cunha Xavier M, Malard PF, Brunel HDSS, Lançoni R. Effectiveness of intraglandular allogeneic mesenchymal stem cell administration for treating chronic vesicular adenitis in bulls. Theriogenology 2025; 241:117419. [PMID: 40188642 DOI: 10.1016/j.theriogenology.2025.117419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 03/27/2025] [Accepted: 03/31/2025] [Indexed: 04/08/2025]
Abstract
This study aimed to evaluate the effects of the application of allogeneic mesenchymal stem cells (MSCs) bilaterally and intraglandularly in the vesicular glands of bulls affected by seminal vesiculitis. Twelve bulls that presented chronic vesiculitis with two or more recurrences were selected at Semen Collection and Processing Centres, based on the presence of pus in the semen, leukocytes on the motility and vigour evaluation slide, reactive to the California Mastitis Test - CMT (one cross or more) and the presence of leukocytes on a slide stained by Diff Quik staining with more than 5 polymorphonuclear cells (PMN) per field. Ultrasound examination of the vesicular glands was performed, and the clinical signs were definitive for the diagnosis. The proposed method of the intraglandular injection of MSCs involved application through the ischiorectal fossa with a long needle measuring 30-35 cm and a guide measuring 25-30 cm in length directly into the affected vesicular glands. The MSCs were cultured and frozen in the Bio Cell Cellular Therapy® laboratory (Brasilia, Brazil) and prepared by washing and centrifugation for intraglandular injection on the day of application. In total, 3x106 MSCs were injected into each vesicular gland. Data were evaluated for normality of residuals using the Shapiro-Wilk test. When the normality of the test was significant (P < 0.05), the data were transformed or outliers were removed and reevaluated. The "T-Test" was applied to identify statistical differences between variables before and after treatment. The probability of P ≤ 0.05 was considered a significant difference. Data were presented as the mean ± standard error of the mean (S.E.M.). Improvements were observed in the initial percent motility from 60.09 ± 4.8 to 69.89 ± 4.6 (P < 0.05), as well as in the post-thawing percent motility from 26.26 ± 6.77 to 42.5 ± 5 0.99 (P < 0.05). The number of doses produced increased significantly after treatment with MSCs, from 95.61 ± 23.31 units to 337.84 ± 67.75 units (P < 0.05) per ejaculate. The number of leukocytes observed per field decreased from 5.83 ± 0.48 to zero, demonstrating the recovery of the inflamed vesicular glands. Based on the results presented, it was concluded that the application of 3x106 MSCs in the vesicular glands of bulls with vesiculitis is safe and efficient, as it improved several parameters evaluated in this research, mainly the production of semen doses per ejaculate.
Collapse
Affiliation(s)
- Neimar Correa Severo
- School of Veterinary Medicine and Animal Science, Federal University of Uberlândia, Uberlândia, MG, Brazil
| | | | | | | | | | | | - Renata Lançoni
- School of Veterinary Medicine and Animal Science, Federal University of Uberlândia, Uberlândia, MG, Brazil.
| |
Collapse
|
19
|
Poudineh M, Mohammadyari F, Parsamanesh N, Jamialahmadi T, Kesharwani P, Sahebkar A. Cell and gene therapeutic approaches in non-alcoholic fatty liver disease. Gene 2025; 956:149466. [PMID: 40189164 DOI: 10.1016/j.gene.2025.149466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 03/14/2025] [Accepted: 03/31/2025] [Indexed: 04/11/2025]
Abstract
Non-Alcoholic Fatty Liver Disease (NAFLD) refers to a range of conditions marked by the buildup of triglycerides in liver cells, accompanied by inflammation, which contributes to liver damage, clinical symptoms, and histopathological alterations. Multiple molecular pathways contribute to NAFLD pathogenesis, including immune dysregulation, endoplasmic reticulum stress, and tissue injury. Both the innate and adaptive immune systems play crucial roles in disease progression, with intricate crosstalk between liver and immune cells driving NAFLD development. Among emerging therapeutic strategies, cell and gene-based therapies have shown promise. This study reviews the pathophysiological mechanisms of NAFLD and explores the therapeutic potential of cell-based interventions, highlighting their immunomodulatory effects, inhibition of hepatic stellate cells, promotion of hepatocyte regeneration, and potential for hepatocyte differentiation. Additionally, we examine gene delivery vectors designed to target NAFLD, focusing on their role in engineering hepatocytes through gene addition or editing to enhance therapeutic efficacy.
Collapse
Affiliation(s)
| | | | - Negin Parsamanesh
- Metabolic Diseases Research Center, Zanjan University of Medical Sciences, Zanjan, Iran; Department of Genetics and Molecular Medicine, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Tananz Jamialahmadi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Medical Toxicology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Prashant Kesharwani
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya, Sagar, Madhya Pradesh 470003, India.
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Centre for Research Impact and Outcome, Chitkara University, Rajpura 140417, Punjab, India; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
20
|
Zhao W, Wang X, Liu Y, Lu L, Ding Y, Zhang T. Timosaponin AⅢ inhibits ectopic lipid deposition and enhances the browning of white adipose tissue. Eur J Pharmacol 2025; 998:177506. [PMID: 40074140 DOI: 10.1016/j.ejphar.2025.177506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 02/15/2025] [Accepted: 03/10/2025] [Indexed: 03/14/2025]
Abstract
Timosaponin AⅢ(TAⅢ), derived from the Chinese medicinal herb Anemarrhena asphodeloides Bunge, has been reported to have a range of pharmacological effects including improvement of learning and memory deficits, anti-tumor, hypoglycemic effect and anti-hypertension. This study explored the therapeutic effects and preliminary mechanisms of TAⅢ in improving insulin resistance in ob/ob mice. We found that treatment with 10 mg kg-1·d-1 of TAⅢ reduced the expression of SREBPs and alleviated ectopic lipid deposition by decreasing DAG accumulation in liver. The decrease of DAG further inhibited the membrane translocation of PKC-ε, releasing its inhibition of phosphorylation at Ser307 of IRS1, and ultimately enhancing the AKT signaling response to insulin stimulation. In addition, TAⅢ promoted the browning of iWAT by activating the PGC1α-UCP1 axis on ob/ob mice, thereby enhancing fatty acid oxidation and increasing energy consumption, thus reducing its interference with insulin signaling. TAⅢ worked by enhancing the function of adipose tissue and inhibited lipid synthesis. These actions collectively ameliorated metabolic disturbances associated with insulin resistance. Therefore, we preliminarily concluded that TAⅢ improved metabolic disturbances related to insulin resistance. However, further research is needed, additional studies are necessary to validate these potential mechanisms.
Collapse
Affiliation(s)
- Wenjun Zhao
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xiaoying Wang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yun Liu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Lu Lu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yue Ding
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; National Innovation Platform for Medical Industry-Education Integration, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Tong Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; National Innovation Platform for Medical Industry-Education Integration, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
21
|
Jing Y, Zheng W, Zhou Z, Yao H, Zhang W, Wu Y, Guo Z, Huang C, Wang X. Recent research advances of c-fos in regulating cell senescence. Arch Biochem Biophys 2025; 769:110423. [PMID: 40246221 DOI: 10.1016/j.abb.2025.110423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 10/25/2024] [Accepted: 04/14/2025] [Indexed: 04/19/2025]
Abstract
c-fos is an immediate early gene (IEG) that forms a heterodimeric activator protein-1 (AP-1) complex with c-Jun. Following stimulation by a variety of factors, it changes the expression of genes and participates in cellular growth, proliferation, differentiation, and apoptosis. Previous studies have reported that c-fos is linked to cellular senescence and is involved in aging-related signaling pathways or damage repair processes. However, there are limited studies related to this topic. This review summarizes the findings of the connection between c-fos and cellular senescence, including the regulatory role of c-fos in the senescence of stem cells and various kinds of somatic cells. In addition, we discussed the involvement of c-fos in the cellular senescence process and related signaling pathways, as well as the importance of regulating DNA damage repair. The current studies have demonstrated that c-fos has important roles in inhibiting stem cell senescence. They can pave the way for a more thorough examination of the aging process and the regeneration of stem cells and provide new therapeutic strategies for aging-related diseases.
Collapse
Affiliation(s)
- Yuxin Jing
- Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Wei Zheng
- Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Zhou Zhou
- Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Haiyang Yao
- Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Wenchuan Zhang
- Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yilun Wu
- Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Zimo Guo
- Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Chenxuan Huang
- Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xianli Wang
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
22
|
Ilgaz C, Casula L, Sarais G, Schlich M, Dessì D, Cardia MC, Sinico C, Kadiroglu P, Lai F. Proniosomal encapsulation of olive leaf extract for improved delivery of oleuropein: Towards the valorization of an agro-industrial byproduct. Food Chem 2025; 479:143877. [PMID: 40106918 DOI: 10.1016/j.foodchem.2025.143877] [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: 10/03/2024] [Revised: 03/10/2025] [Accepted: 03/11/2025] [Indexed: 03/22/2025]
Abstract
Olive leaf, a by-product of the olive oil industry, is rich in bioactive compounds, including the antioxidant and anti-inflammatory oleuropein. Olive leaf extracts have been explored for nutraceutical applications, but oleuropein's low bioavailability and stability limit its use in food and supplements. This work aimed to mitigate these issues by nano-encapsulating the olive leaf extract in proniosomes-free-flowing powders that form niosomes upon hydration. These niosomes can then be further processed into dosage forms or incorporated into functional foods. Proniosomes based on lactose or mannitol were developed and characterized. Hydration of the proniosomes yielded niosomes with high oleuropein loading and antioxidant activity. These niosomes controlled oleuropein release in simulated gastric and intestinal fluids, protecting it from degradation. Furthermore, niosomal encapsulation enhanced protection against oxidative stress in intestinal cells compared to the unformulated extract, suggesting improved intracellular delivery and making this formulation a suitable candidate as a functional food ingredient.
Collapse
Affiliation(s)
- Ceren Ilgaz
- Food Engineering Department, Adana Alparslan Türkeş Science and Technology University, Adana, Turkey
| | - Luca Casula
- Department of Life and Environmental Sciences, University of Cagliari, Cagliari, Italy
| | - Giorgia Sarais
- Department of Life and Environmental Sciences, University of Cagliari, Cagliari, Italy
| | - Michele Schlich
- Department of Life and Environmental Sciences, University of Cagliari, Cagliari, Italy
| | - Debora Dessì
- Department of Life and Environmental Sciences, University of Cagliari, Cagliari, Italy; Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Maria Cristina Cardia
- Department of Life and Environmental Sciences, University of Cagliari, Cagliari, Italy
| | - Chiara Sinico
- Department of Life and Environmental Sciences, University of Cagliari, Cagliari, Italy
| | - Pınar Kadiroglu
- Food Engineering Department, Adana Alparslan Türkeş Science and Technology University, Adana, Turkey
| | - Francesco Lai
- Department of Life and Environmental Sciences, University of Cagliari, Cagliari, Italy.
| |
Collapse
|
23
|
Tian M, Zhou Y, Guo Y, Xia Q, Wang Z, Zheng X, Shen J, Guo J, Duan S, Wang L. MicroRNAs in adipose tissue fibrosis: Mechanisms and therapeutic potential. Genes Dis 2025; 12:101287. [PMID: 40242037 PMCID: PMC12002615 DOI: 10.1016/j.gendis.2024.101287] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Accepted: 03/07/2024] [Indexed: 04/18/2025] Open
Abstract
Adipose tissue fibrosis, characterized by abnormal extracellular matrix deposition within adipose tissue, signifies a crucial indicator of adipose tissue malfunction, potentially leading to organ tissue dysfunction. Various factors, including a high-fat diet, non-alcoholic fatty liver disease, and insulin resistance, coincide with adipose tissue fibrosis. MicroRNAs (miRNAs) represent a class of small non-coding RNAs with significant influence on tissue fibrosis through diverse signaling pathways. For instance, in response to a high-fat diet, miRNAs can modulate signaling pathways such as TGF-β/Smad, PI3K/AKT, and PPAR-γ to impact adipose tissue fibrosis. Furthermore, miRNAs play roles in inhibiting fibrosis in different contexts: suppressing corneal fibrosis via the TGF-β/Smad pathway, mitigating cardiac fibrosis through the VEGF signaling pathway, reducing wound fibrosis via regulation of the MAPK signaling pathway, and diminishing fibrosis post-fat transplantation via involvement in the PDGFR-β signaling pathway. Notably, the secretome released by miRNA-transfected adipose-derived stem cells facilitates targeted delivery of miRNAs to evade host immune rejection, enhancing their anti-fibrotic efficacy. Hence, this study endeavors to elucidate the role and mechanism of miRNAs in adipose tissue fibrosis and explore the mechanisms and advantages of the secretome released by miRNA-transfected adipose-derived stem cells in combating fibrotic diseases.
Collapse
Affiliation(s)
- Mei Tian
- College of Pharmacy, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
- Geriatric Medicine Center, Department of Endocrinology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang 310015, China
| | - Yang Zhou
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang 310015, China
| | - Yitong Guo
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang 310015, China
| | - Qing Xia
- College of Pharmacy, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang 310015, China
| | - Zehua Wang
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang 310015, China
| | - Xinying Zheng
- College of Pharmacy, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang 310015, China
| | - Jinze Shen
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang 310015, China
| | - Junping Guo
- Rainbowfish Rehabilitation and Nursing School, Hangzhou Vocational & Technical College, Hangzhou, Zhejiang 310018, China
- Department of Clinical Medicine, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang 310015, China
| | - Shiwei Duan
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang 310015, China
| | - Lijun Wang
- Geriatric Medicine Center, Department of Endocrinology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| |
Collapse
|
24
|
Park KS, Lässer C, Lötvall J. Extracellular vesicles and the lung: from disease pathogenesis to biomarkers and treatments. Physiol Rev 2025; 105:1733-1821. [PMID: 40125970 DOI: 10.1152/physrev.00032.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 10/14/2024] [Accepted: 03/12/2025] [Indexed: 03/25/2025] Open
Abstract
Nanosized extracellular vesicles (EVs) are released by all cells to convey cell-to-cell communication. EVs, including exosomes and microvesicles, carry an array of bioactive molecules, such as proteins and RNAs, encapsulated by a membrane lipid bilayer. Epithelial cells, endothelial cells, and various immune cells in the lung contribute to the pool of EVs in the lung microenvironment and carry molecules reflecting their cellular origin. EVs can maintain lung health by regulating immune responses, inducing tissue repair, and maintaining lung homeostasis. They can be detected in lung tissues and biofluids such as bronchoalveolar lavage fluid and blood, offering information about disease processes, and can function as disease biomarkers. Here, we discuss the role of EVs in lung homeostasis and pulmonary diseases such as asthma, chronic obstructive pulmonary disease, cystic fibrosis, pulmonary fibrosis, and lung injury. The mechanistic involvement of EVs in pathogenesis and their potential as disease biomarkers are discussed. Finally, the pulmonary field benefits from EVs as clinical therapeutics in severe pulmonary inflammatory disease, as EVs from mesenchymal stem cells attenuate severe respiratory inflammation in multiple clinical trials. Further, EVs can be engineered to carry therapeutic molecules for enhanced and broadened therapeutic opportunities, such as the anti-inflammatory molecule CD24. Finally, we discuss the emerging opportunity of using different types of EVs for treating severe respiratory conditions.
Collapse
Affiliation(s)
- Kyong-Su Park
- Krefting Research Centre, Institute of Medicine at the Sahlgrenska Academy, University of Gothenburg, Göteborg, Sweden
| | - Cecilia Lässer
- Krefting Research Centre, Institute of Medicine at the Sahlgrenska Academy, University of Gothenburg, Göteborg, Sweden
| | - Jan Lötvall
- Krefting Research Centre, Institute of Medicine at the Sahlgrenska Academy, University of Gothenburg, Göteborg, Sweden
| |
Collapse
|
25
|
García-Briega MI, Plá-Salom J, Clara-Trujillo S, Tolosa L, Cordón L, Sempere A, Ribelles JLG. Co-culture of multiple myeloma cell lines and bone marrow mesenchymal stem cells in a 3D microgel environment. BIOMATERIALS ADVANCES 2025; 172:214243. [PMID: 40054228 DOI: 10.1016/j.bioadv.2025.214243] [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: 11/04/2024] [Revised: 12/20/2024] [Accepted: 02/24/2025] [Indexed: 03/17/2025]
Abstract
This study reproduces the complex relationships between tumour plasma cells and their bone marrow microenvironment in multiple myeloma in vitro. These relationships are established both with other cells and with the extracellular matrix and are key factors in tumour progression, generating resistance to antitumour drugs in the cellular and non-cellular environments. This paper proposes a 3D microenvironment model designed to capture the main components of the multiple myeloma tumour microenvironment. Multiple myeloma cells (MMCs) were dispersed in a microgel medium formed by gel-textured microspheres. The proteins and polysaccharides considered important in the interaction of the MMCs with their non-cellular environment were successfully grafted onto the surface of the microspheres, while human mesenchymal stem cells (MSCs) were cultured in a pellet with non-functionalised microspheres. The MSCs pellet was placed in the well plate together with the microgel and the MMCs and orbitally shaken to maintain them in suspension. The viability, cell cycle and proliferation of the RPMI8226, MM1S and U266 multiple myeloma cell lines and the direct adhesion of MMCs to the MSC pellet were quantified. The results revealed that all three cell lines are able to grow satisfactorily. In addition, the normal behaviour of the MMCs is not modified in any of the culture conditions studied.
Collapse
Affiliation(s)
- M Inmaculada García-Briega
- Centre for Biomaterials and Tissue Engineering, CBIT, Universitat Politècnica de València, 46022, Valencia, Spain; Centro de Investigación Biomédica en Red de Bioingeniería Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, Spain.
| | - Júlia Plá-Salom
- Centre for Biomaterials and Tissue Engineering, CBIT, Universitat Politècnica de València, 46022, Valencia, Spain
| | - Sandra Clara-Trujillo
- Centre for Biomaterials and Tissue Engineering, CBIT, Universitat Politècnica de València, 46022, Valencia, Spain; Centro de Investigación Biomédica en Red de Bioingeniería Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, Spain; Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat de València-Universitat Politècnica de València, Camino de Vera s/n, Valencia 46022, Spain; Unidad Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades y Nanomedicina, Centro de Investigación Príncipe Felipe, Universitat Politècnica de València, Valencia 46013, Spain
| | - Laia Tolosa
- Centro de Investigación Biomédica en Red de Bioingeniería Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, Spain; Experimental Hepatology Unit, Health Research Institute La Fe (IIS La Fe), Valencia 46026, Spain
| | - Lourdes Cordón
- Hematology Research Group, Instituto de Investigación Sanitaria La Fe (IISLAFE), 46022, Valencia, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto Carlos III, 28029 Madrid, Spain
| | - Amparo Sempere
- Hematology Research Group, Instituto de Investigación Sanitaria La Fe (IISLAFE), 46022, Valencia, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto Carlos III, 28029 Madrid, Spain; Haematology Department, Hospital Universitari i Politècnic La Fe, 46026, Valencia, Spain
| | - José Luís Gómez Ribelles
- Centre for Biomaterials and Tissue Engineering, CBIT, Universitat Politècnica de València, 46022, Valencia, Spain; Centro de Investigación Biomédica en Red de Bioingeniería Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, Spain
| |
Collapse
|
26
|
Moghassemi S, Nikanfar S, Dadashzadeh A, Sousa MJ, Wan Y, Sun F, Colson A, De Windt S, Kwaspen L, Kanbar M, Sobhani K, Yang J, Vlieghe H, Li Y, Debiève F, Wyns C, Amorim CA. The revolutionary role of placental derivatives in biomedical research. Bioact Mater 2025; 49:456-485. [PMID: 40177109 PMCID: PMC11964572 DOI: 10.1016/j.bioactmat.2025.03.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 03/11/2025] [Accepted: 03/14/2025] [Indexed: 04/05/2025] Open
Abstract
The human placenta is a transient yet crucial organ that plays a key role in sustaining the relationship between the maternal and fetal organisms. Despite its historical classification as "biowaste," placental tissues have garnered increasing attention since the early 1900s for their significant medical potential, particularly in wound repair and surgical application. As ethical considerations regarding human placental derivatives have largely been assuaged in many countries, they have gained significant attention due to their versatile applications in various biomedical fields, such as biomedical engineering, regenerative medicine, and pharmacology. Moreover, there is a substantial trend toward various animal product substitutions in laboratory research with human placental derivatives, reflecting a broader commitment to advancing ethical and sustainable research methodologies. This review provides a comprehensive examination of the current applications of human placental derivatives, explores the mechanisms behind their therapeutic effects, and outlines the future potential and directions of this rapidly advancing field.
Collapse
Affiliation(s)
- Saeid Moghassemi
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Saba Nikanfar
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Arezoo Dadashzadeh
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Maria João Sousa
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Yuting Wan
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Fengxuan Sun
- Department of Obstetrics, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Arthur Colson
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
- Department of Obstetrics, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Sven De Windt
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
- Department of Gynecology and Andrology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Lena Kwaspen
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
- Department of Gynecology and Andrology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Marc Kanbar
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
- Department of Gynecology and Andrology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Keyvan Sobhani
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Jie Yang
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Hanne Vlieghe
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Yongqian Li
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Frédéric Debiève
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
- Department of Obstetrics, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Christine Wyns
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
- Department of Gynecology and Andrology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Christiani A. Amorim
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| |
Collapse
|
27
|
Akhtar M, Nashwan AJ. Evaluating Wharton’s jelly-derived stem cell therapy in autism: Insights from a case study. World J Methodol 2025; 15:100074. [DOI: 10.5662/wjm.v15.i2.100074] [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: 08/06/2024] [Revised: 09/26/2024] [Accepted: 09/30/2024] [Indexed: 11/27/2024] Open
Abstract
Autism spectrum disorder (ASD) is a complex neurodevelopmental disorder affecting over 2% of the global population, marked by social communication deficits and repetitive behaviors. Kabatas et al explored the efficacy and safety of Wharton’s jelly-derived mesenchymal stem cell (WJ-MSC) therapy in a 4-year-old child with ASD. Using the childhood autism rating scale and Denver II developmental screening test, significant improvements were seen after six WJ-MSC sessions, with no adverse events over 2 years. Despite promising results, the study’s single-case design limits generalizability. Larger, multi-center trials are needed to validate the findings and assess long-term effects of WJ-MSC therapy in ASD.
Collapse
Affiliation(s)
- Muzamil Akhtar
- College of Medicine, Gujranwala Medical College, Gujranwala 52250, Punjab, Pakistan
| | - Abdulqadir J Nashwan
- Department of Nursing and Midwifery Research, Hamad Medical Corporation, Doha 3050, Qatar
| |
Collapse
|
28
|
Isildar B, Ozkan S, Neccar D, Koyuturk M. Preconditioning and post-preconditioning states of mesenchymal stem cells with deferoxamine: A comprehensive analysis. Eur J Pharmacol 2025; 996:177574. [PMID: 40180273 DOI: 10.1016/j.ejphar.2025.177574] [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: 01/29/2025] [Revised: 03/17/2025] [Accepted: 03/31/2025] [Indexed: 04/05/2025]
Abstract
Mesenchymal stem cells (MSCs) derive their therapeutic potential from their secretomes, which can be modulated by external stimuli. Hypoxia is one such stimulus, and research on preconditioning MSCs with hypoxia-mimetic agents is rising. However, the effects of these preconditioning processes and the resulting metabolic status require further investigation. This study evaluated the effects of deferoxamine (DFX), a hypoxia-mimetic agent, preconditioning on MSCs in serum and serum-free environments. The influence of hypoxia on cell metabolism was examined during and after preconditioning by assessing cytotoxicity, proliferation, migration, secretomes, senescence, autophagy, and apoptosis mechanisms. The optimal DFX dose and duration for preconditioning were determined as 150 μM and 24 h based on cytotoxicity testing. Accordingly, DFX preconditioning significantly upregulated HIF-1α expression, increasing protein secretion and reducing total oxidant status. DFX appears to enhance the therapeutic potential of MSCs by increasing their secretome and antioxidant capacity. However, upon DFX removal, HIF-1α levels returned to normal, and the associated positive effects diminished. Autophagy was markedly enhanced during DFX preconditioning, potentially improving metabolic activity, preserving cellular integrity, and preparing MSCs for ischemic environments. Autophagy returned to baseline after DFX withdrawal, indicating a temporary hypoxia-mimetic response. In a serum-containing medium, specific effects of preconditioning were relatively weak to be observed. This study demonstrates that DFX-preconditioning increases MSCs' metabolic activity and enhances their adaptive cellular response. However, the effect may be transient, which provides insights into the behavior of MSCs in ischemic environments and emphasizes the need to evaluate the long-term effects of hypoxia-mimetic agents.
Collapse
Affiliation(s)
- Basak Isildar
- Balıkesir University, Histology and Embryology Department, Balıkesir, Turkey.
| | - Serbay Ozkan
- İzmir Katip Çelebi University, Histology and Embryology Department, Izmir, Turkey.
| | - Duygu Neccar
- İstanbul University-Cerrahpaşa, Cerrahpaşa Faculty of Medicine, Department of Histology and Embryology, Istanbul, Turkey.
| | - Meral Koyuturk
- İstanbul University-Cerrahpaşa, Cerrahpaşa Faculty of Medicine, Department of Histology and Embryology, Istanbul, Turkey.
| |
Collapse
|
29
|
Gao Q, Peng J, Xiong M, Zhou S, Wang X, Lu J, Guo Y, Peng Q, Zeng M, Song H. Sinomenine alleviates experimental autoimmune uveitis in rats: Possible involvement of PI3K/AKT and NF-κB signaling pathways. Eur J Pharmacol 2025; 996:177571. [PMID: 40180267 DOI: 10.1016/j.ejphar.2025.177571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 03/23/2025] [Accepted: 03/31/2025] [Indexed: 04/05/2025]
Abstract
Uveitis is an inflammatory ocular condition characterized by an autoimmune etiology. Sinomenine (SIN), the main active constituent of the rhizome of Sinomenium acutum (Thunb.) Rehd. et Wils., exhibits both anti-inflammatory and immunosuppressive properties. The present study sought to investigate the therapeutic effects of SIN on experimental autoimmune uveitis (EAU) in rats and to elucidate its underlying mechanisms. A bioinformatics analysis was conducted to identify signaling pathways implicated in the pathogenesis of uveitis, leading to the identification of the PI3K/AKT and NF-κB pathways for further experimental validation. An EAU model was subsequently established, and the ocular surface morphology was examined using slit lamp microscopy and hematoxylin-eosin staining. Immunofluorescence was utilized to measure the protein expression and distribution. Enzyme-linked immunosorbent assay was used to determine the expression of inflammatory cytokines. Experimental findings demonstrated that SIN significantly decreased ocular inflammation scores. Further validation revealed that SIN significantly elevated levels of interleukin-10 (IL-10) while reducing levels of IL-17, tumor necrosis factor-α (TNF-α), and IL-1β in EAU rats. SIN significantly suppressed the expression of phosphorylated proteins in the PI3K/AKT and NF-κB pathways. In addition, it reduced the expression of RORγt while enhancing the expression of Foxp3, the transcription factors associated with Th17 cells and Tregs, respectively. In summary, our data demonstrate that SIN alleviates EAU inflammation by inhibiting the activation of the PI3K/AKT and NF-κB signaling pathways and restoring the balance between Th17 and Tregs. These findings highlight SIN as a promising therapeutic agent for the treatment of uveitis.
Collapse
Affiliation(s)
- Qing Gao
- Hunan Provincial Key Laboratory of Traditional Chinese Medicine Diagnostics, Hunan University of Chinese Medicine, Changsha, Hunan Province, China; School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Jun Peng
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Meng Xiong
- Hunan Provincial Key Laboratory of Traditional Chinese Medicine Diagnostics, Hunan University of Chinese Medicine, Changsha, Hunan Province, China; School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Shunhua Zhou
- Hunan Provincial Key Laboratory of Traditional Chinese Medicine Diagnostics, Hunan University of Chinese Medicine, Changsha, Hunan Province, China; School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Xiaojuan Wang
- Hunan Provincial Key Laboratory of Traditional Chinese Medicine Diagnostics, Hunan University of Chinese Medicine, Changsha, Hunan Province, China; School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Jing Lu
- School of Medicine, Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Yuanyi Guo
- School of Stomatology, Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Qinghua Peng
- Hunan Provincial Key Laboratory of Traditional Chinese Medicine Diagnostics, Hunan University of Chinese Medicine, Changsha, Hunan Province, China; School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Meiyan Zeng
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan Province, China.
| | - Houpan Song
- Hunan Provincial Key Laboratory of Traditional Chinese Medicine Diagnostics, Hunan University of Chinese Medicine, Changsha, Hunan Province, China; School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan Province, China.
| |
Collapse
|
30
|
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] [MESH Headings] [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
|
31
|
Li Y, Xue J, Ma Y, Ye K, Zhao X, Ge F, Zheng F, Liu L, Gao X, Wang D, Xia Q. The complex roles of m 6 A modifications in neural stem cell proliferation, differentiation, and self-renewal and implications for memory and neurodegenerative diseases. Neural Regen Res 2025; 20:1582-1598. [PMID: 38845217 PMCID: PMC11688559 DOI: 10.4103/nrr.nrr-d-23-01872] [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: 11/13/2023] [Revised: 02/19/2024] [Accepted: 03/25/2024] [Indexed: 08/07/2024] Open
Abstract
N6-methyladenosine (m 6 A), the most prevalent and conserved RNA modification in eukaryotic cells, profoundly influences virtually all aspects of mRNA metabolism. mRNA plays crucial roles in neural stem cell genesis and neural regeneration, where it is highly concentrated and actively involved in these processes. Changes in m 6 A modification levels and the expression levels of related enzymatic proteins can lead to neurological dysfunction and contribute to the development of neurological diseases. Furthermore, the proliferation and differentiation of neural stem cells, as well as nerve regeneration, are intimately linked to memory function and neurodegenerative diseases. This paper presents a comprehensive review of the roles of m 6 A in neural stem cell proliferation, differentiation, and self-renewal, as well as its implications in memory and neurodegenerative diseases. m 6 A has demonstrated divergent effects on the proliferation and differentiation of neural stem cells. These observed contradictions may arise from the time-specific nature of m 6 A and its differential impact on neural stem cells across various stages of development. Similarly, the diverse effects of m 6 A on distinct types of memory could be attributed to the involvement of specific brain regions in memory formation and recall. Inconsistencies in m 6 A levels across different models of neurodegenerative disease, particularly Alzheimer's disease and Parkinson's disease, suggest that these disparities are linked to variations in the affected brain regions. Notably, the opposing changes in m 6 A levels observed in Parkinson's disease models exposed to manganese compared to normal Parkinson's disease models further underscore the complexity of m 6 A's role in neurodegenerative processes. The roles of m 6 A in neural stem cell proliferation, differentiation, and self-renewal, and its implications in memory and neurodegenerative diseases, appear contradictory. These inconsistencies may be attributed to the time-specific nature of m 6 A and its varying effects on distinct brain regions and in different environments.
Collapse
Affiliation(s)
- Yanxi Li
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
- College of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Jing Xue
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
- College of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Yuejia Ma
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
- College of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Ke Ye
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
- College of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Xue Zhao
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
- College of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Fangliang Ge
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
- College of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Feifei Zheng
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
- College of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Lulu Liu
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
- College of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Xu Gao
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
- College of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
- Basic Medical Institute, Heilongjiang Academy of Medical Sciences, Harbin, Heilongjiang Province, China
- Key Laboratory of Heilongjiang Province for Genetically Modified Animals, Harbin Medical University, Harbin, Heilongjiang Province, China
- Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, Heilongjiang Province, China
| | - Dayong Wang
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
- College of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
- Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, Heilongjiang Province, China
| | - Qing Xia
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
32
|
Zeng L, Chen C, Xiong Y, Liu Y, Huang M, Ye J, Zhong J, Peng W. Acetylation of H3K18 activated by p300 promotes osteogenesis in human adipose-derived mesenchymal stem cells. Biochem Pharmacol 2025; 236:116901. [PMID: 40164340 DOI: 10.1016/j.bcp.2025.116901] [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: 11/09/2024] [Revised: 02/26/2025] [Accepted: 03/25/2025] [Indexed: 04/02/2025]
Abstract
Human adipose-derived mesenchymal stem cells (hAD-MSCs) have garnered significant interest as a viable alternative source of stem cells for applications in bone tissue engineering due to their high and ease availability. At present, the limited studies on potential epigenetic regulatory mechanism in hAD-MSCs greatly hinders its clinical application in bone repair. Histone acetylation has been identified as a critical regulator of the osteogenic differentiation of mesenchymal stem cells (MSCs), with increased levels of histone acetylation sites frequently correlating with enhanced osteogenic differentiation. However, their specific roles in MSCs osteogenesis remain unclear. In this study, we observed a significant up-regulation of H3K18 acetylation (H3K18ac) during the osteogenic induction of hAD-MSCs. This modification was notably enriched in the promoter regions of genes associated with osteogenesis, thereby facilitating osteogenic differentiation. Furthermore, the treatment of histone acetyltransferases p300 inhibitor A-485 in hAD-MSCs resulted in a reduction of H3K18 acetylation levels during the osteogenic differentiation, which corresponded with a diminished osteoblast phenotype and function. These results indicated that p300-mediated acetylation of H3K18 enhances the osteogenic differentiation of hAD-MSCs. It provides a novel insight into understanding the mechanism of osteogenic differentiation of hAD-MSCs and promoting its application in bone tissue engineering.
Collapse
Affiliation(s)
- Liping Zeng
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang 330006, China; Jiangxi Province Key Laboratory of Biomaterials and Biofabrication for Tissue Engineering, Gannan Medical University, Ganzhou 341000, China; Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou 341000, China
| | - Chen Chen
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang 330006, China; Jiangxi Province Key Laboratory of Biomaterials and Biofabrication for Tissue Engineering, Gannan Medical University, Ganzhou 341000, China; Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou 341000, China
| | - Yafei Xiong
- Jiangxi Province Key Laboratory of Biomaterials and Biofabrication for Tissue Engineering, Gannan Medical University, Ganzhou 341000, China; Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou 341000, China; School of Medical Information Engineering, Gannan Medical University, Ganzhou 341000, China
| | - Yinan Liu
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang 330006, China; Jiangxi Province Key Laboratory of Biomaterials and Biofabrication for Tissue Engineering, Gannan Medical University, Ganzhou 341000, China; Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou 341000, China
| | - Miao Huang
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou 341000, China
| | - Junsong Ye
- Jiangxi Province Key Laboratory of Biomaterials and Biofabrication for Tissue Engineering, Gannan Medical University, Ganzhou 341000, China; Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou 341000, China; Subcenter for Stem Cell Clinical Translation, The First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
| | - Jianing Zhong
- Jiangxi Province Key Laboratory of Biomaterials and Biofabrication for Tissue Engineering, Gannan Medical University, Ganzhou 341000, China; Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou 341000, China.
| | - Weijie Peng
- Jiangxi Province Key Laboratory of Biomaterials and Biofabrication for Tissue Engineering, Gannan Medical University, Ganzhou 341000, China; Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou 341000, China; School of Pharmaceutics, Nanchang Medical College, Nanchang, 330000, China.
| |
Collapse
|
33
|
Li J, Wang D, Li F, Liao X, Yin D, Wei T, Le Y, Cui X, Yang J, Wei R, Hong T. Mulberry twig (Sangzhi) alkaloids induce pancreatic α-to-β-cell phenotypic conversion in type 2 diabetic mice. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156685. [PMID: 40220417 DOI: 10.1016/j.phymed.2025.156685] [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: 09/12/2024] [Revised: 03/19/2025] [Accepted: 03/23/2025] [Indexed: 04/14/2025]
Abstract
BACKGROUND Pancreatic β-cell regeneration represents a promising therapeutic strategy for diabetes, yet safe and effective treatments remain elusive. PURPOSE We aimed to investigate whether and how mulberry twig (Sangzhi) alkaloids (SZ-A), a newly approved anti-diabetic Chinese medicine, promoted β-cell regeneration. METHODS Diabetic db/db mice and pancreatic α-cell lineage-tracing mice were treated with SZ-A, acarbose, or vehicle daily via intragastric gavage. Blood glucose and plasma insulin levels were measured. The areas of islets, α-cells and β-cells were quantified. Cell transdifferentiation was assessed by double-immunostaining of glucagon or α-cell lineage-tracing marker with β-cell-specific markers. Mouse α-cells were incubated with SZ-A or its three main components, and the mRNA levels of cell transdifferentiation-related genes were detected. RNA-sequencing was performed to screen potential targets. The activities of five mitochondrial complexes were detected following treatment, and specific inhibitor was utilized to validate the involvement. RESULTS Both SZ-A and acarbose improved glycemic control, but only SZ-A enlarged islet and β-cell areas in the diabetic mice. SZ-A induced α-to-β-cell conversion, as indicated by glucagon and insulin double-immunostaining and confirmed by α-cell lineage-tracing. In cultured α-cells, SZ-A and its main component 1-deoxynojirimycin (DNJ) downregulated the expressions of α-cell-specific markers, while upregulated the expressions of β-cell-specific markers. DNJ-induced differentially expressed genes were enriched in the mitochondrial protein complex term. DNJ inhibited mitochondrial complex I activity, and the complex inhibitor induced α-to-β-cell conversion. CONCLUSION SZ-A, especially its main component DNJ, induces α-to-β-cell transdifferentiation via inhibiting mitochondrial complex I. Our finding provides a potential strategy for β-cell regeneration and diabetes treatment.
Collapse
Affiliation(s)
- Jian Li
- Department of Endocrinology and Metabolism, State Key Laboratory of Female Fertility Promotion, Peking University Third Hospital, Beijing 100191, PR China
| | - Dandan Wang
- Department of Endocrinology and Metabolism, State Key Laboratory of Female Fertility Promotion, Peking University Third Hospital, Beijing 100191, PR China
| | - Fei Li
- Department of Endocrinology and Metabolism, State Key Laboratory of Female Fertility Promotion, Peking University Third Hospital, Beijing 100191, PR China
| | - Xinyue Liao
- Department of Endocrinology and Metabolism, State Key Laboratory of Female Fertility Promotion, Peking University Third Hospital, Beijing 100191, PR China
| | - Deshan Yin
- Department of Endocrinology and Metabolism, State Key Laboratory of Female Fertility Promotion, Peking University Third Hospital, Beijing 100191, PR China
| | - Tianjiao Wei
- Department of Endocrinology and Metabolism, State Key Laboratory of Female Fertility Promotion, Peking University Third Hospital, Beijing 100191, PR China
| | - Yunyi Le
- Department of Endocrinology and Metabolism, State Key Laboratory of Female Fertility Promotion, Peking University Third Hospital, Beijing 100191, PR China
| | - Xiaona Cui
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Endocrinology and Metabolism, Peking University Third Hospital, Beijing 100191, PR China
| | - Jin Yang
- Department of Endocrinology and Metabolism, State Key Laboratory of Female Fertility Promotion, Peking University Third Hospital, Beijing 100191, PR China
| | - Rui Wei
- Department of Endocrinology and Metabolism, State Key Laboratory of Female Fertility Promotion, Peking University Third Hospital, Beijing 100191, PR China; NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Peking University, Beijing 100191, PR China.
| | - Tianpei Hong
- Department of Endocrinology and Metabolism, State Key Laboratory of Female Fertility Promotion, Peking University Third Hospital, Beijing 100191, PR China; NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Peking University, Beijing 100191, PR China.
| |
Collapse
|
34
|
Andersen C, Jacobsen S, Uvebrant K, Griffin JF, Vonk LA, Walters M, Berg LC, Lundgren-Åkerlund E, Lindegaard C. Integrin α10β1-Selected Mesenchymal Stem Cells Reduce Pain and Cartilage Degradation and Increase Immunomodulation in an Equine Osteoarthritis Model. Cartilage 2025; 16:250-264. [PMID: 37990503 DOI: 10.1177/19476035231209402] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2023] Open
Abstract
ObjectiveIntegrin α10β1-selected mesenchymal stem cells (integrin α10-MSCs) have previously shown potential in treating cartilage damage and osteoarthritis (OA) in vitro and in animal models in vivo. The aim of this study was to further investigate disease-modifying effects of integrin α10-MSCs.DesignOA was surgically induced in 17 horses. Eighteen days after surgery, horses received 2 × 107 integrin α10-MSCs intra-articularly or were left untreated. Lameness and response to carpal flexion was assessed weekly along with synovial fluid (SF) analysis. On day 52 after treatment, horses were euthanized, and carpi were evaluated by computed tomography (CT), MRI, histology, and for macroscopic pathology and integrin α10-MSCs were traced in the joint tissues.ResultsLameness and response to carpal flexion significantly improved over time following integrin α10-MSC treatment. Treated horses had milder macroscopic cartilage pathology and lower cartilage histology scores than the untreated group. Prostaglandin E2 and interleukin-10 increased in the SF after integrin α10-MSC injection. Integrin α10-MSCs were found in SF from treated horses up to day 17 after treatment, and in the articular cartilage and subchondral bone from 5 of 8 treated horses after euthanasia at 52 days after treatment. The integrin α10-MSC injection did not cause joint flare.ConclusionThis study demonstrates that intra-articular (IA) injection of integrin α10-MSCs appears to be safe, alleviate pathological changes in the joint, and improve joint function in an equine post-traumatic osteoarthritis (PTOA) model. The results suggest that integrin α10-MSCs hold promise as a disease-modifying osteoarthritis drug (DMOAD).
Collapse
Affiliation(s)
- Camilla Andersen
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Taastrup, Denmark
- Xintela AB, Lund, Sweden
| | - Stine Jacobsen
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Taastrup, Denmark
| | | | - John F Griffin
- Department of Large Animal Clinical Sciences, Texas A&M University, College Station, TX, USA
| | | | - Marie Walters
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Taastrup, Denmark
| | - Lise Charlotte Berg
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Taastrup, Denmark
| | | | - Casper Lindegaard
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Taastrup, Denmark
| |
Collapse
|
35
|
Abd Rahman F, Azwa FN. Comparative Dental Pulp Stem Cells (DPSCs) and Periodontal Ligament Stem Cells (PDLSCs): Difference in effect of aspirin on osteoblast potential of PDLSCs and DPSCs. Tissue Cell 2025; 94:102776. [PMID: 40022908 DOI: 10.1016/j.tice.2025.102776] [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: 10/14/2024] [Revised: 01/27/2025] [Accepted: 02/01/2025] [Indexed: 03/04/2025]
Abstract
Periodontal Ligament Stem Cells (PDLSCs) and Dental Pulp Stem Cells (DPSCs) are mesenchymal stem cells with the ability to self-renew and differentiate into three lineages. One significant advantage of dental stem cells, such as PDLSCs and DPSCs, is their ease of harvest compared to other types of mesenchymal stem cells (MSCs). While MSCs are highly valued in bone tissue engineering, MSCs sourced from dental tissues, such as PDLSCs and DPSCs, offer promising options for periodontal regeneration because they are more easily accessible and can be collected through minimally invasive methods. Currently, PDLSCs and DPSCs exhibit a strong ability to undergo osteogenic differentiation when stimulated by factors such as growth factors, chemicals, and paracrine signaling. It has been shown that aspirin (ASA) can enhance the osteoblastic potential of PDLSCs and DPSCs, although the exact mechanism remains unclear. This article examines the origin and features of mesenchymal stem cells, the bone regeneration potential of DPSCs and PDLSCs, the factors that enhance their osteogenic differentiation, and a comparison of PDLSCs and DPSCs regarding their proliferation and differentiation abilities. Additionally, we will examine the effects of aspirin on PDLSCs and DPSCs. In conclusion, PDLSCs show a greater effect on osteoblast differentiation.
Collapse
Affiliation(s)
- Fazliny Abd Rahman
- School of Dentistry (SoD), Management & Science University (MSU), University Drive, Off Persiaran Olahraga, 40100 Shah Alam, Selangor.
| | - Fatin Nur Azwa
- Faculty of Dentistry, Oral Cancer Research Centre (ORCC), University of Malaya (UM), Wilayah Persekutuan, Kuala Lumpur 50603, Malaysia
| |
Collapse
|
36
|
Maeda T, Sobajima S, Matsumoto T, Tsubosaka M, Matsushita T, Iwaguro H, Kuroda R. Comparison of short-term clinical outcomes of intra-articular injection of micro-fragmented adipose tissue and stromal vascular fraction cells for knee osteoarthritis treatment: A retrospective single-center cohort study. Regen Ther 2025; 29:91-99. [PMID: 40129683 PMCID: PMC11932757 DOI: 10.1016/j.reth.2025.02.013] [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: 01/23/2025] [Revised: 02/15/2025] [Accepted: 02/27/2025] [Indexed: 03/26/2025] Open
Abstract
Background Stromal vascular fraction (SVF) cells and micro-fragmented adipose tissue (MFAT) have potential for treating knee osteoarthritis (OA), but their efficacy has not been compared. This study aimed to compare the clinical outcomes of SVF and MFAT for knee OA. We hypothesized that SVF provides stronger short-term effects, while MFAT offers more sustained benefits. Methods A retrospective single-center cohort study was conducted on patients with knee OA, with 36 SVF and 36 MFAT cases selected through propensity score matching between September 2017 and February 2022. Patients with KL grades I-IV varus knee OA, significant pain (VAS ≥40), and functional impairment despite conservative treatments were included. Those with knee trauma, severe bony defects, infections, genu valgus, osteonecrosis, rheumatoid arthritis, or severe deformities were excluded. Clinical outcomes were assessed using the visual analog scale, KOOS, knee range of motion, extension/flexion strength, and MRI T2 mapping. Results SVF and MFAT groups demonstrated significant improvements in VAS (p < 0.01 for both groups). Both groups showed notable improvements in extension angle, extension/flexion muscle strength, and KOOS, with no significant differences between them. However, the MFAT group demonstrated significantly greater improvement in flexion angle compared to the SVF group (p = 0.03). No serious adverse events were reported. T2 mapping showed significant improvements in cartilage quality in both groups, with the MFAT group demonstrating superior improvements in specific lateral regions. Responder rate in SVF group initially improved but declined over time; however, the MFAT group showed sustained improvement from six months onward. Conclusion T2 mapping revealed that MFAT had better cartilage preservation than that of SVF cells in less-loaded areas, with a potentially longer-lasting therapeutic effect. These findings offer important insights for clinicians to tailor treatment strategies based on patient needs and disease progression.
Collapse
Affiliation(s)
- Takuma Maeda
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
- Department of Orthopaedic Surgery, Sobajima Clinic, Higashiosaka, Japan
| | - Satoshi Sobajima
- Department of Orthopaedic Surgery, Sobajima Clinic, Higashiosaka, Japan
| | - Tomoyuki Matsumoto
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Masanori Tsubosaka
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Takehiko Matsushita
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Hideki Iwaguro
- Department of Orthopaedic Surgery, Sobajima Clinic, Higashiosaka, Japan
| | - Ryosuke Kuroda
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|
37
|
Saeedi P, Nilchiani LS, Zand B, Hajimirghasemi M, Halabian R. An overview of stem cells and cell products involved in trauma injury. Regen Ther 2025; 29:60-76. [PMID: 40143930 PMCID: PMC11938091 DOI: 10.1016/j.reth.2025.02.011] [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: 11/14/2024] [Revised: 02/01/2025] [Accepted: 02/20/2025] [Indexed: 03/28/2025] Open
Abstract
Trauma injuries represent a significant public health burden worldwide, often leading to long-term disability and reduced quality of life. This review provides a comprehensive overview of the therapeutic potential of stem cells and cell products for traumatic injuries. The extraordinary characteristics of stem cells, such as self-renewal and transdifferentiation, make them definitive candidates for tissue regeneration. Mesenchymal stem cells (MSCs), neural stem cells (NSCs), and hematopoietic stem cells (HSCs) have been tested in preclinical studies for treating distinct traumatic injuries. Stem cell mechanisms of action are addressed through paracrine signaling, immunomodulation, differentiation, and neuroprotection. Cell products such as conditioned media, exosomes, and secretomes offer cell-free resources, thereby avoiding the risks of live cell transplantation. Clinical trials have reported many effective outcomes; however, variability exists across trauma types. Some challenges include tumorigenicity, standardized protocols, and regulatory issues. Collaboration and interdisciplinary research are being conducted to harness stem cells and products for trauma treatment. This emerging field is promising for improving patient recovery and quality of life after traumatic injuries.
Collapse
Affiliation(s)
- Pardis Saeedi
- Research Center for Health Management in Mass Gathering, Red Crescent Society of the Islamic Republic of Iran, Tehran, Iran
- Applied Microbiology Research Center, Biomedicine Technologies Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Leila Sadat Nilchiani
- Department of Molecular and Cell Biology, Faculty of Advanced Sciences and Technology, Islamic Azad University Tehran Medical Sciences, Tehran, Iran
| | - Bita Zand
- Department of Molecular and Cell Biology, Faculty of Advanced Sciences and Technology, Islamic Azad University Tehran Medical Sciences, Tehran, Iran
| | - Maryam Hajimirghasemi
- Department of Internal Medicine, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Raheleh Halabian
- Applied Microbiology Research Center, Biomedicine Technologies Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
38
|
Zununi Vahed S, Hejazian SM, Bakari WN, Landon R, Gueguen V, Meddahi-Pellé A, Anagnostou F, Barzegari A, Pavon-Djavid G. Milking mesenchymal stem cells: Updated protocols for cell lysate, secretome, and exosome extraction, and comparative analysis of their therapeutic potential. Methods 2025; 238:40-60. [PMID: 40058715 DOI: 10.1016/j.ymeth.2025.03.004] [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: 11/05/2024] [Revised: 02/28/2025] [Accepted: 03/04/2025] [Indexed: 03/21/2025] Open
Abstract
The potential of the cell lysate, secretome, and extracellular vesicles (EVs) of mesenchymal stem cells (MSCs) to modulate the immune response and promote tissue regeneration has positioned them as a promising option for cell-free therapy. Currently, many clinical trials in stem cells-derived EVs and secretome are in progress various diseases and sometimes the results are failing. The major challenge on this roadmap is the lack of a standard extraction method for exosome, secretome, and lysate. The most optimal method for obtaining the secretome of MSCs for clinical utilization involves a comprehensive approach that includes non-destructive collection methods, time optimization, multiple collection rounds, optimization of culture conditions, and quality control measures. Further research and clinical studies are warranted to validate and refine these methods for safe and effective utilization of the MSC exosome, secretome, and lysate in various clinical applications. To address these challenges, it is imperative to establish a standardized and unified methodology to ensure reliable evaluation of these extractions in clinical trials. This review seeks to outline the pros and cons of methods for the preparation of MSCs-derived exosome, and secretome/lysate, and comparative analysis of their therapeutic potential.
Collapse
Affiliation(s)
| | | | - William Ndjidda Bakari
- Université Sorbonne Paris Nord, INSERM U1148, Laboratory for Vascular Translational Science, Nanotechnologies for Vascular Medicine and Imaging Team, 99 Av. Jean-Baptiste Clément 93430 Villetaneuse, France; Université Paris Cité, CNRS UMR7052, INSERM U1271, ENVA, B3OA, F-75010 Paris, France
| | - Rebecca Landon
- Université Paris Cité, CNRS UMR7052, INSERM U1271, ENVA, B3OA, F-75010 Paris, France
| | - Virginie Gueguen
- Université Sorbonne Paris Nord, INSERM U1148, Laboratory for Vascular Translational Science, Nanotechnologies for Vascular Medicine and Imaging Team, 99 Av. Jean-Baptiste Clément 93430 Villetaneuse, France
| | - Anne Meddahi-Pellé
- Université Sorbonne Paris Nord, INSERM U1148, Laboratory for Vascular Translational Science, Nanotechnologies for Vascular Medicine and Imaging Team, 99 Av. Jean-Baptiste Clément 93430 Villetaneuse, France
| | - Fani Anagnostou
- Université Paris Cité, CNRS UMR7052, INSERM U1271, ENVA, B3OA, F-75010 Paris, France
| | - Abolfazl Barzegari
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran; Research Center for Pharmaceutical Nanotechnology (RCPN), Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Graciela Pavon-Djavid
- Université Sorbonne Paris Nord, INSERM U1148, Laboratory for Vascular Translational Science, Nanotechnologies for Vascular Medicine and Imaging Team, 99 Av. Jean-Baptiste Clément 93430 Villetaneuse, France.
| |
Collapse
|
39
|
Chen D, Zhao Z, Zhang S, Chen S, Wu X, Shi J, Liu N, Pan C, Tang Y, Meng C, Zhao X, Tao B, Liu W, Chen D, Ding H, Zhang P, Tang Z. Evolving Therapeutic Landscape of Intracerebral Hemorrhage: Emerging Cutting-Edge Advancements in Surgical Robots, Regenerative Medicine, and Neurorehabilitation Techniques. Transl Stroke Res 2025; 16:975-989. [PMID: 38558011 PMCID: PMC12045821 DOI: 10.1007/s12975-024-01244-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 03/06/2024] [Accepted: 03/19/2024] [Indexed: 04/04/2024]
Abstract
Intracerebral hemorrhage (ICH) is the most serious form of stroke and has limited available therapeutic options. As knowledge on ICH rapidly develops, cutting-edge techniques in the fields of surgical robots, regenerative medicine, and neurorehabilitation may revolutionize ICH treatment. However, these new advances still must be translated into clinical practice. In this review, we examined several emerging therapeutic strategies and their major challenges in managing ICH, with a particular focus on innovative therapies involving robot-assisted minimally invasive surgery, stem cell transplantation, in situ neuronal reprogramming, and brain-computer interfaces. Despite the limited expansion of the drug armamentarium for ICH over the past few decades, the judicious selection of more efficacious therapeutic modalities and the exploration of multimodal combination therapies represent opportunities to improve patient prognoses after ICH.
Collapse
Affiliation(s)
- Danyang Chen
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhixian Zhao
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shenglun Zhang
- School of Mechanical Science and Engineering, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shiling Chen
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xuan Wu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jian Shi
- School of Mechanical Science and Engineering, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Na Liu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Chao Pan
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yingxin Tang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Cai Meng
- School of Astronautics, Beihang University, Beijing, China
| | - Xingwei Zhao
- School of Mechanical Science and Engineering, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Bo Tao
- School of Mechanical Science and Engineering, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Wenjie Liu
- Beijing WanTeFu Medical Instrument Co., Ltd., Beijing, China
| | - Diansheng Chen
- Institute of Robotics, School of Mechanical Engineering and Automation, Beihang University, Beijing, China
| | - Han Ding
- School of Mechanical Science and Engineering, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ping Zhang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| | - Zhouping Tang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
40
|
Izawa H, Xiang C, Ogawa S, Hisanaga I, Yoshimoto T. Amelioration of female menopausal syndrome by intravenous administration of autologous menstrual blood-derived stem cells. Regen Ther 2025; 29:192-201. [PMID: 40225052 PMCID: PMC11992397 DOI: 10.1016/j.reth.2025.03.009] [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: 01/10/2025] [Revised: 01/29/2025] [Accepted: 03/18/2025] [Indexed: 04/15/2025] Open
Abstract
Introduction Menopausal syndrome is characterized by a wide range of physical and psychological symptoms in women aged 40s-50s as a result of hormonal fluctuations and age-related decline. Various treatments have been used to manage the symptoms, including hormone replacement therapy, but no effective causal therapies have yet been identified. Regenerative medicine has gained considerable attention as a promising approach to age-related problems, and mesenchymal stem cell therapies have been extensively studied. Recently, menstrual blood has emerged as a novel cell source of stem cells, called menstrual blood-derived stem cells (MenSCs), due to its non-invasive, regular and consistent collection from women. In this study, we have investigated the therapeutic potential of intravenous administration of autologous MenSCs on female menopausal syndromes. Methods Menstrual blood was collected from 15 patients aged 30s-60s with ovarian dysfunction using a menstrual cup, and MenSCs were isolated, cultured and expanded. Patients received either 3 × 107 cells or 1 × 108 cells intravenously 1 to 5 times at intervals of more than 1 month. Patient-reported symptoms were assessed using the Simplified Menopausal Index at pre-treatment and after 1, 3, 6, and 12 months, and safety assessments were performed. Serum estradiol and follicle-stimulating hormone levels were also measured by immunoassay. Results Almost all patients who received MenSCs experienced a sharp reduction in menopausal symptoms, including vasomotor, neuropsychiatric, and motor symptoms, one month after the first administration, and these symptoms remained low for 6 months. The Simplified Menopausal Index score was significantly reduced after treatment. The reducing potency of 1 × 108 MenSCs was greater than that of 3 × 107 MenSCs. Patients who received a higher number of MenSCs showed an increasing trend in estradiol levels and a decreasing trend in follicle-stimulating hormone levels. When MenSCs were administered to postmenopausal patients, this trend was more pronounced. Overall, no apparent serious adverse events were observed during these treatments. Conclusions The present results suggest that the administration of MenSCs improved menopausal symptoms and regulated hormonal balance without any serious adverse events. This is the first report on the promising therapeutic potential of cell-based therapy using autologous MenSCs for female menopausal syndrome.
Collapse
Affiliation(s)
- Hiromi Izawa
- Jingu-Gaien Woman Life Clinic, Shibuya-ku, Tokyo, Japan
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, Shinjuku-ku, Tokyo, Japan
| | - Charlie Xiang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Research Units of Infectious Disease and Microecology, Chinese Academy of Medical Sciences, Hangzhou, Beijing, China
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, China
| | - Seiji Ogawa
- Jingu-Gaien Woman Life Clinic, Shibuya-ku, Tokyo, Japan
- Fujita Medical Innovation Center Tokyo, Reproduction Center, Ota-ku, Tokyo, Japan
| | - Ichiro Hisanaga
- Jingu-Gaien Woman Life Clinic, Shibuya-ku, Tokyo, Japan
- Dai Nippon Printing Co., Ltd., Human Engineering Laboratory, Shinjuku-ku, Tokyo, Japan
- Ritsumeikan University, Art Research Center, Kyoto-shi, Kyoto, Japan
| | - Takayuki Yoshimoto
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, Shinjuku-ku, Tokyo, Japan
| |
Collapse
|
41
|
Dalal D, Singh L, Singh A. Calycosin and kidney health: a molecular perspective on its protective mechanisms. Pharmacol Rep 2025; 77:658-669. [PMID: 40249500 DOI: 10.1007/s43440-025-00728-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 04/09/2025] [Accepted: 04/11/2025] [Indexed: 04/19/2025]
Abstract
Kidney diseases encompass a diverse group of pathological conditions characterized by the progressive loss of renal function, leading to systemic complications and increased morbidity. Their global prevalence increasing, posing a substantial public health challenge. The underlying pathophysiology involves complex molecular interactions that drive inflammation, fibrosis, and tissue injury. Notably, the AGE/RAGE axis activates NF-κB, a pivotal transcription factor responsible for pro-inflammatory cytokine production. This response is further intensified by NLRP3-inflammasome activation, which detects cellular stress and promotes IL-1β release. Additionally, TGF-β signaling through SMADs and MAPK pathways induces ECM accumulation, contributing to tissue fibrosis. Besides this, oxidative stress-induced ferroptosis and apoptosis also play critical roles in disease progression. Given the multifactorial nature of kidney diseases, agents with multi-targeted actions are promising for effective renoprotection. Significant research interest has emerged in exploring calycosin's protective effects against kidney-related pathologies, owing to its diverse pharmacological properties, including anti-inflammatory, antioxidant, anti-apoptotic, and anti-fibrotic effects. Calycosin is a naturally occurring isoflavone primarily found in Astragalus membranaceus, a well-known medicinal herb in traditional Chinese medicine. Several studies have demonstrated that calycosin exerts its renoprotective effects by modulating key molecular mediators, including RAGE, NF-κB, TGF-β, MAPKs, NLRP3-inflammasome, Nrf-2, PPARγ, and Sirtuin-3, among others, thereby providing a multitargeted defense against kidney diseases. Considering the potential of calycosin in modulating these mediators, the present study was conceptualized to study the mechanistic interplay underlying its renoprotective effects. By investigating these interconnected pathways, this study will provide foundational insights that will enable future researchers to address existing gaps and further elucidate calycosin's potential in renal disorders.
Collapse
Affiliation(s)
- Diksha Dalal
- University Institute of Pharma Sciences, Chandigarh University, Mohali, Punjab, India
| | - Lovedeep Singh
- University Institute of Pharma Sciences, Chandigarh University, Mohali, Punjab, India.
| | - Anish Singh
- University Institute of Pharma Sciences, Chandigarh University, Mohali, Punjab, India
| |
Collapse
|
42
|
Canale MG, Muñoz FL, Muñoz SE, Diaz MDP. Favorable trends in lung cancer incidence with unfavorable survival prognosis: A spatiotemporal analysis by histology in Córdoba, Argentina. Cancer Epidemiol 2025; 96:102796. [PMID: 40081021 DOI: 10.1016/j.canep.2025.102796] [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: 03/03/2025] [Indexed: 03/15/2025]
Abstract
Lung cancer (LC) represents the leading cause of cancer-related death and the third in incidence in Argentina. Survival rates are low. OBJECTIVE To analyze the spatial distribution of LC incidence in Córdoba-Argentina (2004-2014), explore trends in histological types, and estimate the probability of survival. METHODS A longitudinal ecological study was conducted using data from the Provincial Cancer Registry. Age-specific and standardized incidence rates for LC (ICD-10: C33-34) were calculated, truncated (35-84 years), and stratified by sex, year (2004-2014), and histology (small cell carcinoma and non-small cell: adenocarcinoma, squamous cells, large cells, and other carcinomas). Temporal analysis employed Joinpoint regression models, estimating annual percentage changes (APC). Median times estimated survival curves and semiparametric Cox regression models were employed for survival. Statistical significance: log-rank tests and proportional hazards tests. Software: Joinpoint-Regression-Program and Stata17. RESULTS From 2004-2014, 8246 LC cases were diagnosed in individuals aged 35-84. The highest incidence occurred in males aged 75-79 and females aged 80-84. The Age-standardized incidence rates for males and females were 57.9 and 23.6 cases per 100,000 person-years, respectively. In both sexes, the temporal incidence trend was decreasing (APC -3.21 %; p = 0.001), more pronounced in males (APC -3.99 %, p = 0.011), with negative APCs in all histological subtypes. The probability of survival decreased to 32 % (95 %CI: 31 %-34 %) within just 12 months (38 % in females, 30 % in males). The risk of death increased proportionally with age (males HR: 1.007, (95 %CI: 1.004-1.01, p = 0.000); females HR: 1.005, (95 %CI: 1.00-1.01, p = 0.031)) and across all histological types, with lower proportional risks in females and disparities based on histology: in males, the highest risk was in large cells (p = 0.008) and SMCC, while in females, it was SCLC (p = 0.055). CONCLUSIONS Despite estimating a favorable trend in LC incidence since 2004, the survival prognosis remains unfavorable one-year post-diagnosis, dependent on sex, age, and histological type.
Collapse
Affiliation(s)
- Marcela Guadalupe Canale
- Instituto de Investigaciones en Ciencias de la Salud (INICSA) CONICET-UNC, Universidad Nacional de Córdoba, Córdoba, Argentina; Escuela de Nutrición, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Fabian Leonardo Muñoz
- Instituto de Investigaciones en Ciencias de la Salud (INICSA) CONICET-UNC, Universidad Nacional de Córdoba, Córdoba, Argentina; Escuela de Nutrición, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Sonia Edith Muñoz
- Instituto de Investigaciones en Ciencias de la Salud (INICSA) CONICET-UNC, Universidad Nacional de Córdoba, Córdoba, Argentina; Instituto de Biología Celular, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Maria Del Pilar Diaz
- Instituto de Investigaciones en Ciencias de la Salud (INICSA) CONICET-UNC, Universidad Nacional de Córdoba, Córdoba, Argentina.
| |
Collapse
|
43
|
Huang D, Li Z, Li G, Zhou F, Wang G, Ren X, Su J. Biomimetic structural design in 3D-printed scaffolds for bone tissue engineering. Mater Today Bio 2025; 32:101664. [PMID: 40206144 PMCID: PMC11979411 DOI: 10.1016/j.mtbio.2025.101664] [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: 02/04/2025] [Revised: 03/11/2025] [Accepted: 03/13/2025] [Indexed: 04/11/2025] Open
Abstract
The rising prevalence of bone diseases in an aging population underscores the urgent need for innovative and clinically translatable solutions in bone tissue engineering. While significant progress has been made in refining the chemical properties of biomaterials, the structural design of scaffolds-a critical determinant of repair success-remains comparatively underexplored. Structural parameters such as porosity, pore size, and interconnectivity are not only essential for achieving mechanical stability but also pivotal in regulating biological processes, including vascularization, osteogenesis, and immune modulation. This review systematically categorizes scaffold architectures documented in the literature and highlights how these design parameters can be optimized to enhance bone regeneration. Advanced fabrication technologies, particularly 3D printing, are emphasized for their transformative potential in creating precise, biomimetic scaffolds that align with the complex functional demands of native bone. Furthermore, this work synthesizes diverse findings to provide a comprehensive framework for designing next-generation scaffolds. By bridging the gap between structural innovation and clinical application, this review delivers actionable strategies and a strategic roadmap for advancing the field toward improved clinical outcomes and transformative breakthroughs in regenerative medicine.
Collapse
Affiliation(s)
- Dan Huang
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- School of Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Zuhao Li
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
- Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Guangfeng Li
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- School of Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
- Department of Trauma Orthopedics, Zhongye Hospital, Shanghai, 200941, China
| | - Fengjin Zhou
- Honghui Hospital, Xi'an Jiao Tong University, Xi'an, 710000, China
| | - Guangchao Wang
- Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Xiaoxiang Ren
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Jiacan Su
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
- Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| |
Collapse
|
44
|
Nguyen TK, Pham DV, Park PH. Leptin impairs the therapeutic efficacy of adipose-derived mesenchymal stem cells by inducing apoptosis through NLRP3 inflammasomes activation. Biochem Pharmacol 2025; 236:116868. [PMID: 40081766 DOI: 10.1016/j.bcp.2025.116868] [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: 10/24/2024] [Revised: 02/11/2025] [Accepted: 03/10/2025] [Indexed: 03/16/2025]
Abstract
Mesenchymal stem cells (MSC) have been widely applied for regenerative medicine and the treatment of immune-disorders due to their multilineage differentiation and potent immunomodulatory properties. The therapeutic application of MSC post transplantation are influenced by various endogenous modulators. Leptin, a hormone primarily derived from adipose tissue, exerts a variety of physiological functions, in addition to the metabolic effects. In this study, we examined the effects of leptin on the viability of adipose-derived mesenchymal stem cells (ADSC) and its underlying molecular mechanisms with a particular focus on NLRP3 inflammasomes, which serve as signaling platform of the innate immune system. Leptin significantly decreased the viability of ADSC and induced apoptosis. Mechanistically, NLRP3 inflammasomes signaling critically contributes to leptin-induced apoptosis of ADSC by upregulating p53 and Puma. In addition, NLRP3 inflammasomes activation by leptin is mediated via ER stress induction and ROS accumulation. Finally, suppression of ADSC therapeutic efficacy by leptin and the critical role of NLRP3 inflammasomes in this phenomenon were confirmed in DSS-induced colitis model. Pre-conditioning with leptin before transplantation impaired the therapeutic efficacy and immunomodulatory function of ADSC, which were restored by treatment with a pharmacological inhibitor of NLRP3 inflammasomes. Taken together, the results suggest that leptin induces apoptotic cell death in ADSC and impairs the therapeutic effectiveness of ADSC by activating NLRP3 inflammasomes.
Collapse
Affiliation(s)
- Thi-Kem Nguyen
- College of Pharmacy, Yeungnam University, Gyeongsan, Republic of Korea
| | - Duc-Vinh Pham
- College of Pharmacy, Yeungnam University, Gyeongsan, Republic of Korea; Department of Pharmacology, Hanoi University of Pharmacy, Hanoi, Vietnam
| | - Pil-Hoon Park
- College of Pharmacy, Yeungnam University, Gyeongsan, Republic of Korea; Research Institute of Cell Culture, Yeungnam University, South Korea.
| |
Collapse
|
45
|
Bian Z, Zhai Y, Zhang Y, Wang T, Li H, Ouyang J, Liu C, Wang S, Hu Z, Chang X, Zhang C, Liu M, Li C. Senescent cartilage endplate stem cells-derived exosomes induce oxidative stress injury in nucleus pulposus cells and aggravate intervertebral disc degeneration by regulating FOXO3. Free Radic Biol Med 2025; 233:39-54. [PMID: 40118349 DOI: 10.1016/j.freeradbiomed.2025.03.027] [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: 02/16/2025] [Revised: 03/15/2025] [Accepted: 03/18/2025] [Indexed: 03/23/2025]
Abstract
Intervertebral disc degeneration (IVDD) is the leading cause of low back pain and associated disability worldwide. The cartilage endplate (CEP) is a critical structure in maintaining the homeostasis of the intervertebral disc, by exosomes (Exos)-mediated intracellular communication between cartilage endplate stem cells (CESCs) and nucleus pulposus cells (NPCs). However, whether the senescence of CESCs influences the functionality of CESCs-derived Exos (CESCs-Exos) and participates in the progress of IVDD remains unclear. In this study, we explored the role and mechanism of the Exos-based intracellular communication between senescent CESCs and NPCs in IVDD. CESCs isolated from aged individuals (S-CESCs) exhibited high levels of senescence compared with CESCs isolated from young individuals (Y-CESCs). Exos from Y-CESCs (Y-Exos) and from S-CESCs (S-Exos) were extracted and identified. Surprisingly, we found that S-Exos lost the therapeutic effects as the Y-Exos exhibited in mitigating IVDD, and even aggravated IVDD by inducing oxidative stress injury in NPCs. MicroRNA-sequencing revealed significant upregulation of miR-29b-3p expression in S-Exos. Through microRNA target prediction, dual luciferase assays, RNA-sequencing, lentivirus-mediated overexpression and suppression, we demonstrated that miR-29b-3p regulates the expression of FOXO3 and downstream antioxidant enzymes to induce oxidative stress injury in NPCs. In vivo experiments further verified that countering miR-29b-3p by antagomir reversed the detrimental effects of S-Exos in exacerbating IVDD. This work elucidates the role and mechanism of senescent CESCs in disrupting redox homeostasis in the nucleus pulposus and exacerbating IVDD by Exos-mediated intracellular communication and offers an experimental foundation for the selection of proper CESCs-Exos to obtain better therapeutic effects in IVDD.
Collapse
Affiliation(s)
- Zhiqun Bian
- Department of Orthopedics, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China; Chongqing Municipal Health Commission Key Laboratory of Precise Orthopedics, Chongqing, 400038, China
| | - Yu Zhai
- Department of Orthopedics, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China; Chongqing Municipal Health Commission Key Laboratory of Precise Orthopedics, Chongqing, 400038, China.
| | - Yuyao Zhang
- Department of Orthopedics, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China; Chongqing Municipal Health Commission Key Laboratory of Precise Orthopedics, Chongqing, 400038, China
| | - Tianling Wang
- Department of Orthopedics, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China; Chongqing Municipal Health Commission Key Laboratory of Precise Orthopedics, Chongqing, 400038, China
| | - Hao Li
- Department of Orthopedics, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China; Chongqing Municipal Health Commission Key Laboratory of Precise Orthopedics, Chongqing, 400038, China
| | - Jian Ouyang
- Department of Orthopedics, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China; Chongqing Municipal Health Commission Key Laboratory of Precise Orthopedics, Chongqing, 400038, China
| | - Chao Liu
- Department of Orthopedics, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China; Chongqing Municipal Health Commission Key Laboratory of Precise Orthopedics, Chongqing, 400038, China
| | - Siya Wang
- College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Zhilei Hu
- Department of Orthopedics, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China; Chongqing Municipal Health Commission Key Laboratory of Precise Orthopedics, Chongqing, 400038, China
| | - Xian Chang
- Department of Orthopedics, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China; Chongqing Municipal Health Commission Key Laboratory of Precise Orthopedics, Chongqing, 400038, China
| | - Chao Zhang
- Department of Orthopedics, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China; Chongqing Municipal Health Commission Key Laboratory of Precise Orthopedics, Chongqing, 400038, China.
| | - Minghan Liu
- Department of Orthopedics, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China; Chongqing Municipal Health Commission Key Laboratory of Precise Orthopedics, Chongqing, 400038, China; State Key Laboratory of Trauma and Chemical Poisoning, Chongqing, 400038, China.
| | - Changqing Li
- Department of Orthopedics, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China; Chongqing Municipal Health Commission Key Laboratory of Precise Orthopedics, Chongqing, 400038, China; State Key Laboratory of Trauma and Chemical Poisoning, Chongqing, 400038, China.
| |
Collapse
|
46
|
Inafuku N, Sowa Y, Kishida T, Sawai S, Ntege EH, Numajiri T, Yamamoto K, Shimizu Y, Mazda O. Investigation of the stemness and wound-healing potential of long-term cryopreserved stromal vascular fraction cells. Regen Ther 2025; 29:128-139. [PMID: 40162021 PMCID: PMC11952815 DOI: 10.1016/j.reth.2025.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 01/26/2025] [Accepted: 02/12/2025] [Indexed: 04/02/2025] Open
Abstract
Introduction Stromal vascular fraction (SVF), a heterogeneous cell population primarily derived from adipose tissue, is widely utilized in regenerative therapies for its wound-healing properties and accessibility. While its immediate availability is advantageous, repeated harvesting can be burdensome, especially for elderly patients, and the regenerative capacity of SVF declines with donor age. Long-term cryopreservation offers a potential solution by allowing the banking of SVF from younger donors for future use; however, the impact of this process on SVF functionality remains elusive. This study investigates the stemness and wound-healing potential of SVF following prolonged cryopreservation. Methods SVF cells were isolated from adipose tissue harvested from twelve patients and cryopreserved for either two months (short-term cryopreserved SVF, S-SVF) or 12-13 years (long-term cryopreserved SVF, L-SVF), with six patients in each group. In vitro assays assessed cell viability and stemness, while in vivo assays evaluated wound-healing ability by administering thawed SVF cells from each group to dorsal wounds in immunodeficient mice, compared with a control group. Non-parametric statistical tests analyzed the differences between groups. Results L-SVF exhibited significantly lower stemness compared to S-SVF. Importantly, the L-SVF group showed significantly improved wound healing compared with the control group, although the wound-healing effect of L-SVF was inferior to that of the S-SVF. Conclusion This study demonstrated that, despite reduced stemness, L-SVF retains partial wound-healing potential after 12-13 years of cryopreservation. These findings highlight the need for optimized cryopreservation protocols to enhance SVF viability and regenerative capacity for clinical applications.
Collapse
Affiliation(s)
- Naoki Inafuku
- Department of Plastic and Reconstructive Surgery, Kyoto Prefectural University of Medicine, Kamigyo, Kyoto, Japan
| | - Yoshihiro Sowa
- Department of Plastic Surgery, Jichi Medical University, Shimotsuke, Tochigi, Japan
| | - Tsunao Kishida
- Department of Immunology, Kyoto Prefectural University of Medicine, Kamigyo, Kyoto, Japan
| | - Seiji Sawai
- Department of Orthopedics, Jyujyo Takeda Rehabilitation Hospital, Kyoto, Japan
| | - Edward Hosea Ntege
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Toshiaki Numajiri
- Department of Plastic and Reconstructive Surgery, Kyoto Prefectural University of Medicine, Kamigyo, Kyoto, Japan
| | - Kenta Yamamoto
- Department of Immunology, Kyoto Prefectural University of Medicine, Kamigyo, Kyoto, Japan
| | - Yusuke Shimizu
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Osam Mazda
- Department of Immunology, Kyoto Prefectural University of Medicine, Kamigyo, Kyoto, Japan
| |
Collapse
|
47
|
Álvarez-Barrera L, Mateos-Nava RA, Hernández-Córdova KN, Lezama-Sánchez E, Alcántara-Mejía VA, Rodríguez-Mercado JJ. Transplacental and genotoxicity effects of thallium(I) during organogenesis in mice. Toxicol Rep 2025; 14:101896. [PMID: 39897402 PMCID: PMC11783430 DOI: 10.1016/j.toxrep.2025.101896] [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: 11/08/2024] [Revised: 12/20/2024] [Accepted: 01/03/2025] [Indexed: 02/04/2025] Open
Abstract
The increased concentration of thallium (Tl) in the environment is a cause for concern because the entire population, including pregnant women, is exposed, and this metal crosses the placenta and reaches the conceptus during development. In biological models such as mice, some abnormalities and delays in ossification occur in the fetuses of mice administered Tl on day 7 of gestation, but exposure to environmental Tl is constant during fetal development; therefore, in this study, the effects of several administrations of TI during organogenesis on the external morphology, skeletal development and genotoxicity of fetuses were evaluated. Four groups of 10 pregnant mice were administered 5.28, 6.16, 7.4 or 9.25 mg/kg body weight Tl(I) acetate intraperitoneally during fetal organogenesis. Additionally, samples were taken from fetuses from pregnant mice treated with 5.28 and 6.16 mg/kg body weight to evaluate the transplacental genotoxicity. The results revealed that the 9.25 mg/kg body weight dose produced maternal and fetal toxicity, and all of the treatment groups presented relatively high percentages of fetuses with external abnormalities, reduced bone ossification, and an increased percentage of liver cells with structural chromosomal aberrations (SCAs) and micronuclei (MNs) in blood cells. These results show that Tl(I) acetate administered during organogenesis produces abnormalities, including a delay in ossification and transplacental genotoxicity, in mouse fetuses. These findings are important because Tl has negative effects on development and may affect the health of offspring in the future because it can damage genetic material.
Collapse
Affiliation(s)
- Lucila Álvarez-Barrera
- Unidad de Investigación en Genética y Toxicología Ambiental (UNIGEN), Laboratorio 5, primer piso, Unidad Multidisciplinaria de Investigación Experimental (UMIEZ-Z). Facultad de Estudios Superiores-Zaragoza, Campus II, UNAM, Ciudad de México, Mexico
- Carrera Médico Cirujano, Ciencias Biomédicas, BQ. FES-Zaragoza, UNAM, Mexico
| | - Rodrigo Aníbal Mateos-Nava
- Unidad de Investigación en Genética y Toxicología Ambiental (UNIGEN), Laboratorio 5, primer piso, Unidad Multidisciplinaria de Investigación Experimental (UMIEZ-Z). Facultad de Estudios Superiores-Zaragoza, Campus II, UNAM, Ciudad de México, Mexico
| | - Keyla Nahomi Hernández-Córdova
- Unidad de Investigación en Genética y Toxicología Ambiental (UNIGEN), Laboratorio 5, primer piso, Unidad Multidisciplinaria de Investigación Experimental (UMIEZ-Z). Facultad de Estudios Superiores-Zaragoza, Campus II, UNAM, Ciudad de México, Mexico
| | - Eduardo Lezama-Sánchez
- Unidad de Investigación en Genética y Toxicología Ambiental (UNIGEN), Laboratorio 5, primer piso, Unidad Multidisciplinaria de Investigación Experimental (UMIEZ-Z). Facultad de Estudios Superiores-Zaragoza, Campus II, UNAM, Ciudad de México, Mexico
| | - Víctor Alan Alcántara-Mejía
- Unidad de Investigación en Genética y Toxicología Ambiental (UNIGEN), Laboratorio 5, primer piso, Unidad Multidisciplinaria de Investigación Experimental (UMIEZ-Z). Facultad de Estudios Superiores-Zaragoza, Campus II, UNAM, Ciudad de México, Mexico
| | - Juan José Rodríguez-Mercado
- Unidad de Investigación en Genética y Toxicología Ambiental (UNIGEN), Laboratorio 5, primer piso, Unidad Multidisciplinaria de Investigación Experimental (UMIEZ-Z). Facultad de Estudios Superiores-Zaragoza, Campus II, UNAM, Ciudad de México, Mexico
| |
Collapse
|
48
|
Fayazi M, Rostami M, Amiri Moghaddam M, Nasiri K, Tadayonfard A, Roudsari MB, Ahmad HM, Parhizgar Z, Majbouri Yazdi A. A state-of-the-art review of the recent advances in drug delivery systems for different therapeutic agents in periodontitis. J Drug Target 2025; 33:612-647. [PMID: 39698877 DOI: 10.1080/1061186x.2024.2445051] [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: 10/07/2024] [Revised: 12/08/2024] [Accepted: 12/12/2024] [Indexed: 12/20/2024]
Abstract
Periodontitis (PD) is a chronic gum illness that may be hard to cure for a number of reasons, including the fact that no one knows what causes it, the side effects of anti-microbial treatment, and how various kinds of bacteria interact with one another. As a result, novel therapeutic approaches for PD treatment must be developed. Additionally, supplementary antibacterial regimens, including local and systemic medication administration of chemical agents, are necessary for deep pockets to assist with mechanical debridement of tooth surfaces. As our knowledge of periodontal disease and drug delivery systems (DDSs) grows, new targeted delivery systems like extracellular vesicles, lipid-based nanoparticles (NPs), metallic NPs, and polymer NPs have been developed. These systems aim to improve the targeting and precision of PD treatments while reducing the systemic side effects of antibiotics. Nanozymes, photodermal therapy, antibacterial metallic NPs, and traditional PD therapies have all been reviewed in this research. Medicinal herbs, antibiotics, photothermal therapy, nanozymes, antibacterial metallic NPs, and conventional therapies for PD have all been examined in this research. After that, we reviewed the key features of many innovative DDSs and how they worked for PD therapy. Finally, we have discussed the advantages and disadvantages of these DDSs.
Collapse
Affiliation(s)
- Mehrnaz Fayazi
- School of Dentistry, Islamic Azad University of Medical Sciences, Tehran, Iran
| | - Mitra Rostami
- School of Dentistry, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Kamyar Nasiri
- Department of Dentistry, Islamic Azad University of Medical Sciences, Tehran, Iran
| | - Azadeh Tadayonfard
- Department of Prosthodontics, Dental Faculty, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammadreza Behnam Roudsari
- Dental Research Center, Research Institute of Dental Sciences, School of Dentistry, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Zahra Parhizgar
- Resident of Periodontology, Department of Periodontics, Mashhad University of Medical Sciences, Mashhad, Iran
| | | |
Collapse
|
49
|
Awano-Kim S, Hosoya S, Yokomizo R, Kishi H, Okamoto A. Novel therapeutic strategies for Asherman's syndrome: Endometrial regeneration using menstrual blood-derived stem cells. Regen Ther 2025; 29:328-340. [PMID: 40242087 PMCID: PMC12002619 DOI: 10.1016/j.reth.2025.03.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 03/06/2025] [Accepted: 03/23/2025] [Indexed: 04/18/2025] Open
Abstract
Endometrium is vital to the establishment of pregnancy through its cyclical regeneration, which, when disrupted, can lead to endometrial thinning and Asherman's syndrome (AS). AS is characterized by infertility, pelvic pain, menstrual irregularities, and placental complications. Currently, treatments such as hysteroscopic adhesiolysis and hormone replacement therapy have demonstrated variable efficacy with limited clinical evidence. Recent developments in cell therapy have introduced menstrual blood-derived mesenchymal stem cells (MenSCs) as a promising alternative therapeutic strategy. Menstrual blood offers a noninvasive, periodically available source of mesenchymal stem cells, MenSCs for endometrial regeneration. This review comprehensively examines the endometrial regenerative process, pathophysiology of AS, and therapeutic prospects of MenSCs, underscoring the need for continued research to optimize treatment strategies.
Collapse
Affiliation(s)
- Sena Awano-Kim
- Department of Obstetrics and Gynecology, The Jikei University School of Medicine, 3-25-8, Nishi-shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Satoshi Hosoya
- Department of Obstetrics and Gynecology, The Jikei University School of Medicine, 3-25-8, Nishi-shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Ryo Yokomizo
- Department of Obstetrics and Gynecology, The Jikei University School of Medicine, 3-25-8, Nishi-shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Hiroshi Kishi
- Department of Obstetrics and Gynecology, The Jikei University School of Medicine, 3-25-8, Nishi-shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Aikou Okamoto
- Department of Obstetrics and Gynecology, The Jikei University School of Medicine, 3-25-8, Nishi-shimbashi, Minato-ku, Tokyo 105-8461, Japan
| |
Collapse
|
50
|
Zhang Y, Chen L, Yang S, Dai R, Sun H, Zhang L. Identification and Validation of Circadian Rhythm-Related Genes Involved in Intervertebral Disc Degeneration and Analysis of Immune Cell Infiltration via Machine Learning. JOR Spine 2025; 8:e70066. [PMID: 40225045 PMCID: PMC11994230 DOI: 10.1002/jsp2.70066] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 03/23/2025] [Accepted: 03/25/2025] [Indexed: 04/15/2025] Open
Abstract
Background Low back pain is a significant burden worldwide, and intervertebral disc degeneration (IVDD) is identified as the primary cause. Recent research has emphasized the significant role of circadian rhythms (CRs) and immunity in affecting intervertebral discs (IVD). However, the influence of circadian rhythms and immunity on the mechanism of IVDD remains unclear. This study aimed to identify and validate key rhythm-related genes in IVDD and analyze their correlation with immune cell infiltration. Methods Two gene expression profiles related to IVDD and rhythm-related genes were obtained from the Gene Expression Omnibus and GeneCards databases to identify differentially expressed rhythm-related genes (DERGs). Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) and gene set enrichment analysis (GSEA) were conducted to explore the biological functions of these genes. LASSO regression and SVM algorithms were employed to identify hub genes. We subsequently investigated the correlation between hub rhythm-related genes and immune cell infiltration. Finally, nucleus pulposus-derived mesenchymal stem cells (NPMSCs) were isolated from normal and degenerative human IVD tissues. Hub rhythm-related genes expression in NPMSCs was confirmed by real-time quantitative PCR (RT-qPCR). Results Six hub genes related to CRs (CCND1, FOXO1, FRMD8, NTRK2, PRRT1, and TFPI) were screened out. Immune infiltration analysis revealed that the IVDD group had significantly more M0 macrophages and significantly fewer follicular helper T cells than those of the control group. Specifically, M0 macrophages were significantly associated with FRMD8, PRRT1, and TFPI. T follicular helper cells were significantly associated with FRDM8, FOXO1, and CCND1. We further confirmed that CCND1, FRMD8, NTRK2, and TFPI were dysrhythmic within NPMSCs from degenerated IVD in vitro. Conclusion Six genes (CCND1, FOXO1, FRMD8, NTRK2, PRRT1 and TFPI) linked to circadian rhythms associated with IVDD progression, together with immunity. The identification of these DEGs may provide new insights for the diagnosis and treatment of IVDD.
Collapse
Affiliation(s)
- Yongbo Zhang
- Department of OrthopedicsNorthern Jiangsu People's Hospital Affiliated to Yangzhou UniversityYangzhouChina
- Department of OrthopedicsThe Yangzhou School of Clinical Medicine of Dalian Medical UniversityYangzhouChina
| | - Liuyang Chen
- Department of OrthopedicsNorthern Jiangsu People's Hospital Affiliated to Yangzhou UniversityYangzhouChina
- Department of OrthopedicsNorthern Jiangsu People's HospitalYangzhouChina
| | - Sheng Yang
- Department of OrthopedicsNorthern Jiangsu People's Hospital Affiliated to Yangzhou UniversityYangzhouChina
- Department of OrthopedicsThe Yangzhou School of Clinical Medicine of Dalian Medical UniversityYangzhouChina
| | - Rui Dai
- Department of OrthopedicsNorthern Jiangsu People's Hospital Affiliated to Yangzhou UniversityYangzhouChina
- Department of OrthopedicsNorthern Jiangsu People's HospitalYangzhouChina
| | - Hua Sun
- Department of OrthopedicsNorthern Jiangsu People's Hospital Affiliated to Yangzhou UniversityYangzhouChina
- Department of OrthopedicsNorthern Jiangsu People's HospitalYangzhouChina
| | - Liang Zhang
- Department of OrthopedicsNorthern Jiangsu People's Hospital Affiliated to Yangzhou UniversityYangzhouChina
- Department of OrthopedicsNorthern Jiangsu People's HospitalYangzhouChina
| |
Collapse
|