1
|
Jeon HH, Salas MCC, Park K, Fisher L, Ha S, Palmer C, Chan F, Graves DT. Comparison of the bone remodeling in the midpalatal suture during maxillary expansion between young and middle-aged mice. Bone 2025; 197:117512. [PMID: 40324615 DOI: 10.1016/j.bone.2025.117512] [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: 05/18/2024] [Revised: 04/17/2025] [Accepted: 05/02/2025] [Indexed: 05/07/2025]
Abstract
Maxillary expansion is a common orthodontic procedure for treating maxillary transverse deficiency. However, the cell responses to mechanical force may vary across different age groups, suggesting the need for age-specific treatment protocols. To compare the age-related responses to the mechanical force, we examined the 6-week- and 12-month-old mice undergoing maxillary expansion with 0.012-in. stainless steel orthodontic wire bonded to the maxillary first and second molars (25 g force). Mice were euthanized on days 0, 3, 7, and 14 for analysis. MicroCT analysis, tartrate-resistant acid phosphatase (TRAP) stain, and immunofluorescence/immunohistochemistry stain using antibodies to RUNX2, alkaline phosphatase (ALP), Gli1 and Ki67 along with the TUNEL assay, were conducted to evaluate suture width, osteoclast activity, new bone formation and mesenchymal stem cell (MSC) proliferation and apoptosis. Both 6-week- and 12-month-old mice exhibited successful midpalatal suture opening, but young mice demonstrated earlier and more intense osteoclast activity, along with higher expression of RUNX2 and ALP. Young mice also exhibited a higher percentage of Gli1+Ki67+ immunopositive cells, while middle-aged mice showed a higher percentage of Gli1+TUNEL+ positive cells on day 3 after maxillary expansion. Our findings suggest that aging negatively impacts mechanical force-induced bone remodeling by reducing osteoclastogenesis, osteogenesis, and MSC proliferation while increasing MSC apoptosis.
Collapse
Affiliation(s)
- Hyeran Helen Jeon
- Department of Orthodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Mary Cruz Contreras Salas
- Department of Orthodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kyungjoon Park
- School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Lindsay Fisher
- School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sara Ha
- School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Caroline Palmer
- School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Fionna Chan
- School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Dana T Graves
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
2
|
Liu X, Lv M, Feng B, Gong Y, Min Q, Wang Y, Wu Q, Chen J, Zhao D, Li J, Zhang W, Zhan Q. SQLE amplification accelerates esophageal squamous cell carcinoma tumorigenesis and metastasis through oncometabolite 2,3-oxidosqualene repressing Hippo pathway. Cancer Lett 2025; 621:217528. [PMID: 39924077 DOI: 10.1016/j.canlet.2025.217528] [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: 10/18/2024] [Revised: 01/17/2025] [Accepted: 02/02/2025] [Indexed: 02/11/2025]
Abstract
Esophageal squamous cell carcinoma (ESCC) is one of the most prevalent cancers worldwide, characterized by a dismal prognosis and elusive therapeutic targets. Dysregulated cholesterol metabolism is a critical hallmark of cancer cells, facilitating tumor progression. Here, we used whole genome sequencing data from several ESCC cohorts to identify the important role of squalene epoxidase (SQLE) in promoting ESCC tumorigenesis and metastasis. Specifically, our findings highlight the significance of 2,3-oxidosqualene, an intermediate metabolite of cholesterol biosynthesis, synthesized by SQLE and metabolized by lanosterol synthase (LSS), as a key regulator of ESCC progression. Mechanistically, the interaction between 2,3-oxidosqualene and vinculin enhances the nuclear accumulation of Yes-associated protein 1 (YAP), thereby increasing YAP/TEAD-dependent gene expression and accelerating both tumor growth and metastasis. In a 4-nitroquinoline 1-oxide (4-NQO)-induced ESCC mouse model, overexpression of Sqle resulted in accelerated tumorigenesis compared to wild-type controls, highlighting the pivotal role of SQLE in vivo. Furthermore, elevated SQLE expression in ESCC patients correlates with a poorer prognoses, suggesting potential therapeutic avenues for treatment. In conclusion, our study elucidates the oncogenic function of 2,3-oxidosqualene as a naturally occurring metabolite and proposes modulation of its levels as a promising therapeutic strategy for ESCC.
Collapse
Affiliation(s)
- Xuesong Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China; Peking University International Cancer Institute, Beijing, 100191, China; Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, 100021, China
| | - Mengzhu Lv
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China; Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, 100021, China
| | - Bicong Feng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China; Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, 100021, China
| | - Ying Gong
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China; Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Breast Oncology, Peking University Cancer Hospital and Institute, Beijing, 100142, China; Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, 100021, China
| | - Qingjie Min
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China; Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, 100021, China
| | - Yan Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China; Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, 100021, China
| | - Qingnan Wu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China; Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, 100021, China
| | - Jie Chen
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China; Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, 100021, China
| | - Dongyu Zhao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China; Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, 100021, China
| | - Jinting Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China; Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, 100021, China
| | - Weimin Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China; Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, 518107, China; Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, 100021, China.
| | - Qimin Zhan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China; Peking University International Cancer Institute, Beijing, 100191, China; Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, 518107, China; Soochow University Cancer Institute, Suzhou, 215127, China; Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, 100021, China.
| |
Collapse
|
3
|
Dai W, Xu Q, Li Q, Wang X, Zhang W, Zhou G, Chen X, Liu W, Wang W. Piezoelectric nanofilms fabricated by coaxial electrospun polycaprolactone/Barium titanate promote Achilles tendon regeneration by reducing IL-17A/NF-κB-mediated inflammation. Bioact Mater 2025; 49:1-22. [PMID: 40110584 PMCID: PMC11914770 DOI: 10.1016/j.bioactmat.2025.02.038] [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: 12/01/2024] [Revised: 02/22/2025] [Accepted: 02/25/2025] [Indexed: 03/22/2025] Open
Abstract
Tendon injuries are often exacerbated by persistent inflammation, which hampers tissue regeneration. In this study, we developed a noninvasive, wirelessly controlled, and self-powered piezoelectric nanofilm fabricated by coaxial electrospinning of polycaprolactone (PCL) and tetragonal barium titanate nanoparticles (BTO), and investigated its roles in modulating inflammation and repairing Achilles tendon defects as well as the mechanism in a rat model. In vitro study and in vivo study upon subcutaneous implantation showed that the piezoelectric PCL/BTO nanofilms could inhibit M1 macrophage polarization and reduce the secretion of inflammatory factors. Moreover, when bridging an Achilles tendon defect, the nanofilms could promote tenogenic gene expression including collagen deposition, and collagen remodeling, facilitate functional tendon recovery and significantly reduce tissue inflammation by suppressing M1 macrophage polarization and promoting M2 polarization. Moreover, the piezoelectric stimulation could also enhance tendon regeneration by inhibiting angiogenesis, reducing lipid deposition, and decreasing ectopic ossification. Mechanistically, the piezoelectric nanofilms reduced tissue inflammation mainly via inhibiting the nuclear factor (NF)-κB signaling pathway that is mediated by interleukin (IL)-17A secreted from CD3+ T cells, and thus to reduce proinflammatory factors, such as IL-1β and IL-6, inducible nitric oxide synthase, monocyte chemoattractant protein-1, and tumor necrosis factor-α. These findings indicate the potential of piezoelectric stimulation in immunomodulation, and in promoting tendon regeneration via IL-17A/NF-κB-mediated pathway.
Collapse
Affiliation(s)
- Wufei Dai
- Department of Plastic and Reconstructive Surgery, Shanghai Key Laboratory of Tissue Engineering, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Department of Medicine 1, University Hospital Erlangen, Friedrich-Alexander University Erlangen Nuremberg, Erlangen, Germany
| | - Qi Xu
- Department of Plastic and Reconstructive Surgery, Shanghai Key Laboratory of Tissue Engineering, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Department of Surgery, Air Force Medical Center, PLA, Beijing, 100142, China
| | - Qinglin Li
- Department of Aesthetic Surgery, Zhumadian Central Hospital, Zhumadian, 463000, China
| | - Xiansong Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Key Laboratory of Tissue Engineering, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Wenjie Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai Key Laboratory of Tissue Engineering, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Guangdong Zhou
- Department of Plastic and Reconstructive Surgery, Shanghai Key Laboratory of Tissue Engineering, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Xu Chen
- Department of Laboratory Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Wei Liu
- Department of Plastic and Reconstructive Surgery, Shanghai Key Laboratory of Tissue Engineering, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Wenbo Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Key Laboratory of Tissue Engineering, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| |
Collapse
|
4
|
Mao J, Xu Y, Wang W, Deng X, Hui Y, Rui M, Tang J, Wang W, Huang Y, Wu L, Xi K, Zhu Y, Gu Y, Chen L. Topological cues of microparticles train stem cells for tissue repair via mechanotransduction. Bioact Mater 2025; 48:531-549. [PMID: 40114729 PMCID: PMC11923629 DOI: 10.1016/j.bioactmat.2025.02.032] [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: 10/22/2024] [Revised: 02/15/2025] [Accepted: 02/19/2025] [Indexed: 03/22/2025] Open
Abstract
Microspheres (MPs) and porous microspheres (PMPs) are the two most widely used microparticles in tissue engineering and stem cell therapy. However, how stem cells perceive the topological differences between them to regulate cell function remains to be unclear. Here, we systematically studied the changes in stem cell function under the action of MPs and PMPs and elucidated the related mechanisms. Our findings show that the porous structure of PMPs can be sensed by focal adhesions (FAs), which triggers the synthesis of F-actin to inhibit the phosphorylation and degradation of Yes-associated protein (YAP), while also transmitting stress to the nucleus through the contraction of F-actin, thereby enhancing the nuclear translocation of YAP protein. The activation of YAP significantly enhances the proliferation, osteogenesis, paracrine and glucose metabolism of BMSCs, making them exhibit stronger bone repair ability in both in vivo and in vitro experiments. In summary, this study provides a comprehensive and reliable understanding of the behavior of BMSCs in response to MPs and PMPs. It also deepens our understanding of the association between microparticles' topological cues and biological functions, which will provide valuable guidance for the construction of bone tissue engineering (BTE) scaffolds.
Collapse
Affiliation(s)
- Jiannan Mao
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, PR China
- Department of Orthopaedics, Wuxi Key Laboratory of Biomaterials for Clinical Application, Department of Central Laboratory, Jiangyin Clinical College of Xuzhou Medical University, No.163 Shoushan Road, Jiang Yin, 214400, PR China
| | - Yichang Xu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, PR China
| | - Wenbo Wang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, PR China
| | - Xiongwei Deng
- Department of Orthopaedics, Wuxi Key Laboratory of Biomaterials for Clinical Application, Department of Central Laboratory, Jiangyin Clinical College of Xuzhou Medical University, No.163 Shoushan Road, Jiang Yin, 214400, PR China
| | - Yujian Hui
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, PR China
- Department of Orthopaedics, Wuxi Key Laboratory of Biomaterials for Clinical Application, Department of Central Laboratory, Jiangyin Clinical College of Xuzhou Medical University, No.163 Shoushan Road, Jiang Yin, 214400, PR China
| | - Min Rui
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, PR China
- Department of Orthopaedics, Wuxi Key Laboratory of Biomaterials for Clinical Application, Department of Central Laboratory, Jiangyin Clinical College of Xuzhou Medical University, No.163 Shoushan Road, Jiang Yin, 214400, PR China
| | - Jincheng Tang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, PR China
| | - Wei Wang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, PR China
| | - Yiyang Huang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, PR China
| | - Liang Wu
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, PR China
| | - Kun Xi
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, PR China
| | - Yunrong Zhu
- Department of Orthopaedics, Wuxi Key Laboratory of Biomaterials for Clinical Application, Department of Central Laboratory, Jiangyin Clinical College of Xuzhou Medical University, No.163 Shoushan Road, Jiang Yin, 214400, PR China
| | - Yong Gu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, PR China
| | - Liang Chen
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, PR China
| |
Collapse
|
5
|
Zhao C, Wang H, Xu C, Fang F, Gao L, Zhai N, Zhong Y, Wang X. The critical role of the Hippo signaling pathway in renal fibrosis. Cell Signal 2025; 130:111661. [PMID: 39988289 DOI: 10.1016/j.cellsig.2025.111661] [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: 11/21/2024] [Revised: 02/12/2025] [Accepted: 02/14/2025] [Indexed: 02/25/2025]
Abstract
Renal fibrosis is a fundamental pathological change in the progression of various chronic kidney diseases to the end stage of renal disease. The Hippo signaling pathway is an evolutionary highly conserved signaling pathway that is involved in the regulation of organ size, tissue regeneration, and human reproduction and development. Currently, many studies have shown that it is closely associated with renal diseases, such as, renal fibrosis, diabetic nephropathy, and renal cancer. Here, we review the current researches on the effect of Hippo signaling pathway on renal fibrosis, which provides new ideas and theoretical basis for clinical therapeutics of renal fibrosis.
Collapse
Affiliation(s)
- Chenchen Zhao
- Hebei Key Laboratory of Liver and Kidney Diseases of Integrated Traditional Chinese and Western Medicine 7th Floor, Scientific Research Building, Hebei University of Traditional Chinese Medicine, Shijiazhuang City, China
| | - Hongshuang Wang
- Hebei Key Laboratory of Liver and Kidney Diseases of Integrated Traditional Chinese and Western Medicine 7th Floor, Scientific Research Building, Hebei University of Traditional Chinese Medicine, Shijiazhuang City, China
| | - Chang Xu
- Hebei Key Laboratory of Liver and Kidney Diseases of Integrated Traditional Chinese and Western Medicine 7th Floor, Scientific Research Building, Hebei University of Traditional Chinese Medicine, Shijiazhuang City, China
| | - Fang Fang
- Hebei Key Laboratory of Liver and Kidney Diseases of Integrated Traditional Chinese and Western Medicine 7th Floor, Scientific Research Building, Hebei University of Traditional Chinese Medicine, Shijiazhuang City, China
| | - Lanjun Gao
- Hebei Key Laboratory of Liver and Kidney Diseases of Integrated Traditional Chinese and Western Medicine 7th Floor, Scientific Research Building, Hebei University of Traditional Chinese Medicine, Shijiazhuang City, China
| | - Nan Zhai
- Hebei Key Laboratory of Liver and Kidney Diseases of Integrated Traditional Chinese and Western Medicine 7th Floor, Scientific Research Building, Hebei University of Traditional Chinese Medicine, Shijiazhuang City, China
| | - Yan Zhong
- Hebei Key Laboratory of Liver and Kidney Diseases of Integrated Traditional Chinese and Western Medicine 7th Floor, Scientific Research Building, Hebei University of Traditional Chinese Medicine, Shijiazhuang City, China.
| | - Xiangting Wang
- Hebei Key Laboratory of Liver and Kidney Diseases of Integrated Traditional Chinese and Western Medicine 7th Floor, Scientific Research Building, Hebei University of Traditional Chinese Medicine, Shijiazhuang City, China.
| |
Collapse
|
6
|
Baban NS, Stubbs CJ, Song YA. MechanoBioCAD: a generalized semi-automated computational tool for mechanobiological studies. LAB ON A CHIP 2025; 25:2632-2643. [PMID: 40070318 DOI: 10.1039/d4lc00843j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2025]
Abstract
Soft micropillar arrays enable detailed studies of cellular mechanotransduction and biomechanics using traditional beam-bending models. However, they often rely on simplified assumptions, leading to significant errors in force estimation. We present MechanoBioCAD (MBC), a finite element method (FEM)-based tool designed specifically for micropillar research and error estimation. Unlike traditional methods, MBC leverages the principle of minimizing total potential energy, avoiding errors associated with beam bending assumptions. MBC automates FEM model generation, analysis, and post-processing, providing accurate force quantification based on deflection input. The tool addresses critical issues such as substrate deformation, interpillar interactions, improper load application heights, and nonlinear effects. Applied to fibroblast cell traction and Caenorhabditis elegans (C. elegans) thrashing cases, MBC recorded 23% and 34% errors in the estimated forces, respectively, compared to traditional methods. As an open-access tool with the Abaqus Student Edition, MBC democratizes rational design, analysis, and error estimation for researchers who are not subject matter experts in FEM.
Collapse
Affiliation(s)
- Navajit S Baban
- Division of Engineering, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates.
| | - Christopher J Stubbs
- School of Computer Sciences and Engineering, Fairleigh Dickinson University, NJ, USA
| | - Yong-Ak Song
- Division of Engineering, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates.
- Department of Chemical and Biomolecular Engineering, New York University, Brooklyn, NY, USA
- Department of Biomedical Engineering, New York University, Brooklyn, NY, USA
| |
Collapse
|
7
|
Bai J, Yang Y. Activation of Hippo/YAP signaling pathway exacerbates vascular remodeling and aggravates hypertension by upregulating Foxm1. J Mol Histol 2025; 56:158. [PMID: 40388085 DOI: 10.1007/s10735-025-10443-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Accepted: 04/29/2025] [Indexed: 05/20/2025]
Abstract
The Hippo/YAP signaling pathway is closely related to the occurrence and development of cardiovascular diseases. However, it's still unclear whether this pathway plays a certain role in hypertension. In this study, aortic morphology and function in spontaneously hypertensive rats (SHR) were comprehensively evaluated using Wistar-Kyoto rats (WKY) as controls. Results indicated that the aorta of SHRs have distinct changes in pathological structure. Furthermore, the proliferative activity of vascular smooth muscle cells (VSMCs) was enhanced, with vascular fibrosis being aggravated. Immunohistochemical analysis revealed that SHRs exhibited high expression of Yes-Associated Protein (YAP). Western Blot analysis showed that cytoplasmic YAP and TAZ expression decreased in hypertensive rats, indicating that YAP/TAZ nuclear transfer increased and Hippo/YAP signaling pathway had been activated. The cell function experiments of VSMCs extracted from rat aorta showed that the cell viability and proliferation ability of VSMCs in SHRs were enhanced, the expression of Fibronectin and collagen I was increased, and vascular fibrosis was aggravated. siRNA-YAP (si-YAP) can reverse the above phenomenon in VSMCs. Knockdown of YAP can inhibit Foxm1 expression. As an inhibitor of large tumor suppressor kinases LAST1/2, GA-107 can inhibit the phosphorylation level of YAP, increase blood pressure, aggravate aortic pathomorphological changes, promote VSMCs proliferation and vascular fibrosis, and thus aggravate hypertension symptoms in SHRs. However, these effects of GA-107 can be antagonized by inhibiting Foxm1 with thiosulfathiazole (Thio). Conclusively, Hippo/YAP signaling pathway promotes vascular remodeling through the regulation of Foxm1 and causes hypertension.
Collapse
Affiliation(s)
- Jing Bai
- Department of Geriatric Cardiovascular Medicine, Shaanxi Provincial People's Hospital, No. 256, Youyi West Road, Xi'an, 710068, China
| | - Yujuan Yang
- Department of Geriatric Cardiovascular Medicine, Shaanxi Provincial People's Hospital, No. 256, Youyi West Road, Xi'an, 710068, China.
| |
Collapse
|
8
|
Sampietro M, Cellani M, Scielzo C. B cell mechanobiology in health and disease: emerging techniques and insights into therapeutic responses. FEBS Lett 2025. [PMID: 40387441 DOI: 10.1002/1873-3468.70071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 04/15/2025] [Accepted: 04/23/2025] [Indexed: 05/20/2025]
Abstract
Cells sense physical cues from their environment and convert them into biochemical responses through mechanotransduction. Unlike solid tumours, the role of such forces in haematological cancers is underexplored. In this context, immune cells experience dynamic mechanical stimuli as they migrate, extravasate and home to specific tissues. Understanding how these forces shape B-cell function and malignancy represents a groundbreaking area of research. This review examines the key mechanosensory pathways and molecules involved in lymphocyte mechanotransduction, beginning with mechanosensory proteins at the plasma membrane, followed by intracellular signal propagation through the cytoskeleton, eventually highlighting the nucleus as a 'signal actuator'. Subsequently, we cover some measurement approaches and advanced systems to investigate tumour biomechanics, highlighting their application in the context of B cells. Finally, we focus on the implications of mechanobiology in leukaemia, identifying molecules involved in B-cell malignancies that could serve as potential 'mechano-targets' for personalised therapies. This review emphasises the need to understand how lymphocytes generate, sense and respond to mechanical stimuli, which could open avenues for future biomedical innovations. Impact statement Our review is particularly valuable in highlighting the underexplored role of mechanobiology in B cell function and malignancies, while also discussing emerging techniques that can advance this research area. It bridges mechanotransduction, immunology, and cancer biology in a way that will be of interest to researchers across these three main fields.
Collapse
Affiliation(s)
- Marta Sampietro
- Unit of Malignant B Cells biology and 3D Modelling, Division of Experimental Oncology, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Marco Cellani
- Unit of Malignant B Cells biology and 3D Modelling, Division of Experimental Oncology, IRCCS Ospedale San Raffaele, Milan, Italy
- Università Vita-Salute San Raffaele, Milan, Italy
| | - Cristina Scielzo
- Unit of Malignant B Cells biology and 3D Modelling, Division of Experimental Oncology, IRCCS Ospedale San Raffaele, Milan, Italy
| |
Collapse
|
9
|
Kim SG, Li J, Hwang JS, Hassan MAU, Sim YE, Lee JY, Mo JS, Kim MO, Lee G, Park S. Synphilin-1 regulates mechanotransduction in rigidity sensing through interaction with zyxin. J Nanobiotechnology 2025; 23:345. [PMID: 40369541 PMCID: PMC12076907 DOI: 10.1186/s12951-025-03429-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Accepted: 05/01/2025] [Indexed: 05/16/2025] Open
Abstract
BACKGROUND Synphilin-1 has been studied extensively in the context of Parkinson's disease pathology. However, the biophysical functions of synphilin-1 remain unexplored. To investigate its novel functionalities herein, cellular traction force and rigidity sensing ability are analyzed based on synphilin-1 overexpression using elastomeric pillar arrays and substrates of varying stiffness. Molecular changes are analyzed using RNA sequencing-based transcriptomic and liquid chromatography-tandem mass spectrometry-based proteomic analyses. RESULTS Synphilin-1 overexpression reduces cell area, with a decline of local contraction on elastomeric pillar arrays. Cells overexpressing synphilin-1 exhibit an impaired ability to respond to substrate rigidity; however, synphilin-1 knockdown restores rigidity sensing abilities. Integrated omics analysis and in silico prediction corroborate the phenotypic alterations induced by synphilin-1 overexpression at a biophysical level. Zyxin emerges as a novel synphilin-1 binding protein, and synphilin-1 overexpression reduces the nuclear translocation of yes-associated protein. CONCLUSION These findings provide novel insights into the biophysical functions of synphilin-1, suggesting a potential protective role to the altered extracellular matrix, which may be relevant to neurodegenerative conditions such as Parkinson's disease.
Collapse
Affiliation(s)
- Seok Gi Kim
- Department of Molecular Science and Technology, Ajou University, 206 World cup‑ro, Suwon, 16499, Republic of Korea
| | - Jinyan Li
- Department of Mechanical Engineering, Sungkyunkwan University, 2066 Seobu-ro, Suwon, 16419, Republic of Korea
| | - Ji Su Hwang
- Department of Molecular Science and Technology, Ajou University, 206 World cup‑ro, Suwon, 16499, Republic of Korea
| | - Muhammad Anwar Ul Hassan
- Department of Mechanical Engineering, Sungkyunkwan University, 2066 Seobu-ro, Suwon, 16419, Republic of Korea
| | - Ye Eun Sim
- Department of Biomedical Sciences, Graduate School, Ajou University School of Medicine, 164 World cup‑ro, Suwon, 16499, Republic of Korea
| | - Ju Yeon Lee
- Digital Omics Research Center, Korea Basic Science Institute, 162 Yeongudanji-ro, Cheongju, 28119, Republic of Korea
| | - Jung-Soon Mo
- Department of Biomedical Sciences, Graduate School, Ajou University School of Medicine, 164 World cup‑ro, Suwon, 16499, Republic of Korea
- Institute of Medical Science, Ajou University School of Medicine, 164 World cup‑ro, Suwon, 16499, Republic of Korea
| | - Myeong Ok Kim
- Division of Life Science and Applied Life Science (BK21 FOUR), College of Natural Sciences, Gyeongsang National University, Jinju, 52828, Republic of Korea
| | - Gwang Lee
- Department of Molecular Science and Technology, Ajou University, 206 World cup‑ro, Suwon, 16499, Republic of Korea.
- Department of Physiology, Ajou University School of Medicine, 164 World Cup‑ro, Suwon, 16499, Republic of Korea.
| | - Sungsu Park
- Department of Mechanical Engineering, Sungkyunkwan University, 2066 Seobu-ro, Suwon, 16419, Republic of Korea.
- Institute of Quantum Biophysics (IQB), Sungkyunkwan University, 2066 Seobu-ro, Suwon, 16419, Republic of Korea.
- Department of MetaBioHealth, Sungkyunkwan University, 2066 Seobu-ro, Suwon, 16419, Republic of Korea.
| |
Collapse
|
10
|
Verbroekken RM, Savchak OK, Alofs TF, Schenning AP, Gumuscu B. Light-Responsive Liquid Crystal Surface Topographies for Dynamic Stimulation of Cells. ACS APPLIED MATERIALS & INTERFACES 2025; 17:27871-27881. [PMID: 40318038 PMCID: PMC12086769 DOI: 10.1021/acsami.5c02526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 04/22/2025] [Accepted: 04/23/2025] [Indexed: 05/07/2025]
Abstract
All biological surfaces possess distinct dynamic surface topographies. Due to their versatility, these topographies play a crucial role in modulating cell behavior and, when intentionally designed, can precisely guide cellular responses. So far, biomechanical responses have predominantly been studied on static surfaces, overlooking the dynamic environment in the body, where cells constantly interact with shifting biomechanical cues. In this work, we designed and fabricated a light-responsive liquid crystal polymer film to study the effect of micrometer-scale, dynamic surface topographies on cells under physiologically relevant conditions. The light-responsive liquid crystal polymers enable on-demand surface topographical changes, reaching pillar heights of 800 nm and grooved topographies with 700 nm height differences at 37 °C in water. The light-induced surface topographies increased mechanosensitive cell signaling by up to 2-fold higher yes-associated protein (YAP) translocation to the nucleus, as well as up to 3-fold more heterogeneity in distribution of focal adhesions, in a topography-related manner. The pillared topography was seen to cause a lower cellular response, while the grooved topography caused an increased mechanical activation, as well as cell alignment due to a more continuous and aligned physical cue that enhances cell organization. Excitingly, we observed that subsequent surface topography changes induced a 3-fold higher YAP nuclear translocation in fibroblast cells, as well as a 5-fold higher vinculin heterogeneity distribution, indicating that multiple cycles of topography exposure ampliated the cell response. Our work emphasizes the potential of light-responsive liquid crystal polymer films generating dynamic biomechanical cues that allow us to modulate and steer cells in vitro.
Collapse
Affiliation(s)
- Ruth M.C. Verbroekken
- Stimuli-Responsive
Functional Materials and Devices, Department of Chemical Engineering
and Chemistry, Eindhoven University of Technology, P.O. Box 513, Eindhoven 5600 MB, The Netherlands
- Institute
for Complex Molecular Systems, Eindhoven
University of Technology, P.O. Box 513, Eindhoven 5600 MB, The Netherlands
| | - Oksana K. Savchak
- Institute
for Complex Molecular Systems, Eindhoven
University of Technology, P.O. Box 513, Eindhoven 5600 MB, The Netherlands
- Biosensors
and Devices Laboratory, Department of Biomedical Engineering, Eindhoven University of Technology, P.O. Box 513, Eindhoven 5600 MB, The Netherlands
| | - Thom F.J. Alofs
- Stimuli-Responsive
Functional Materials and Devices, Department of Chemical Engineering
and Chemistry, Eindhoven University of Technology, P.O. Box 513, Eindhoven 5600 MB, The Netherlands
| | - Albert P.H.J. Schenning
- Stimuli-Responsive
Functional Materials and Devices, Department of Chemical Engineering
and Chemistry, Eindhoven University of Technology, P.O. Box 513, Eindhoven 5600 MB, The Netherlands
- Institute
for Complex Molecular Systems, Eindhoven
University of Technology, P.O. Box 513, Eindhoven 5600 MB, The Netherlands
| | - Burcu Gumuscu
- Institute
for Complex Molecular Systems, Eindhoven
University of Technology, P.O. Box 513, Eindhoven 5600 MB, The Netherlands
- Biosensors
and Devices Laboratory, Department of Biomedical Engineering, Eindhoven University of Technology, P.O. Box 513, Eindhoven 5600 MB, The Netherlands
| |
Collapse
|
11
|
Houtekamer RM, van der Net MC, Vliem MJ, Noordzij TEJC, van Uden L, van Es RM, Sim JY, Deguchi E, Terai K, Hopcroft MA, Vos HR, Pruitt BL, Matsuda M, Pannekoek WJ, Gloerich M. E-cadherin mechanotransduction activates EGFR-ERK signaling in epithelial monolayers by inducing ADAM-mediated ligand shedding. Sci Signal 2025; 18:eadr7926. [PMID: 40359261 DOI: 10.1126/scisignal.adr7926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 04/15/2025] [Indexed: 05/15/2025]
Abstract
The behavior of cells is governed by signals originating from their local environment, including mechanical forces exerted on the cells. Forces are transduced by mechanosensitive proteins, which can impinge on signaling cascades that are also activated by growth factors. We investigated the cross-talk between mechanical and biochemical signals in the regulation of intracellular signaling networks in epithelial monolayers. Phosphoproteomic and transcriptomic analyses on epithelial monolayers subjected to mechanical strain revealed the activation of extracellular signal-regulated kinase (ERK) downstream of the epidermal growth factor receptor (EGFR) as a predominant strain-induced signaling event. Strain-induced EGFR-ERK signaling depended on mechanosensitive E-cadherin adhesions. Proximity labeling showed that the metalloproteinase ADAM17, an enzyme that mediates shedding of soluble EGFR ligands, was closely associated with E-cadherin. A probe that we developed to monitor ADAM-mediated shedding demonstrated that mechanical strain induced ADAM activation. Mechanically induced ADAM activation was essential for mechanosensitive, E-cadherin-dependent EGFR-ERK signaling. Together, our data demonstrate that mechanical strain transduced by E-cadherin adhesion triggers the shedding of EGFR ligands that stimulate downstream ERK activity. Our findings illustrate how mechanical signals and biochemical ligands can operate within a linear signaling cascade.
Collapse
Affiliation(s)
- Ronja M Houtekamer
- Center for Molecular Medicine, University Medical Center Utrecht and Utrecht University, Utrecht, Netherlands
| | - Mirjam C van der Net
- Center for Molecular Medicine, University Medical Center Utrecht and Utrecht University, Utrecht, Netherlands
| | - Marjolein J Vliem
- Center for Molecular Medicine, University Medical Center Utrecht and Utrecht University, Utrecht, Netherlands
| | - Tomas E J C Noordzij
- Center for Molecular Medicine, University Medical Center Utrecht and Utrecht University, Utrecht, Netherlands
| | - Lisa van Uden
- Center for Molecular Medicine, University Medical Center Utrecht and Utrecht University, Utrecht, Netherlands
| | - Robert M van Es
- Center for Molecular Medicine, University Medical Center Utrecht and Utrecht University, Utrecht, Netherlands
- Oncode Institute, Utrecht, Netherlands
| | - Joo Yong Sim
- Department of Bioengineering, University of California Santa Barbara, Santa Barbara, CA, USA
- Department of Mechanical Engineering, University of California Santa Barbara, Santa Barbara, CA, USA
| | - Eriko Deguchi
- Department of Pathology and Biology of Diseases, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kenta Terai
- Department of Pathology and Biology of Diseases, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Matthew A Hopcroft
- Department of Bioengineering, University of California Santa Barbara, Santa Barbara, CA, USA
- Department of Mechanical Engineering, University of California Santa Barbara, Santa Barbara, CA, USA
| | - Harmjan R Vos
- Center for Molecular Medicine, University Medical Center Utrecht and Utrecht University, Utrecht, Netherlands
- Oncode Institute, Utrecht, Netherlands
| | - Beth L Pruitt
- Department of Bioengineering, University of California Santa Barbara, Santa Barbara, CA, USA
- Department of Mechanical Engineering, University of California Santa Barbara, Santa Barbara, CA, USA
| | - Michiyuki Matsuda
- Department of Pathology and Biology of Diseases, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Willem-Jan Pannekoek
- Center for Molecular Medicine, University Medical Center Utrecht and Utrecht University, Utrecht, Netherlands
| | - Martijn Gloerich
- Center for Molecular Medicine, University Medical Center Utrecht and Utrecht University, Utrecht, Netherlands
| |
Collapse
|
12
|
Li H, Li S, Kanamori Y, Liu S, Moroishi T. Auranofin resensitizes ferroptosis-resistant lung cancer cells to ferroptosis inducers. Biochem Biophys Res Commun 2025; 770:151992. [PMID: 40373379 DOI: 10.1016/j.bbrc.2025.151992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2025] [Revised: 03/23/2025] [Accepted: 05/09/2025] [Indexed: 05/17/2025]
Abstract
Lung cancer, a major cause of cancer-related mortality, has limited therapeutic options, especially for advanced cases. Ferroptosis, an iron-dependent form of cell death, is a potential therapeutic strategy for this disease; however, resistance mechanisms in the tumor microenvironment impede its effectiveness. Therefore, in this study, we aimed to investigate the efficacy of sulfasalazine (SAS), a ferroptosis inducer, and auranofin (AUR), a Food and Drug Administration-approved anti-inflammatory agent, combination to counteract ferroptosis resistance in lung cancer. SAS induced ferroptosis in vitro; however, its efficacy in vivo was limited, possibly because of factors, such as nutrient deprivation and high cell density, in the microenvironment that suppressed the activities of Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ), key regulators of ferroptosis resistance. Screening of 2483 drugs revealed AUR as a compound resensitizing the YAP/TAZ-deficient lung cancer cells to ferroptosis. Moreover, SAS and AUR combination significantly enhanced lipid peroxidation and reactive oxygen species accumulation, further driving ferroptosis in cells. This combination effectively inhibited tumor growth and enhanced survival in a murine lung cancer model. Overall, our findings suggest that AUR potentiates ferroptosis-based therapies, serving as an effective candidate to overcome ferroptosis resistance in lung cancer.
Collapse
Affiliation(s)
- Hao Li
- Department of Molecular and Medical Pharmacology, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto, 860-8556, Japan
| | - Shuran Li
- Department of Molecular and Medical Pharmacology, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto, 860-8556, Japan
| | - Yohei Kanamori
- Department of Molecular and Medical Pharmacology, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto, 860-8556, Japan
| | - Saisai Liu
- Department of Molecular and Medical Pharmacology, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto, 860-8556, Japan
| | - Toshiro Moroishi
- Department of Molecular and Medical Pharmacology, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto, 860-8556, Japan; Center for Metabolic Regulation of Healthy Aging, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto, 860-8556, Japan; Division of Cellular Dynamics, Medical Research Laboratory, Institute of Integrated Research, Institute of Science Tokyo, 1-5-45 Yushima, Tokyo, 113-8510, Japan.
| |
Collapse
|
13
|
Chalifoux M, Avdeeva M, Posfai E. Geometric, cell cycle and maternal-to-zygotic transition-associated YAP dynamics during preimplantation embryo development. Dev Biol 2025; 524:105-115. [PMID: 40349907 DOI: 10.1016/j.ydbio.2025.05.006] [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: 02/26/2025] [Revised: 05/07/2025] [Accepted: 05/08/2025] [Indexed: 05/14/2025]
Abstract
During the first cell fate decision in mammalian embryos, the inner cell mass cells, which will give rise to the embryo proper and other extraembryonic tissues, segregate from the trophectoderm cells, the precursors of the placenta. Cell fate segregation proceeds in a gradual manner encompassing two rounds of cell division, as well as cell positional and morphological changes. While it is known that the activity of the Hippo signaling pathway and the subcellular localization of its downstream effector YAP dictate lineage specific gene expression, the response of YAP to these dynamic cellular changes remains incompletely understood. Here we address these questions by quantitative live imaging of endogenously tagged YAP while simultaneously monitoring geometric cellular features and cell cycle progression throughout cell fate segregation. We apply a probabilistic model to our dynamic data, providing a quantitative characterization of the mutual effects of YAP and cellular relative exposed area, which has previously been shown to correlate with subcellular YAP localization in fixed samples. Additionally, we study how nuclear YAP levels are influenced by other factors, such as the decreasing pool of maternally provided YAP that is partitioned to daughter cells through cleavage divisions, cell cycle-associated nuclear volume changes, and a delay after divisions in adjusting YAP levels to new cell positions. Interestingly, we find that establishing low nuclear YAP levels required for the inner cell mass fate is largely achieved by passive cell cycle-associated mechanisms. Moreover, contrary to expectations, we find that mechanical perturbations that result in cell and nuclear shape changes do not influence YAP localization in the embryo. Together our work identifies how various inputs are integrated over a dynamic developmental time course to shape the levels of a key molecular determinant of the first cell fate choice.
Collapse
Affiliation(s)
- Madeleine Chalifoux
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA; Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ, USA
| | - Maria Avdeeva
- Center for Computational Biology, Flatiron Institute, Simons Foundation, New York, USA
| | - Eszter Posfai
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA.
| |
Collapse
|
14
|
Wang L, Wang X, Wu J, Chen J, He Z, Wang J, Zhang X. Magnesium Ions Induce Endothelial Cell Differentiation into Tip Cell and Enhance Vascularized Bone Regeneration. Adv Healthc Mater 2025:e2500274. [PMID: 40346783 DOI: 10.1002/adhm.202500274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 04/07/2025] [Indexed: 05/12/2025]
Abstract
Vascularization has been considered an essential strategy for bone regeneration and can be promoted by magnesium ions (Mg2+). During angiogenesis, the differentiation of endothelial cells (ECs) into tip cell is a critical step since it controls the growth direction and pattern of new vascular sprouts. While several studies have noted the pro-angiogenic effects of Mg2+, however, their specific influence on tip cell formation is unclear. Therefore, this research seeks to examine the impact of Mg2+ on tip cells and elucidate the potential mechanisms involved. The results reveal that Mg2+ shows good compatibility and stimulates ECs to migrate and invade in vitro. Moreover, Mg2+ enhances EC spheroids sprouting and elevates the expression of genes linked to tip cells. The underlying mechanisms are that Mg2+ facilitates tip cell differentiation via the VEGFA-VEGFR2/Notch1 signaling pathway crosstalk and promotes migration and filopodia formation of tip cells and proliferation of stalk cells by inducing YAP nuclear translocation, culminating in the maturation of vascular networks. Furthermore, EC spheroids stimulated by Mg2+ load in hydrogel enhance vascularized bone regeneration in vivo. These findings enrich the understanding of how Mg2+ influence blood vessel formation and provide practical strategies for the development and design of magnesium-based biomaterials.
Collapse
Affiliation(s)
- Liang Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Xu Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Jicenyuan Wu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Junyu Chen
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Zihan He
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
- Department of Prosthodontics and Implantology, The Affiliated Stomatological Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, China
| | - Jian Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Xin Zhang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| |
Collapse
|
15
|
Yang Y, Akdemir AR, Rashik RA, Shihadeh Khater OA, Weng Z, Wang L, Zhong Y, Gallant ND. Guided neural stem cell differentiation by dynamic loading of 3D printed elastomeric scaffolds. J Mech Behav Biomed Mater 2025; 165:106940. [PMID: 39955829 DOI: 10.1016/j.jmbbm.2025.106940] [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: 12/10/2024] [Revised: 01/22/2025] [Accepted: 02/09/2025] [Indexed: 02/18/2025]
Abstract
The limited regenerative ability of "permanent" cells is a major barrier to treating conditions like spinal cord injury (SCI) and myocardial infarction (MI). The delivery of stem cells, which can generate various cell types, offer potential for personalized therapy with reduced immunoreaction and recovery time. However, restoring function to these tissues also requires new or replacement cells to align properly. Neurons, for example, must organize and extend parallel axons, mimicking their natural structure for directional signal propagation. Current stem cell differentiation methods lack guidance, resulting in randomly distributed axons and limited repair effectiveness. Advancing methods and materials to guide stem cell differentiation into functional, aligned nerve bundles is crucial for improving SCI treatment outcomes. This study aimed to develop an in vitro system to promote aligned neural differentiation by applying cyclic uniaxial tension to PC-12 stem cells adhered to 3D-printed elastic scaffolds. We created a simple loading device which can apply cyclic and controllable stretching force to a scaffold, which in turn transmits uniaxial tension to cells adhered to the scaffold during their differentiation. An elastomer ink for 3D printing scaffolds was formulated and surface treatment processes were investigated to enhance the cell-scaffold adhesion to support the dynamic loading. It was revealed that a corona discharge treatment while the scaffold is soaked with type I collagen can significantly enhance cell adhesion. A range of strain magnitudes and frequencies were revealed to enhance the differentiation of neural tissue derived PC-12 cells to neuron cells and increase the length of their neurites up to 76%. The combination of 3% maximum strain and 1 Hz loading frequency maximized differentiation and neurite extension. These findings demonstrate that dynamic mechanical stimulation enhances neural differentiation and organization, offering an alternative approach for regenerative therapies targeting SCI and similar conditions.
Collapse
Affiliation(s)
- Yi Yang
- Department of Mechanical Engineering, University of South Florida, 4202 E. Fowler Ave, Tampa, FL, 33620, USA
| | - Abdullah Revaha Akdemir
- Department of Mechanical Engineering, University of South Florida, 4202 E. Fowler Ave, Tampa, FL, 33620, USA
| | - Rafsan Ahmed Rashik
- Department of Mechanical Engineering, University of South Florida, 4202 E. Fowler Ave, Tampa, FL, 33620, USA
| | - Omar Ahmad Shihadeh Khater
- Department of Mechanical Engineering, University of South Florida, 4202 E. Fowler Ave, Tampa, FL, 33620, USA
| | - Zijian Weng
- Department of Mechanical Engineering, University of South Florida, 4202 E. Fowler Ave, Tampa, FL, 33620, USA
| | - Long Wang
- Department of Civil and Environmental Engineering, California Polytechnic State University, San Luis Obispo, CA, 93407, USA
| | - Ying Zhong
- Sauvage Laboratory for Smart Materials, School of Integrated Circuits, Harbin Institute of Technology (Shenzhen), University Town of Shenzhen, Shenzhen, Guangdong, 518055, China.
| | - Nathan D Gallant
- Department of Mechanical Engineering, University of South Florida, 4202 E. Fowler Ave, Tampa, FL, 33620, USA.
| |
Collapse
|
16
|
Basak S, Packirisamy G. Impact of Magneto-Mechanical Actuation on Cell Differentiation: A Study Using Wireless, 3D-Printed Device and a Porous Ferrogel. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2412112. [PMID: 39776212 DOI: 10.1002/smll.202412112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Indexed: 01/11/2025]
Abstract
Cells perceive external and internally generated forces of different kinds, significantly impacting their cellular biology. In the relatively nascent field of mechanobiology, the impact of such forces is studied and further utilized to broaden the knowledge of cellular developmental pathways, disease progression, tissue engineering, and developing novel regenerative strategies. However, extensive considerations of mechanotransduction pathways for biomedical applications are still broadly limited due to a lack of affordable technologies in terms of devices and simple magnetic actuatable materials. Herein, synthesizing a monophasic, macroporous, in situ-fabricated gelatin-based ferrogel is reported using polyethylene glycol (PEG) coated-iron oxide (magnetite) particles with high magnetization. Developing a 3D printed, compact, and wireless device capable of providing a wide range of magneto-mechanical actuation using magnetic field susceptible materials in a noncontact manner is reported. Using the device and ferrogel, C2C12 myoblast differentiation is studied under magnetic field actuation, and significant differences in the myogenin, a differentiation marker, expression behavior are observed. Due to careful design considerations, robust component selection, and easy availability of low-cost precursor for magnetic responsive material fabrication, the device-ferrogel combination can be easily adapted to routine biological studies, thereby helping mechanobiology to be utilized for developing exciting new biomedical strategies.
Collapse
Affiliation(s)
- Soumyadeep Basak
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, 247667, India
| | - Gopinath Packirisamy
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, 247667, India
- Centre for Nanotechnology, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, 247667, India
| |
Collapse
|
17
|
Karlinski Zur M, Bhattacharya B, Solomonov I, Ben Dror S, Savidor A, Levin Y, Prior A, Sapir T, Harris T, Olender T, Schmidt R, Schwarz JM, Sagi I, Buxboim A, Reiner O. Altered extracellular matrix structure and elevated stiffness in a brain organoid model for disease. Nat Commun 2025; 16:4094. [PMID: 40312467 PMCID: PMC12045990 DOI: 10.1038/s41467-025-59252-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 04/15/2025] [Indexed: 05/03/2025] Open
Abstract
The viscoelastic properties of tissues influence their morphology and cellular behavior, yet little is known about changes in these properties during brain malformations. Lissencephaly, a severe cortical malformation caused by LIS1 mutations, results in a smooth cortex. Here, we show that human-derived brain organoids with LIS1 mutation exhibit increased stiffness compared to controls at multiple developmental stages. This stiffening correlates with abnormal extracellular matrix (ECM) expression and organization, as well as elevated water content, measured by diffusion-weighted MRI. Short-term MMP9 treatment reduces both stiffness and water diffusion levels to control values. Additionally, a computational microstructure mechanical model predicts mechanical changes based on ECM organization. These findings suggest that LIS1 plays a critical role in ECM regulation during brain development and that its mutation leads to significant viscoelastic alterations.
Collapse
Grants
- AARG-NTF-21-849529 Alzheimer's Association
- We express our gratitude for the help of Dr. Arpan Parichha and Alfredo Isaac Ponce Arias. Orly Reiner is an incumbent of the Berstein-Mason professorial chair of Neurochemistry and the Head of the M. Judith Ruth Institute for Preclinical Brain Research. Our research has been supported by a research grant from Ethel Lena Levy, the Selsky Memory Research Project, the Gladys Monroy and Larry Marks Center for Brain Disorders, the Advantage Trust, the Nella and Leon Benoziyo Center for Neurological Diseases, the David and Fela Shapell Family Center for Genetic Disorders Research, the Abish-Frenkel RNA center, the Brenden- Mann Women's Innovation Impact Fund, The Irving B. Harris Fund for New Directions in Brain Research, the Irving Bieber, M.D. and Toby Bieber, M.D. Memorial Research Fund, The Leff Family, Barbara & Roberto Kaminitz, Sergio & Sônia Lozinsky, Debbie Koren, Jack and Lenore Lowenthal, and the Dears Foundation. A research grant from the Estates of Ethel H. Smith, Gerald Alexander, Mr. and Mrs. George Zbeda, David A. Fishstrom, Norman Fidelman, Hermine Miller, Olga Klein Astrachan, Hermine Miller, and The Maurice and Vivienne Wohl Biology Endowment, Supported by a research grant from Emily Merjan, the ISF grant (545/21), and the United States-Israel Binational Science Foundation (BSF; Grant No. 2023009).
Collapse
Affiliation(s)
- Maayan Karlinski Zur
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
- Department of Molecular Neuroscience, Weizmann Institute, Rehovot, Israel
| | - Bidisha Bhattacharya
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
- Department of Molecular Neuroscience, Weizmann Institute, Rehovot, Israel
| | - Inna Solomonov
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Sivan Ben Dror
- The Institute of Life Sciences, The Hebrew University of Jerusalem, The Edmond J. Safra Campus, Jerusalem, Israel
| | - Alon Savidor
- The De Botton Protein Profiling Institute of the Nancy and Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot, Israel
| | - Yishai Levin
- The De Botton Protein Profiling Institute of the Nancy and Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot, Israel
| | - Amir Prior
- The De Botton Protein Profiling Institute of the Nancy and Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot, Israel
| | - Tamar Sapir
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
- Department of Molecular Neuroscience, Weizmann Institute, Rehovot, Israel
| | - Talia Harris
- Department of Chemical Research Support, Weizmann Institute of Science, Rehovot, Israel
| | - Tsviya Olender
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Rita Schmidt
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel
- The Azrieli National Institute for Human Brain Imaging and Research, Weizmann Institute of Science, Rehovot, Israel
| | - J M Schwarz
- Physics Department, Syracuse University, Syracuse, NY, USA
| | - Irit Sagi
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel.
| | - Amnon Buxboim
- The Institute of Life Sciences, The Hebrew University of Jerusalem, The Edmond J. Safra Campus, Jerusalem, Israel.
- School of Computer Science and Engineering, The Hebrew University of Jerusalem, The Edmond J. Safra Campus, Jerusalem, Israel.
- The Alexender Grass Center for Bioengineering, The Hebrew University of Jerusalem, The Edmond J. Safra Campus, Jerusalem, Israel.
| | - Orly Reiner
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel.
- Department of Molecular Neuroscience, Weizmann Institute, Rehovot, Israel.
| |
Collapse
|
18
|
Wang Y, Ruf S, Wang L, Heimerl T, Bange G, Groeger S. The Dual Roles of Lamin A/C in Macrophage Mechanotransduction. Cell Prolif 2025; 58:e13794. [PMID: 39710429 PMCID: PMC12099221 DOI: 10.1111/cpr.13794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 11/12/2024] [Accepted: 12/04/2024] [Indexed: 12/24/2024] Open
Abstract
Cellular mechanotransduction is a complex physiological process that integrates alterations in the external environment with cellular behaviours. In recent years, the role of the nucleus in mechanotransduction has gathered increased attention. Our research investigated the involvement of lamin A/C, a component of the nuclear envelope, in the mechanotransduction of macrophages under compressive force. We discovered that hydrostatic compressive force induces heterochromatin formation, decreases SUN1/SUN2 levels, and transiently downregulates lamin A/C. Notably, downregulated lamin A/C increased nuclear permeability to yes-associated protein 1 (YAP1), thereby amplifying certain effects of force, such as inflammation induction and proliferation inhibition. Additionally, lamin A/C deficiency detached the linker of nucleoskeleton and cytoskeleton (LINC) complex from nuclear envelope, consequently reducing force-induced DNA damage and IRF4 expression. In summary, lamin A/C exerted dual effects on macrophage responses to mechanical compression, promoting certain outcomes while inhibiting others. It operated through two distinct mechanisms: enhancing nuclear permeability and impairing intracellular mechanotransmission. The results of this study support the understanding of the mechanisms of intracellular mechanotransduction and may assist in identifying potential therapeutic targets for mechanotransduction-related diseases.
Collapse
Affiliation(s)
- Yao Wang
- Department of Orthodontics, Faculty of MedicineJustus Liebig UniversityGiessenGermany
| | - Sabine Ruf
- Department of Orthodontics, Faculty of MedicineJustus Liebig UniversityGiessenGermany
| | - Lei Wang
- Department of Orthodontics, Faculty of MedicineJustus Liebig UniversityGiessenGermany
- Department of Oral and Maxillofacial Surgery, Affiliated Stomatological HospitalSouthwest Medical UniversityLuzhouP. R. China
| | - Thomas Heimerl
- Center for Synthetic Microbiology (SYNMIKRO)Philipps‐University MarburgMarburgGermany
| | - Gert Bange
- Center for Synthetic Microbiology (SYNMIKRO)Philipps‐University MarburgMarburgGermany
| | - Sabine Groeger
- Department of Orthodontics, Faculty of MedicineJustus Liebig UniversityGiessenGermany
| |
Collapse
|
19
|
Li T, Huang L, Guo C, Ren J, Chen X, Ke Y, Xun Z, Hu W, Qi Y, Wang H, Gong Z, Liang X, Xue X. Massage-Mimicking Nanosheets Mechanically Reorganize Inter-organelle Contacts to Restore Mitochondrial Functions in Parkinson's Disease. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2413376. [PMID: 40223359 PMCID: PMC12120710 DOI: 10.1002/advs.202413376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 03/11/2025] [Indexed: 04/15/2025]
Abstract
Parkinson's disease (PD) is exacerbated by dysfunction of inter-organelle contact, which depends on cellular responses to the mechanical microenvironment and can be regulated by external mechanical forces. Delivering dynamic mechanical forces to neural cells proves challenging due to the skull. Inspired by the effects of massage; here PEGylated black phosphorus nanosheets (PEG-BPNS), known for their excellent biocompatibility, biodegradability, specific surface area, mechanical strength, and flexibility, are introduced, which are capable of adhering to neural cell membrane and generating mechanical stimulation with their lateral size of 200 nm, exhibiting therapeutic potential in a 1-methyl-4-phenyl-1,2,3,6-te-trahydropyridine-induced PD mouse model by regulating inter-organelle contacts. Specifically, it is found that 200 nm PEG-BPNS, acting as "NanoMassage," significantly increase plasma membrane tension, as evidenced by fluorescent lipid tension reporter fluorescence lifetime analysis. This mechanical force modulates actin reorganization, subsequently regulating the contacts between actin, mitochondria, and endoplasmic reticulum, further controlling mitochondrial fission and mitigating mitochondrial dysfunction in PD, exhibiting therapeutic efficacy via intranasal administration. These findings provide a noninvasive strategy for applying mechanical stimulation to deep brain areas and elucidate the mechanism of NanoMassage mediating inter-organelle contacts, suggesting the rational design of "NanoMassage" to remodel inter-organelle communications in neurodegenerative disease treatment.
Collapse
Affiliation(s)
- Tianqi Li
- State Key Laboratory of Medicinal Chemical BiologyCollege of PharmacyNankai UniversityTianjin300350P. R. China
| | - Liwen Huang
- State Key Laboratory of Medicinal Chemical BiologyCollege of PharmacyNankai UniversityTianjin300350P. R. China
- Present address:
Department of ChemistryShanghai Key Laboratory of Molecular Catalysis and Innovative MaterialsState Key Laboratory of Molecular Engineering of Polymers and iChemFudan UniversityShanghai200438P. R. China
| | - Chenxiao Guo
- State Key Laboratory of Medicinal Chemical BiologyCollege of PharmacyNankai UniversityTianjin300350P. R. China
| | - Jing Ren
- State Key Laboratory of Medicinal Chemical BiologyCollege of PharmacyNankai UniversityTianjin300350P. R. China
| | - Xi Chen
- State Key Laboratory of Medicinal Chemical BiologyCollege of PharmacyNankai UniversityTianjin300350P. R. China
| | - Yachu Ke
- State Key Laboratory of Medicinal Chemical BiologyCollege of PharmacyNankai UniversityTianjin300350P. R. China
| | - Zengyu Xun
- State Key Laboratory of Medicinal Chemical BiologyCollege of PharmacyNankai UniversityTianjin300350P. R. China
| | - Wenzhuo Hu
- State Key Laboratory of Medicinal Chemical BiologyCollege of PharmacyNankai UniversityTianjin300350P. R. China
| | - Yilin Qi
- State Key Laboratory of Medicinal Chemical BiologyCollege of PharmacyNankai UniversityTianjin300350P. R. China
| | - Heping Wang
- State Key Laboratory of Medicinal Chemical BiologyCollege of PharmacyNankai UniversityTianjin300350P. R. China
- Present address:
State Key Laboratory of Advanced Medical Materials and DevicesTianjin Key Laboratory of Radiation Medicine and Molecular Nuclear MedicineKey Laboratory of Radiopharmacokinetics for Innovative DrugsTianjin Institutes of Health ScienceInstitute of Radiation MedicineChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjin300192P. R. China
| | - Zhongying Gong
- Department of NeurologyTianjin First Central HospitalSchool of MedicineNankai UniversityTianjin300192P. R. China
| | - Xing‐Jie Liang
- Laboratory of Controllable NanopharmaceuticalsChinese Academy of Sciences (CAS) Center for Excellence in Nanoscience and CAS Key Laboratory for Biomedical Effects of Nanomaterials and NanosafetyNational Center for Nanoscience and TechnologyBeijing100190P. R. China
- University of Chinese Academy of SciencesBeijing101408P. R. China
| | - Xue Xue
- State Key Laboratory of Medicinal Chemical BiologyCollege of PharmacyNankai UniversityTianjin300350P. R. China
| |
Collapse
|
20
|
Chen R, Zhang R, Ke F, Guo X, Zeng F, Liu Q. Mechanisms of breast cancer metastasis: the role of extracellular matrix. Mol Cell Biochem 2025; 480:2771-2796. [PMID: 39652293 DOI: 10.1007/s11010-024-05175-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 11/23/2024] [Indexed: 05/03/2025]
Abstract
The components of the extracellular matrix (ECM) are dynamic, and they mediate mechanical signals that modulate cellular behaviors. Disruption of the ECM can induce the migration and invasion of cancer cells via specific signaling pathways and cytokines. Metastasis is a leading cause of high mortality in malignancies, and early intervention can improve survival rates. However, breast cancer is frequently diagnosed subsequent to metastasis, resulting in poor prognosis and distant metastasis poses substantial hurdles in therapy. In breast cancer, there is notable tissue remodeling of ECM proteins, with several identified as essential components for metastasis. Moreover, specific ECM molecules, receptors, enzymes, and various signaling pathways play crucial roles in breast cancer metastasis, drug treatment, and resistance. The in-depth consideration of these elements could provide potential therapeutic targets to enhance the survival rates and quality of life for breast cancer patients. This review explores the mechanisms by which alterations in the ECM contribute to breast cancer metastasis and discusses current clinical applications targeting ECM in breast cancer treatment, offering valuable perspectives for future ECM-based therapies.
Collapse
Affiliation(s)
- Rui Chen
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
- Laboratory of Biochemistry and Molecular Biology, School of Basic Medical Science, Southwest Medical University, Luzhou, 646000, China
| | - Ranqi Zhang
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
- Laboratory of Biochemistry and Molecular Biology, School of Basic Medical Science, Southwest Medical University, Luzhou, 646000, China
| | - Famin Ke
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Xiurong Guo
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Fancai Zeng
- Laboratory of Biochemistry and Molecular Biology, School of Basic Medical Science, Southwest Medical University, Luzhou, 646000, China.
| | - Qiuyu Liu
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China.
- Laboratory of Biochemistry and Molecular Biology, School of Basic Medical Science, Southwest Medical University, Luzhou, 646000, China.
| |
Collapse
|
21
|
Han J, Halwachs K, West T, Larsen B, Sacks MS, Rosales AM, Zoldan J. Matrix Stiffness Regulates Mechanotransduction and Vascular Network Formation of hiPSC-Derived Endothelial Progenitors Encapsulated in 3D Hydrogels. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.11.648340. [PMID: 40291699 PMCID: PMC12027365 DOI: 10.1101/2025.04.11.648340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
The mechanical properties of the extracellular matrix (ECM), particularly stiffness, regulate endothelial progenitor responses during vascular development, yet their behavior in physiologically compliant matrices (<1 kPa) remains underexplored. Using norbornene-modified hyaluronic acid (NorHA) hydrogels with tunable stiffness (190-884 Pa), we investigated how hydrogel stiffness influences cell morphology, endothelial maturation, mechanotransduction, and microvascular network formation in human induced pluripotent stem cell-derived endothelial progenitors (hiPSC-EPs). Our findings reveal a stiffness-dependent tradeoff between mechanotransduction and vascular network formation. At intermediate stiffness (551 Pa), cells exhibited the greatest increase in endothelial marker CD31 expression and Yes-associated protein (YAP)/ transcriptional coactivator with PDZ-binding motif (TAZ) nuclear translocation, indicating enhanced mechanotransduction and endothelial maturation. However, this did not translate to superior plexus formation. Instead, the most compliant matrix (190 Pa) supported greater vascular connectivity, characterized by longer branches (∼0.03/volume vs. 0.015 at 551 Pa) and enhanced actin remodeling. 3D cell contraction measurements revealed a 15.6-fold higher basal displacement in compliant hydrogels, suggesting that cell-generated forces and matrix deformability collectively drive vascular morphogenesis. Unlike prior studies focusing on pathological stiffness ranges (>10 kPa), our results emphasize that vascularization is not solely driven by the most mechanotransductive environment but rather by a balance of compliance, contractility, and cell-induced remodeling. These findings underscore the need to design hydrogels that provide sufficient mechanotransduction for endothelial maturation while maintaining compliance to support dynamic vascular morphogenesis. This work provides a mechanically tuned framework for optimizing microenvironments to balance endothelial differentiation and vascular network formation in tissue engineering and regenerative medicine.
Collapse
|
22
|
Terrell JA, Chen C. Extracellular Matrix Microstructures Modulate Hepatic Methionine Cycle and Methylations. Biomacromolecules 2025. [PMID: 40298277 DOI: 10.1021/acs.biomac.4c01748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
The field of mechanobiology has grown in the past decade, but limited studies investigate how the extracellular matrix affects the cell metabolome. The methionine cycle involves the catabolism and regeneration of methionine through the donation and recovery of a single methyl group; this methyl group can methylate DNA, RNA, and proteins to alter gene expression and protein-protein interactions. Through studying cells cultured on fibrous (mimicking healthy extracellular matrice (ECM)) and flat (mimicking severely fibrotic ECM) substrates, we observed an increase in methionine cycle enzyme expression in cells on the flat substrate. We also present how the methionine cycle is modulated by the ECM through transmembrane protein integrin β1. By inhibiting integrin activation through the ligand-mimicking peptide RGD, we observed that the methionine cycle was protected from alteration. The results presented provide insight into possible therapeutic targets for fibrotic diseases and knowledge of mechanisms by which the ECM alters cell processes.
Collapse
Affiliation(s)
- John A Terrell
- Department of Chemistry and Biochemistry, University of Maryland Baltimore County, Baltimore, Maryland 21250, United States
| | - Chengpeng Chen
- Department of Chemistry and Biochemistry, University of Maryland Baltimore County, Baltimore, Maryland 21250, United States
| |
Collapse
|
23
|
Kofler M, Venugopal S, Gill G, Di Ciano-Oliveira C, Kapus A. M-Motif, a potential non-conventional NLS in YAP/TAZ and other cellular and viral proteins that inhibits classic protein import. iScience 2025; 28:112105. [PMID: 40224012 PMCID: PMC11986988 DOI: 10.1016/j.isci.2025.112105] [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: 03/12/2024] [Revised: 08/22/2024] [Accepted: 02/21/2025] [Indexed: 04/15/2025] Open
Abstract
Multiple mechanisms were proposed to mediate the nuclear import of TAZ/YAP, transcriptional co-activators regulating organ growth and regeneration. Our earlier observations showed that TAZ/YAP harbor a C-terminal, unconventional nuclear localization signal (NLS). Here, we show that this sequence, necessary and sufficient for basal, ATP-independent nuclear import, contains an indispensable central methionine flanked by negatively charged residues. Based on these features, we define the M-motif and propose that it is a new class of NLS, also present and import-competent in other cellular (STAT1 and cyclin B1) and viral (ORF6 of SARS-CoV2, VSV-M) proteins. Accordingly, ORF6 SARS-Cov2 competitively inhibits TAZ/YAP uptake, while TAZ abrogates STAT1 import. Similar to viral M-motif proteins, TAZ binds RAE1 and inhibits classic nuclear protein import, including that of antiviral factors (IRF3 and NF-κB). However, RAE1 is dispensable for TAZ import itself. Thus, the TAZ/YAP NLS has a dual function: it mediates unconventional nuclear import and inhibits classic import, contributing to the suppression of antiviral responses.
Collapse
Affiliation(s)
- Michael Kofler
- Keenan Research Centre for Biomedical Science of the St. Michael’s Hospital, Toronto, ON, Canada
| | - Shruthi Venugopal
- Keenan Research Centre for Biomedical Science of the St. Michael’s Hospital, Toronto, ON, Canada
| | - Gary Gill
- Keenan Research Centre for Biomedical Science of the St. Michael’s Hospital, Toronto, ON, Canada
| | | | - András Kapus
- Keenan Research Centre for Biomedical Science of the St. Michael’s Hospital, Toronto, ON, Canada
- Department Surgery, University of Toronto, Toronto, ON M5B 1T8, Canada
- Department Biochemistry, University of Toronto, Toronto, ON M5B 1T8, Canada
| |
Collapse
|
24
|
Kuhn MR, Wolcott EA, Langer EM. Developments in gastrointestinal organoid cultures to recapitulate tissue environments. Front Bioeng Biotechnol 2025; 13:1521044. [PMID: 40313639 PMCID: PMC12043594 DOI: 10.3389/fbioe.2025.1521044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 03/21/2025] [Indexed: 05/03/2025] Open
Abstract
Culture platforms that closely mimic the spatial architecture, cellular diversity, and extracellular matrix composition of native tissues can serve as invaluable tools for a range of scientific discovery and biomedical applications. Organoids have emerged as a promising alternative to both traditional 2D cell culture and animal models, offering a physiologically relevant 3D culture system for studying human cell biology. Organoids provide a manipulable platform to investigate organ development and function as well as to model patient-specific phenotypes. This mini review examines various methods used for culturing organoids to model normal and disease conditions in gastrointestinal tissues. We focus on how the matrix composition and media formulations can impact cell signaling, altering the baseline cellular phenotypes as well as response to perturbations. We discuss future directions for optimizing organoid culture conditions to improve basic and translational potential.
Collapse
Affiliation(s)
- Madeline R. Kuhn
- Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, United States
- Division of Oncological Sciences, Oregon Health and Science University, Portland, OR, United States
| | - Emma A. Wolcott
- Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, United States
- Division of Oncological Sciences, Oregon Health and Science University, Portland, OR, United States
| | - Ellen M. Langer
- Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, United States
- Division of Oncological Sciences, Oregon Health and Science University, Portland, OR, United States
- Brenden-Colson Center for Pancreatic Care, Oregon Health and Science University, Portland, OR, United States
| |
Collapse
|
25
|
Neyazi B, Swiatek VM, Karimpour MA, Stassen S, Stein KP, Rashidi A, Dumitru CA, Sandalcioglu IE. The Role of Hippo Signaling in Brain Arteriovenous Malformations: Molecular Insights into Post-Embolization Remodeling. Int J Mol Sci 2025; 26:3791. [PMID: 40332417 PMCID: PMC12028238 DOI: 10.3390/ijms26083791] [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: 02/28/2025] [Revised: 04/12/2025] [Accepted: 04/14/2025] [Indexed: 05/08/2025] Open
Abstract
Brain arteriovenous malformations (bAVMs) are complex vascular lesions with significant clinical risks. The Hippo signaling pathway, particularly its downstream effector YAP, plays a crucial role in angiogenesis and vascular remodeling. This study investigates the role of YAP and related molecular markers in bAVMs, focusing on the effects of embolization. Immunohistochemical analysis was conducted on tissue samples from bAVM patients (n = 127), as well as on healthy blood vessels (n = 17). YAP, HIF-1α, FGFR1, CTGF, and CYR61 expression were quantified and correlated with clinical parameters. Results: In healthy vessels, YAP exhibited nuclear localization in (sub)endothelial cells and the tunica media, while CTGF and CYR61 were detected in the cytoplasm and extracellular matrix. The expression of YAP, CTGF, and CYR61 was significantly lower in bAVM tissues. Embolized bAVMs exhibited significantly higher expression of YAP, CTGF, and CYR61 compared to non-embolized tissues, suggesting a link between embolization and pro-angiogenic signaling. Additionally, FGFR1 was upregulated in embolized tissues. These results suggest that upregulation of YAP expression via the Hippo pathway might play a key role in bAVM pathophysiology. Embolization may further promote vascular remodeling. Dysregulation of YAP and related molecules in bAVMs warrants further studies to explore potential therapeutic strategies targeting the Hippo pathway.
Collapse
Affiliation(s)
- Belal Neyazi
- Department of Neurosurgery, Otto-von-Guericke University, 39120 Magdeburg, Germany; (V.M.S.); (M.A.K.)
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Jiang M, Ding H, Huang Y, Lau CW, Guo Y, Luo J, Shih YT, Xia Y, Yao X, Chiu JJ, Wang L, Chien S, Huang Y. Endothelial Serotonin Receptor 1B Acts as a Mechanosensor to Drive Atherosclerosis. Circ Res 2025; 136:887-901. [PMID: 40071330 DOI: 10.1161/circresaha.124.325453] [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: 08/26/2024] [Revised: 03/02/2025] [Accepted: 03/04/2025] [Indexed: 04/12/2025]
Abstract
BACKGROUND Atherosclerosis is characterized by the accumulation of fatty and fibrotic plaques, which preferentially develop at curvatures and branches along the arterial trees that are exposed to disturbed flow. However, the mechanisms by which endothelial cells sense disturbed flow are still unclear. METHODS The partial carotid ligation mouse model was used to investigate disturbed flow-induced atherogenesis. In vitro experiments were performed using the ibidi system to generate oscillatory shear stress and laminar shear stress. ApoE-/- mice with endothelium-specific knockout or overexpression of 5-HT1B (serotonin receptor 1B) were used to investigate the role of endothelial 5-HT1B in atherosclerosis. RNA sequencing analysis, immunofluorescence analysis, and molecular biological techniques were used to explore the role of 5-HT1B in mechanotransduction and endothelial activation. RESULTS The data showed that human endothelial cells express a high level of 5-HT1B, which is a serotonin receptor subtype. Endothelial 5-HT1B is upregulated in atherosclerotic areas of both humans and rodents and is increased by disturbed flow both in vivo and in vitro. Endothelium-specific overexpression of 5-HT1B exacerbates, whereas knockout or knockdown of 5-HT1B in endothelium inhibits disturbed flow-induced endothelial inflammation and atherogenesis in both male and female ApoE-/- mice. We reveal a previously unknown role of 5-HT1B as a mechanosensor in endothelial cells in response to mechanical stimuli. Upon activation by oscillatory shear stress, 5-HT1B recruits β-arrestin, orchestrates RhoA (ras homolog family member A), and then activates mechanosensitive YAP (yes-associated protein), thereby enhancing endothelial inflammation and monocyte infiltration. Pharmacological blockade of 5-HT1B suppresses endothelial activation and atherogenesis via inhibition of YAP. CONCLUSIONS Taken together, these results uncover that endothelial 5-HT1B acts as a mechanosensor for disturbed flow and contributes to atherogenesis. Inhibition of 5-HT1B could be a promising therapeutic strategy for atherosclerosis.
Collapse
MESH Headings
- Animals
- Humans
- Mechanotransduction, Cellular
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Atherosclerosis/genetics
- Receptor, Serotonin, 5-HT1B/metabolism
- Receptor, Serotonin, 5-HT1B/genetics
- Mice
- Male
- Mice, Knockout, ApoE
- Mice, Inbred C57BL
- Endothelial Cells/metabolism
- Stress, Mechanical
- Cells, Cultured
- Female
- Human Umbilical Vein Endothelial Cells/metabolism
- Apolipoproteins E/genetics
- Mice, Knockout
- Disease Models, Animal
Collapse
Affiliation(s)
- Minchun Jiang
- Department of Endocrinology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China (M.J.)
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China (M.J., H.D., C.W.L., Y.X., X.Y., Yu Huang)
| | - Huanyu Ding
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China (M.J., H.D., C.W.L., Y.X., X.Y., Yu Huang)
| | - Yuhong Huang
- Department of Biomedical Sciences (Yuhong Huang, L.W., Yu Huang), City University of Hong Kong, Hong Kong, China
| | - Chi Wai Lau
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China (M.J., H.D., C.W.L., Y.X., X.Y., Yu Huang)
| | - Ying Guo
- Department of Endocrinology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China (Y.G.)
| | - Jianfang Luo
- Department of Cardiology, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China (J.L.)
| | - Yu-Tsung Shih
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli, Taiwan (Y.-T.S., J.-J.C.)
| | - Yin Xia
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China (M.J., H.D., C.W.L., Y.X., X.Y., Yu Huang)
| | - Xiaoqiang Yao
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China (M.J., H.D., C.W.L., Y.X., X.Y., Yu Huang)
| | - Jeng-Jiann Chiu
- College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan (J.-J.C.)
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli, Taiwan (Y.-T.S., J.-J.C.)
| | - Li Wang
- Department of Biomedical Sciences (Yuhong Huang, L.W., Yu Huang), City University of Hong Kong, Hong Kong, China
| | - Shu Chien
- Departments of Bioengineering and Medicine, and Institute of Engineering in Medicine, University of California, San Diego, CA (S.C.)
| | - Yu Huang
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China (M.J., H.D., C.W.L., Y.X., X.Y., Yu Huang)
- Department of Biomedical Sciences (Yuhong Huang, L.W., Yu Huang), City University of Hong Kong, Hong Kong, China
- Tung Biomedical Sciences Centre (Yu Huang), City University of Hong Kong, Hong Kong, China
| |
Collapse
|
27
|
Chen F, Kang NW, Wungcharoen T, Jiang L, Basco CA, Garcia-Sanchez J, Lin D, Seo YA, Jang K, Myung D, Liu WW. Repurposing verteporfin and hyaluronic acid gel for ocular surface treatment to prevent corneal scarring. J Control Release 2025; 380:1141-1151. [PMID: 39986473 DOI: 10.1016/j.jconrel.2025.02.051] [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: 10/09/2024] [Revised: 02/13/2025] [Accepted: 02/18/2025] [Indexed: 02/24/2025]
Abstract
There is an unmet need for point-of-care therapies to prevent scarring and promote corneal clarity after injury, which is essential for maintaining vision. Verteporfin, an inhibitor of Yes-associated protein (YAP), has been shown to prevent fibrosis in several organs. Visudyne (VP) is an FDA-approved liposomal formulation of verteporfin used to treat abnormal blood vessels in the eye. Here, we showed that VP reduces myofibroblast formation in corneal stromal fibroblasts. To prolong the residence time of verteporfin on the ocular surface, the cohesive viscoelastic ProVisc® hyaluronic acid (HA) gel was hybridized to VP. This formulation is readily translatable because both VP and ProVisc® HA gel are FDA-approved agents. The ProVisc® HA gel increased the residence of subconjunctivally injected verteporfin 12-fold at 24 h after injection compared with pure VP. A single subconjunctival administration of VP hybridized within ProVisc® HA gel (VP/HA hydrogel) significantly reduced YAP activation, corneal fibrosis, neovascularization, and inflammation, leading to reduced opacity without compromising epithelial wound healing in mechanically injured rat corneas. This work demonstrated that VP hybridized with a viscoelastic HA gel can be readily repurposed to promote scar-less healing in the cornea.
Collapse
Affiliation(s)
- Fang Chen
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA 94303, USA
| | - Nae-Won Kang
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA 94303, USA
| | - Thitima Wungcharoen
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA 94303, USA
| | - Li Jiang
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA 94303, USA
| | - Chris A Basco
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA 94303, USA
| | - Julian Garcia-Sanchez
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA 94303, USA
| | - Danting Lin
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA 94303, USA
| | - Youngyoon Amy Seo
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA 94303, USA
| | - Kyeongwoo Jang
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA 94303, USA
| | - David Myung
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA; Division of Ophthalmology, Department of Veterans Affairs, Palo Alto Health Care System, Palo Alto, CA 94304, USA.
| | - Wendy W Liu
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA 94303, USA.
| |
Collapse
|
28
|
Zhang M, Zhang B. Extracellular matrix stiffness: mechanisms in tumor progression and therapeutic potential in cancer. Exp Hematol Oncol 2025; 14:54. [PMID: 40211368 PMCID: PMC11984264 DOI: 10.1186/s40164-025-00647-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Accepted: 03/23/2025] [Indexed: 04/14/2025] Open
Abstract
Tumor microenvironment (TME) is a complex ecosystem composed of both cellular and non-cellular components that surround tumor tissue. The extracellular matrix (ECM) is a key component of the TME, performing multiple essential functions by providing mechanical support, shaping the TME, regulating metabolism and signaling, and modulating immune responses, all of which profoundly influence cell behavior. The quantity and cross-linking status of stromal components are primary determinants of tissue stiffness. During tumor development, ECM stiffness not only serves as a barrier to hinder drug delivery but also promotes cancer progression by inducing mechanical stimulation that activates cell membrane receptors and mechanical sensors. Thus, a comprehensive understanding of how ECM stiffness regulates tumor progression is crucial for identifying potential therapeutic targets for cancer. This review examines the effects of ECM stiffness on tumor progression, encompassing proliferation, migration, metastasis, drug resistance, angiogenesis, epithelial-mesenchymal transition (EMT), immune evasion, stemness, metabolic reprogramming, and genomic stability. Finally, we explore therapeutic strategies that target ECM stiffness and their implications for tumor progression.
Collapse
Affiliation(s)
- Meiling Zhang
- School of Basic Medicine, China Three Gorges University, 8 Daxue Road, Yichang, 443002, Hubei, China
- Central Laboratory, The First Affiliated Hospital of Jinan University, No. 613 Huangpu West Road, Tianhe District, Guangzhou, 510627, Guangdong, China
| | - Bin Zhang
- School of Basic Medicine, China Three Gorges University, 8 Daxue Road, Yichang, 443002, Hubei, China.
- Central Laboratory, The First Affiliated Hospital of Jinan University, No. 613 Huangpu West Road, Tianhe District, Guangzhou, 510627, Guangdong, China.
| |
Collapse
|
29
|
Lu J, Wang Z, Zhang L. Single-cell transcriptome analysis revealing mechanotransduction via the Hippo/YAP pathway in promoting fibroblast-to-myofibroblast transition and idiopathic pulmonary fibrosis development. Gene 2025; 943:149271. [PMID: 39855369 DOI: 10.1016/j.gene.2025.149271] [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: 09/27/2024] [Revised: 12/12/2024] [Accepted: 01/20/2025] [Indexed: 01/27/2025]
Abstract
OBJECTIVE Idiopathic pulmonary fibrosis (IPF) is an irreversible and fatal interstitial lung disease, characterized by excessive extracellular matrix (ECM) secretion that disrupts normal alveolar structure. This study aims to explore the potential molecular mechanisms underlying the promotion of IPF development. METHODS Firstly, we compared the transcriptome and single-cell sequencing data from lung tissue samples of patients with IPF and healthy individuals. Subsequently, we conducted Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses on the differentially expressed genes (DEGs). Furthermore, we employed sodium alginate hydrogels with varying degrees of crosslinking to provide differential mechanical stress, mimicking the mechanical microenvironment in vivo during lung fibrosis. On this basis, we examined cytoskeletal remodeling in fibroblasts MRC-5, mRNA expression of multiple related genes, immunofluorescence localization, and cellular proliferation capacity. RESULTS Bioinformatics analysis revealed a series of DEGs associated with IPF. Further functional and pathway enrichment analyses indicated that these DEGs were primarily enriched in ECM-related biological processes. Single-cell sequencing data revealed that fibroblasts and myofibroblasts are the main contributors to excessive ECM secretion and suggested activation of mechanotransduction and the Hippo/YAP signaling pathway in myofibroblasts. Cellular experiments demonstrated that sodium alginate hydrogels with different stiffness can simulate different mechanical stress environments, thereby affecting cytoskeletal rearrangement and Hippo/YAP pathway activity in MRC-5 lung fibroblasts. Notably, high levels of mechanical stress promoted YAP nuclear translocation, increased expression of type I collagen and α-SMA, and enhanced proliferative capacity. Additionally, we also found that fibroblasts primarily participate in mechanotransduction through the Rho/ROCK and Integrin/FAK pathways under high mechanical stress conditions, ultimately upregulating the gene expression of CCNE1/2, CTGF, and FGF1. CONCLUSION Our study uncovers the crucial role of cytoskeletal mechanotransduction in myofibroblast transformation and IPF development through activation of the Hippo/YAP pathway, providing new insights into understanding the pathogenesis of IPF.
Collapse
Affiliation(s)
- Jiaqi Lu
- Department of Oncology, Xinxiang Central Hospital, The Fourth Clinical College of Xinxiang Medical University, China.
| | - Zhenhua Wang
- Department of Oncology, Xinxiang Central Hospital, The Fourth Clinical College of Xinxiang Medical University, China
| | - Liguo Zhang
- Department of Oncology, Xinxiang Central Hospital, The Fourth Clinical College of Xinxiang Medical University, China
| |
Collapse
|
30
|
Whited A, Elkhalil A, Clark G, Ghose P. CDH-3/Cadherin, YAP-1/YAP and EGL-44/TEAD promote SYX-2/Syntaxin and EFF-1 fusogen-mediated phagosome closure. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.02.646655. [PMID: 40236144 PMCID: PMC11996554 DOI: 10.1101/2025.04.02.646655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Physical interactions between cells, such as cell-cell junctions, can profoundly impact cell fate. A vital cell fate for normal development and homeostasis is programmed cell death. Cells fated to die must be efficiently cleared away via phagocytosis, and defects are associated with a variety of diseased states. Whether cell-cell physical associations affect programmed cell elimination has not been well-explored. Here we describe, in vivo, a cell-cell adhesion-driven signaling pathway that ensures compartment-specific cell clearance during development. We previously described the specialized cell death program "Compartmentalized Cell Elimination" (CCE) in the C. elegans embryo. During CCE, the tail-spike cell (TSC), a polarized epithelial cell, undergoes a tripartite, ordered, and organized death sequence, allowing for the study of three distinct death modalities in a single cell setting. Prior to its demise, the TSC serves as a scaffold for the tail tip, formed by the hyp10 epithelial cell which develops along the TSC process. The hyp10 cell in turn also serves as the phagocyte for the dying TSC process. Here we present data suggesting that the physical association between the dying TSC and hyp10 phagocyte via CDH-3/cadherin mediates function of the mechanosensitive transcriptional coactivator YAP-1/YAP and its partner EGL-44/TEAD in the hyp10 phagocyte to promote localization of hyp10 SYX-2/Syntaxin around the dying TSC remnant. This pathway facilitates the phagocytic function of EFF-1/fusogen, which we have previously shown to be required for phagosome sealing during CCE. Our work sheds additional light on a poorly understood step of phagocytosis and implicates adhesive forces and signaling between cells as important in cell uptake.
Collapse
|
31
|
Zhurenkov KE, Svirskis D, Connor B, Malmström J. Actuated Hydrogel Platforms To Study Brain Cell Behavior. Adv Healthc Mater 2025; 14:e2404484. [PMID: 40091290 PMCID: PMC12004428 DOI: 10.1002/adhm.202404484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 02/16/2025] [Indexed: 03/19/2025]
Abstract
The human brain is a highly complex organ characterized by intricate neural networks, biochemical signaling, and unique mechanical properties. The soft and dynamic viscoelastic extracellular matrix (ECM) plays a crucial role in supporting different types of brain cells and influencing their behavior. Understanding how brain cells respond to mechanical stimuli within this complex environment is essential for unraveling fundamental mechanisms of healthy, unhealthy, and regenerative functions within the central nervous system. This requires the development of advanced materials and techniques to study the interplay between mechanical cues and cell responses. Hydrogels have become essential in this research, mimicking the brain's ECM in both chemical composition and mechanical behavior. Conventional hydrogels, while helpful, are static and lack dynamic stimulation. On the other hand, dynamic hydrogels provide reversible, dynamic stimulation, closely replicating the brain's ECM properties. This review discusses current hydrogel platforms used to investigate brain function in health and disease, focusing on traumatic brain injury (TBI)-like conditions and brain tumors. These dynamic materials offer sophisticated tools for understanding brain cell mechanobiology and developing new therapeutic approaches.
Collapse
Affiliation(s)
- Kirill E. Zhurenkov
- Department of Chemical and Materials EngineeringThe University of AucklandAuckland1010New Zealand
- MacDiarmid Institute for Advanced Materials and NanotechnologyWellington6140New Zealand
| | - Darren Svirskis
- School of PharmacyFaculty of Medical and Health SciencesThe University of AucklandAuckland1023New Zealand
| | - Bronwen Connor
- Department of Pharmacology and Clinical PharmacologySchool of Medical Sciences, Faculty of Medical and Health SciencesThe University of AucklandAuckland1023New Zealand
| | - Jenny Malmström
- Department of Chemical and Materials EngineeringThe University of AucklandAuckland1010New Zealand
- MacDiarmid Institute for Advanced Materials and NanotechnologyWellington6140New Zealand
| |
Collapse
|
32
|
Passi M, Stöckl JB, Fröhlich T, Moser S, Vollmar AM, Zahler S. CDK5 interacts with MST2 and modulates the Hippo signalling pathway. FEBS Open Bio 2025; 15:647-660. [PMID: 39739588 PMCID: PMC11961382 DOI: 10.1002/2211-5463.13962] [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: 10/31/2024] [Accepted: 12/16/2024] [Indexed: 01/02/2025] Open
Abstract
MST2 (STK3) is a major upstream kinase in the Hippo signalling pathway, an evolutionary conserved pathway in regulation of organ size, self-renewal and tissue homeostasis. Its downstream effectors are the transcriptional regulators YAP and TAZ. This pathway is regulated by a variety of factors, such as substrate stiffness or cell-cell contacts. Using a yeast two-hybrid screen, we detected a novel interaction between the kinases MST2 and CDK5, which we further confirmed by co-immunoprecipitation experiments. Cyclin-dependent kinase 5 (CDK5) is an unusual member of the family of cyclin-dependent kinases, involved in tumour growth and angiogenesis. Although a link between CDK5 and Hippo has been previously postulated, the mode of action is still elusive. Here, we show that knockdown of CDK5 causes reduced transcriptional activity of YAP and that CDK5 influences the phosphorylation levels of the Hippo upstream kinase LATS1. Moreover, a phosphoproteomics approach revealed that CDK5 interferes with the phosphorylation of DLG5, another upstream kinase, which regulates the Hippo pathway. Hence, CDK5 seems to act as a signalling hub for integrating the Hippo pathway and other signalling cascades. These interactions might have important implications for the use of CDK5 inhibitors, which are already in clinical use for tumour diseases.
Collapse
Affiliation(s)
- Mehak Passi
- Center for Drug ResearchLudwig‐Maximilians‐University MunichGermany
| | - Jan B. Stöckl
- Laboratory for Functional Genome Analysis, Gene Center MunichLudwig‐Maximilians‐University MunichGermany
| | - Thomas Fröhlich
- Laboratory for Functional Genome Analysis, Gene Center MunichLudwig‐Maximilians‐University MunichGermany
| | - Simone Moser
- Center for Drug ResearchLudwig‐Maximilians‐University MunichGermany
- Institute of PharmacyUniversity of InnsbruckAustria
| | | | - Stefan Zahler
- Center for Drug ResearchLudwig‐Maximilians‐University MunichGermany
| |
Collapse
|
33
|
Mohr-Allen SR, Gleghorn JP, Varner VD. Fluid secretion and luminal pressure control lateral branching morphogenesis in the embryonic avian lung. Dev Biol 2025; 520:251-263. [PMID: 39870322 DOI: 10.1016/j.ydbio.2025.01.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 12/20/2024] [Accepted: 01/23/2025] [Indexed: 01/29/2025]
Abstract
During lung development, the embryonic airway originates as a wishbone-shaped epithelial tube, which undergoes a series of branching events to build the bronchial tree. This process depends crucially on cell proliferation and is thought to involve distinct branching modes: lateral branching, wherein daughter branches emerge along the length of a parent branch, and bifurcations, wherein the tip of a parent branch splits to form two new daughter branches. The developing airway is fluid-filled, and previous studies have shown that altered luminal pressure can influence rates of branching morphogenesis. However, it is not clear if altered tissue mechanics influence patterns of proliferation along the embryonic airway epithelium nor if individual branching modes are affected differently by changes in luminal pressure. Here, we focused on mechanisms of lateral branching and used as a model system the embryonic avian lung, which forms exclusively via this branching mode during early development. We used microinjected fluid droplets or pharmacological modulators of fluid secretion to alter luminal fluid pressure either locally or globally within cultured embryonic lungs. Somewhat surprisingly, we found both local and global increases in luminal pressure to suppress the formation of new lateral branches while also promoting increased epithelial proliferation. In a consistent manner, decreased luminal pressure led to an increase in lateral branching morphogenesis. Morphometric analysis of airway branching patterns revealed that altered luminal pressure shifts the overall branching program, rather than simply changing rates of morphogenesis. Taken together, these results highlight the importance of mechanical forces during airway branching and suggest that different branching modes may be affected differently by luminal fluid pressure.
Collapse
Affiliation(s)
- Shelby R Mohr-Allen
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, USA; Department of Biomedical Engineering, UT Southwestern Medical Center, Dallas, TX, USA
| | - Jason P Gleghorn
- Department of Biomedical Engineering, University of Delaware, Newark, DE, 19716, USA
| | - Victor D Varner
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, USA; Department of Biomedical Engineering, UT Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
34
|
Liu OX, Lin LB, Bunk S, Chew T, Wu SK, Motegi F, Low BC. A ZO-2 scaffolding mechanism regulates the Hippo signalling pathway. FEBS J 2025; 292:1587-1601. [PMID: 39462647 DOI: 10.1111/febs.17304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 09/06/2024] [Accepted: 10/08/2024] [Indexed: 10/29/2024]
Abstract
Contact inhibition of proliferation is a critical cell density control mechanism governed by the Hippo signalling pathway. The biochemical signalling underlying cell density-dependent cues regulating Hippo signalling and its downstream effectors, YAP, remains poorly understood. Here, we reveal that the tight junction protein ZO-2 is required for the contact-mediated inhibition of proliferation. We additionally determined that the well-established molecular players of this process, namely Hippo kinase LATS1 and YAP, are regulated by ZO-2 and that the scaffolding function of ZO-2 promotes the interaction with and phosphorylation of YAP by LATS1. Mechanistically, YAP is phosphorylated when ZO-2 brings LATS1 and YAP together via its SH3 and PDZ domains, respectively, subsequently leading to the cytoplasmic retention and inactivation of YAP. In conclusion, we demonstrate that ZO-2 maintains Hippo signalling pathway activation by promoting the stability of LATS1 to inactivate YAP.
Collapse
Affiliation(s)
- Olivia Xuan Liu
- Mechanobiology Institute, National University of Singapore, Singapore
- Department of Biological Sciences, National University of Singapore, Singapore
| | | | - Soumya Bunk
- Mechanobiology Institute, National University of Singapore, Singapore
- Department of Biological Sciences, National University of Singapore, Singapore
| | - Tiweng Chew
- Mechanobiology Institute, National University of Singapore, Singapore
| | - Selwin K Wu
- Mechanobiology Institute, National University of Singapore, Singapore
- Department of Biological Sciences, National University of Singapore, Singapore
| | - Fumio Motegi
- Mechanobiology Institute, National University of Singapore, Singapore
- Department of Biological Sciences, National University of Singapore, Singapore
- Temasek Life-Sciences Laboratory, Singapore, Singapore
- Institute for Genetic Medicine, Hokkaido University, Japan
| | - Boon Chuan Low
- Mechanobiology Institute, National University of Singapore, Singapore
- Department of Biological Sciences, National University of Singapore, Singapore
- NUS College, National University of Singapore, Singapore
| |
Collapse
|
35
|
Zheng Y, Nützl M, Schackel T, Chen J, Weidner N, Müller R, Puttagunta R. Biomaterial scaffold stiffness influences the foreign body reaction, tissue stiffness, angiogenesis and neuroregeneration in spinal cord injury. Bioact Mater 2025; 46:134-149. [PMID: 39760066 PMCID: PMC11700269 DOI: 10.1016/j.bioactmat.2024.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 11/25/2024] [Accepted: 12/04/2024] [Indexed: 01/07/2025] Open
Abstract
Biomaterial scaffold engineering presents great potential in promoting axonal regrowth after spinal cord injury (SCI), yet persistent challenges remain, including the surrounding host foreign body reaction and improper host-implant integration. Recent advances in mechanobiology spark interest in optimizing the mechanical properties of biomaterial scaffolds to alleviate the foreign body reaction and facilitate seamless integration. The impact of scaffold stiffness on injured spinal cords has not been thoroughly investigated. Herein, we introduce stiffness-varied alginate anisotropic capillary hydrogel scaffolds implanted into adult rat C5 spinal cords post-lateral hemisection. Four weeks post-implantation, scaffolds with a stiffness approaching that of the spinal cord effectively minimize the host foreign body reaction via yes-associated protein (YAP) nuclear translocation. Concurrently, the softest scaffolds maximize cell infiltration and angiogenesis, fostering significant axonal regrowth but limiting the rostral-caudal linear growth. Furthermore, as measured by atomic force microscopy (AFM), the surrounding spinal cord softens when in contact with the stiffest scaffold while maintaining a physiological level in contact with the softest one. In conclusion, our findings underscore the pivotal role of stiffness in scaffold engineering for SCI in vivo, paving the way for the optimal development of efficacious biomaterial scaffolds for tissue engineering in the central nervous system.
Collapse
Affiliation(s)
- Yifeng Zheng
- Laboratory of Experimental Neuroregeneration, Spinal Cord Injury Center, Heidelberg University Hospital, 69118, Heidelberg, Germany
- Department of Neurosurgery, Neurosurgery Research Institute, the First Affiliated Hospital of Fujian Medical University, 350005, Fuzhou, China
| | - Maximilian Nützl
- Department of Physical and Theoretical Chemistry, University of Regensburg, 93053, Regensburg, Germany
| | - Thomas Schackel
- Laboratory of Experimental Neuroregeneration, Spinal Cord Injury Center, Heidelberg University Hospital, 69118, Heidelberg, Germany
| | - Jing Chen
- Laboratory of Experimental Neuroregeneration, Spinal Cord Injury Center, Heidelberg University Hospital, 69118, Heidelberg, Germany
| | - Norbert Weidner
- Spinal Cord Injury Center, Heidelberg University Hospital, 69118, Heidelberg, Germany
| | - Rainer Müller
- Department of Physical and Theoretical Chemistry, University of Regensburg, 93053, Regensburg, Germany
| | - Radhika Puttagunta
- Laboratory of Experimental Neuroregeneration, Spinal Cord Injury Center, Heidelberg University Hospital, 69118, Heidelberg, Germany
| |
Collapse
|
36
|
Rippe C, Bastrup JA, Holmberg J, Kawka K, Arévalo Martinez M, Albinsson S, Jepps TA, Swärd K. Declining activity of serum response factor in aging aorta in relation to aneurysm progression. J Biol Chem 2025; 301:108400. [PMID: 40081573 PMCID: PMC12002835 DOI: 10.1016/j.jbc.2025.108400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Accepted: 03/05/2025] [Indexed: 03/16/2025] Open
Abstract
Age is a critical determinant of arterial disease, including aneurysm formation. Here, to understand the impact of aging on the arterial transcriptome, we leveraged RNA-sequencing data to define transcripts that change with advancing age in human arteries. Among the most repressed transcripts in aged individuals were those that are relevant for actomyosin structure and organization, including both myosin light chain kinase (MYLK) and smooth muscle γ-actin (ACTG2). This was associated with a reduction of serum response factor (SRF), which controls these transcripts via defined promoter elements. To determine the consequences of isolated Srf depletion, we conditionally deleted Srf in vascular smooth muscle of young mice (i8-SRF-KO mice). This led to a reduction of the SRF regulon, including Mylk and Actg2, and impaired arterial contractility, but left endothelial-dependent dilatation unaffected. Srf-depletion also increased aortic diameter and Alcian blue staining of the aortic media, which are cardinal features of aortopathy, such as aortic aneurysmal disease. Despite this, i8-SRF-KO mice were protected from aortic lesions elicited by angiotensin II (AngII). Proteomics demonstrated that Srf-depletion mimicked a protein signature of AngII treatment involving increases of the mechanoresponsive transcriptional coactivators YAP and TAZ and reduction of the Hippo kinase Lats2. Protection from aortopathy could be overcome by changing the order of KO induction and AngII administration resulting in advanced aneurysms in both i8-SRF-KO and control mice. Our work provides important insights into the molecular underpinnings of age-dependent changes in aortic function and mechanisms of adaptation in hypertension.
Collapse
Affiliation(s)
- Catarina Rippe
- Vascular Physiology Environment, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Joakim Armstrong Bastrup
- Vascular Biology Group, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Johan Holmberg
- Vascular Physiology Environment, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Katarzyna Kawka
- Vascular Physiology Environment, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | | | - Sebastian Albinsson
- Vascular Physiology Environment, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Thomas A Jepps
- Vascular Biology Group, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Karl Swärd
- Vascular Physiology Environment, Department of Experimental Medical Science, Lund University, Lund, Sweden.
| |
Collapse
|
37
|
Ye W, Meng X, Xu S. [Research progress on collagen secretion mechanisms in scarring]. Zhejiang Da Xue Xue Bao Yi Xue Ban 2025; 54:266-278. [PMID: 40194913 PMCID: PMC12062945 DOI: 10.3724/zdxbyxb-2024-0535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 12/12/2024] [Accepted: 02/22/2025] [Indexed: 04/09/2025]
Abstract
Scar formation is characterized by dynamic alterations in collagen secretion, which critically determine scar morphology and pathological progression. In fibroblasts, collagen secretion is initiated through the activation of cytokine- and integrin-mediated signaling pathways, which promote collagen gene transcription. The procollagen polypeptide α chains undergo extensive post-translational modifications, including hydroxylation and glycosylation, within the endoplasmic reticulum (ER), followed by folding and assembly into triple-helical procollagen. Subsequent intracellular trafficking involves the sequential transport of procollagen through the ER, Golgi apparatus, and plasma membrane, accompanied by further structural refinements prior to extracellular secretion. Once secreted, procollagen is enzymatically processed to form mature collagen fibrils, which drive scar tissue remodeling. Recent advances in elucidating regulation of collagen secretion have identified pivotal molecular targets, such as transforming growth factor-beta 1 (TGF-β1), prolyl 4-hydroxylase (P4H), heat shock protein 47 (HSP47), and transport and Golgi organization protein 1 (TANGO1), providing novel therapeutic strategies to mitigate pathological scar hyperplasia and improve regenerative outcomes. This review provides a comprehensive analysis of the molecular mechanisms governing collagen secretion during scar formation, with emphasis on signaling cascades, procollagen biosynthesis, intracellular transport dynamics, and post-translational modifications, thereby offering a framework for developing targeted anti-scar therapies.
Collapse
Affiliation(s)
- Wenkai Ye
- Center of Stem Cell and Regenerative Medicine, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou 310058, China.
| | - Xinan Meng
- The Fourth Affiliated Hospital, Zhejiang University School of Medicine, International Institutes of Medicine, Zhejiang University, Center for Membrane Receptors and Brain Medicine, International School of Medicine, Zhejiang University, Yiwu 322000, Zhejiang Province, China
| | - Suhong Xu
- Center of Stem Cell and Regenerative Medicine, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou 310058, China.
| |
Collapse
|
38
|
Kamal KY, Trombetta-Lima M. Mechanotransduction and Skeletal Muscle Atrophy: The Interplay Between Focal Adhesions and Oxidative Stress. Int J Mol Sci 2025; 26:2802. [PMID: 40141444 PMCID: PMC11943188 DOI: 10.3390/ijms26062802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 03/15/2025] [Accepted: 03/18/2025] [Indexed: 03/28/2025] Open
Abstract
Mechanical unloading leads to profound musculoskeletal degeneration, muscle wasting, and weakness. Understanding the specific signaling pathways involved is essential for uncovering effective interventions. This review provides new perspectives on mechanotransduction pathways, focusing on the critical roles of focal adhesions (FAs) and oxidative stress in skeletal muscle atrophy under mechanical unloading. As pivotal mechanosensors, FAs integrate mechanical and biochemical signals to sustain muscle structural integrity. When disrupted, these complexes impair force transmission, activating proteolytic pathways (e.g., ubiquitin-proteasome system) that accelerate atrophy. Oxidative stress, driven by mitochondrial dysfunction and NADPH oxidase-2 (NOX2) hyperactivation, exacerbates muscle degeneration through excessive reactive oxygen species (ROS) production, impaired repair mechanisms, and dysregulated redox signaling. The interplay between FA dysfunction and oxidative stress underscores the complexity of muscle atrophy pathogenesis: FA destabilization heightens oxidative damage, while ROS overproduction further disrupts FA integrity, creating a self-amplifying vicious cycle. Therapeutic strategies, such as NOX2 inhibitors, mitochondrial-targeted antioxidants, and FAK-activating compounds, promise to mitigate muscle atrophy by preserving mechanotransduction signaling and restoring redox balance. By elucidating these pathways, this review advances the understanding of muscle degeneration during unloading and identifies promising synergistic therapeutic targets, emphasizing the need for combinatorial approaches to disrupt the FA-ROS feedback loop.
Collapse
Affiliation(s)
- Khaled Y. Kamal
- Department of Kinesiology, Iowa State University, Ames, IA 50011, USA
| | - Marina Trombetta-Lima
- Department of Pharmaceutical Technology and Biopharmacy, Groningen Research Institute of Pharmacy, University of Groningen, 9700 Groningen, The Netherlands;
| |
Collapse
|
39
|
Zhang Z, Liu X, Sha B, Zhang Y, Zhao L, Zhao G, Feng J, Zhang Y, Yang J, Wang Z, Xu F, Lu TJ, Lin M. Tunable Integrin-Ligand Coupling Strength Modulates Cellular Adaptive Mechanosensing. NANO LETTERS 2025; 25:4170-4179. [PMID: 40052581 DOI: 10.1021/acs.nanolett.4c05199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
Cells sense and respond to the matrix by exerting traction force through binding of integrins to an integrin-specific ligand. Here, Arg-Gly-Asp (RGD) peptide is covalently conjugated to the double-stranded DNA (dsDNA) and stem-loop DNA (slDNA) tethers with a tension tolerance of 43pN and immobilized on a PEG substrate. Unlike dsDNA, which is ruptured under high tension, leading to the removal of RGD, slDNA remains bound even when ruptured. Our results suggest that cells adapt their adhesion state by modulating actin filament polymerization and cofilin phosphorylation, effectively balancing the talin conformation to prevent dsDNA rupture and maintain normal adhesion. This phenomenon, termed integrin-ligand coupling strength, mediated cellular adaptive mechanosensing. Furthermore, we demonstrate that positive durotaxis can shift to negative durotaxis, depending on the integrin-ligand coupling strength. This study highlights the significance of the coupling strength in cell-extracellular matrix (ECM) interactions and offers new insights into designing biomaterials with tunable adhesive properties for cell-based applications.
Collapse
Affiliation(s)
- Zheng Zhang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, P.R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, P.R. China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, P.R. China
| | - Xiaoxi Liu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, P.R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, P.R. China
| | - Baoyong Sha
- School of Basic Medical Science, Xi'an Medical University, Xi'an 710021, P.R. China
| | - Yu Zhang
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, P.R. China
| | - Lingzhu Zhao
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, P.R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, P.R. China
| | - Guoqing Zhao
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, P.R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, P.R. China
| | - Jinteng Feng
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, P.R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, P.R. China
- Department of Thoracic Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, P.R. China
| | - Ying Zhang
- Xijing 986 Hospital Department, Fourth Military Medical University, Xi'an 710054, P.R. China
| | - Jin Yang
- Department of Medical Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Zheng Wang
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, P.R. China
- Pancreatic Disease Treatment Center, Department of Medical Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, P.R. China
| | - Feng Xu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, P.R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, P.R. China
| | - Tian Jian Lu
- State Key Laboratory of Mechanics and Control of Mechanical Structures, Nanjing University of Aeronautics and Astronautics, Nanjing 210016, P.R. China
| | - Min Lin
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, P.R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, P.R. China
| |
Collapse
|
40
|
Wang D, Silvani G, Schroeter L, Brynn R, Chou J, Poole K. The mechanosensitive channel ELKIN1 regulates cellular adaptations to simulated microgravity. NPJ Microgravity 2025; 11:10. [PMID: 40090965 PMCID: PMC11911437 DOI: 10.1038/s41526-025-00466-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 03/05/2025] [Indexed: 03/19/2025] Open
Abstract
In conditions of microgravity the human body undergoes extensive alterations in physiological function. However, it has proven challenging to determine how these changes are mediated at the molecular and cellular level. Here, we investigated whether ELKIN1, a mechanically activated ion channel, regulates changes in cellular and molecular structures in conditions of simulated microgravity. Deletion of ELKIN1 inhibited the simulated microgravity-induced alterations of cellular structure and attachment. In addition, cells lacking ELKIN1 did not exhibit changes in focal adhesion structures and redistribution of the YAP1 transcription factor in response to simulated microgravity, consistent with wild type cells. Finally, melanoma cell invasion of a collagen gel, from organotypic spheroids, was reduced in simulated microgravity, in an ELKIN1 dependent manner. Thus, the force sensing molecule, ELKIN1, modulates the impact of microgravity at both the molecular and cellular levels, revealing one of the molecular mechanisms that underpins cellular adaptations to conditions of microgravity.
Collapse
Affiliation(s)
- Daphne Wang
- School of Biomedical Sciences, Faculty of Medicine & Health, University of New South Wales, 2052, Sydney, NSW, Australia
- Eye Genetics Research Unit, Children's Medical Research Institute, The Children's Hospital at Westmead, Save Sight Institute, University of Sydney, Sydney, NSW, Australia
| | - Giulia Silvani
- School of Biomedical Engineering, Faculty of Engineering and Information Technology, University of Technology Sydney, Ultimo, NSW, Australia
- Laboratory for Advanced Biomaterials & Matrix Engineering, School of Chemistry and School of Materials Science and Engineering, University of New South Wales, 2052, Sydney, NSW, Australia
| | - Lioba Schroeter
- School of Biomedical Sciences, Faculty of Medicine & Health, University of New South Wales, 2052, Sydney, NSW, Australia
| | - Remi Brynn
- School of Biomedical Sciences, Faculty of Medicine & Health, University of New South Wales, 2052, Sydney, NSW, Australia
| | - Joshua Chou
- School of Biomedical Engineering, Faculty of Engineering and Information Technology, University of Technology Sydney, Ultimo, NSW, Australia
- EXPLOR Biologics, NSW, 2000, Sydney, Australia
| | - Kate Poole
- School of Biomedical Sciences, Faculty of Medicine & Health, University of New South Wales, 2052, Sydney, NSW, Australia.
| |
Collapse
|
41
|
Maupérin M, Sun Y, Glandorf T, Oswald TA, Klatt N, Geil B, Mutero-Maeda A, Méan I, Jond L, Janshoff A, Yan J, Citi S. A feedback circuitry involving γ-actin, β-actin and nonmuscle myosin-2 A controls tight junction and apical cortex mechanics. Nat Commun 2025; 16:2514. [PMID: 40082413 PMCID: PMC11906862 DOI: 10.1038/s41467-025-57428-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 02/21/2025] [Indexed: 03/16/2025] Open
Abstract
Cytoplasmic β- and γ-actin isoforms, along with non-muscle myosin 2 isoforms, are tightly regulated in epithelial cells and compose the actomyosin cytoskeleton at the apical junctional complex. However, their specific role in regulating the mechanics of the membrane cortex and the organization of junctions, and which biomechanical circuitries modulate their expression remain poorly understood. Here, we show that γ-actin depletion in MDCK and other epithelial cells results in increased expression and junctional accumulation of β-actin and increased tight junction membrane tortuosity, both dependent on nonmuscle myosin-2A upregulation. The knock-out of γ-actin also decreases apical membrane stiffness and increases dynamic exchange of the cytoplasmic tight junction proteins like ZO-1 and cingulin, without affecting tight junction organization and barrier function. In summary, our findings uncover a biomechanical circuitry linking γ-actin to β-actin expression through nonmuscle myosin-2A and reveal γ-actin as a key regulator of tight junction and apical membrane cortex mechanics, and the dynamics of cytoskeleton-associated tight junction proteins in epithelial cells.
Collapse
Affiliation(s)
- Marine Maupérin
- Department of Molecular and Cellular Biology, University of Geneva, Geneva, Switzerland
| | - Yuze Sun
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
| | - Thomas Glandorf
- Georg-August Universität, Institute for Physical Chemistry, Göttingen, Germany
| | - Tabea Anne Oswald
- Georg-August Universität, Institute for Organic and Biomolecular Chemistry, Göttingen, Germany
| | - Niklas Klatt
- Georg-August Universität, Institute for Physical Chemistry, Göttingen, Germany
| | - Burkhard Geil
- Georg-August Universität, Institute for Physical Chemistry, Göttingen, Germany
| | - Annick Mutero-Maeda
- Department of Molecular and Cellular Biology, University of Geneva, Geneva, Switzerland
| | - Isabelle Méan
- Department of Molecular and Cellular Biology, University of Geneva, Geneva, Switzerland
| | - Lionel Jond
- Department of Molecular and Cellular Biology, University of Geneva, Geneva, Switzerland
| | - Andreas Janshoff
- Georg-August Universität, Institute for Physical Chemistry, Göttingen, Germany
| | - Jie Yan
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
| | - Sandra Citi
- Department of Molecular and Cellular Biology, University of Geneva, Geneva, Switzerland.
| |
Collapse
|
42
|
Joshi H, Anaya E, Addanki A, Almgren-Bell A, Todd EM, Morley SC. Mechanosensitivity of macrophage polarization: comparing small molecule leukadherin-1 to substrate stiffness. Front Immunol 2025; 16:1420325. [PMID: 40114914 PMCID: PMC11922956 DOI: 10.3389/fimmu.2025.1420325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 02/21/2025] [Indexed: 03/22/2025] Open
Abstract
Macrophages sustain tissue homeostasis through host defense and wound repair. To promote host defense, macrophages upregulate surface markers associated with antigen processing and secrete pro-inflammatory mediators such as IL-6 and IL-1β. After pathogen clearance, macrophages shift phenotype to promote wound repair. Shifts in phenotypes are termed "polarization" and have historically been modeled by exposure to soluble mediators such as LPS+IFNγ (host defense) or IL-4+IL-13 (tissue repair). Greater emphasis is now being placed on understanding how the mechanical environment of macrophages, such as tissue compliance, regulates macrophages responses. Here, we compare incubation of primary macrophages on collagen-coated silica gels of varying stiffness to treatment with the small molecule integrin activator, leukadherin-1 (LA1), to examine how substrate stiffness alters macrophage polarization in response to multiple stimuli. LA1 was developed as an immunomodulator to treat inflammatory diseases by impairing trafficking of inflammatory cells. A recent clinical trial examining LA1 as an immunomodulator in solid tumors was terminated early because no benefit was observed. We hypothesized that LA1 treatment may exert additional, unexpected effects on macrophage polarization by replicating mechanotransduction. Specifically, we hypothesized that LA1 would mimic effects of incubation on stiffer substrates, as both conditions would be predicted to activate integrins. Our results show that soft substrate (0.2 kPa) trends towards upregulation of host defense molecules, in contrast to prior reports using different experimental systems. We further show that soft substrates enhance NLRP3-mediated IL-1β production, compared to stiff, in both primary mouse and human macrophages. LA1 mimicked incubation on stiff substrates in inhibiting NLRP3 activation and in regulating expression of several surface markers but differed by reducing IL-6 production. Our results show that macrophage inflammatory responses are regulated by adhesion-based, integrin-mediated mechanical signaling. Modulation of NLRP3-mediated IL-1β production by LA1 supports the possibility of repurposing LA1 to treat NLRP3-dependent inflammatory diseases.
Collapse
Affiliation(s)
- Hemant Joshi
- Division of Infectious Diseases, Department of Pediatrics, Washington School of Medicine in St. Louis, St. Louis, MO, United States
| | - Edgar Anaya
- Division of Infectious Diseases, Department of Pediatrics, Washington School of Medicine in St. Louis, St. Louis, MO, United States
| | - Anvitha Addanki
- Division of Infectious Diseases, Department of Pediatrics, Washington School of Medicine in St. Louis, St. Louis, MO, United States
| | - Alison Almgren-Bell
- Division of Infectious Diseases, Department of Pediatrics, Washington School of Medicine in St. Louis, St. Louis, MO, United States
| | - Elizabeth M. Todd
- Division of Infectious Diseases, Department of Pediatrics, Washington School of Medicine in St. Louis, St. Louis, MO, United States
| | - Sharon Celeste Morley
- Division of Infectious Diseases, Department of Pediatrics, Washington School of Medicine in St. Louis, St. Louis, MO, United States
- Division of Immunobiology, Department of Pathology and Immunology, Washington School of Medicine in St. Louis, St. Louis, MO, United States
| |
Collapse
|
43
|
Chen N, Zhang X, Yang P, He X. Inhibition of autophagy promotes ultrasound‑targeted microbubble destruction-induced apoptosis of pancreatic cancer cells. Int J Med Sci 2025; 22:1708-1719. [PMID: 40093805 PMCID: PMC11905268 DOI: 10.7150/ijms.106509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 02/21/2025] [Indexed: 03/19/2025] Open
Abstract
In therapeutic studies of pancreatic cancer, ultrasound-targeted microbubble destruction (UTMD) has shown potential in promoting apoptosis as a safe and non-invasive adjuvant therapy. Autophagy, a regulatory mechanism for cellular stress response and survival, plays a dual role in tumor development, progression, and treatment. However, the role of autophagy in UTMD-induced apoptosis in pancreatic cancer cells remains unclear. In this study, chloroquine (CQ), an autophagy inhibitor, was combined with UTMD to treat pancreatic cancer both in vitro and in vivo, with changes in apoptosis assessed through Western blot and TUNEL staining. The results showed that UTMD induced both apoptosis and autophagy in pancreatic cancer cells. Notably, inhibiting autophagy significantly enhanced UTMD-induced apoptosis, while the inhibition of apoptosis did not affect UTMD-induced autophagy. These findings suggest that autophagy reduces the effectiveness of UTMD in treating pancreatic cancer. This study offers a new perspective on UTMD for treating pancreatic cancer, suggesting that combining autophagy inhibitors could be a promising strategy to enhance the effectiveness of pancreatic cancer therapy.
Collapse
Affiliation(s)
- Nan Chen
- Department of Ultrasound, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Xiaoyu Zhang
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Ping Yang
- Department of Ultrasound, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Xuemei He
- Department of Ultrasound, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
44
|
Chen X, Ji X, Lao Z, Pan B, Qian Y, Yang W. Role of YAP/TAZ in bone diseases: A transductor from mechanics to biology. J Orthop Translat 2025; 51:13-23. [PMID: 39902099 PMCID: PMC11787699 DOI: 10.1016/j.jot.2024.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 10/24/2024] [Accepted: 12/09/2024] [Indexed: 02/05/2025] Open
Abstract
Wolff's Law and the Mechanostat Theory elucidate how bone tissues detect and convert mechanical stimuli into biological signals, crucial for maintaining bone equilibrium. Abnormal mechanics can lead to diseases such as osteoporosis, osteoarthritis, and nonunion fractures. However, the detailed molecular mechanisms by which mechanical cues are transformed into biological responses in bone remain underexplored. Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ), key regulators of bone homeostasis, are instrumental in this process. Emerging research highlights bone cells' ability to sense various mechanical stimuli and relay these signals intracellularly. YAP/TAZ are central in receiving these mechanical cues and converting them into signals that influence bone cell behavior. Abnormal YAP/TAZ activity is linked to several bone pathologies, positioning these proteins as promising targets for new treatments. Thus, this review aims to provide an in-depth examination of YAP/TAZ's critical role in the interpretation of mechanical stimuli to biological signals, with a special emphasis on their involvement in bone cell mechanosensing, mechanotransduction, and mechanoresponse. The translational potential of this article: Clinically, appropriate stress stimulation promotes fracture healing, while bed rest can lead to disuse osteoporosis and excessive stress can cause osteoarthritis or bone spurs. Recent advancements in the understanding of YAP/TAZ-mediated mechanobiological signal transduction in bone diseases have been significant, yet many aspects remain unknown. This systematic review summarizes current research progress, identifies unaddressed areas, and highlights potential future research directions. Advancements in this field facilitate a deeper understanding of the molecular mechanisms underlying bone mechanics regulation and underscore the potential of YAP/TAZ as therapeutic targets for bone diseases such as fractures, osteoporosis, and osteoarthritis.
Collapse
Affiliation(s)
- Xin Chen
- Department of Orthopedics Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, 310006, China
| | - Xing Ji
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, Department of Clinical Medicine, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
| | - Zhaobai Lao
- Department of Orthopedics Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, 310006, China
| | - Bin Pan
- Department of Orthopedics Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, 310006, China
| | - Yu Qian
- Department of Orthopedics Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, 310006, China
| | - Wanlei Yang
- Department of Orthopedics Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, 310006, China
| |
Collapse
|
45
|
Lv H, Xu X, Wu Z, Lin Y, Liu Y, Liu M, Xu L, Wang X, Sun N, Abdel-Shafy H, Abdelrahman M, Alsaegh AA, Ahmed AE, Yang L, Hua G. Yes-associated protein 1 is essential for maintaining lactation via regulating mammary epithelial cell dynamics and secretion capacity. Int J Biol Macromol 2025; 293:139290. [PMID: 39743110 DOI: 10.1016/j.ijbiomac.2024.139290] [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: 10/31/2024] [Revised: 12/08/2024] [Accepted: 12/27/2024] [Indexed: 01/04/2025]
Abstract
Understanding the physiology and molecular mechanisms of lactogenesis is crucial for enhancing mammalian milk production. Yes-associated protein 1 (YAP1) regulated mammary epithelial cell survival during pregnancy, but its role in lactation maintenance remains unclear. We found that YAP1 was highly expressed in mammary gland across specie, with elevated expression levels during murine gestation and lactation, particularly localized in alveoli epithelial cells. In vivo administration of a YAP1 inhibitor impaired murine milk yield, mammary gland weight, alveolar structure, and mammary epithelial cell dynamics. In vitro, YAP1 positively affected mammary epithelial cell growth and the synthesis of triglyceride and α-casein. Notably, the primary lactogenesis hormone Prolactin induced cell growth and triglyceride secretion while enhancing YAP1 expression and activity. In contrast, Melatonin inhibited cell growth and triglyceride synthesis, decreasing YAP1 expression and activity. YAP1 knockdown compromised prolactin induced effects, whereas YAP1 overexpression partially rescued cell functions inhibited by melatonin. Finally, Bioinformatics analyses revealed that YAP1 regulated multiple biological processes related to lactogenesis, including cell cycle, apoptosis, endoplasmic reticulum, amino acid transport and biosynthesis, etc. These finding indicated that YAP1 is essential for mammary epithelial cells growth and secretion and played an essential role in the lactating endocrine network by mediating key hormone functions.
Collapse
Affiliation(s)
- Haimiao Lv
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518120, China; Shenzhen Institute of Nutrition and Health, Huazhong Agricultural University, Shenzhen 518038, China
| | - Xiaoling Xu
- Laboratory of Animal Reproduction, Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, BJ, China
| | - Zihui Wu
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Yuxin Lin
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Yan Liu
- Laboratory of Animal Reproduction, Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, BJ, China
| | - Miaoyu Liu
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Linghua Xu
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Xiaojie Wang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Nan Sun
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Hamdy Abdel-Shafy
- Department of Animal Production, Faculty of Agriculture, Cairo University, El-Gamma Street, 12613 Giza, Egypt
| | - Mohamed Abdelrahman
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; Animal Production Department, Faculty of Agriculture, Assuit University, Asyut, Egypt
| | - Aiman A Alsaegh
- Department of Laboratory Medicine, Faculty of Applied Medical Sciences, Umm Al-Qura University, Saudi Arabia
| | - Ahmed Ezzat Ahmed
- Department of Biology, College of Science, King Khalid University, P.O. Box 9004, 61413 Abha, Saudi Arabia; Department of Theriogenology, Faculty of Veterinary Medicine, South Valley University, 83523 Qena, Egypt
| | - Liguo Yang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; National Center for International Research on Animal Genetics, Breeding and Reproduction, Frontiers Science Center for Animal Breeding and Sustainable Production, Huazhong Agricultural University, Wuhan 430070, China
| | - Guohua Hua
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518120, China; Shenzhen Institute of Nutrition and Health, Huazhong Agricultural University, Shenzhen 518038, China; National Center for International Research on Animal Genetics, Breeding and Reproduction, Frontiers Science Center for Animal Breeding and Sustainable Production, Huazhong Agricultural University, Wuhan 430070, China.
| |
Collapse
|
46
|
Zhong B, Du J, Liu F, Sun S. The Role of Yes-Associated Protein in Inflammatory Diseases and Cancer. MedComm (Beijing) 2025; 6:e70128. [PMID: 40066231 PMCID: PMC11892025 DOI: 10.1002/mco2.70128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 02/02/2025] [Accepted: 02/11/2025] [Indexed: 03/17/2025] Open
Abstract
Yes-associated protein (YAP) plays a central role in the Hippo pathway, primarily governing cell proliferation, differentiation, and apoptosis. Its significance extends to tumorigenesis and inflammatory conditions, impacting disease initiation and progression. Given the increasing relevance of YAP in inflammatory disorders and cancer, this study aims to elucidate its pathological regulatory functions in these contexts. Specifically, we aim to investigate the involvement and molecular mechanisms of YAP in various inflammatory diseases and cancers. We particularly focus on how YAP activation, whether through Hippo-dependent or independent pathways, triggers the release of inflammation and inflammatory mediators in respiratory, cardiovascular, and digestive inflammatory conditions. In cancer, YAP not only promotes tumor cell proliferation and differentiation but also modulates the tumor immune microenvironment, thereby fostering tumor metastasis and progression. Additionally, we provide an overview of current YAP-targeted therapies. By emphasizing YAP's role in inflammatory diseases and cancer, this study aims to enhance our understanding of the protein's pivotal involvement in disease processes, elucidate the intricate pathological mechanisms of related diseases, and contribute to future drug development strategies targeting YAP.
Collapse
Affiliation(s)
- Bing Zhong
- Department of Otolaryngology‐Head and Neck SurgeryWest China HospitalSichuan UniversityChengduSichuanChina
| | - Jintao Du
- Department of Otolaryngology‐Head and Neck SurgeryWest China HospitalSichuan UniversityChengduSichuanChina
| | - Feng Liu
- Department of Otolaryngology‐Head and Neck SurgeryWest China HospitalSichuan UniversityChengduSichuanChina
| | - Silu Sun
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesChinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and ManagementWest China Hospital of StomatologySichuan UniversityChengduSichuanChina
| |
Collapse
|
47
|
Zhao L, Gui Y, Deng X. Focus on mechano-immunology: new direction in cancer treatment. Int J Surg 2025; 111:2590-2602. [PMID: 39764598 DOI: 10.1097/js9.0000000000002224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 11/29/2024] [Indexed: 03/16/2025]
Abstract
The immune response is modulated by a diverse array of signals within the tissue microenvironment, encompassing biochemical factors, mechanical forces, and pressures from adjacent tissues. Furthermore, the extracellular matrix and its constituents significantly influence the function of immune cells. In the case of carcinogenesis, changes in the biophysical properties of tissues can impact the mechanical signals received by immune cells, and these signals c1an be translated into biochemical signals through mechano-transduction pathways. These mechano-transduction pathways have a profound impact on cellular functions, influencing processes such as cell activation, metabolism, proliferation, and migration, etc. Tissue mechanics may undergo temporal changes during the process of carcinogenesis, offering the potential for novel dynamic levels of immune regulation. Here, we review advances in mechanoimmunology in malignancy studies, focusing on how mechanosignals modulate the behaviors of immune cells at the tissue level, thereby triggering an immune response that ultimately influences the development and progression of malignant tumors. Additionally, we have also focused on the development of mechano-immunoengineering systems, with the help of which could help to further understand the response of tumor cells or immune cells to alterations in the microenvironment and may provide new research directions for overcoming immunotherapeutic resistance of malignant tumors.
Collapse
Affiliation(s)
- Lin Zhao
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Clinical Medical Research Center for Cancer Pathogenic Genes Testing and Diagnosis, Changsha, Human, China
| | - Yajun Gui
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Clinical Medical Research Center for Cancer Pathogenic Genes Testing and Diagnosis, Changsha, Human, China
| | - Xiangying Deng
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Clinical Medical Research Center for Cancer Pathogenic Genes Testing and Diagnosis, Changsha, Human, China
- Institute of Medical Sciences, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
48
|
Jiu J, Liu H, Li D, Li X, Zhang J, Yan L, Fan Z, Li S, Du G, Li JJ, Wu A, Liu W, Du Y, Zhao B, Wang B. 3D Mechanical Response Stem Cell Complex Repairs Spinal Cord Injury by Promoting Neurogenesis and Regulating Tissue Homeostasis. Adv Healthc Mater 2025; 14:e2404925. [PMID: 39853962 DOI: 10.1002/adhm.202404925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Indexed: 01/26/2025]
Abstract
Spinal cord injury (SCI) leads to acute tissue damage that disrupts the microenvironmental homeostasis of the spinal cord, inhibiting cell survival and function, and thereby undermining treatment efficacy. Traditional stem cell therapies have limited success in SCI, due to the difficulties in maintaining cell survival and inducing sustained differentiation into neural lineages. A new solution may arise from controlling the fate of stem cells by creating an appropriate mechanical microenvironment. In this study, mechanical response stem cell complex (MRSCC) is created as an innovative therapeutic strategy for SCI, utilizing 3D bioprinting technology and gelatin microcarriers (GM) loaded with mesenchymal stem cells (MSCs). GM creates an optimal microenvironment for MSCs growth and paracrine activity. Meanwhile, 3D bioprinting allows accurate control of spatial pore architecture and mechanical characteristics of the cell construct to encourage neuroregeneration. The mechanical microenvironment created by MRSCC is found to activate the Piezo1 channel and prevent excessive nuclear translocation of YAP, thereby increasing neural-related gene expression in MSCs. Transplanting MRSCC in rats with spinal cord injuries boosts sensory and motor recovery, reduces inflammation, and stimulates the regeneration of neurons and glial cells. The MRSCC offers a new tissue engineering solution that can promote spinal cord repair.
Collapse
Affiliation(s)
- Jingwei Jiu
- Department of Orthopaedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
- Department of Orthopedics, The Second Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Haifeng Liu
- Department of Orthopaedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
- Department of Orthopedics, The Second Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Dijun Li
- Department of Orthopedics, Affiliated Renhe Hospital of China Three Gorges University, Yichang, 443000, China
| | - Xiaoke Li
- Department of Orthopedics, The Second Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Jing Zhang
- Department of Emergency Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550001, China
| | - Lei Yan
- Department of Orthopedics, The Second Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Zijuan Fan
- Department of Health Statistics, School of Public Health, Shanxi Medical University, Taiyuan, 030001, China
| | - Songyan Li
- Department of Orthopaedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Guangyuan Du
- Department of Orthopaedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Jiao Jiao Li
- School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - Aimin Wu
- Department of Orthopaedics, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Wei Liu
- Development of Research, Beijing Hua Niche Biotechnology Co., LTD, Beijing, 100084, China
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Yanan Du
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Bin Zhao
- Department of Orthopedics, The Second Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Bin Wang
- Department of Orthopaedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| |
Collapse
|
49
|
Li R, Wang J, Lin Q, Yin Z, Zhou F, Chen X, Tan H, Su J. Mechano-Responsive Biomaterials for Bone Organoid Construction. Adv Healthc Mater 2025; 14:e2404345. [PMID: 39740101 DOI: 10.1002/adhm.202404345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/08/2024] [Indexed: 01/02/2025]
Abstract
Mechanical force is essential for bone development, bone homeostasis, and bone fracture healing. In the past few decades, various biomaterials have been developed to provide mechanical signals that mimic the natural bone microenvironment, thereby promoting bone regeneration. Bone organoids, emerging as a novel research approach, are 3D micro-bone tissues that possess the ability to self-renew and self-organize, exhibiting biomimetic spatial characteristics. Incorporating mechano-responsive biomaterials in the construction of bone organoids presents a promising avenue for simulating the mechanical bone microenvironment. Therefore, this review commences by elucidating the impact of mechanical force on bone health, encompassing both cellular interactions and alterations in bone structure. Furthermore, the most recent applications of mechano-responsive biomaterials within the realm of bone tissue engineering are highlighted. Three different types of mechano-responsive biomaterials are introduced with a focus on their responsive mechanisms, construction strategies, and efficacy in facilitating bone regeneration. Based on a comprehensive overview, the prospective utilization and future challenges of mechano-responsive biomaterials in the construction of bone organoids are discussed. As bone organoid technology advances, these biomaterials are poised to become powerful tools in bone regeneration.
Collapse
Affiliation(s)
- Ruiyang Li
- Department of Orthopedics, Trauma Orthopedics Center, Institute of Musculoskeletal Injury and Translational Medicine of Organoids, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, P. R. China
- Institute of Translational Medicine, National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai, 200444, P. R. China
| | - Jian Wang
- Department of Orthopedics, Trauma Orthopedics Center, Institute of Musculoskeletal Injury and Translational Medicine of Organoids, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, P. R. China
- Institute of Translational Medicine, National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai, 200444, P. R. China
| | - Qiushui Lin
- Department of Spine Surgery, First Affiliated Hospital of Naval Medical University, Shanghai, 200433, P. R. China
| | - Zhifeng Yin
- Department of Orthopedics, Shanghai Zhongye Hospital, Shanghai, 200941, P. R. China
| | - Fengjin Zhou
- Department of Orthopedics, Honghui Hospital, Xi'an Jiao Tong University, Xi'an, 710000, P. R. China
| | - Xiao Chen
- Department of Orthopedics, Trauma Orthopedics Center, Institute of Musculoskeletal Injury and Translational Medicine of Organoids, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, P. R. China
| | - Hongbo Tan
- Department of Orthopedics, The 920th Hospital of Joint Logistics Support Force, Yunnan, 650020, P. R. China
| | - Jiacan Su
- Department of Orthopedics, Trauma Orthopedics Center, Institute of Musculoskeletal Injury and Translational Medicine of Organoids, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, P. R. China
- Institute of Translational Medicine, National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai, 200444, P. R. China
| |
Collapse
|
50
|
Leng Y, Wang W, Lu J, Chen J, Chen X, Li Y, Wang J, Liu Y, Tan Q, Yang W, Jiang Y, Huang P, Cai J, Yuan H, Weng L, Xu Q, Lu Y. Endothelial TRIM35-Regulated MMP10 Release Exacerbates Calcification of Vascular Grafts. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2409641. [PMID: 39865905 PMCID: PMC11923891 DOI: 10.1002/advs.202409641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 12/28/2024] [Indexed: 01/28/2025]
Abstract
Vascular calcification is a highly regulated process in cardiovascular disease (CVD) and is strongly correlated with morbidity and mortality, especially in the adverse stage of vascular remodeling after coronary artery bypass graft surgery (CABG). However, the pathogenesis of vascular graft calcification, particularly the role of endothelial-smooth muscle cell interaction, is still unclear. To test how ECs interact with SMCs in artery grafts, single-cell analysis of wild-type mice is first performed using an arterial isograft mouse model and found robust cytokine-mediated signaling pathway activation and SMC proliferation, together with upregulated endothelial tripartite motif 35 (TRIM35) expression. Unexpectedly, severe SMC calcification in artery grafts is found in TRIM35 conditional endothelial knockout (cKO) mice. Calcified medium (comprising calcium chloride and beta-glycerophosphate)-induced calcium deposition in vitro is also found in SMCs cocultured with TRIM35 knockout endothelium. This extraordinary phenomenon is further confirmed to be induced by increased MMP10 secretion. Mechanistically, endothelial TRIM35 inhibits MMP10 expression and secretion by promoting K63-linked ubiquitination of RelB and maintaining its nuclear localization, consequently inhibiting nuclear transcription of MMP10 through the noncanonical NF-κB signaling pathway. Targeting MMP10 in situ in arterial isografts can effectively alleviate vascular calcification caused by conditional endothelial TRIM35 knockout. These findings demonstrated that TRIM35 inhibited vascular calcification during arterial isograft remodeling, a process that is driven by the aberrant secretion of endothelial MMP10. Targeting MMP10 pathway may be a potential therapeutic strategy for vascular calcification in vessel grafts.
Collapse
Affiliation(s)
- Yiming Leng
- Clinical Research CenterPostdoctoral Station of Clinical MedicineThe Third Xiangya HospitalCentral South UniversityChangsha410013P. R. China
| | - Wei Wang
- Clinical Research CenterPostdoctoral Station of Clinical MedicineThe Third Xiangya HospitalCentral South UniversityChangsha410013P. R. China
| | - Jun Lu
- Department of Laboratory MedicineThe Third Xiangya HospitalCentral South UniversityChangsha410013P. R. China
| | - Jingyuan Chen
- Clinical Research CenterPostdoctoral Station of Clinical MedicineThe Third Xiangya HospitalCentral South UniversityChangsha410013P. R. China
| | - Xuliang Chen
- Department of Cardiovascular SurgeryXiangya HospitalCentral South UniversityChangsha410028P. R. China
| | - Yalan Li
- Clinical Research CenterPostdoctoral Station of Clinical MedicineThe Third Xiangya HospitalCentral South UniversityChangsha410013P. R. China
| | - Jie Wang
- Clinical Research CenterPostdoctoral Station of Clinical MedicineThe Third Xiangya HospitalCentral South UniversityChangsha410013P. R. China
| | - Yuanyuan Liu
- Clinical Research CenterPostdoctoral Station of Clinical MedicineThe Third Xiangya HospitalCentral South UniversityChangsha410013P. R. China
| | - Qian Tan
- Clinical Research CenterPostdoctoral Station of Clinical MedicineThe Third Xiangya HospitalCentral South UniversityChangsha410013P. R. China
| | - Wenjing Yang
- Clinical Research CenterPostdoctoral Station of Clinical MedicineThe Third Xiangya HospitalCentral South UniversityChangsha410013P. R. China
| | - Youxiang Jiang
- Clinical Research CenterPostdoctoral Station of Clinical MedicineThe Third Xiangya HospitalCentral South UniversityChangsha410013P. R. China
| | - Peiyuan Huang
- MRC Integrative Epidemiology Unit (IEU)Bristol Medical SchoolUniversity of BristolOakfield House, Oakfield GroveBristolBS8 2BNUK
| | - Jingjing Cai
- Clinical Research CenterPostdoctoral Station of Clinical MedicineThe Third Xiangya HospitalCentral South UniversityChangsha410013P. R. China
| | - Hong Yuan
- Clinical Research CenterPostdoctoral Station of Clinical MedicineThe Third Xiangya HospitalCentral South UniversityChangsha410013P. R. China
| | - Liang Weng
- Department of PathologySchool of Basic Medical SciencesPeking University Third HospitalPeking University Health Science CenterBeijing100083P. R. China
| | - Qingbo Xu
- Department of Cardiology, the First Affiliated HospitalSchool of MedicineZhejiang UniversityHangzhou310058P. R. China
| | - Yao Lu
- Clinical Research CenterPostdoctoral Station of Clinical MedicineThe Third Xiangya HospitalCentral South UniversityChangsha410013P. R. China
- Life Sciences & MedicineKing's College LondonLondonSE1 8WAUK
| |
Collapse
|