1
|
Li X, Su K, Zhao L, Zhang H, Yang Q, Du P, Chen X, Pan H. Unraveling osteogenesis mechanisms of the empowered VitaFlux adaptive regeneration biomaterials for bone tissue engineering: Insights into the role of BBGs/BSBGs. Bioact Mater 2025; 49:271-290. [PMID: 40130079 PMCID: PMC11932761 DOI: 10.1016/j.bioactmat.2025.03.006] [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: 11/09/2024] [Revised: 02/07/2025] [Accepted: 03/06/2025] [Indexed: 03/26/2025] Open
Abstract
Bone tissue engineering materials are crucial for bone repair, but existing repair materials still face many challenges, including poor biocompatibility and bioactivity, slow self-repair processes, limited adaptability, inability to promote angiogenesis and so on. To address these issues, the development of third-generation bone repair materials, which are being designed to stimulate specific cellular responses at the molecular level, such as borate and borosilicate bioactive glasses (BBGs/BSBGs) that activate cells and genes, offers new potential for promoting bone tissue self-renewing. Their unique characteristic lies in a flow of life-giving energy, releasing beneficial ions such as boron, calcium and silicon to stimulate cell proliferation and differentiation, accelerating the regeneration of bones. Through this dynamic repair mechanism, these VitaFlux glasses operate like a "living system" within the body, not only speeding up the healing of damaged tissues but also interacting seamlessly with surrounding tissues during the repair process. In this review, we provide a comprehensive analysis of the current understanding of the osteogenesis mechanisms of BBGs/BSBGs, emphasizing their interactions with cells, including ion release and exchange, protein adsorption, and cell adhesion. We also examine key osteogenic signaling pathways related to the alkaline and ionic microenvironments of BBGs/BSBGs, such as the cell cycle, Wnt, MAPK, and BMP signaling pathways, along with macrophage polarization and angiogenesis. Additionally, strategies and future prospects for advancing BBGs/BSBGs research are discussed. Special attention is given to the NaBC1 and GPCR-mediated signaling pathways, which require further investigation.
Collapse
Affiliation(s)
- Xian Li
- Shenzhen Key Laboratory of Marine Biomedical Materials, CAS-HK Joint Lab of Biomaterials, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, PR China
| | - Kun Su
- Shenzhen Key Laboratory of Marine Biomedical Materials, CAS-HK Joint Lab of Biomaterials, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, PR China
| | - Limin Zhao
- Shenzhen Healthemes Biotechnology Co. Ltd, Shenzhen, 518102, PR China
- Geriatric Medicine Department and General Medicine Department, Shenzhen Longhua District Central Hospital, Shenzhen, 518000, PR China
| | - Hao Zhang
- Shenzhen Key Laboratory of Marine Biomedical Materials, CAS-HK Joint Lab of Biomaterials, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, PR China
| | - Qiang Yang
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, 300211, PR China
| | - Ping Du
- Shenzhen Key Laboratory of Marine Biomedical Materials, CAS-HK Joint Lab of Biomaterials, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, PR China
| | - Xiaofeng Chen
- National Engineering Research Center for Tissue Restoration and Reconstruction, Guangzhou, 510006, PR China
- Department of Biomedical Engineering, School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510641, PR China
- School of Medicine, Foshan University, Foshan, 528000, PR China
| | - Haobo Pan
- Shenzhen Key Laboratory of Marine Biomedical Materials, CAS-HK Joint Lab of Biomaterials, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, PR China
- Shenzhen Healthemes Biotechnology Co. Ltd, Shenzhen, 518102, PR China
| |
Collapse
|
2
|
Kim K, Min S, Thangam R, Tag KR, Lee HJ, Heo J, Jung H, Swe TT, Zare I, Song G, Najafabadi AH, Lee J, Jung HD, Kim JS, Hur S, Song HC, Park SG, Zhang K, Zhao P, Bian L, Kim SH, Yoon J, Ahn JP, Kim HK, Kang H. Dynamic hierarchical ligand anisotropy for competing macrophage regulation in vivo. Bioact Mater 2025; 47:121-135. [PMID: 39897585 PMCID: PMC11787691 DOI: 10.1016/j.bioactmat.2025.01.009] [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: 11/02/2024] [Revised: 12/30/2024] [Accepted: 01/10/2025] [Indexed: 02/04/2025] Open
Abstract
Diverse connective tissues exhibit hierarchical anisotropic structures that intricately regulate homeostasis and tissue functions for dynamic immune response modulation. In this study, remotely manipulable hierarchical nanostructures are tailored to exhibit multi-scale ligand anisotropy. Hierarchical nanostructure construction involves coupling liganded nanoscale isotropic/anisotropic Au (comparable to few integrin molecules-scale) to the surface of microscale isotropic/anisotropic magnetic Fe3O4 (comparable to integrin cluster-scale) and then elastically tethering them to a substrate. Systematic independent tailoring of nanoscale or microscale ligand isotropy versus anisotropy in four different hierarchical nanostructures with constant liganded surface area demonstrates similar levels of integrin molecule bridging and macrophage adhesion on the nanoscale ligand isotropy versus anisotropy. Conversely, the levels of integrin cluster bridging across hierarchical nanostructures and macrophage adhesion are significantly promoted by microscale ligand anisotropy compared with microscale ligand isotropy. Furthermore, microscale ligand anisotropy dominantly activates the host macrophage adhesion and pro-regenerative M2 polarization in vivo over the nanoscale ligand anisotropy, which can be cyclically reversed by substrate-proximate versus substrate-distant magnetic manipulation. This unprecedented scale-specific regulation of cells can be diversified by unlimited tuning of the scale, anisotropy, dimension, shape, and magnetism of hierarchical structures to decipher scale-specific dynamic cell-material interactions to advance immunoengineering strategies.
Collapse
Affiliation(s)
- Kanghyeon Kim
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Sunhong Min
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Ramar Thangam
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Kyong-Ryol Tag
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
- Advanced Analysis Center, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Hyun-Jeong Lee
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
- Advanced Analysis Center, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Jeongyun Heo
- Center for Theragnosis, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Hwapyung Jung
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Thet Thet Swe
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Iman Zare
- Research and Development Department, Sina Medical Biochemistry Technologies Co., Ltd., Shiraz, 7178795844, Iran
| | - Guosheng Song
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, China
| | | | - Junmin Lee
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), Pohang, 790-784, Republic of Korea
| | - Hyun-Do Jung
- Division of Materials Science and Engineering, Hanyang University, Seoul, 04763, Republic of Korea
| | - Jong Seung Kim
- Department of Chemistry, Korea University, Seoul, 02841, Republic of Korea
| | - Sunghoon Hur
- Electronic Materials Research Center, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- KHU-KIST Department of Converging Science and Technology, Kyung Hee University, Yongin, 17104, Republic of Korea
| | - Hyun-Cheol Song
- Electronic Materials Research Center, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- School of Advanced Materials Science and Engineering, Sungkyunkwan University (SKKU), Suwon, 16419, Republic of Korea
- KIST-SKKU Carbon-Neutral Research Center, Sungkyunkwan University (SKKU), Suwon, 16419, Republic of Korea
| | - Sung-Gyu Park
- Department of Nano-Bio Convergence, Korea Institute of Materials Science (KIMS), Changwon, Gyeongnam, 51508, Republic of Korea
- Department of Future Convergence Materials, Korea University, Seoul, 02841, Republic of Korea
| | - Kunyu Zhang
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, China
| | - Pengchao Zhao
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, China
| | - Liming Bian
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, China
| | - Se Hoon Kim
- Center for Theragnosis, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
| | - Juyoung Yoon
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul, 03760, Republic of Korea
- Graduate Program in Innovative Biomaterials Convergence, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Jae-Pyoung Ahn
- Advanced Analysis Center, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Hong-Kyu Kim
- Advanced Analysis Center, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Heemin Kang
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
- Department of Future Convergence Materials, Korea University, Seoul, 02841, Republic of Korea
- College of Medicine, Korea University, Seoul, 02841, Republic of Korea
| |
Collapse
|
3
|
Sun Q, Qiu T, Liu X, Wei Q. Cellular Spatial Sensing Determines Cell Mechanotransduction Activity on the Aligned Nanofibers. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2410351. [PMID: 39967397 DOI: 10.1002/smll.202410351] [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: 11/03/2024] [Revised: 02/06/2025] [Indexed: 02/20/2025]
Abstract
The geometric properties of extracellular matrix (ECM) fibers play a crucial role in regulating cellular behaviors and functions. Although extensive research has examined the effects of fiber alignment, conflicting results have often arisen, leaving the precise mechanisms by which electrospun fiber alignment affects cellular behavior still unclear. This study investigates how the arrangement of polycaprolactone (PCL) electrospun fiber substrates affects cellular mechanosensing by modulating cell positioning. Larger cells, whose width on a coverslip exceeds 5 times the width of the aligned fiber gaps (≈8 µm in this study) and that span multiple aligned fibers, demonstrate enhanced spreading and mechanotransduction. Conversely, smaller cells, whose width is less than or equal to 2.5 times the width of the aligned fiber gaps and are confined within fiber interstices, exhibit limited mechanotransductive signaling. These findings are further supported by manipulating cell size and, more importantly, have led to the fabrication of semi-aligned fiber networks that enhance both cell spreading and mechanotransduction. This research emphasizes the importance of optimizing fiber architecture to improve cellular interactions, offering valuable insights for the design of biomimetic scaffolds in tissue regeneration.
Collapse
Affiliation(s)
- Qian Sun
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials and Engineering, Sichuan University, Chengdu, 610065, P. R. China
| | - Tiecheng Qiu
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials and Engineering, Sichuan University, Chengdu, 610065, P. R. China
| | - Xiaojing Liu
- Department of Pediatric Dentistry, School and Hospital of Stomatology, Cheeloo College of Medicine, and Shandong Key Laboratory of Oral Tissue Regeneration, Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration, and Shandong Provincial Clinical Research Center for Oral Diseases, Shandong University, Jinan, 250012, P. R. China
| | - Qiang Wei
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials and Engineering, Sichuan University, Chengdu, 610065, P. R. China
| |
Collapse
|
4
|
Yang Q, Chen A, Zhang X, Wu Z, Zhang C. Functional poly(ether-ketone-ketone) composite scaffold with enhanced cell-material interaction, anti-inflammatory and osteogenesis for facilitating osteointegration and bone regeneration. Mater Today Bio 2025; 31:101533. [PMID: 39974817 PMCID: PMC11835654 DOI: 10.1016/j.mtbio.2025.101533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 01/27/2025] [Accepted: 01/28/2025] [Indexed: 02/21/2025] Open
Abstract
Bone defects resulting from trauma or disease remain a significant challenge in clinical practice, often requiring prolonged treatment. Poly(ether-ketone-ketone) (PEKK) is a commonly used implant material due to its excellent biocompatibility and mechanical properties, which are similar to those of bone. However, its biological inertness leads to poor anti-inflammatory and osteointegration properties, significantly hindering the bone repair process. In this study, a cryogel filled - PEKK/bioglass (BG) composite scaffold (SPBC) was prepared via 3D printing to provide immunomodulatory and bone integration performance. Compared with untreated PEKK, SPBC exhibited significant enhancements in surface properties, including higher hydrophilicity and roughness. Additionally, SPBC enhanced the adsorption of fibronectin and vitronectin on the scaffold surface and regulated the maturation of cytoskeleton and adhesion plaques by increasing the phosphorylation level of FAK at Y397, thereby promoting cell adhesion and spreading. Due to the release of bioactive ions, SPBC can significantly promote the polarization of RAW264.7 cells towards M2 and the secretion of anti-inflammatory cytokines, while also enhancing the proliferation and differentiation of rat mesenchymal stem cells (rMSCs) in vitro. Furthermore, the in vivo results confirmed the enhanced anti-inflammatory properties and the integration of SPBC with the host tissue. In summary, after surface modification and cryogel filling, SPBC demonstrated excellent anti-inflammatory and bone integration abilities, presenting potential for clinical application as an orthopedic implant scaffold.
Collapse
Affiliation(s)
- Qianwen Yang
- School of Biomedical Engineering, Shenzhen Campus, Sun Yat-Sen University, Shenzhen, Guangdong, 518107, China
| | - Anbei Chen
- School of Biomedical Engineering, Shenzhen Campus, Sun Yat-Sen University, Shenzhen, Guangdong, 518107, China
| | - Xin Zhang
- School of Biomedical Engineering, Shenzhen Campus, Sun Yat-Sen University, Shenzhen, Guangdong, 518107, China
| | - Zhaoying Wu
- School of Biomedical Engineering, Shenzhen Campus, Sun Yat-Sen University, Shenzhen, Guangdong, 518107, China
| | - Chao Zhang
- School of Biomedical Engineering, Shenzhen Campus, Sun Yat-Sen University, Shenzhen, Guangdong, 518107, China
| |
Collapse
|
5
|
Park YS, Choi Y, Lee JS. Focal adhesion dynamics-mediated cell migration and proliferation on silica bead arrays. Biomater Sci 2025; 13:1849-1857. [PMID: 40012335 DOI: 10.1039/d4bm01659a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2025]
Abstract
Interactions between cells and the extracellular matrix (ECM) alter cellular behaviors, including adhesion, migration, proliferation, and differentiation via focal adhesions that link the ECM to the actin cytoskeleton as an intracellular signaling pathway. Although nanomaterials with various mechanical, geometrical, and topographical features have been used to provide a variety of cell-ECM interactions, it remains unclear how their nanostructured surfaces affect cellular behavior. In this study, we investigated focal adhesion dynamics during the migration and proliferation of HeLa cells on silica bead (SB) arrays with various nanotopographies. Cell adhesion was altered according to the surface curvature and pinhole size of the SB arrays, and cell morphology was determined by the ratio of the adhesive and non-adhesive areas of cells on the SB arrays. In turn, this triggered different focal adhesion dynamics in cells. In addition, we demonstrated the rapid migration and high proliferation characteristics of rounded cells with weak adhesion based on confocal microscopy analysis and migration trajectory on SB arrays, indicating focal adhesion dynamics-mediated cell migration and proliferation on nanostructured surfaces.
Collapse
Affiliation(s)
- Yi-Seul Park
- Department of Chemistry, Sookmyung Women's University, Seoul 04310, Korea.
| | - Yerin Choi
- Department of Chemistry and Research Institute for Convergence of Basic Sciences, Hanyang University, Seoul 04763, South Korea.
| | - Jin Seok Lee
- Department of Chemistry and Research Institute for Convergence of Basic Sciences, Hanyang University, Seoul 04763, South Korea.
| |
Collapse
|
6
|
Wang A, Mizejewski GJ, Zhang C. Growth inhibitory peptides: a potential novel therapeutic approach to cancer treatment. Eur J Pharmacol 2025; 996:177554. [PMID: 40147579 DOI: 10.1016/j.ejphar.2025.177554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 03/02/2025] [Accepted: 03/24/2025] [Indexed: 03/29/2025]
Abstract
Cancer remains a major global public health concern, with considerable interest in exploring biological molecules for cancer treatment and prevention. Growth inhibitory peptide (GIP), a promising new class of biological therapeutics, has drawn attention for its distinct anti-tumor properties. Derived from human alpha-fetoprotein (HAFP), this synthetic 34-amino-acid peptide has demonstrated substantial anti-tumor effects across various cancer cell lines, effectively inhibiting tumor cell proliferation, migration, and metastasis. Studies reveal that GIP mediates its effects through a range of mechanisms, including interactions with G protein-coupled receptors (GPCRs), anti-cell adhesion activities, inhibition of cell spreading and metastatic processes, morphological alterations, platelet aggregation inhibition, immune enhancement, cell membrane disruption, ion channel blockade, and cell cycle arrest. While GIP has exhibited promising anti-tumor activity in both in vitro and in vivo models, further investigation is essential to advance its development as a therapeutic drug, particularly regarding pharmacokinetics, safety profiles, storage stability, and clinical efficacy.
Collapse
Affiliation(s)
- Aixin Wang
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing, PR China; Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China
| | - G J Mizejewski
- Division of Translational Medicine, Molecular Diagnostics Laboratory, Wadsworth Center, New York State Department of Health Biggs Laboratory, Empire State Plaza Albany, NY 12237, USA
| | - Chao Zhang
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing, PR China; Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China.
| |
Collapse
|
7
|
Chen Z, Dong L. Decoupling of Density-Dependent Migration/Proliferation Dichotomy on Surface Potential Gradient. ACS APPLIED MATERIALS & INTERFACES 2025; 17:16468-16478. [PMID: 40036071 DOI: 10.1021/acsami.4c18787] [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: 03/06/2025]
Abstract
The reciprocal connection between cell migration and proliferation relies on the intertwined contributions from substrate-associated and intercellular cues in the microenvironment. However, how cells perceive the substrates, make contact with their neighbors, and switch phenotypes under different trade-off conditions are still not fully understood. Here, we designed a distinct heterogeneous electric surface potential gradient of piezoelectric biomaterials to decouple the density-dependent migration/proliferation dichotomy. We found that the surface potential gradient accelerated both individual and collective cell migration but reduced proliferation through G0/G1 cell cycle arrest via the integrin/cytoskeleton signaling axis in low density. Interestingly, the initial cell density encodes the proliferative potential independent of the substrate feature. While in high density, the surface potential gradient ceased cell proliferation mainly via the E-cadherin/β-catenin signaling axis. Taken together, these results shed light on the underlying mechanism of the intertwined contributions of cell-material and cell-cell cross-links on migration and proliferation and also provide a new paradigm of materiobiology.
Collapse
Affiliation(s)
- Zejun Chen
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Province Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310006, China
| | - Lingqing Dong
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Province Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310006, China
| |
Collapse
|
8
|
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
|
9
|
Ye S, Shi D, Li X, Yang Y, Pan X, Wang L, Wu H. Development and bioevaluation of 18F-labeled bivalent cyclic peptides for PET imaging of αvβ6 integrin overexpression. Bioorg Chem 2025; 159:108362. [PMID: 40096805 DOI: 10.1016/j.bioorg.2025.108362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 03/01/2025] [Accepted: 03/07/2025] [Indexed: 03/19/2025]
Abstract
Integrin αvβ6 has emerged as a critical target in cancer diagnostics and therapeutics. In this study, we developed two bivalent ligands, NOTA-(SDM17)2 and NOTA-(AvB6)2, for positron emission tomography (PET) imaging of αvβ6 integrins. Surface plasmon resonance (SPR) revealed affinities for NOTA-(SDM17)2 and NOTA-(AvB6)2 with KD values of 2.15 μM and 5.21 μM, respectively. Micro-PET imaging demonstrated significantly higher uptake of [18F]AlF-NOTA-(SDM17)2 and [18F]AlF-NOTA-(AvB6)2 in H2009 tumors (αvβ6-positive) compared to MDA-MB-231 tumors (αvβ6-negative) ([18F]AlF-NOTA-(SDM17)2: 3.2 ± 0.3 vs. 0.3 ± 0.07 %ID/g; [18F]AlF-NOTA-(AvB6)2: 6.4 ± 0.5 vs. 1.0 ± 0.2 %ID/g at 60 min p.i., P < 0.05). Both bivalent tracers exhibited enhanced tumor uptake and retention relative to their monovalent counterparts ([18F]AlF-NOTA-SDM17 and [18F]AlF-NOTA-AvB6) at 60 min p.i., (P < 0.05). Notably, [18F]AlF-NOTA-(SDM17)2 demonstrated a superior tumor-to-liver ratio (13.24 vs. 5.93, P = 0.029) and longer retention, as confirmed by in vivo biodistribution studies. These findings highlight the potential of [18F]AlF-NOTA-(SDM17)2 and [18F]AlF-NOTA-(AvB6)2 as bivalent PET tracers to enhance tumor uptake and prolong retention. Among them, [18F]AlF-NOTA-(SDM17)2 shows particular promise for clinical translation due to its higher tumor-to-non-tumor ratio and prolonged retention.
Collapse
Affiliation(s)
- Shimin Ye
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong Province 510515, China
| | - Dazhi Shi
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong Province 510515, China
| | - Xuefei Li
- Central Research Institute, United Imaging Healthcare, 2258 Chengbei Road, Shanghai 201807, China
| | - Yali Yang
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong Province 510515, China
| | - Xingzhu Pan
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong Province 510515, China
| | - Lijuan Wang
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong Province 510515, China.
| | - Hubing Wu
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong Province 510515, China.
| |
Collapse
|
10
|
Zhang Y, Fu Q, Sun W, Yue Q, He P, Niu D, Zhang M. Mechanical forces in the tumor microenvironment: roles, pathways, and therapeutic approaches. J Transl Med 2025; 23:313. [PMID: 40075523 PMCID: PMC11899831 DOI: 10.1186/s12967-025-06306-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 02/23/2025] [Indexed: 03/14/2025] Open
Abstract
Tumors often exhibit greater stiffness compared to normal tissues, primarily due to increased deposition within the tumor stroma. Collagen, proteoglycans, laminin, and fibronectin are key components of the extracellular matrix (ECM), interacting to facilitate ECM assembly. Enhanced fiber density and cross-linking within the ECM result in elevated matrix stiffness and interstitial fluid pressure, subjecting tumors to significant physical stress during growth. This mechanical stress is transduced intracellularly via integrins, the Rho signaling pathway, and the Hippo signaling pathway, thereby promoting tumor invasion. Additionally, mechanical pressure fosters glycolysis in tumor cells, boosting energy production to support metastasis. Mechanical cues also regulate macrophage polarization, maintaining an inflammatory microenvironment conducive to tumor survival. In summary, mechanical signals within tumors play a crucial role in tumor growth and invasion. Understanding these signals and their involvement in tumor progression is essential for advancing our knowledge of tumor biology and enhancing therapeutic approaches.
Collapse
Affiliation(s)
- Yanli Zhang
- Key Laboratory for Molecular Genetic Mechanisms and Intervention Research on High Altitude Disease of Tibet Autonomous Region, School of Medicine, Xizang Minzu University, Xianyang, 712082, Shaanxi Province, China.
| | - Qi Fu
- Key Laboratory for Molecular Genetic Mechanisms and Intervention Research on High Altitude Disease of Tibet Autonomous Region, School of Medicine, Xizang Minzu University, Xianyang, 712082, Shaanxi Province, China
| | - Wenyue Sun
- Key Laboratory for Molecular Genetic Mechanisms and Intervention Research on High Altitude Disease of Tibet Autonomous Region, School of Medicine, Xizang Minzu University, Xianyang, 712082, Shaanxi Province, China
| | - Qiujuan Yue
- Key Laboratory for Molecular Genetic Mechanisms and Intervention Research on High Altitude Disease of Tibet Autonomous Region, School of Medicine, Xizang Minzu University, Xianyang, 712082, Shaanxi Province, China
| | - Ping He
- Key Laboratory for Molecular Genetic Mechanisms and Intervention Research on High Altitude Disease of Tibet Autonomous Region, School of Medicine, Xizang Minzu University, Xianyang, 712082, Shaanxi Province, China
| | - Dong Niu
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Min Zhang
- Key Laboratory for Molecular Genetic Mechanisms and Intervention Research on High Altitude Disease of Tibet Autonomous Region, School of Medicine, Xizang Minzu University, Xianyang, 712082, Shaanxi Province, China.
| |
Collapse
|
11
|
Zhang Q, Li T, Jiang H, Cao J, Wang H, Wang Z, Tang Q, Yang N, Zhao J, Wang F. Transcriptomic Insights Into Electroacupuncture Using Different Acupoint Combinations to Repair Mucosal Inflammatory Injury Induced in a Rat Model of Gastric Ulcer. J Inflamm Res 2025; 18:3399-3417. [PMID: 40093956 PMCID: PMC11910035 DOI: 10.2147/jir.s504930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 03/04/2025] [Indexed: 03/19/2025] Open
Abstract
Background Electroacupuncture (EA) is a promising treatment for gastrointestinal disorders, yet the efficacy of different acupoint combinations remains mechanistically undefined. We evaluated the therapeutic effects of different acupoint combinations on mucosal inflammatory injury induced in a rat model of gastric ulcer (GU) and dissected its molecular mechanisms through transcriptomic profiling. Methods A GU rat model was established using hypothermic restrained water immersion stress. EA therapy was administered to the He-Mu (ST36-CV12), Shu-Mu (BL21-CV12), and Yuan-Luo (ST42- ST40) acupoint combinations for 5 days. EA therapeutic effects were evaluated by coat score, fecal moisture percentage, pain threshold, body mass, organ index, histopathological changes, serum level of oxidative stress, and inflammatory cytokine levels in gastric tissue. A transcriptome analysis identified the related differentially expressed genes (DEGs) and central signaling pathway. Real-time quantitative PCR and Western blot were performed to verify the mRNA and protein expression levels of the main genes in the central pathway. Results EA using different acupoint combinations differentially alleviated gastric mucosal injury in GU rats, with the He-Mu group exhibiting superior tissue damage alleviation, as well as inflammation and oxidative stress reductions. A Venn diagram transcriptome analysis revealed a shared central pathway among the three groups, corresponding to focal adhesion. Quantitative validation confirmed that the mRNA, protein, and phosphorylated protein expression of FAK, VCL, and EGFR-the core signal transduction factors of the focal adhesion pathway activated in gastric tissue after EA treatment-were upregulated, consistent with their therapeutic efficacy. Conclusion Our results demonstrated that the He-Mu acupoint combination exhibited superior therapeutic efficacy among the three acupoint combinations. EA using different acupoint combinations improved gastric mucosal injury to varying degrees, and was related to the focal adhesion pathway. The FAK, VCL, and EGFR are promising targets, and further studies are needed to elucidate their functional consequences in GU.
Collapse
Affiliation(s)
- Qi Zhang
- School of Acupuncture and Massage, Changchun University of Chinese Medicine, Changchun City, Jilin Province, People's Republic of China
- School of Acupuncture and Orthopedics, Hubei University of Chinese Medicine, Wuhan City, Hubei Province, People's Republic of China
| | - Tie Li
- School of Acupuncture and Massage, Changchun University of Chinese Medicine, Changchun City, Jilin Province, People's Republic of China
| | - Hailin Jiang
- School of Acupuncture and Massage, Changchun University of Chinese Medicine, Changchun City, Jilin Province, People's Republic of China
| | - Jiazhen Cao
- School of Nursing, Changchun University of Chinese Medicine, Changchun City, Jilin Province, People's Republic of China
| | - He Wang
- School of Acupuncture and Massage, Changchun University of Chinese Medicine, Changchun City, Jilin Province, People's Republic of China
| | - Zhongke Wang
- School of Acupuncture and Massage, Changchun University of Chinese Medicine, Changchun City, Jilin Province, People's Republic of China
| | - Qingqing Tang
- School of Acupuncture and Massage, Changchun University of Chinese Medicine, Changchun City, Jilin Province, People's Republic of China
| | - Ning Yang
- School of Acupuncture and Massage, Changchun University of Chinese Medicine, Changchun City, Jilin Province, People's Republic of China
| | - Jinying Zhao
- School of Acupuncture and Massage, Changchun University of Chinese Medicine, Changchun City, Jilin Province, People's Republic of China
| | - Fuchun Wang
- School of Acupuncture and Massage, Changchun University of Chinese Medicine, Changchun City, Jilin Province, People's Republic of China
| |
Collapse
|
12
|
Li B, Ming H, Qin S, Nice EC, Dong J, Du Z, Huang C. Redox regulation: mechanisms, biology and therapeutic targets in diseases. Signal Transduct Target Ther 2025; 10:72. [PMID: 40050273 PMCID: PMC11885647 DOI: 10.1038/s41392-024-02095-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 10/09/2024] [Accepted: 11/21/2024] [Indexed: 03/09/2025] Open
Abstract
Redox signaling acts as a critical mediator in the dynamic interactions between organisms and their external environment, profoundly influencing both the onset and progression of various diseases. Under physiological conditions, oxidative free radicals generated by the mitochondrial oxidative respiratory chain, endoplasmic reticulum, and NADPH oxidases can be effectively neutralized by NRF2-mediated antioxidant responses. These responses elevate the synthesis of superoxide dismutase (SOD), catalase, as well as key molecules like nicotinamide adenine dinucleotide phosphate (NADPH) and glutathione (GSH), thereby maintaining cellular redox homeostasis. Disruption of this finely tuned equilibrium is closely linked to the pathogenesis of a wide range of diseases. Recent advances have broadened our understanding of the molecular mechanisms underpinning this dysregulation, highlighting the pivotal roles of genomic instability, epigenetic modifications, protein degradation, and metabolic reprogramming. These findings provide a foundation for exploring redox regulation as a mechanistic basis for improving therapeutic strategies. While antioxidant-based therapies have shown early promise in conditions where oxidative stress plays a primary pathological role, their efficacy in diseases characterized by complex, multifactorial etiologies remains controversial. A deeper, context-specific understanding of redox signaling, particularly the roles of redox-sensitive proteins, is critical for designing targeted therapies aimed at re-establishing redox balance. Emerging small molecule inhibitors that target specific cysteine residues in redox-sensitive proteins have demonstrated promising preclinical outcomes, setting the stage for forthcoming clinical trials. In this review, we summarize our current understanding of the intricate relationship between oxidative stress and disease pathogenesis and also discuss how these insights can be leveraged to optimize therapeutic strategies in clinical practice.
Collapse
Affiliation(s)
- Bowen Li
- Department of Biotherapy, Institute of Oxidative Stress Medicine, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, PR China
| | - Hui Ming
- Department of Biotherapy, Institute of Oxidative Stress Medicine, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, PR China
| | - Siyuan Qin
- Department of Biotherapy, Institute of Oxidative Stress Medicine, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, PR China
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, PR China
| | - Edouard C Nice
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
| | - Jingsi Dong
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
- Lung Cancer Center/Lung Cancer Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Zhongyan Du
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
- Key Laboratory of Blood-stasis-toxin Syndrome of Zhejiang Province, Hangzhou, China.
| | - Canhua Huang
- Department of Biotherapy, Institute of Oxidative Stress Medicine, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, PR China.
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, PR China.
| |
Collapse
|
13
|
Lai W, Geliang H, Bin X, Wang W. Effects of hydrogel stiffness and viscoelasticity on organoid culture: a comprehensive review. Mol Med 2025; 31:83. [PMID: 40033190 DOI: 10.1186/s10020-025-01131-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 02/14/2025] [Indexed: 03/05/2025] Open
Abstract
As an emerging technology, organoids are promising new tools for basic and translational research in disease. Currently, the culture of organoids relies mainly on a type of unknown composition scaffold, namely Matrigel, which may pose problems in studying the effect of mechanical properties on organoids. Hydrogels, a new material with adjustable mechanical properties, can adapt to current studies. In this review, we summarized the synthesis of recent advance in developing definite hydrogel scaffolds for organoid culture and identified the critical parameters for regulating mechanical properties. In addition, classified by different mechanical properties like stiffness and viscoelasticity, we concluded the effect of mechanical properties on the development of organoids and tumor organoids. We hope this review enhances the understanding of the development of organoids by hydrogels and provides more practical approaches to investigating them.
Collapse
Affiliation(s)
- Wei Lai
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Hu Geliang
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Xu Bin
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Wei Wang
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
14
|
Ren WW, Kawahara R, Suzuki KG, Dipta P, Yang G, Thaysen-Andersen M, Fujita M. MYO18B promotes lysosomal exocytosis by facilitating focal adhesion maturation. J Cell Biol 2025; 224:e202407068. [PMID: 39751400 PMCID: PMC11697975 DOI: 10.1083/jcb.202407068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 10/17/2024] [Accepted: 12/02/2024] [Indexed: 01/04/2025] Open
Abstract
Many cancer cells exhibit increased amounts of paucimannose glycans, which are truncated N-glycan structures rarely found in mammals. Paucimannosidic proteins are proposedly generated within lysosomes and exposed on the cell surface through a yet uncertain mechanism. In this study, we revealed that paucimannosidic proteins are produced by lysosomal glycosidases and secreted via lysosomal exocytosis. Interestingly, lysosomal exocytosis preferentially occurred in the vicinity of focal adhesions, protein complexes connecting the actin cytoskeleton to the extracellular matrix. Through genome-wide knockout screening, we identified that MYO18B, an actin crosslinker, is required for focal adhesion maturation, facilitating lysosomal exocytosis and the release of paucimannosidic lysosomal proteins to the extracellular milieu. Moreover, a mechanosensitive cation channel PIEZO1 locally activated at focal adhesions imports Ca2+ necessary for lysosome-plasma membrane fusion. Collectively, our study unveiled an intimate relationship between lysosomal exocytosis and focal adhesion, shedding light on the unexpected interplay between lysosomal activities and cellular mechanosensing.
Collapse
Affiliation(s)
- Wei-Wei Ren
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
- Institute for Glyco-core Research (iGCORE), Gifu University, Gifu, Japan
| | - Rebeca Kawahara
- Institute for Glyco-core Research (iGCORE), Nagoya University, Aichi, Japan
| | - Kenichi G.N. Suzuki
- Institute for Glyco-core Research (iGCORE), Gifu University, Gifu, Japan
- Division of Advanced Bioimaging, National Cancer Center Research Institute, Tokyo, Japan
| | - Priya Dipta
- School of Natural Sciences, Macquarie University, Sydney, Australia
| | - Ganglong Yang
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, China
| | - Morten Thaysen-Andersen
- Institute for Glyco-core Research (iGCORE), Nagoya University, Aichi, Japan
- School of Natural Sciences, Macquarie University, Sydney, Australia
| | - Morihisa Fujita
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
- Institute for Glyco-core Research (iGCORE), Gifu University, Gifu, Japan
| |
Collapse
|
15
|
Li XL, Zhao YQ, Miao L, An YX, Wu F, Han JY, Han JY, Tay FR, Mu Z, Jiao Y, Wang J. Strategies for promoting neurovascularization in bone regeneration. Mil Med Res 2025; 12:9. [PMID: 40025573 PMCID: PMC11874146 DOI: 10.1186/s40779-025-00596-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Accepted: 01/26/2025] [Indexed: 03/04/2025] Open
Abstract
Bone tissue relies on the intricate interplay between blood vessels and nerve fibers, both are essential for many physiological and pathological processes of the skeletal system. Blood vessels provide the necessary oxygen and nutrients to nerve and bone tissues, and remove metabolic waste. Concomitantly, nerve fibers precede blood vessels during growth, promote vascularization, and influence bone cells by secreting neurotransmitters to stimulate osteogenesis. Despite the critical roles of both components, current biomaterials generally focus on enhancing intraosseous blood vessel repair, while often neglecting the contribution of nerves. Understanding the distribution and main functions of blood vessels and nerve fibers in bone is crucial for developing effective biomaterials for bone tissue engineering. This review first explores the anatomy of intraosseous blood vessels and nerve fibers, highlighting their vital roles in bone embryonic development, metabolism, and repair. It covers innovative bone regeneration strategies directed at accelerating the intrabony neurovascular system over the past 10 years. The issues covered included material properties (stiffness, surface topography, pore structures, conductivity, and piezoelectricity) and acellular biological factors [neurotrophins, peptides, ribonucleic acids (RNAs), inorganic ions, and exosomes]. Major challenges encountered by neurovascularized materials during their clinical translation have also been highlighted. Furthermore, the review discusses future research directions and potential developments aimed at producing bone repair materials that more accurately mimic the natural healing processes of bone tissue. This review will serve as a valuable reference for researchers and clinicians in developing novel neurovascularized biomaterials and accelerating their translation into clinical practice. By bridging the gap between experimental research and practical application, these advancements have the potential to transform the treatment of bone defects and significantly improve the quality of life for patients with bone-related conditions.
Collapse
Affiliation(s)
- Xin-Ling Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Oral Implants, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Yu-Qing Zhao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Oral Implants, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Li Miao
- Department of Stomatology, The Seventh Medical Center of PLA General Hospital, Beijing, 100700, China
| | - Yan-Xin An
- Department of General Surgery, The First Affiliated Hospital of Xi'an Medical University, Xi'an, 710077, China
| | - Fan Wu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Oral Implants, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Jin-Yu Han
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Oral Implants, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Jing-Yuan Han
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Oral Implants, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Franklin R Tay
- Graduate School of Augusta University, Augusta, GA, 30912, USA
| | - Zhao Mu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China.
| | - Yang Jiao
- Department of Stomatology, The Seventh Medical Center of PLA General Hospital, Beijing, 100700, China.
| | - Jing Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Oral Implants, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
16
|
Matrullo G, Filomeni G, Rizza S. Redox regulation of focal adhesions. Redox Biol 2025; 80:103514. [PMID: 39879736 PMCID: PMC11810850 DOI: 10.1016/j.redox.2025.103514] [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: 11/04/2024] [Revised: 01/07/2025] [Accepted: 01/23/2025] [Indexed: 01/31/2025] Open
Abstract
Focal adhesions (FAs), multi-protein complexes that link the extracellular matrix to the intracellular cytoskeleton, are key mediators of cell adhesion, migration, and proliferation. These dynamic structures act as mechanical sensors, transmitting stimuli from the extracellular to intracellular environment activating in this way signaling pathways and enabling cells to adapt to environmental changes. As such, FAs are critical for tissue organization and serve as hubs governing cell spatial arrangement within the organism. The assembly, reactivity, and functional regulation of FAs are tightly controlled by post-translational modifications, including redox modulation by reactive oxygen and nitrogen species. Increasing evidence suggests that redox signaling plays a pivotal role in both the physiological and pathological functions of FAs and their downstream processes. Redox regulation affects various components of the FA complex, including integrins, focal adhesion kinase 1 (FAK1), SRC, adapter proteins, and cytoskeletal elements. In this review, we provide an updated overview of the complex interplay between redox signaling and post-translational modifications in FAs. We explore how redox reactions influence the structure, dynamics, and function of FAs, shedding light on their broader implications in health and disease.
Collapse
Affiliation(s)
- Gianmarco Matrullo
- Department of Biology, University of Rome "Tor Vergata", 00100, Rome, Italy
| | - Giuseppe Filomeni
- Department of Biology, University of Rome "Tor Vergata", 00100, Rome, Italy; Redox Biology Group, Danish Cancer Institute, 2100, Copenhagen, Denmark
| | - Salvatore Rizza
- Redox Biology Group, Danish Cancer Institute, 2100, Copenhagen, Denmark.
| |
Collapse
|
17
|
Liu W, Lu D, Jia S, Yang Y, Meng F, Du Y, Yang Y, Yuan L, Nan Y. Molecular mechanism of Gancao Xiexin Decoction regulating EMT and suppressing hepatic metastasis of gastric cancer via the TGF-β1/SMAD pathway. JOURNAL OF ETHNOPHARMACOLOGY 2025; 342:119430. [PMID: 39900270 DOI: 10.1016/j.jep.2025.119430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 01/16/2025] [Accepted: 01/28/2025] [Indexed: 02/05/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Gastric cancer (GC) is a highly malignant tumor of the digestive tract, posing a significant menace to human health. Gancao Xiexin Decoction (GCXXD), being a traditional Chinese medicine (TCM), has a good effect on inhibiting the proliferation and metastasis of GC. However, its mechanisms still need further investigation. AIM OF STUDY To investigate the mechanism by which GCXXD inhibits GC metastasis through network pharmacology, and to verify through in vivo and in vitro experiments. MATERIALS AND METHODS The TCMSP and GEO databases, in combination with UPLC-MS/MS techniques, were employed to identify the hub genes, active ingredients, and critical pathways of GCXXD in the treatment of GC. Subsequently, molecular docking was conducted on both the hub genes and the core components. Finally, based on the results of the bioinformatics analysis, the role of GCXXD in inhibiting liver metastasis of GC was elucidated through in vivo and in vitro experiments, including scratch assays, Transwell assays, HE staining, immunohistochemistry, in vivo live imaging, qRT-PCR, and Western blotting. RESULTS Utilizing UPLC-MS/MS and network pharmacology, we identified 20 active ingredients and 5 hub targets in the treatment of GC by GCXXD. Through KEGG analyses, GCXXD treatment of GC could through the TGF-beta pathway. In vivo and in vitro experiments, GCXXD downregulated the mRNA and protein expression level of hub genes involved in the TGF-β1/SMAD pathway and the EMT process. Additionally, GCXXD significantly reduced the incidence of liver metastases in GC. CONCLUSION GCXXD inhibited EMT via blocking the TGF-β1/SMAD pathway, which suppressed GC cell growth and liver metastasis. This study provides data to support the treatment of liver metastasis in GC with TCM and holds significant importance for the research and development of new anticancer drugs.
Collapse
Affiliation(s)
- Wenjing Liu
- Key Laboratory of Ningxia Ethnomedicine Modernization, Ministry of Education, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Doudou Lu
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Shumin Jia
- Key Laboratory of Ningxia Ethnomedicine Modernization, Ministry of Education, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Yating Yang
- The Second Hospital of Chinese Medicine of BAO JI City, Baoji, 721300, Xian, China
| | - Fandi Meng
- Traditional Chinese Medicine College, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Yuhua Du
- College of Pharmacy, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Yi Yang
- College of Pharmacy, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Ling Yuan
- College of Pharmacy, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Yi Nan
- Key Laboratory of Ningxia Ethnomedicine Modernization, Ministry of Education, Ningxia Medical University, Yinchuan, 750004, Ningxia, China; Traditional Chinese Medicine College, Ningxia Medical University, Yinchuan, 750004, Ningxia, China.
| |
Collapse
|
18
|
Villari G, Gioelli N, Gino M, Zhang H, Hodge K, Cordero F, Zanivan S, Zhu J, Serini G. Luminescent sensing of conformational integrin activation in living cells. Cell Rep 2025; 44:115319. [PMID: 39964812 PMCID: PMC11861568 DOI: 10.1016/j.celrep.2025.115319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 11/29/2024] [Accepted: 01/27/2025] [Indexed: 02/20/2025] Open
Abstract
Integrins are major receptors for secreted extracellular matrix, playing crucial roles in physiological and pathological contexts, such as angiogenesis and cancer. Regulation of the transition between inactive and active conformation is key for integrins to fulfill their functions, and pharmacological control of those dynamics may have therapeutic applications. We create and validate a prototypic luminescent β1 integrin activation sensor (β1IAS) by introducing a split luciferase into an activation reporting site between the βI and the hybrid domains. As a recombinant protein in both solution and living cells, β1IAS accurately reports β1 integrin activation in response to (bio)chemical and physical stimuli. A short interfering RNA (siRNA) high-throughput screening on live β1IAS knockin endothelial cells unveils hitherto unknown regulators of β1 integrin activation, such as β1 integrin inhibitors E3 ligase Pja2 and vascular endothelial growth factor B (VEGF-B). This split-luciferase-based strategy provides an in situ label-free measurement of integrin activation and may be applicable to other β integrins and receptors.
Collapse
Affiliation(s)
- Giulia Villari
- Department of Oncology, University of Torino School of Medicine, Candiolo, TO, Italy; Candiolo Cancer Institute - Fondazione del Piemonte per l'Oncologia (FPO) Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Candiolo, TO, Italy
| | - Noemi Gioelli
- Candiolo Cancer Institute - Fondazione del Piemonte per l'Oncologia (FPO) Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Candiolo, TO, Italy
| | - Marta Gino
- Department of Oncology, University of Torino School of Medicine, Candiolo, TO, Italy; Candiolo Cancer Institute - Fondazione del Piemonte per l'Oncologia (FPO) Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Candiolo, TO, Italy
| | - Heng Zhang
- Thrombosis and Hemostasis Program, Versiti Blood Research Institute, Milwaukee, WI, USA
| | - Kelly Hodge
- Cancer Research UK Scotland Institute, Glasgow, UK
| | | | - Sara Zanivan
- Cancer Research UK Scotland Institute, Glasgow, UK; School of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Jieqing Zhu
- Thrombosis and Hemostasis Program, Versiti Blood Research Institute, Milwaukee, WI, USA; Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Guido Serini
- Department of Oncology, University of Torino School of Medicine, Candiolo, TO, Italy; Candiolo Cancer Institute - Fondazione del Piemonte per l'Oncologia (FPO) Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Candiolo, TO, Italy.
| |
Collapse
|
19
|
Geng J, Zheng K, Wang P, Su B, Wei Q, Liu X. Focal Adhesion Regulation as a Strategy against Kidney Fibrosis. ACS Chem Biol 2025; 20:464-478. [PMID: 39818722 DOI: 10.1021/acschembio.4c00776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2025]
Abstract
Chronic kidney fibrosis poses a significant global health challenge with effective therapeutic strategies remaining elusive. While cell-extracellular matrix (ECM) interactions are known to drive fibrosis progression, the specific role of focal adhesions (FAs) in kidney fibrosis is not fully understood. In this study, we investigated the role of FAs in kidney tubular epithelial cell fibrosis by employing precise nanogold patterning to modulate integrin distribution. We demonstrate that increasing ligand spacing disrupts integrin clustering, thereby inhibiting FA formation and attenuating fibrosis. Importantly, enhanced FA activity is associated with kidney fibrosis in both human disease specimens and murine models. Mechanistically, FAs regulate fibrosis through mechanotransduction pathways, and our in vivo experiments show that suppressing mechanotransduction significantly mitigates kidney fibrosis in mice. These findings highlight the potential of targeting FAs as a therapeutic strategy, offering new insights into clinical intervention in kidney fibrosis.
Collapse
Affiliation(s)
- Jiwen Geng
- Department of Nephrology, Kidney Research Institute, West China Hospital, Sichuan University, Chengdu 610041, China
- College of Polymer Science and Engineering, College of Biomedical Engineering, State Key Laboratory of Polymer Materials and Engineering Sichuan University, Chengdu 610065, China
| | - Kaikai Zheng
- College of Polymer Science and Engineering, College of Biomedical Engineering, State Key Laboratory of Polymer Materials and Engineering Sichuan University, Chengdu 610065, China
| | - Peng Wang
- College of Polymer Science and Engineering, College of Biomedical Engineering, State Key Laboratory of Polymer Materials and Engineering Sichuan University, Chengdu 610065, China
| | - Baihai Su
- Department of Nephrology, Kidney Research Institute, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qiang Wei
- College of Polymer Science and Engineering, College of Biomedical Engineering, State Key Laboratory of Polymer Materials and Engineering Sichuan University, Chengdu 610065, China
| | - Xiaojing Liu
- Department of Pediatric Dentistry, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, and Shandong Key Laboratory of Oral Tissue Regeneration, Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration, Shandong Provincial Clinical Research Center for Oral Diseases, Jinan 250012, China
| |
Collapse
|
20
|
Wang Q, Herrmann J, Worthington KS, Sander EA. Cyclic mechanical loading of photopolymerized methacrylated hydrogels for probing interdependent effects of strain, stiffness, and substrate composition in pulmonary fibrogenesis. Sci Rep 2025; 15:5997. [PMID: 39966483 PMCID: PMC11836278 DOI: 10.1038/s41598-025-90753-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 02/14/2025] [Indexed: 02/20/2025] Open
Abstract
Pulmonary fibrosis is characterized by excessive deposition of extracellular matrix (ECM), stiffening of the lung tissue, and impaired gas exchange. Our current understanding of fibrogenesis generally focuses on the individual roles of mechanical and biochemical stimuli in driving disease progression. However, many mechano-chemical pathways are interrelated, so dissecting the interactive effects of mechanical and biochemical signals is an important knowledge gap. To address this gap, we investigated lung fibroblast behavior on static and cyclically strained photopolymerizable hydrogels consisting of different ratios of methacrylated gelatin, methacrylated hyaluronan, and non-methacrylated gelatin to create substrates with tunable stiffness and chemistry, representative of both healthy and fibrotic lung ECM properties. We observed that higher stiffness gels amplified the impact of strain, resulting in distinct differences in expression of MMP1, CTGF, Rho/ROCK, and ECM deposition genes. Substrates with hyaluronan demonstrated a capacity to modulate strain-induced fibrogenic responses, suggesting a buffering effect of hyaluronan on fibrotic disease progression. Overall, our results highlight mechanotransductive changes in gene expression in response to substrate composition, stiffness, and cyclic mechanical strain. Through the controlled study of mechanical and biochemical cues, our findings contribute to a deeper understanding of the pathogenesis of pulmonary fibrosis.
Collapse
Affiliation(s)
- Q Wang
- Roy J. Carver Department of Biomedical Engineering, College of Engineering, The University of Iowa, Iowa City, IA, USA
| | - J Herrmann
- Roy J. Carver Department of Biomedical Engineering, College of Engineering, The University of Iowa, Iowa City, IA, USA
| | - K S Worthington
- Roy J. Carver Department of Biomedical Engineering, College of Engineering, The University of Iowa, Iowa City, IA, USA
| | - E A Sander
- Roy J. Carver Department of Biomedical Engineering, College of Engineering, The University of Iowa, Iowa City, IA, USA.
- Department of Orthopedics and Rehabilitation, The University of Iowa, 5629 Seamans Center, Iowa City, IA, 52242, USA.
| |
Collapse
|
21
|
Yin Y, Liao L, Xu Q, Xie S, Yuan L, Zhou R. Insight into the post-translational modifications in pregnancy and related complications†. Biol Reprod 2025; 112:204-224. [PMID: 39499652 DOI: 10.1093/biolre/ioae149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 09/19/2024] [Indexed: 11/07/2024] Open
Abstract
Successful pregnancy is dependent on a number of essential events, including embryo implantation, decidualization, and placentation. Failure of the above process may lead to pregnancy-related complications, including preeclampsia, gestational diabetes mellitus, preterm birth, and fetal growth restriction, may affect 15% of pregnancies, and lead to increased mortality and morbidity of pregnant women and perinatal infants, as well as the occurrence of short-term and long-term diseases. These complications have distinct etiology and pathogenesis, and the present comprehension is still lacking. Post-translational modifications are important events in epigenetics, altering the properties of proteins through protein hydrolysis or the addition of modification groups to one or more amino acids, with different modification states regulating subcellular localization, protein degradation, protein-protein interaction, signal transduction, and gene transcription. In this review, we focus on the impact of various post-translational modifications on the progress of embryo and placenta development and pregnancy-related complications, which will provide important experimental bases for exploring new insights into the physiology of pregnancy and pathogenesis associated with pregnancy complications.
Collapse
Affiliation(s)
- Yangxue Yin
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University) of Ministry of Education, Chengdu, China
- National Health Commission Key Laboratory of Chronobiology, Sichuan University, Chengdu, China
| | - Lingyun Liao
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University) of Ministry of Education, Chengdu, China
- National Health Commission Key Laboratory of Chronobiology, Sichuan University, Chengdu, China
| | - Qin Xu
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University) of Ministry of Education, Chengdu, China
- National Health Commission Key Laboratory of Chronobiology, Sichuan University, Chengdu, China
| | - Shuangshuang Xie
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University) of Ministry of Education, Chengdu, China
- National Health Commission Key Laboratory of Chronobiology, Sichuan University, Chengdu, China
| | - Liming Yuan
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University) of Ministry of Education, Chengdu, China
- National Health Commission Key Laboratory of Chronobiology, Sichuan University, Chengdu, China
| | - Rong Zhou
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University) of Ministry of Education, Chengdu, China
- National Health Commission Key Laboratory of Chronobiology, Sichuan University, Chengdu, China
| |
Collapse
|
22
|
Sharip A, Kunz J. Mechanosignaling via Integrins: Pivotal Players in Liver Fibrosis Progression and Therapy. Cells 2025; 14:266. [PMID: 39996739 PMCID: PMC11854242 DOI: 10.3390/cells14040266] [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: 12/09/2024] [Revised: 02/05/2025] [Accepted: 02/05/2025] [Indexed: 02/26/2025] Open
Abstract
Liver fibrosis, a consequence of chronic liver injury, represents a major global health burden and is the leading cause of liver failure, morbidity, and mortality. The pathological hallmark of this condition is excessive extracellular matrix deposition, driven primarily by integrin-mediated mechanotransduction. Integrins, transmembrane heterodimeric proteins that serve as primary ECM receptors, orchestrate complex mechanosignaling networks that regulate the activation, differentiation, and proliferation of hepatic stellate cells and other ECM-secreting myofibroblasts. These mechanical signals create self-reinforcing feedback loops that perpetuate the fibrotic response. Recent advances have provided insight into the roles of specific integrin subtypes in liver fibrosis and revealed their regulation of key downstream effectors-including transforming growth factor beta, focal adhesion kinase, RhoA/Rho-associated, coiled-coil containing protein kinase, and the mechanosensitive Hippo pathway. Understanding these mechanotransduction networks has opened new therapeutic possibilities through pharmacological manipulation of integrin-dependent signaling.
Collapse
Affiliation(s)
- Aigul Sharip
- Department of Biomedical Sciences, Nazarbayev University School of Medicine, Astana 020000, Kazakhstan;
- Laboratory of Bioinformatics and Systems Biology, National Laboratory Astana, Astana 020000, Kazakhstan
| | - Jeannette Kunz
- Department of Biomedical Sciences, Nazarbayev University School of Medicine, Astana 020000, Kazakhstan;
| |
Collapse
|
23
|
Wang Y, Wang Y, Chen Y, Lu W, Tong X, Li J, Gao W, Huang R, Hu W, Feng Y. Distinct mechanisms of electroacupuncture and manual acupuncture in modulating hypothalamic GnRH-tanycyte unit function of polycystic ovary syndrome. Chin Med 2025; 20:18. [PMID: 39910658 DOI: 10.1186/s13020-025-01068-3] [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: 09/04/2024] [Accepted: 01/20/2025] [Indexed: 02/07/2025] Open
Abstract
BACKGROUND Polycystic ovary syndrome (PCOS) is a complex neuroendocrine disorder characterized by dysregulation of the hypothalamus. Both electroacupuncture (EA) and manual acupuncture (MA) have demonstrated therapeutic efficacy in the treatment of PCOS through improvements in hypothalamic function. However, the underlying mechanisms remain poorly understood. Gonadotropin-releasing hormone (GnRH) neurons are pivotal in regulating hypothalamic endocrine function, whereas tanycyte, a specialized glial cell type, potentially contribute to this process. METHODS A dihydrotestosterone (DHT)-induced PCOS-like mouse model was used to investigate the effects of acupuncture. Tissue clearing and three-dimensional (3D) imaging were employed to visualize the hypothalamic GnRH neuronal network and assess postacupuncture modifications. Transcriptome sequencing was performed to identify changes in the gene profiles associated with EA and MA. Rax-CreERT2 transgenic mice were utilized to investigate the molecular targets of EA in tanycytes. RESULTS EA significantly alleviated neuroendocrine dysfunction in PCOS-like mice by restoring the density and coverage of GnRH axonal projections. MA displayed similar therapeutic effects but had less pronounced effects on GnRH axons. Transcriptome analysis revealed distinct mechanisms for these two approaches: EA primarily regulates neuroglial plasticity, whereas MA predominantly targets neurotransmitter regulation. Both EA and MA share a common therapeutic target in the integrin family. Functional studies in Rax-CreERT2 transgenic mice confirmed that Itgb1 plays a critical role in maintaining the balance of hypothalamic GnRH-tanycyte unit during EA treatment. CONCLUSIONS EA exerts therapeutic effects on PCOS by targeting hypothalamic GnRH-tanycyte unit, with Itgb1 identified as a key factor. MA primarily functions through neurotransmitter regulation. These findings highlight potential hypothalamic targets and provide new insights into the distinct mechanisms of EA and MA.
Collapse
Affiliation(s)
- Yu Wang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science , Fudan University, Shanghai, 200032, China
- Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Shanghai Institute of Acupuncture and Moxibustion, Fudan University, Shanghai, 200433, China
| | - Yicong Wang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science , Fudan University, Shanghai, 200032, China
- Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Shanghai Institute of Acupuncture and Moxibustion, Fudan University, Shanghai, 200433, China
| | - Yuning Chen
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science , Fudan University, Shanghai, 200032, China
- Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Shanghai Institute of Acupuncture and Moxibustion, Fudan University, Shanghai, 200433, China
| | - Wenhan Lu
- Department of Ophthalmology & Visual Science, Eye & ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xiaoyu Tong
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science , Fudan University, Shanghai, 200032, China
- Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Shanghai Institute of Acupuncture and Moxibustion, Fudan University, Shanghai, 200433, China
| | - Jiajia Li
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science , Fudan University, Shanghai, 200032, China
- Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Shanghai Institute of Acupuncture and Moxibustion, Fudan University, Shanghai, 200433, China
| | - Wenhao Gao
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science , Fudan University, Shanghai, 200032, China
- Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Shanghai Institute of Acupuncture and Moxibustion, Fudan University, Shanghai, 200433, China
| | - Rui Huang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science , Fudan University, Shanghai, 200032, China
- Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Shanghai Institute of Acupuncture and Moxibustion, Fudan University, Shanghai, 200433, China
| | - Wei Hu
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science , Fudan University, Shanghai, 200032, China.
- Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Shanghai Institute of Acupuncture and Moxibustion, Fudan University, Shanghai, 200433, China.
| | - Yi Feng
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science , Fudan University, Shanghai, 200032, China.
- Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Shanghai Institute of Acupuncture and Moxibustion, Fudan University, Shanghai, 200433, China.
| |
Collapse
|
24
|
Kim SHJ, Lee H, Gingras A, Ley K, Spangler JB, Ginsberg MH. A CD25-CCR7 complex initiates non-canonical IL-2 signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.03.636356. [PMID: 39975281 PMCID: PMC11838579 DOI: 10.1101/2025.02.03.636356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
IL-2, a central regulator of immune function, binds to its receptor subunit CD25 (IL-2Rα), promoting IL-2 interaction with β and γ subunits to trigger the canonical IL-2 signaling pathway. An anti-mouse CD25 antibody, PC61, triggers alternative IL-2 signaling, leading to integrin activation. PC61 induces a complex formed by the IL-2-dependent association of CD25 with CCR7, suggesting that the formation of this complex initiates alternative IL-2 signaling. Here, we used structure-based design together with combinatorial screening to identify an IL-2 mutant (denoted IL-2(E52K)) that spares canonical IL-2 signaling but disrupts both PC61-induced complex formation and integrin activation while retaining the full CD25 affinity of the parent molecule. We also report that heparan sulfate (HS), a physiological ligand of IL-2 that triggers alternative signaling, induced IL-2-dependent CD25-CCR7 association, whereas IL-2(E52K) failed to support both HS-induced CD25-CCR7 complex formation and integrin activation. Thus, both anti-CD25 antibody and HS require common features of IL-2 needed for CD25-CCR7 complex assembly and resulting integrin activation. Collectively, these data show that IL-2 promotes CD25 interaction with CCR7, thereby forming the signal initiating complex. Furthermore, canonical and alternative IL-2 signaling can be decoupled by an IL-2 mutation, creating a tool to specify the biological role of alternative IL-2 signaling in immune responses.
Collapse
Affiliation(s)
- Sarah Hyun Ji Kim
- Department of Medicine, University of California San Diego School of Medicine, La Jolla, CA
| | - Hosup Lee
- Department of Medicine, University of California San Diego School of Medicine, La Jolla, CA
| | - Alexandre Gingras
- Department of Medicine, University of California San Diego School of Medicine, La Jolla, CA
| | - Klaus Ley
- Department of Physiology, Augusta University, Augusta, GA
| | - Jamie B. Spangler
- Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, MD
- Department of Chemical & Biomolecular Engineering, Johns Hopkins School of Medicine, Baltimore, MD
| | - Mark H. Ginsberg
- Department of Medicine, University of California San Diego School of Medicine, La Jolla, CA
| |
Collapse
|
25
|
Qiao M, Li Y, Yan S, Zhang RJ, Dong H. Modulation of arterial wall remodeling by mechanical stress: Focus on abdominal aortic aneurysm. Vasc Med 2025:1358863X241309836. [PMID: 39895313 DOI: 10.1177/1358863x241309836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
The rupture of an abdominal aortic aneurysm (AAA) poses a significant threat, with a high mortality rate, and the mechanical stability of the arterial wall determines both its growth and potential for rupture. Owing to extracellular matrix (ECM) degradation, wall-resident cells are subjected to an aberrant mechanical stress environment. In response to stress, the cellular mechanical signaling pathway is activated, initiating the remodeling of the arterial wall to restore stability. A decline in mechanical signal responsiveness, coupled with inadequate remodeling, significantly contributes to the AAA's progressive expansion and eventual rupture. In this review, we summarize the main stresses experienced by the arterial wall, emphasizing the critical role of the ECM in withstanding stress and the importance of stress-exposed cells in maintaining mechanical stability. Furthermore, we will discuss the application of biomechanical analyses as a predictive tool for assessing AAA stability.
Collapse
Affiliation(s)
- Maolin Qiao
- Shanxi Medical University Second Affiliated Hospital, Taiyuan, Shanxi, China
| | - Yaling Li
- Shanxi Medical University Second Affiliated Hospital, Taiyuan, Shanxi, China
| | - Sheng Yan
- Shanxi Medical University Second Affiliated Hospital, Taiyuan, Shanxi, China
| | - Rui Jing Zhang
- Shanxi Medical University Second Affiliated Hospital, Taiyuan, Shanxi, China
| | - Honglin Dong
- Shanxi Medical University Second Affiliated Hospital, Taiyuan, Shanxi, China
| |
Collapse
|
26
|
Wu J, Han B, Ai S, Wang A, Song Y, Jin M, Qu X, Wang X. Injectable double network hydrogel with adjustable stiffness for modulation of macrophage polarization. POLYMER TESTING 2025; 143:108685. [DOI: 10.1016/j.polymertesting.2024.108685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
|
27
|
Zhang C, Song W, Guo X, Li Z, Kong Y, Du J, Hou L, Feng Y, Wang Y, Zhang M, Liang L, Huang Y, Li J, Zhu D, Liu Q, Tan Y, Zhao Z, Zhao Y, Fu X, Huang S. Piezoelectric nanocomposite electrospun dressings: Tailoring mechanics for scar-free wound recovery. BIOMATERIALS ADVANCES 2025; 167:214119. [PMID: 39556886 DOI: 10.1016/j.bioadv.2024.214119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 10/31/2024] [Accepted: 11/11/2024] [Indexed: 11/20/2024]
Abstract
Rational wound management and enhancing healing quality are critical in clinical practice. Electrical stimulation therapy (EST) has emerged as a valuable adjunctive treatment due to its safety and cost-effectiveness. Integrating piezoelectric materials into dressings offers a way to miniaturize and personalize electrotherapy, enhancing convenience. To address the impact of physical factors of dressings on wound healing, a nanocomposite piezoelectric electrospun dressing using poly(L-lactic acid) (PLLA) and barium titanate (BaTiO3) was developed. We intentionally exaggerated design flaws to mimic the characteristics of scar extracellular matrix (ECM), including the oriented thick fibers and high Young's modulus. Initially, these dressings promoted fibrosis and hindered functional regeneration. However, when the piezoelectric effect was triggered by ultrasound, the fibrotic phenotype was reversed, leading to scar-free healing with well-regenerated functional structures. This study highlights the significant therapeutic potential of piezoelectric dressings in skin wound treatment and underscores the importance of carefully designing the static physical properties of dressings for optimal efficacy.
Collapse
Affiliation(s)
- Chao Zhang
- School of Medicine, Nankai University, Tianjin 300071, China; Research Center for Wound Repair and Tissue Regeneration, Medical Innovation Research Department, Chinese PLA General Hospital, Beijing 100048, China; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing 100048, China
| | - Wei Song
- Research Center for Wound Repair and Tissue Regeneration, Medical Innovation Research Department, Chinese PLA General Hospital, Beijing 100048, China; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing 100048, China
| | - Xu Guo
- Research Center for Wound Repair and Tissue Regeneration, Medical Innovation Research Department, Chinese PLA General Hospital, Beijing 100048, China; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing 100048, China; College of Graduate, Tianjin Medical University, Tianjin 300203, China
| | - Zhao Li
- Research Center for Wound Repair and Tissue Regeneration, Medical Innovation Research Department, Chinese PLA General Hospital, Beijing 100048, China; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing 100048, China
| | - Yi Kong
- Research Center for Wound Repair and Tissue Regeneration, Medical Innovation Research Department, Chinese PLA General Hospital, Beijing 100048, China; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing 100048, China
| | - Jinpeng Du
- Research Center for Wound Repair and Tissue Regeneration, Medical Innovation Research Department, Chinese PLA General Hospital, Beijing 100048, China; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing 100048, China
| | - Linhao Hou
- Research Center for Wound Repair and Tissue Regeneration, Medical Innovation Research Department, Chinese PLA General Hospital, Beijing 100048, China; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing 100048, China
| | - Yu Feng
- Research Center for Wound Repair and Tissue Regeneration, Medical Innovation Research Department, Chinese PLA General Hospital, Beijing 100048, China; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing 100048, China
| | - Yuzhen Wang
- Research Center for Wound Repair and Tissue Regeneration, Medical Innovation Research Department, Chinese PLA General Hospital, Beijing 100048, China; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing 100048, China
| | - Mengde Zhang
- Research Center for Wound Repair and Tissue Regeneration, Medical Innovation Research Department, Chinese PLA General Hospital, Beijing 100048, China; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing 100048, China
| | - Liting Liang
- Research Center for Wound Repair and Tissue Regeneration, Medical Innovation Research Department, Chinese PLA General Hospital, Beijing 100048, China; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing 100048, China
| | - Yuyan Huang
- Research Center for Wound Repair and Tissue Regeneration, Medical Innovation Research Department, Chinese PLA General Hospital, Beijing 100048, China; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing 100048, China
| | - Jianjun Li
- Research Center for Wound Repair and Tissue Regeneration, Medical Innovation Research Department, Chinese PLA General Hospital, Beijing 100048, China; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing 100048, China
| | - Dongzhen Zhu
- Research Center for Wound Repair and Tissue Regeneration, Medical Innovation Research Department, Chinese PLA General Hospital, Beijing 100048, China; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing 100048, China
| | - Qinghua Liu
- Research Center for Wound Repair and Tissue Regeneration, Medical Innovation Research Department, Chinese PLA General Hospital, Beijing 100048, China; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing 100048, China
| | - Yaxin Tan
- Research Center for Wound Repair and Tissue Regeneration, Medical Innovation Research Department, Chinese PLA General Hospital, Beijing 100048, China; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing 100048, China
| | - Ziteng Zhao
- Senior Department of Orthopedics, the Fourth Medical Center of PLA General Hospital, Beijing 100048, China
| | - Yantao Zhao
- Senior Department of Orthopedics, the Fourth Medical Center of PLA General Hospital, Beijing 100048, China
| | - Xiaobing Fu
- School of Medicine, Nankai University, Tianjin 300071, China; Research Center for Wound Repair and Tissue Regeneration, Medical Innovation Research Department, Chinese PLA General Hospital, Beijing 100048, China; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing 100048, China; College of Graduate, Tianjin Medical University, Tianjin 300203, China.
| | - Sha Huang
- Research Center for Wound Repair and Tissue Regeneration, Medical Innovation Research Department, Chinese PLA General Hospital, Beijing 100048, China; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing 100048, China.
| |
Collapse
|
28
|
Chen H, Yang J, Yang Q, Jia Y, Guo X. Protein prenylation in mechanotransduction: implications for disease and therapy. Trends Pharmacol Sci 2025; 46:163-179. [PMID: 39818521 DOI: 10.1016/j.tips.2024.12.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 12/05/2024] [Accepted: 12/18/2024] [Indexed: 01/18/2025]
Abstract
The process by which cells translate external mechanical cues into intracellular biochemical signals involves intricate mechanisms that remain unclear. In recent years, research into post-translational modifications (PTMs) has offered valuable insights into this field, spotlighting protein prenylation as a crucial mechanism in cellular mechanotransduction and various human diseases. Protein prenylation, which involves the covalent attachment of isoprenoid groups to specific substrate proteins, profoundly affects the functions of key mechanotransduction proteins such as Rho, Ras, and lamins. This review provides the first comprehensive examination of the connections between prenylation and mechanotransduction, exploring both the mechanistic details and its impact on mechanosensitive cellular behaviors. We further highlight recent evidence linking protein prenylation to diseases associated with disrupted mechanical homeostasis, and outline emerging targeted therapeutic strategies.
Collapse
Affiliation(s)
- Heng Chen
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, China
| | - Jian Yang
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Qingzhen Yang
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, China; MOE Key Laboratory of Biomedical Information Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Yuanbo Jia
- Department of Hepatobiliary Surgery and Liver Transplantation, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, P.R. China; Key Laboratory of Surgical Critical Care and Life Support (Xi'an Jiaotong University), Ministry of Education, Xi'an 710004, P.R. China; TFX Group-Xi'an Jiaotong University Institute of Life Health, Xi'an 710049, P.R. China.
| | - Xiaogang Guo
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China.
| |
Collapse
|
29
|
Chastney MR, Kaivola J, Leppänen VM, Ivaska J. The role and regulation of integrins in cell migration and invasion. Nat Rev Mol Cell Biol 2025; 26:147-167. [PMID: 39349749 DOI: 10.1038/s41580-024-00777-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/23/2024] [Indexed: 01/29/2025]
Abstract
Integrin receptors are the main molecular link between cells and the extracellular matrix (ECM) as well as mediating cell-cell interactions. Integrin-ECM binding triggers the formation of heterogeneous multi-protein assemblies termed integrin adhesion complexes (IACs) that enable integrins to transform extracellular cues into intracellular signals that affect many cellular processes, especially cell motility. Cell migration is essential for diverse physiological and pathological processes and is dysregulated in cancer to favour cell invasion and metastasis. Here, we discuss recent findings on the role of integrins in cell migration with a focus on cancer cell dissemination. We review how integrins regulate the spatial distribution and dynamics of different IACs, covering classical focal adhesions, emerging adhesion types and adhesion regulation. We discuss the diverse roles integrins have during cancer progression from cell migration across varied ECM landscapes to breaching barriers such as the basement membrane, and eventual colonization of distant organs.
Collapse
Affiliation(s)
- Megan R Chastney
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Jasmin Kaivola
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Veli-Matti Leppänen
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Johanna Ivaska
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland.
- Department of Life Technologies, University of Turku, Turku, Finland.
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland.
- Western Finnish Cancer Center (FICAN West), University of Turku, Turku, Finland.
- Foundation for the Finnish Cancer Institute, Helsinki, Finland.
| |
Collapse
|
30
|
Dutta Gupta S, Pal N, Ta M. Vitronectin regulates focal adhesion turnover and migration of human placenta-derived MSCs under nutrient stress. Eur J Cell Biol 2025; 104:151477. [PMID: 39893799 DOI: 10.1016/j.ejcb.2025.151477] [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: 10/07/2024] [Revised: 01/23/2025] [Accepted: 01/23/2025] [Indexed: 02/04/2025] Open
Abstract
At sites of tissue damage and wound healing, the mesenchymal stem cells (MSCs) are often challenged by nutrient availability due to blood supply disruption. Thus, it becomes critical to identify novel factors and their mechanism of action in regulating the adhesion and migration of MSCs under nutrient stress condition for successful clinical application. In human placenta-derived MSCs (PL-MSCs), we demonstrated an increase in cell spread area, along with increased adhesion and reduced migration of the cells, when cultured under nutrient stress condition. Correspondingly, an increase in the total number per cell and size of focal adhesions (FAs), together with prominent stress fibers were observed in nutrient-stressed PL-MSCs compared to control PL-MSCs. The FAs were demonstrated to be more stable, exhibiting slower turnover and longer lifespan. Vitronectin (VTN), an ECM glycoprotein, was upregulated under nutrient stress condition. Knockdown of VTN in PL-MSCs led to a significant reduction in the total number per cell and size of FAs, along with their faster turnover and shorter lifespan. Subsequently, a reversal in the cell spread area, adhesion and migration properties of the nutrient-stressed PL-MSCs were noted. Additionally, our findings indicated that VTN, as an upstream regulator, stimulated the phosphorylation of myosin light chain, which possibly promoted the maturation and stability of FAs along with assembly of stress fibers, thereby leading to increased adhesion and reduced migration of the cells. Overall, our study defines a distinct role of VTN as a critical regulator of migration in PL-MSCs under nutrient stress condition.
Collapse
Affiliation(s)
- Srishti Dutta Gupta
- Indian Institute of Science Education and Research, Kolkata (IISER Kolkata), India.
| | - Nitish Pal
- Indian Institute of Science Education and Research, Kolkata (IISER Kolkata), India.
| | - Malancha Ta
- Indian Institute of Science Education and Research, Kolkata (IISER Kolkata), India.
| |
Collapse
|
31
|
Katoh K. Integrin and Its Associated Proteins as a Mediator for Mechano-Signal Transduction. Biomolecules 2025; 15:166. [PMID: 40001469 PMCID: PMC11853369 DOI: 10.3390/biom15020166] [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: 11/21/2024] [Revised: 01/11/2025] [Accepted: 01/20/2025] [Indexed: 02/27/2025] Open
Abstract
Mechano-signal transduction is a process in which cells perceive extracellular mechanical signals, convert them into intracellular biochemical signals, and produce a response. Integrins are cell surface receptors that sense the extracellular mechanical cues and bind to the extracellular matrix (ECM). This binding induces integrin clustering and activation. Cytoplasmic tails of activated integrins interact and induce cytoskeleton tensions via several adaptor proteins. Integrins monitor extracellular stiffness via cytoskeleton tensions and modulate ECM stiffness via downstream signaling pathways regulating the expression of genes of ECM components. Integrin-mediated mechano-transduction is very crucial for the cell as it regulates the cell physiology both in normal and diseased conditions according to extracellular mechanical cues. It regulates cell proliferation, survival, and migration. Abnormal mechanical cues such as extreme and prolonged mechanical stress result in pathological conditions including fibrosis, cancers, skin, and autoimmune disorders. This paper aims to explore the role of integrins and their associated proteins in mechano-signal transduction. It highlights the integrins and their associated proteins as targets for therapy development. Furthermore, it also presents the challenges to the targeted drug development, which can be drug resistance and cytotoxicity. It is concluded in this paper that research on integrin-mediated mechano-signal transduction and its relationship with cell physiology and pathologies will be an important step towards the development of effective therapies.
Collapse
Affiliation(s)
- Kazuo Katoh
- Laboratory of Human Anatomy and Cell Biology, Faculty of Health Sciences, Tsukuba University of Technology, Tsukuba 305-8521, Japan
| |
Collapse
|
32
|
Xiao J, Ang JW, Zhong X, Wong DCP, T T, Yow I, Lee CJM, Foo RSY, Kanchanawong P, Low BC. Coordination of Focal Adhesion Nanoarchitecture and Dynamics in Mechanosensing for Cardiomyoblast Differentiation. ACS APPLIED MATERIALS & INTERFACES 2025; 17:4463-4479. [PMID: 39778877 PMCID: PMC11758775 DOI: 10.1021/acsami.4c15459] [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: 09/10/2024] [Revised: 11/21/2024] [Accepted: 11/21/2024] [Indexed: 01/11/2025]
Abstract
Focal adhesions (FAs) are force-bearing multiprotein complexes, whose nanoscale organization and signaling are essential for cell growth and differentiation. However, the specific organization of FA components to exert spatiotemporal activation of FA proteins for force sensing and transduction remains unclear. In this study, we unveil the intricacies of FA protein nanoarchitecture and that its dynamics are coordinated by a molecular scaffold protein, BNIP-2, to initiate downstream signal transduction for cardiomyoblast differentiation. Within the FAs, BNIP-2 regulates the nano-organization of focal adhesion kinase (FAK), and the dynamics of FAK, paxillin, and vinculin. Depletion of BNIP-2 resulted in altered focal adhesion numbers and sizes per cell, reduced traction force, and decreased FA sensitivity for mechanosensing. At the molecular level, the loss of BNIP-2 disrupted the FAK-paxillin signaling axis, where FAK inhibition reproduces the effects of BNIP-2 loss by impairing the phosphorylation of both FAK and paxillin. Mechanistically, BNIP-2 preferentially binds to constitutively active FAK and acts as a molecular scaffold to mediate interactions between FAK and paxillin and between paxillin and vinculin. We have validated BNIP-2's role in the FAK-paxillin signaling axis in human embryonic stem cells (hESC). Furthermore, we showed that depletion of BNIP-2 resulted in changes in signature gene targets at the cardiac progenitor stage of differentiation. In summary, we showed that the intricate interplay of FA nanoarchitecture and dynamics, governed by BNIP-2, is crucial for force transduction and biochemical signaling in driving cardiomyoblast differentiation.
Collapse
Affiliation(s)
- Jingwei Xiao
- Mechanobiology
Institute Singapore, National University
of Singapore, Singapore 117411, Singapore
| | - Jing Wen Ang
- Mechanobiology
Institute Singapore, National University
of Singapore, Singapore 117411, Singapore
| | - Xueying Zhong
- Mechanobiology
Institute Singapore, National University
of Singapore, Singapore 117411, Singapore
| | - Darren Chen Pei Wong
- Mechanobiology
Institute Singapore, National University
of Singapore, Singapore 117411, Singapore
- Department
of Biological Sciences, National University
of Singapore, Singapore 117558, Singapore
| | - Thivakar T
- Mechanobiology
Institute Singapore, National University
of Singapore, Singapore 117411, Singapore
| | - Ivan Yow
- Mechanobiology
Institute Singapore, National University
of Singapore, Singapore 117411, Singapore
| | - Chang Jie Mick Lee
- Institute
of Molecular and Cell Biology, A*STAR, Singapore 138673, Singapore
- Department
of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
- Cardiovascular
Metabolic Disease Translational Research Programme, National University Health System, Centre for Translational Medicine, Singapore 117599, Singapore
| | - Roger S-Y Foo
- Institute
of Molecular and Cell Biology, A*STAR, Singapore 138673, Singapore
- Department
of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
- Cardiovascular
Metabolic Disease Translational Research Programme, National University Health System, Centre for Translational Medicine, Singapore 117599, Singapore
| | - Pakorn Kanchanawong
- Mechanobiology
Institute Singapore, National University
of Singapore, Singapore 117411, Singapore
- Department
of Biomedical Engineering, National University
of Singapore, Singapore 117583, Singapore
| | - Boon Chuan Low
- Mechanobiology
Institute Singapore, National University
of Singapore, Singapore 117411, Singapore
- Department
of Biological Sciences, National University
of Singapore, Singapore 117558, Singapore
- NUS
College, National University of Singapore, Singapore 138593, Singapore
| |
Collapse
|
33
|
Li Z, Tang J, Zhou L, Mao J, Wang W, Huang Z, Zhang L, Wu J, Jiang X, Ding Z, Xi K, Cai F, Gu Y, Chen L. MicroSphere 3D Structures Delay Tissue Senescence through Mechanotransduction. ACS NANO 2025; 19:2695-2714. [PMID: 39787443 DOI: 10.1021/acsnano.4c14874] [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: 01/12/2025]
Abstract
The extracellular matrix (ECM) stores signaling molecules and facilitates mechanical and biochemical signaling in cells. However, the influence of biomimetic "rejuvenation" ECM structures on aging- and degeneration-related cellular activities and tissue repair is not well understood. We combined physical extrusion and precise "on-off" alternating cross-linking methods to create anisotropic biomaterial microgels (MicroRod and MicroSphere) and explored how they regulate the cell activities of the nucleus pulposus (NP) and their potential antidegenerative effects on intervertebral discs. NP cells exhibited aligned growth along the surface of the MicroRod, enhanced proliferation, and reduced apoptosis. This suggests an adaptive cellular response involving adhesion and mechanosensing, which causes cytoskeletal extension via environmental cues. NP cells maintain nuclear membrane integrity through the YAP/TAZ pathway, which activates the cGAS-STING pathway to rectify the aging mechanisms. In vivo, MicroRod carries NP cells and reduces inflammatory factor and protease secretion in degenerated intervertebral discs, inhibiting degeneration and promoting NP tissue regeneration. Our findings highlight the role of mechanical stress in maintaining cellular activity and antiaging effects in harsh environments, providing a foundation for further research and development of antidegenerative biomaterials.
Collapse
Affiliation(s)
- Ziang Li
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Soochow, Jiangsu 215000, China
| | - Jincheng Tang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Soochow, Jiangsu 215000, China
| | - Liang Zhou
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Soochow, Jiangsu 215000, China
| | - Jiannan Mao
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Soochow, Jiangsu 215000, China
| | - Wei Wang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Soochow, Jiangsu 215000, China
| | - Ziyan Huang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Soochow, Jiangsu 215000, China
| | - Lichen Zhang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Soochow, Jiangsu 215000, China
| | - Jie Wu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Soochow, Jiangsu 215000, China
| | - Xinzhao Jiang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Soochow, Jiangsu 215000, China
| | - Zhouye Ding
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Soochow, Jiangsu 215000, China
| | - Kun Xi
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Soochow, Jiangsu 215000, China
| | - Feng Cai
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Soochow, Jiangsu 215000, China
| | - Yong Gu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Soochow, Jiangsu 215000, China
| | - Liang Chen
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Soochow, Jiangsu 215000, China
| |
Collapse
|
34
|
Jain K, Kishan K, Minhaj RF, Kanchanawong P, Sheetz MP, Changede R. Immobile Integrin Signaling Transit and Relay Nodes Organize Mechanosignaling through Force-Dependent Phosphorylation in Focal Adhesions. ACS NANO 2025; 19:2070-2088. [PMID: 39760672 DOI: 10.1021/acsnano.4c03214] [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] [Indexed: 01/07/2025]
Abstract
Transmembrane signaling receptors, such as integrins, organize as nanoclusters that provide several advantages, including increasing avidity, sensitivity (increasing the signal-to-noise ratio), and robustness (signaling threshold) of the signal in contrast to signaling by single receptors. Furthermore, compared to large micron-sized clusters, nanoclusters offer the advantage of rapid turnover for the disassembly of the signal. However, whether nanoclusters function as signaling hubs remains poorly understood. Here, we employ fluorescence nanoscopy combined with photoactivation and photobleaching at subdiffraction limited resolution of ∼100 nm length scale within a focal adhesion to examine the dynamics of diverse focal adhesion proteins. We show that (i) subregions of focal adhesions are enriched in an immobile population of integrin β3 organized as nanoclusters, which (ii) in turn serve to organize nanoclusters of associated key adhesome proteins-vinculin, focal adhesion kinase (FAK) and paxillin, demonstrating that signaling proceeds by formation of nanoclusters rather than through individual proteins. (iii) Distinct focal adhesion protein nanoclusters exhibit distinct protein dynamics, which is closely correlated to their function in signaling. (iv) Long-lived nanoclusters function as signaling hubs─wherein immobile integrin nanoclusters organize phosphorylated FAK to form stable nanoclusters in close proximity to them, which are disassembled in response to inactivation signal by removal of force and in turn activation of phosphatase PTPN12. (v) Signaling takes place in response to external signals such as force or geometric arrangement of the nanoclusters and when the signal is removed, these nanoclusters disassemble. We term these functional nanoclusters as integrin signaling transit and relay nodes (STARnodes). Taken together, these results demonstrate that integrin STARnodes seed signaling downstream of the integrin receptors by organizing hubs of signaling proteins (FAK, paxillin, vinculin) to relay the incoming signal intracellularly and bring about robust function.
Collapse
Affiliation(s)
- Kashish Jain
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore
| | - Kishan Kishan
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore
- Neurobit Inc., New York, New York 10036, United States
| | - Rida F Minhaj
- Department of Biomedical Engineering, National University of Singapore, Singapore 117583, Singapore
| | - Pakorn Kanchanawong
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore
- Department of Biomedical Engineering, National University of Singapore, Singapore 117583, Singapore
| | - Michael P Sheetz
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore
- Molecular Mechanomedicine Program, Biochemistry and Molecular Biology Department, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - Rishita Changede
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore
- Teora Pte. Ltd, Singapore 139955, Singapore
| |
Collapse
|
35
|
Tran JC, Kuffner CJ, Marzilli AM, Miller RE, Silfen ZE, McMahan JB, Sloas DC, Chen CS, Ngo JT. Fluorescein-based SynNotch adaptors for regulating gene expression responses to diverse extracellular and matrix-based cues. Nat Commun 2025; 16:852. [PMID: 39833147 PMCID: PMC11756391 DOI: 10.1038/s41467-025-56148-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 01/09/2025] [Indexed: 01/22/2025] Open
Abstract
Synthetic Notch (SynNotch) receptors function like natural Notch proteins and can be used to install customized sense-and-respond capabilities into mammalian cells. Here, we introduce an adaptor-based strategy for regulating SynNotch activity via fluorescein isomers and analogs. Using an optimized fluorescein-binding SynNotch receptor, we describe ways to chemically control SynNotch signaling, including an approach based on a bio-orthogonal chemical ligation and a spatially controllable strategy via the photo-patterned uncaging of an o-nitrobenzyl-caged fluorescein conjugate. We further show that fluorescein-conjugated extracellular matrix (ECM)-binding peptides can be used to regulate SynNotch activity depending on the folding state of collagen-based ECM networks. To demonstrate the utility of these tools, we apply them to activate dose-dependent gene expression responses and to induce myogenic-like phenotypes in multipotent fibroblasts with spatiotemporal and microenvironmental control. Overall, we introduce an optimized fluorescein-binding SynNotch as a versatile tool for regulating transcriptional responses to ligands based on the clinically-approved fluorescein dye.
Collapse
Affiliation(s)
- Jeremy C Tran
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
- Biological Design Center, Boston University, Boston, MA, USA
| | - Christopher J Kuffner
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
- Biological Design Center, Boston University, Boston, MA, USA
| | - Alexander M Marzilli
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
- Biological Design Center, Boston University, Boston, MA, USA
| | - Ryan Emily Miller
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
- Biological Design Center, Boston University, Boston, MA, USA
| | - Zachary E Silfen
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
- Biological Design Center, Boston University, Boston, MA, USA
| | - Jeffrey B McMahan
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
- Biological Design Center, Boston University, Boston, MA, USA
| | - D Christopher Sloas
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
- Biological Design Center, Boston University, Boston, MA, USA
| | - Christopher S Chen
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
- Biological Design Center, Boston University, Boston, MA, USA
- Center for Multiscale & Translational Mechanobiology, Boston University, Boston, MA, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - John T Ngo
- Department of Biomedical Engineering, Boston University, Boston, MA, USA.
- Biological Design Center, Boston University, Boston, MA, USA.
- Center for Multiscale & Translational Mechanobiology, Boston University, Boston, MA, USA.
| |
Collapse
|
36
|
Yu C, Chen Y, Luo H, Lin W, Lin X, Jiang Q, Liu H, Liu W, Yang J, Huang Y, Fang J, He D, Han Y, Zheng S, Ren H, Xia X, Yu J, Chen L, Zeng C. NAT10 promotes vascular remodelling via mRNA ac4C acetylation. Eur Heart J 2025; 46:288-304. [PMID: 39453784 DOI: 10.1093/eurheartj/ehae707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 07/19/2024] [Accepted: 10/01/2024] [Indexed: 10/27/2024] Open
Abstract
BACKGROUND AND AIMS Vascular smooth muscle cell (VSMC) phenotype switching is a pathological hallmark in various cardiovascular diseases. N4-acetylcytidine (ac4C) catalyzed by N-acetyltransferase 10 (NAT10) is well conserved in the enzymatic modification of ribonucleic acid (RNA). NAT10-mediated ac4C acetylation is involved in various physiological and pathological processes, including cardiac remodelling. However, the biological functions and underlying regulatory mechanisms of mRNA ac4C modifications in vascular diseases remain elusive. METHODS By combining in-vitro and in-vivo vascular injury models, NAT10 was identified as a crucial protein involved in the promotion of post-injury neointima formation, as well as VSMC phenotype switching. The potential mechanisms of NAT10 in the vascular neointima formation were clarified by RNA sequence (RNA-seq), acetylated mRNA immunoprecipitation sequence (acRIP-seq), and RNA binding protein immunoprecipitation sequence (RIP-seq). RESULTS NAT10 and ac4C modifications were upregulated in injured human and rodent arteries. Deletion of NAT10 in VSMCs effectively reduced post-injury neointima formation and VSMC phenotype switching. Further RNA-seq, RIP-seq, and acRIP-seq revealed that NAT10, by its ac4C modification, directly interacts with genes, including integrin-β1 (ITGB1) and collagen type I alpha 2 chain (Col1a2) mRNAs. Taking ITGB1 as one example, it showed that NAT10-mediated ac4C consequently increased ITGB1 mRNA stability and its downstream focal adhesion kinase (FAK) signaling, directly influencing the proliferation of VSMCs and vascular remodelling. The regulation of NAT10 on the VSMC phenotype is of translational significance because the administration of Remodelin, a NAT10 inhibitor, effectively prevents neointima formation by suppressing VSMC proliferation and downregulating ITGB1 expression and deactivating its FAK signaling. CONCLUSIONS This study reveals that NAT10 promotes vascular remodelling via mRNA ac4C acetylation, which may be a promising therapeutic target against vascular remodelling.
Collapse
Affiliation(s)
- Cheng Yu
- Department of Cardiology, Fujian Medical Center for Cardiovascular Diseases, Fujian Institute of Coronary Heart Disease, Fujian Medical University Union Hospital, Fuzhou, P.R. China
- Department of Cardiology, Daping Hospital, The Third Military Medical University, 10th Changjiangzhilu Road, Yuzhong District, Chongqing 400042, P.R. China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Ministry of Education of China, Chongqing Key Laboratory for Hypertension Research, State Key Laboratory of Trauma and Chemical Poisoning, Daping Hospital, The Third Military Medical University, 10th Changjiangzhilu Road, Yuzhong District, Chongqing 400042, P.R. China
- Chongqing Institute of Cardiology, 10th Changjiangzhilu Road, Yuzhong District, Chongqing 400042, P.R. China
| | - Yue Chen
- Department of Cardiology, Daping Hospital, The Third Military Medical University, 10th Changjiangzhilu Road, Yuzhong District, Chongqing 400042, P.R. China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Ministry of Education of China, Chongqing Key Laboratory for Hypertension Research, State Key Laboratory of Trauma and Chemical Poisoning, Daping Hospital, The Third Military Medical University, 10th Changjiangzhilu Road, Yuzhong District, Chongqing 400042, P.R. China
- Chongqing Institute of Cardiology, 10th Changjiangzhilu Road, Yuzhong District, Chongqing 400042, P.R. China
| | - Hao Luo
- Department of Cardiology, Daping Hospital, The Third Military Medical University, 10th Changjiangzhilu Road, Yuzhong District, Chongqing 400042, P.R. China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Ministry of Education of China, Chongqing Key Laboratory for Hypertension Research, State Key Laboratory of Trauma and Chemical Poisoning, Daping Hospital, The Third Military Medical University, 10th Changjiangzhilu Road, Yuzhong District, Chongqing 400042, P.R. China
- Chongqing Institute of Cardiology, 10th Changjiangzhilu Road, Yuzhong District, Chongqing 400042, P.R. China
| | - Weihong Lin
- Department of Cardiology, Fujian Medical Center for Cardiovascular Diseases, Fujian Institute of Coronary Heart Disease, Fujian Medical University Union Hospital, Fuzhou, P.R. China
| | - Xin Lin
- Department of Cardiology, Daping Hospital, The Third Military Medical University, 10th Changjiangzhilu Road, Yuzhong District, Chongqing 400042, P.R. China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Ministry of Education of China, Chongqing Key Laboratory for Hypertension Research, State Key Laboratory of Trauma and Chemical Poisoning, Daping Hospital, The Third Military Medical University, 10th Changjiangzhilu Road, Yuzhong District, Chongqing 400042, P.R. China
- Chongqing Institute of Cardiology, 10th Changjiangzhilu Road, Yuzhong District, Chongqing 400042, P.R. China
| | - Qiong Jiang
- Department of Cardiology, Fujian Medical Center for Cardiovascular Diseases, Fujian Institute of Coronary Heart Disease, Fujian Medical University Union Hospital, Fuzhou, P.R. China
| | - Hongjin Liu
- Department of Cardiovascular Surgery, Fujian Medical University Union Hospital, Key Laboratory of Cardio-Thoracic Surgery, Fujian Province University, Fuzhou, P.R. China
| | - Wenkun Liu
- Department of Cardiology, Fujian Medical Center for Cardiovascular Diseases, Fujian Institute of Coronary Heart Disease, Fujian Medical University Union Hospital, Fuzhou, P.R. China
| | - Jing Yang
- Department of Cardiology, Fujian Medical Center for Cardiovascular Diseases, Fujian Institute of Coronary Heart Disease, Fujian Medical University Union Hospital, Fuzhou, P.R. China
| | - Yu Huang
- Department of Cardiology, Fujian Medical Center for Cardiovascular Diseases, Fujian Institute of Coronary Heart Disease, Fujian Medical University Union Hospital, Fuzhou, P.R. China
| | - Jun Fang
- Department of Cardiology, Fujian Medical Center for Cardiovascular Diseases, Fujian Institute of Coronary Heart Disease, Fujian Medical University Union Hospital, Fuzhou, P.R. China
| | - Duofen He
- Department of Cardiology, Daping Hospital, The Third Military Medical University, 10th Changjiangzhilu Road, Yuzhong District, Chongqing 400042, P.R. China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Ministry of Education of China, Chongqing Key Laboratory for Hypertension Research, State Key Laboratory of Trauma and Chemical Poisoning, Daping Hospital, The Third Military Medical University, 10th Changjiangzhilu Road, Yuzhong District, Chongqing 400042, P.R. China
- Chongqing Institute of Cardiology, 10th Changjiangzhilu Road, Yuzhong District, Chongqing 400042, P.R. China
| | - Yu Han
- Department of Cardiology, Daping Hospital, The Third Military Medical University, 10th Changjiangzhilu Road, Yuzhong District, Chongqing 400042, P.R. China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Ministry of Education of China, Chongqing Key Laboratory for Hypertension Research, State Key Laboratory of Trauma and Chemical Poisoning, Daping Hospital, The Third Military Medical University, 10th Changjiangzhilu Road, Yuzhong District, Chongqing 400042, P.R. China
- Chongqing Institute of Cardiology, 10th Changjiangzhilu Road, Yuzhong District, Chongqing 400042, P.R. China
| | - Shuo Zheng
- Department of Cardiology, Daping Hospital, The Third Military Medical University, 10th Changjiangzhilu Road, Yuzhong District, Chongqing 400042, P.R. China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Ministry of Education of China, Chongqing Key Laboratory for Hypertension Research, State Key Laboratory of Trauma and Chemical Poisoning, Daping Hospital, The Third Military Medical University, 10th Changjiangzhilu Road, Yuzhong District, Chongqing 400042, P.R. China
- Chongqing Institute of Cardiology, 10th Changjiangzhilu Road, Yuzhong District, Chongqing 400042, P.R. China
| | - Hongmei Ren
- Department of Cardiology, Daping Hospital, The Third Military Medical University, 10th Changjiangzhilu Road, Yuzhong District, Chongqing 400042, P.R. China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Ministry of Education of China, Chongqing Key Laboratory for Hypertension Research, State Key Laboratory of Trauma and Chemical Poisoning, Daping Hospital, The Third Military Medical University, 10th Changjiangzhilu Road, Yuzhong District, Chongqing 400042, P.R. China
- Chongqing Institute of Cardiology, 10th Changjiangzhilu Road, Yuzhong District, Chongqing 400042, P.R. China
| | - Xuewei Xia
- Department of Cardiology, Daping Hospital, The Third Military Medical University, 10th Changjiangzhilu Road, Yuzhong District, Chongqing 400042, P.R. China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Ministry of Education of China, Chongqing Key Laboratory for Hypertension Research, State Key Laboratory of Trauma and Chemical Poisoning, Daping Hospital, The Third Military Medical University, 10th Changjiangzhilu Road, Yuzhong District, Chongqing 400042, P.R. China
- Chongqing Institute of Cardiology, 10th Changjiangzhilu Road, Yuzhong District, Chongqing 400042, P.R. China
| | - Junyi Yu
- Department of Cardiology, Daping Hospital, The Third Military Medical University, 10th Changjiangzhilu Road, Yuzhong District, Chongqing 400042, P.R. China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Ministry of Education of China, Chongqing Key Laboratory for Hypertension Research, State Key Laboratory of Trauma and Chemical Poisoning, Daping Hospital, The Third Military Medical University, 10th Changjiangzhilu Road, Yuzhong District, Chongqing 400042, P.R. China
- Chongqing Institute of Cardiology, 10th Changjiangzhilu Road, Yuzhong District, Chongqing 400042, P.R. China
| | - Lianglong Chen
- Department of Cardiology, Fujian Medical Center for Cardiovascular Diseases, Fujian Institute of Coronary Heart Disease, Fujian Medical University Union Hospital, Fuzhou, P.R. China
| | - Chunyu Zeng
- Department of Cardiology, Fujian Medical Center for Cardiovascular Diseases, Fujian Institute of Coronary Heart Disease, Fujian Medical University Union Hospital, Fuzhou, P.R. China
- Department of Cardiology, Daping Hospital, The Third Military Medical University, 10th Changjiangzhilu Road, Yuzhong District, Chongqing 400042, P.R. China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Ministry of Education of China, Chongqing Key Laboratory for Hypertension Research, State Key Laboratory of Trauma and Chemical Poisoning, Daping Hospital, The Third Military Medical University, 10th Changjiangzhilu Road, Yuzhong District, Chongqing 400042, P.R. China
- Chongqing Institute of Cardiology, 10th Changjiangzhilu Road, Yuzhong District, Chongqing 400042, P.R. China
- Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, P.R. China
| |
Collapse
|
37
|
Pally D, Kapoor N, Naba A. The novel ECM protein SNED1 mediates cell adhesion via the RGD-binding integrins α5β1 and αvβ3. J Cell Sci 2025; 138:JCS263479. [PMID: 39713860 PMCID: PMC11828466 DOI: 10.1242/jcs.263479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 12/15/2024] [Indexed: 12/24/2024] Open
Abstract
The extracellular matrix (ECM) is a complex meshwork comprising over 100 proteins. It serves as an adhesive substrate for cells and, hence, plays crucial roles in health and disease. We have recently identified a novel ECM protein, SNED1, and have found that it is required for neural crest cell migration and craniofacial morphogenesis during development and in breast cancer, where it is necessary for the metastatic dissemination of tumor cells. Interestingly, both processes involve the dynamic remodeling of cell-ECM adhesions via cell surface receptors. Sequence analysis revealed that SNED1 contains two amino acid motifs, RGD and LDV, known to bind integrins, the largest class of ECM receptors. We thus sought to investigate the role of SNED1 in cell adhesion. Here, we report that SNED1 mediates breast cancer and neural crest cell adhesion via its RGD motif. We further demonstrate that cell adhesion to SNED1 is mediated by the RGD integrins α5β1 and αvβ3. These findings are a first step toward identifying the signaling pathways activated downstream of the SNED1-integrin interactions guiding craniofacial morphogenesis and breast cancer metastasis.
Collapse
Affiliation(s)
- Dharma Pally
- Department of Physiology and Biophysics, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Nandini Kapoor
- Department of Physiology and Biophysics, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Alexandra Naba
- Department of Physiology and Biophysics, University of Illinois Chicago, Chicago, IL 60612, USA
- University of Illinois Cancer Center, Chicago, IL 60612, USA
| |
Collapse
|
38
|
Wang C, Quan Y, Jiang J, Yu H, Liu J, Tang W, Li X, Wang S, Huo D, Jiang GL, Yang Y, Ding Q. Protein Coronation-Induced Cancer Staging-Dependent Multilevel Cytotoxicity: An All-Humanized Study in Blood Vessel Organoids. ACS NANO 2025; 19:345-368. [PMID: 39743836 DOI: 10.1021/acsnano.4c07783] [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: 01/04/2025]
Abstract
The protein corona effect refers to the phenomenon wherein nanomaterials in the bloodstream are coated by serum proteins, yet how protein coronated nanomaterials interact with blood vessels and its toxicity implications remain poorly understood. In this study, we investigated protein corona-related vessel toxicity by using an all-humanized assay integrating blood vessel organoids and patient-derived serum. Initially, we screened various nanomaterials to discern how parameters including size, morphology, hydrophobicity, surface charge, and chirality-dependent protein corona difference influence their uptake by vessel organoids. For nanomaterials showing substantial differences in vessel uptake, their protein corona was analyzed by using label-free mass spectra. Our findings revealed the involvement of cancer staging-related cytoskeleton components in mediating preferential uptake by cells, including endothelial and mural cells. Additionally, a transcriptome study was conducted to elucidate the influence of nanomaterials. We confirmed that protein coronated nanomaterials provoke remodeling at both transcriptional and translational levels, impacting pathways such as PI3K-Akt/Hippo/Wnt, and membraneless organelle integrity, respectively. Our study further demonstrated that the remodeling potential of patient-derived protein coronated nanomaterials can be harnessed to synergize with antiangiogenesis therapeutics to improve the outcomes. We anticipate that this study will provide guidance for the safe use of nanomedicine in the future.
Collapse
Affiliation(s)
- Chan Wang
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing 211166, P. R. China
| | - Yingyi Quan
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing 211166, P. R. China
| | - Jiang Jiang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing 211166, P. R. China
| | - Han Yu
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing 211166, P. R. China
| | - Jia Liu
- Department of Medicinal Chemistry, School of Pharmacy, Nanjing Medical University, Nanjing 211166, P. R. China
| | - Wei Tang
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing 211166, P. R. China
| | - Xinyue Li
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing 211166, P. R. China
| | - Shouju Wang
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 211166, P. R. China
| | - Da Huo
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Department of Pharmaceutics, Nanjing Medical University, Nanjing 211169, P. R. China
| | - Guang-Liang Jiang
- Department of Urology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P. R. China
| | - Yang Yang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing 211166, P. R. China
| | - Qingqing Ding
- Department of Geriatric Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 211166, P. R. China
| |
Collapse
|
39
|
Liu J, Zhang B, Huang B, Zhang K, Guo F, Wang Z, Shang D. A stumbling block in pancreatic cancer treatment: drug resistance signaling networks. Front Cell Dev Biol 2025; 12:1462808. [PMID: 39872846 PMCID: PMC11770040 DOI: 10.3389/fcell.2024.1462808] [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: 07/10/2024] [Accepted: 12/30/2024] [Indexed: 01/30/2025] Open
Abstract
The primary node molecules in the cell signaling network in cancer tissues are maladjusted and mutated in comparison to normal tissues, which promotes the occurrence and progression of cancer. Pancreatic cancer (PC) is a highly fatal cancer with increasing incidence and low five-year survival rates. Currently, there are several therapies that target cell signaling networks in PC. However, PC is a "cold tumor" with a unique immunosuppressive tumor microenvironment (poor effector T cell infiltration, low antigen specificity), and targeting a single gene or pathway is basically ineffective in clinical practice. Targeted matrix therapy, targeted metabolic therapy, targeted mutant gene therapy, immunosuppressive therapy, cancer vaccines, and other emerging therapies have shown great therapeutic potential, but results have been disappointing. Therefore, we summarize the identified and potential drug-resistant cell signaling networks aimed at overcoming barriers to existing PC therapies.
Collapse
Affiliation(s)
- Jinming Liu
- Department of General Surgery, Pancreas and Biliary Center, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Biao Zhang
- Department of General Surgery, Pancreas and Biliary Center, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Bingqian Huang
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Department of Clinical Pharmacy, Affiliated Hangzhou First People’s Hospital, Westlake University, Hangzhou, China
| | - Kexin Zhang
- Central Laboratory, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Fujia Guo
- Central Laboratory, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Zhizhou Wang
- Department of General Surgery, Pancreas and Biliary Center, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Dong Shang
- Department of General Surgery, Pancreas and Biliary Center, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, China
| |
Collapse
|
40
|
Narain R, Muncie-Vasic JM, Weaver VM. Forcing the code: tension modulates signaling to drive morphogenesis and malignancy. Genes Dev 2025; 39:163-181. [PMID: 39638568 PMCID: PMC11789492 DOI: 10.1101/gad.352110.124] [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] [Indexed: 12/07/2024]
Abstract
Development and disease are regulated by the interplay between genetics and the signaling pathways stimulated by morphogens, growth factors, and cytokines. Experimental data highlight the importance of mechanical force in regulating embryonic development, tissue morphogenesis, and malignancy. Force not only sculpts tissue movements to drive embryogenesis and morphogenesis but also modifies the context of biochemical signaling and gene expression to regulate cell and tissue fate. Not surprisingly, experiments have demonstrated that perturbations in cell tension drive malignancy and metastasis by altering biochemical signaling and gene expression through modifications in cytoskeletal tension, transmembrane receptor structure and function, and organelle phenotype that enhance cell growth and survival, alter metabolism, and foster cell migration and invasion. At the tissue level, tumor-associated forces disrupt cell-cell adhesions to perturb tissue organization, compromise vascular integrity to induce hypoxia, and interfere with antitumor immunity to foster metastasis and treatment resistance. Exciting new approaches now exist with which to clarify the relationship between mechanotransduction, biochemical signaling, and gene expression in development and disease. Indeed, gaining insight into these interactions is essential to unravel molecular mechanisms that regulate development and clarify the molecular basis of cancer.
Collapse
Affiliation(s)
- Radhika Narain
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, California 94143, USA
- Graduate Program in Bioengineering, University of California, San Francisco and University of California, Berkeley, Berkeley, California 94720, USA
| | | | - Valerie M Weaver
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, California 94143, USA;
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, California 94143, USA
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, California 94143, USA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, San Francisco, California 94143
- UCSF Helen Diller Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California 94143, USA
| |
Collapse
|
41
|
Amouzadeh Tabrizi M, Ali AA, Singuru MMR, Mi L, Bhattacharyya P, You M. A portable and sensitive DNA-based electrochemical sensor for detecting piconewton-scale cellular forces. Anal Chim Acta 2025; 1333:343392. [PMID: 39615910 PMCID: PMC11609405 DOI: 10.1016/j.aca.2024.343392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 10/31/2024] [Accepted: 11/02/2024] [Indexed: 02/12/2025]
Abstract
BACKGROUND Cell-generated forces are a key player in cell biology, especially during cellular shape formation, migration, cancer development, and immune response. The measurement of forces exerted and experienced by cells is fundamental in understanding these mechanosensitive cellular behaviors. While cell-generated forces can now be detected based on techniques like fluorescence microscopy, atomic force microscopy, optical/magnetic tweezers, however, most of these approaches rely on complicated instruments or materials, as well as skilled operators, which could limit their potential broad applications in regular biological laboratories. RESULTS A new type of smartphone-based electrochemical sensor is developed here for cellular force measurement. In this system, a double-stranded DNA-based force probe, known as tension gauge tether, is attached to the surface of a gold screen-printed electrode, which is then incorporated into a portable smartphone-based electrochemical device. Cellular force-induced DNA detachment on the sensor surface results in multiple redox reporters to reach the surface of the electrode and generate enhanced electrochemical signals. To further improve the sensitivity, a CRISPR-Cas12a system has also been incorporated to cleave the remaining surface-attached anchor DNA strand. Using integrin-mediated tension as an example, piconewton-scale adhesion forces generated by ≤ 10 HeLa cells could now be reliably detected. Meanwhile, the threshold forces of these electrochemical sensors can also be modularly tuned to detect different levels of cellular forces. SIGNIFICANCE These novel DNA-based highly sensitive, portable, cost-efficient, and easy-to-use electrochemical sensors can be potentially powerful tools for detecting different cell-generated molecular forces. Functioning as complementary tools with traction force microscopy and fluorescent probes, these electrochemical sensors can be straightforwardly applied in regular biological laboratories for understanding the basic mechanical principles of cell signaling and for developing novel strategies and materials in tissue engineering, regenerative medicine, and cell therapy.
Collapse
Affiliation(s)
| | - Ahsan Ausaf Ali
- Department of Chemistry, University of Massachusetts, Amherst, MA, 01003, USA
| | | | - Lan Mi
- Department of Chemistry, University of Massachusetts, Amherst, MA, 01003, USA
| | | | - Mingxu You
- Department of Chemistry, University of Massachusetts, Amherst, MA, 01003, USA; Molecular and Cellular Biology Program, University of Massachusetts, Amherst, MA, 01003, USA.
| |
Collapse
|
42
|
Gonçalves M, Lopes C, Alégot H, Osswald M, Bosveld F, Ramos C, Richard G, Bellaiche Y, Mirouse V, Morais-de-Sá E. The Dystrophin-Dystroglycan complex ensures cytokinesis efficiency in Drosophila epithelia. EMBO Rep 2025; 26:307-328. [PMID: 39548266 PMCID: PMC11772804 DOI: 10.1038/s44319-024-00319-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 10/21/2024] [Accepted: 10/24/2024] [Indexed: 11/17/2024] Open
Abstract
Cytokinesis physically separates daughter cells at the end of cell division. This step is particularly challenging for epithelial cells, which are connected to their neighbors and to the extracellular matrix by transmembrane protein complexes. To systematically evaluate the impact of the cell adhesion machinery on epithelial cytokinesis efficiency, we performed an RNAi-based modifier screen in the Drosophila follicular epithelium. Strikingly, this unveiled adhesion molecules and transmembrane receptors that facilitate cytokinesis completion. Among these is Dystroglycan, which connects the extracellular matrix to the cytoskeleton via Dystrophin. Live imaging revealed that Dystrophin and Dystroglycan become enriched in the ingressing membrane, below the cytokinetic ring, during and after ring constriction. Using multiple alleles, including Dystrophin isoform-specific mutants, we show that Dystrophin/Dystroglycan localization is linked with unanticipated roles in regulating cytokinetic ring contraction and in preventing membrane regression during the abscission period. Altogether, we provide evidence that, rather than opposing cytokinesis completion, the machinery involved in cell-cell and cell-matrix interactions has also evolved functions to ensure cytokinesis efficiency in epithelial tissues.
Collapse
Affiliation(s)
- Margarida Gonçalves
- IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135, Porto, Portugal
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135, Porto, Portugal
- Programa Doutoral em Biologia Molecular e Celular (MCBiology), Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Catarina Lopes
- IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135, Porto, Portugal
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135, Porto, Portugal
| | - Hervé Alégot
- Université Clermont Auvergne - iGReD (Institute of Genetics, Reproduction and Development), UMR CNRS 6293 - INSERM U1103, Faculté de Médecine, Clermont-Ferrand, France
| | - Mariana Osswald
- IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135, Porto, Portugal
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135, Porto, Portugal
| | - Floris Bosveld
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR3215, INSERM U934, Genetics and Developmental Biology, 75005, Paris, France
| | - Carolina Ramos
- IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135, Porto, Portugal
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135, Porto, Portugal
| | - Graziella Richard
- Université Clermont Auvergne - iGReD (Institute of Genetics, Reproduction and Development), UMR CNRS 6293 - INSERM U1103, Faculté de Médecine, Clermont-Ferrand, France
| | - Yohanns Bellaiche
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR3215, INSERM U934, Genetics and Developmental Biology, 75005, Paris, France
| | - Vincent Mirouse
- Université Clermont Auvergne - iGReD (Institute of Genetics, Reproduction and Development), UMR CNRS 6293 - INSERM U1103, Faculté de Médecine, Clermont-Ferrand, France
| | - Eurico Morais-de-Sá
- IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135, Porto, Portugal.
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135, Porto, Portugal.
| |
Collapse
|
43
|
Zihni C. Phagocytosis by the retinal pigment epithelium: New insights into polarized cell mechanics. Bioessays 2025; 47:e2300197. [PMID: 39663766 DOI: 10.1002/bies.202300197] [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: 10/12/2023] [Accepted: 10/21/2024] [Indexed: 12/13/2024]
Abstract
The retinal pigment epithelium (RPE) is a specialized epithelium at the back of the eye that carries out a variety of functions essential for visual health. Recent studies have advanced our molecular understanding of one of the major functions of the RPE; phagocytosis of spent photoreceptor outer segments (POS). Notably, a mechanical link, formed between apical integrins bound to extracellular POS and the intracellular actomyosin cytoskeleton, is proposed to drive the internalization of POS. The process may involve a "nibbling" action, as an initial step, to sever outer segment tips. These insights have led us to hypothesize an "integrin adhesome-like" network, atypically assembled at apical membrane RPE-POS contacts. I propose that this hypothetical network orchestrates the complex membrane remodeling events required for particle internalization. Therefore, its analysis and characterization will likely lead to a more comprehensive understanding of the molecular mechanisms that control POS phagocytosis.
Collapse
Affiliation(s)
- Ceniz Zihni
- Faculty of Health & Life Sciences, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| |
Collapse
|
44
|
Tomos I, Kanellopoulou P, Nastos D, Aidinis V. Pharmacological targeting of ECM homeostasis, fibroblast activation and invasion for the treatment of pulmonary fibrosis. Expert Opin Ther Targets 2025; 29:43-57. [PMID: 39985559 DOI: 10.1080/14728222.2025.2471579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 01/24/2025] [Accepted: 02/20/2025] [Indexed: 02/24/2025]
Abstract
INTRODUCTION Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive interstitial lung disease with a dismal prognosis. While the standard-of-care (SOC) drugs approved for IPF represent a significant advancement in antifibrotic therapies, they primarily slow disease progression and have limited overall efficacy and many side effects. Consequently, IPF remains a condition with high unmet medical and pharmacological needs. AREAS COVERED A wide variety of molecules and mechanisms have been implicated in the pathogenesis of IPF, many of which have been targeted in clinical trials. In this review, we discuss the latest therapeutic targets that affect extracellular matrix (ECM) homeostasis and the activation of lung fibroblasts, with a specific focus on ECM invasion. EXPERT OPINION A promising new approach involves targeting ECM invasion by fibroblasts, a process that parallels cancer cell behavior. Several cancer drugs are now being tested in IPF for their ability to inhibit ECM invasion, offering significant potential for future treatments. The delivery of these therapies by inhalation is a promising development, as it may enhance local effectiveness and minimize systemic side effects, thereby improving patient safety and treatment efficacy.
Collapse
Affiliation(s)
- Ioannis Tomos
- 5th Department of Respiratory Medicine, 'SOTIRIA' Chest Diseases Hospital of Athens, Athens, Greece
| | - Paraskevi Kanellopoulou
- Institute for Fundamental Biomedical Research, Biomedical Sciences Research Center Alexander Fleming, Athens, Greece
| | - Dimitris Nastos
- Institute for Fundamental Biomedical Research, Biomedical Sciences Research Center Alexander Fleming, Athens, Greece
| | - Vassilis Aidinis
- Institute for Fundamental Biomedical Research, Biomedical Sciences Research Center Alexander Fleming, Athens, Greece
| |
Collapse
|
45
|
Li S, Liu Y, Luo X, Hong W. Systematic Evaluation of Extracellular Coating Matrix on the Differentiation of Human-Induced Pluripotent Stem Cells to Cortical Neurons. Int J Mol Sci 2024; 26:230. [PMID: 39796088 PMCID: PMC11720352 DOI: 10.3390/ijms26010230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/20/2024] [Accepted: 12/23/2024] [Indexed: 01/30/2025] Open
Abstract
Induced pluripotent stem cell (iPSC)-derived neurons (iNs) have been widely used as models of neurodevelopment and neurodegenerative diseases. Coating cell culture vessels with extracellular matrixes (ECMs) gives structural support and facilitates cell communication and differentiation, ultimately enhances neuronal functions. However, the relevance of different ECMs to the natural environment and their impact on neuronal differentiation have not been fully characterized. In this study, we report the use of four commonly used extracellular matrixes, poly-D-lysine (PDL), poly-L-ornithine (PLO), Laminin and Matrigel, which we applied to compare the single-coating and double-coating conditions on iNs differentiation and maturation. Using the IncuCyte live-cell imaging system, we found that iNs cultured on single Matrigel- and Laminin-coated vessels have significantly higher density of neurite outgrowth and branch points than PLO or PDL but produce abnormal highly straight neurite outgrowth and larger cell body clumps. All the four double-coating conditions significantly reduced the clumping of neurons, in which the combination of PDL+Matrigel also enhanced neuronal purity. Double coating with PDL+Matrigel also tended to improve dendritic and axonal development and the distribution of pre and postsynaptic markers. These results demonstrate that the extracellular matrix contributes to the differentiation of cultured neurons and that double coating with PDL+Matrigel gives the best outcomes. Our study indicates that neuronal differentiation and maturation can be manipulated, to a certain extent, by adjusting the ECM recipe, and provides important technical guidance for the use of the ECM in neurological studies.
Collapse
Affiliation(s)
| | | | | | - Wei Hong
- Shenzhen Key Laboratory of Neuroimmunomodulation for Neurological Diseases, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; (S.L.); (Y.L.); (X.L.)
| |
Collapse
|
46
|
Horisaka H, Yokawa S, Suzuki R, Emoto R, Maeda R, Furuno T. Suppression of FcεRI-evoked Degranulation in RBL-2H3 Cells on Gelatin Methacryloyl Hydrogel. Cell Biochem Biophys 2024:10.1007/s12013-024-01657-3. [PMID: 39731647 DOI: 10.1007/s12013-024-01657-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/18/2024] [Indexed: 12/30/2024]
Abstract
Cell-extracellular matrix (ECM) interactions play multiple roles in developmental, physiological, and pathological processes. ECM stiffness substantially affects cellular morphology, migration, and function. In this study, we investigated the effect of ECM comprising gelatin methacryloyl (GelMA) on the activation of rat basophilic leukemia (RBL-2H3) cells, a model mast cell line. Maintenance of intracellular Ca2+ concentration ([Ca2+]i) elevation and subsequent degranulation, evoked by crosslinking the high-affinity IgE receptors (FcεRI), were significantly suppressed in RBL-2H3 cells on collagen-coated GelMA hydrogel than those on collagen-coated glass dishes and plastic wells. Thapsigargin and phorbol myristate acetate caused sustained [Ca2+]i increase and degranulation to a similar extent in cells on both GelMA hydrogel and plastic wells/glass dishes. F-actin was clearly accumulated along the periphery of RBL-2H3 cells in plane attached to glass, but not GelMA hydrogel, suggesting that the loose actin cytoskeleton of RBL-2H3 cells on GelMA hydrogel caused suppressive degranulation through unstable FcεRI aggregation.
Collapse
Affiliation(s)
- Haruna Horisaka
- School of Pharmacy, Aichi Gakuin University, 1-100 Kusumoto-cho, Chikusa-ku, Nagoya, 464-8650, Japan
| | - Satoru Yokawa
- School of Pharmacy, Aichi Gakuin University, 1-100 Kusumoto-cho, Chikusa-ku, Nagoya, 464-8650, Japan
| | - Ruriko Suzuki
- School of Pharmacy, Aichi Gakuin University, 1-100 Kusumoto-cho, Chikusa-ku, Nagoya, 464-8650, Japan
| | - Rin Emoto
- School of Pharmacy, Aichi Gakuin University, 1-100 Kusumoto-cho, Chikusa-ku, Nagoya, 464-8650, Japan
| | - Rino Maeda
- School of Pharmacy, Aichi Gakuin University, 1-100 Kusumoto-cho, Chikusa-ku, Nagoya, 464-8650, Japan
| | - Tadahide Furuno
- School of Pharmacy, Aichi Gakuin University, 1-100 Kusumoto-cho, Chikusa-ku, Nagoya, 464-8650, Japan.
| |
Collapse
|
47
|
Wu M, Yang H, Liu S, Jiang L, Liang T, Wang Y, Zhu M, Song X, Liu H, Shen J, Wang S, Zhu X, Qu CK, Cheng L, Jiang H, Ni F. Enhanced engraftment of human haematopoietic stem cells via mechanical remodelling mediated by the corticotropin-releasing hormone. Nat Biomed Eng 2024:10.1038/s41551-024-01316-1. [PMID: 39715892 DOI: 10.1038/s41551-024-01316-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 11/01/2024] [Indexed: 12/25/2024]
Abstract
The engraftment of haematopoietic stem and progenitor cells (HSPCs), particularly in cord-blood transplants, remains challenging. Here we report the role of the corticotropin-releasing hormone (CRH) in enhancing the homing and engraftment of human-cord-blood HSPCs in bone marrow through mechanical remodelling. By using microfluidics, intravital two-photon imaging and long-term-engraftment assays, we show that treatment with CRH substantially enhances HSPC adhesion, motility and mechanical remodelling, ultimately leading to improved bone-marrow homing and engraftment in immunodeficient mice. CRH induces Ras homologue gene family member A (RhoA)-dependent nuclear translocation of the yes-associated protein (YAP), which upregulates the expression of genes encoding extracellular-matrix proteins (notably, thrombospondin-2 (THBS2)). This process guides the mechanical remodelling of HSPCs via modulation of the actin cytoskeleton and the extracellular matrix, with THBS2 interacting with the integrin αvβ3 and coordinating the nuclear translocation of YAP upon CRH/CRH-receptor-1 (CRH/CRHR1) signalling. Overall, the CRH/CRHR1/RhoA/YAP/THBS2/αvβ3 axis has a central role in modulating HSPC behaviour via a mechanical feedback loop involving THBS2, αvβ3, the actin cytoskeleton and YAP signalling. Our findings may suggest avenues for optimizing the transplantation of HSPCs.
Collapse
Affiliation(s)
- Mingming Wu
- Department of Hematology, The First Affiliated Hospital of USTC, Key Laboratory of Immune Response and Immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Institute of Blood and Cell Therapy and Anhui Provincial Key Laboratory of Blood Research and Applications, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute of Immunology, The CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Haoxiang Yang
- The CAS Key Laboratory of Mechanical Behavior and Design of Materials, University of Science and Technology of China, Hefei, China
| | - Senquan Liu
- Department of Hematology, The First Affiliated Hospital of USTC, Key Laboratory of Immune Response and Immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Institute of Blood and Cell Therapy and Anhui Provincial Key Laboratory of Blood Research and Applications, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Lai Jiang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Tingting Liang
- Department of Hematology, The First Affiliated Hospital of USTC, Key Laboratory of Immune Response and Immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Institute of Blood and Cell Therapy and Anhui Provincial Key Laboratory of Blood Research and Applications, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute of Immunology, The CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yan Wang
- Department of Hematology, The First Affiliated Hospital of USTC, Key Laboratory of Immune Response and Immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Institute of Blood and Cell Therapy and Anhui Provincial Key Laboratory of Blood Research and Applications, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute of Immunology, The CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Mingming Zhu
- Department of Hematology, The First Affiliated Hospital of USTC, Key Laboratory of Immune Response and Immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Institute of Blood and Cell Therapy and Anhui Provincial Key Laboratory of Blood Research and Applications, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute of Immunology, The CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Xian Song
- Department of Hematology, The First Affiliated Hospital of USTC, Key Laboratory of Immune Response and Immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Institute of Blood and Cell Therapy and Anhui Provincial Key Laboratory of Blood Research and Applications, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute of Immunology, The CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Hao Liu
- The CAS Key Laboratory of Mechanical Behavior and Design of Materials, University of Science and Technology of China, Hefei, China
| | - Jinghao Shen
- The CAS Key Laboratory of Mechanical Behavior and Design of Materials, University of Science and Technology of China, Hefei, China
| | - Shuangzi Wang
- The CAS Key Laboratory of Mechanical Behavior and Design of Materials, University of Science and Technology of China, Hefei, China
| | - Xiaoyu Zhu
- Department of Hematology, The First Affiliated Hospital of USTC, Key Laboratory of Immune Response and Immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Institute of Blood and Cell Therapy and Anhui Provincial Key Laboratory of Blood Research and Applications, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Cheng-Kui Qu
- Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Winship Cancer Institute, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA
| | - Linzhao Cheng
- Department of Hematology, The First Affiliated Hospital of USTC, Key Laboratory of Immune Response and Immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Institute of Blood and Cell Therapy and Anhui Provincial Key Laboratory of Blood Research and Applications, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| | - Hongyuan Jiang
- The CAS Key Laboratory of Mechanical Behavior and Design of Materials, University of Science and Technology of China, Hefei, China.
| | - Fang Ni
- Department of Hematology, The First Affiliated Hospital of USTC, Key Laboratory of Immune Response and Immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Institute of Blood and Cell Therapy and Anhui Provincial Key Laboratory of Blood Research and Applications, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
- Institute of Immunology, The CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| |
Collapse
|
48
|
Jorge-Cruz CY, Roa-Espitia AL, Hernández-González EO. Guinea pig spermatozoa adhesion to an immobilized fibronectin matrix alters their physiology and increases their survival†. Biol Reprod 2024; 111:1202-1219. [PMID: 39427254 DOI: 10.1093/biolre/ioae150] [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: 03/21/2024] [Revised: 08/16/2024] [Accepted: 10/17/2024] [Indexed: 10/21/2024] Open
Abstract
Isthmus is the region of the oviduct considered a reservoir for spermatozoa, where they are retained and released synchronously with ovulation. Integrins mediate this interaction, and it is suggested that they regulate the viability and capacitation of spermatozoa. Spermatozoa retained in the oviductal epithelial cells show specific characteristics: normal morphology, intact acrosome and plasma membrane, no DNA fragmentation, and low levels of intracellular Ca2+, and protein phosphorylation at Tyr. This work aimed to define spermatozoa's ability to adhere to an immobilized fibronectin matrix and its effects on their viability and capacitation. We found that guinea pig spermatozoa showed a high affinity for adhering to an immobilized fibronectin matrix but not to those made up of type 1 collagen or laminin. This interaction was mediated by integrins that recognize the RGD domain. Spermatozoa adhered to an immobilized fibronectin matrix were maintained in a state of low capacitation: low levels of intracellular concentration of Ca2+, protein phosphorylation in Tyr, and F-actin. Also, spermatozoa kept their plasma membrane and acrosome intact, flagellum beating and showed low activation of caspases 3/7. The spermatozoa adhered to the immobilized fibronectin matrix, gradually detached, forming rosettes and did not undergo a spontaneous acrosomal reaction but were capable of experiencing a progesterone-induced acrosomal reaction. In conclusion, the adhesion of spermatozoa to an immobilized fibronectin matrix alters the physiology of the spermatozoa, keeping them in a steady state of capacitation, increasing their viability in a similar way to what was reported for spermatozoa adhered to oviductal epithelial cells.
Collapse
Affiliation(s)
- Coral Y Jorge-Cruz
- Dept. of Cell Biology Center of Research and Advanced Studies of the National Polytechnic Institute, Mexico city, Mexico Av. Instituto Politecnico Nacional 2508, CP 07360
| | - Ana L Roa-Espitia
- Dept. of Cell Biology Center of Research and Advanced Studies of the National Polytechnic Institute, Mexico city, Mexico Av. Instituto Politecnico Nacional 2508, CP 07360
| | - Enrique O Hernández-González
- Dept. of Cell Biology Center of Research and Advanced Studies of the National Polytechnic Institute, Mexico city, Mexico Av. Instituto Politecnico Nacional 2508, CP 07360
| |
Collapse
|
49
|
Lu X, Zhao Y, Peng X, Lu C, Wu Z, Xu H, Qin Y, Xu Y, Wang Q, Hao Y, Geng D. Comprehensive Overview of Interface Strategies in Implant Osseointegration. ADVANCED FUNCTIONAL MATERIALS 2024. [DOI: 10.1002/adfm.202418849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Indexed: 01/05/2025]
Abstract
AbstractWith the improvement of implant design and the expansion of application scenarios, orthopedic implants have become a common surgical option for treating fractures and end‐stage osteoarthritis. Their common goal is rapidly forming and long‐term stable osseointegration. However, this fixation effect is limited by implant surface characteristics and peri‐implant bone tissue activity. Therefore, this review summarizes the strategies of interface engineering (osteogenic peptides, growth factors, and metal ions) and treatment methods (porous nanotubes, hydrogel embedding, and other load‐release systems) through research on its biological mechanism, paving the way to achieve the adaptation of both and coordination between different strategies. With the transition of the osseointegration stage, interface engineering strategies have demonstrated varying therapeutic effects. Especially, the activity of osteoblasts runs almost through the entire process of osseointegration, and their physiological activities play a dominant role in bone formation. Furthermore, diseases impacting bone metabolism exacerbate the difficulty of achieving osseointegration. This review aims to assist future research on osseointegration engineering strategies to improve implant‐bone fixation, promote fracture healing, and enhance post‐implantation recovery.
Collapse
Affiliation(s)
- Xiaoheng Lu
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| | - Yuhu Zhao
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| | - Xiaole Peng
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
- Department of Orthopedics The First Affiliated Hospital of Chongqing Medical University 1 Youyi Street Chongqing 400016 China
| | - Chengyao Lu
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| | - Zebin Wu
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| | - Hao Xu
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| | - Yi Qin
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| | - Yaozeng Xu
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| | - Qing Wang
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| | - Yuefeng Hao
- Orthopedics and Sports Medicine Center The Affiliated Suzhou Hospital of Nanjing Medical University 242 Guangji Street Suzhou Jiangsu 215006 China
| | - Dechun Geng
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| |
Collapse
|
50
|
Valeo M, Marie S, Rémy M, Menguy T, Le Coz C, Molinari M, Feuillie C, Granier F, Durrieu MC. Bioactive hydrogels based on lysine dendrigrafts as crosslinkers: tailoring elastic properties to influence hMSC osteogenic differentiation. J Mater Chem B 2024; 12:12508-12522. [PMID: 39576239 DOI: 10.1039/d4tb01578a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2024]
Abstract
Dendrigrafts are multivalent macromolecules with less ordered topology and higher branching than dendrimers. Exhibiting a high density of terminal amines, poly-L-lysine dendrigrafts of the fifth generation (DGL G5) allow hydrogel formation with tailorable crosslinking density and surface modification. This work presents DGL G5 as multifunctional crosslinkers in biomimetic PEG hydrogels to favour the osteogenic differentiation of human mesenchymal stem cells (hMSCs). DGL G5 reaction with dicarboxylic-acid PEG chains yielded amide networks of variable stiffness, measured at the macro and surface nanoscale. Oscillatory rheometry and compression afforded consistent values of Young's modulus, increasing from 8 to more than 30 kPa and correlating with DGL G5 concentration. At the surface level, AFM measurements showed the same tendency but higher E values, from approximately 15 to more than 100 kPa, respectively. To promote cell adhesion and differentiation, the hydrogels were functionalised with a GRGDSPC peptide and a biomimetic of the bone morphogenetic protein 2 (BMP-2), ensuring the same grafting concentrations (between 2.15 ± 0.54 and 2.28 ± 0.23 pmols mm-2) but different hydrogel stiffness. 6 h after seeding on functionalised hydrogels in serum-less media, hMSC showed nascent adhesions on the stiffer gels and greater spreading than on glass controls with serum. After two weeks in osteogenic media, hMSC seeded on the stiffer gels showed greater spreading, more polygonal morphologies and increased levels of osteopontin, an osteoblast marker, compared to controls, which peaked on 22 kPa-gels. Together, these results demonstrate that DGL G5-PEG hydrogel bioactivity can influence the adhesion, spreading and early commitment of hMSCs.
Collapse
Affiliation(s)
- Michele Valeo
- Université de Bordeaux, CNRS, Bordeaux INP, CBMN, UMR 5248, F-33600 Pessac, France.
| | | | - Murielle Rémy
- Université de Bordeaux, CNRS, Bordeaux INP, CBMN, UMR 5248, F-33600 Pessac, France.
| | | | - Cédric Le Coz
- Université de Bordeaux, CNRS, Bordeaux INP, LCPO, ENSMAC, F-33600 Pessac, France
| | - Michael Molinari
- Université de Bordeaux, CNRS, Bordeaux INP, CBMN, UMR 5248, F-33600 Pessac, France.
| | - Cécile Feuillie
- Université de Bordeaux, CNRS, Bordeaux INP, CBMN, UMR 5248, F-33600 Pessac, France.
| | | | | |
Collapse
|