1
|
Isogai T, Hirosawa KM, Kanno M, Sho A, Kasai RS, Komura N, Ando H, Furukawa K, Ohmi Y, Furukawa K, Yokota Y, Suzuki KG. Extracellular vesicles adhere to cells primarily by interactions of integrins and GM1 with laminin. J Cell Biol 2025; 224:e202404064. [PMID: 40304687 PMCID: PMC12042775 DOI: 10.1083/jcb.202404064] [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: 04/12/2024] [Revised: 12/09/2024] [Accepted: 03/11/2025] [Indexed: 05/02/2025] Open
Abstract
Tumor-derived extracellular vesicles (EVs) have attracted significant attention, yet the molecular mechanisms that govern their specific binding to recipient cells remain elusive. Our in vitro study utilizing single-particle tracking demonstrated that integrin heterodimers comprising α6β4 and α6β1 and ganglioside, GM1, are responsible for the binding of small EV (sEV) subtypes to laminin. EVs derived from four distinct tumor cell lines, regardless of size, exhibited high binding affinities for laminin but not for fibronectin, although fibronectin receptors are abundant in EVs and have functional roles in EV-secreting cells. Our findings revealed that integrins in EVs bind to laminin via the conventional molecular interface, facilitated by CD151 rather than by inside-out signaling of talin-1 and kindlin-2. Super-resolution movie observation revealed that sEV integrins bind only to laminin on living recipient cells. Furthermore, sEVs bound to HUVEC and induced cell branching morphogenesis in a laminin-dependent manner. Thus, we demonstrated that EVs predominantly bind to laminin on recipient cells, which is indispensable for cell responses.
Collapse
Affiliation(s)
- Tatsuki Isogai
- The United Graduate School of Agricultural Science, Gifu University, Gifu, Japan
| | | | - Miki Kanno
- Graduate School of Natural Science and Technology, Gifu University, Gifu, Japan
| | - Ayano Sho
- Faculty of Applied Biological Sciences, Gifu University, Gifu, Japan
| | - Rinshi S. Kasai
- Division of Advanced Bioimaging, National Cancer Center Research Institute (NCCRI), Tokyo, Japan
| | - Naoko Komura
- Institute for Glyco-core Research (iGCORE), Gifu University, Gifu, Japan
| | - Hiromune Ando
- The United Graduate School of Agricultural Science, Gifu University, Gifu, Japan
- Institute for Glyco-core Research (iGCORE), Gifu University, Gifu, Japan
- Graduate School of Natural Science and Technology, Gifu University, Gifu, Japan
- Faculty of Applied Biological Sciences, Gifu University, Gifu, Japan
- Innovation Research Center for Quantum Medicine, Graduate School of Medicine, Gifu University, Gifu, Japan
| | - Keiko Furukawa
- Department of Biomedical Sciences, Chubu University, Kasugai, Japan
| | - Yuhsuke Ohmi
- Department of Biomedical Sciences, Chubu University, Kasugai, Japan
| | - Koichi Furukawa
- Department of Biomedical Sciences, Chubu University, Kasugai, Japan
| | - Yasunari Yokota
- Department of Information Science, Faculty of Engineering, Gifu University, Gifu, Japan
| | - Kenichi G.N. Suzuki
- The United Graduate School of Agricultural Science, Gifu University, Gifu, Japan
- Institute for Glyco-core Research (iGCORE), Gifu University, Gifu, Japan
- Graduate School of Natural Science and Technology, Gifu University, Gifu, Japan
- Faculty of Applied Biological Sciences, Gifu University, Gifu, Japan
- Division of Advanced Bioimaging, National Cancer Center Research Institute (NCCRI), Tokyo, Japan
- Innovation Research Center for Quantum Medicine, Graduate School of Medicine, Gifu University, Gifu, Japan
| |
Collapse
|
2
|
Pijning AE, Hogg PJ. Disulfide bond control of platelet αIIbβ3 integrin. Thromb Res 2025; 250:109320. [PMID: 40300222 DOI: 10.1016/j.thromres.2025.109320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Revised: 03/25/2025] [Accepted: 04/15/2025] [Indexed: 05/01/2025]
Abstract
The platelet αIIbβ3 integrin is the most abundant platelet receptor, orchestrating platelet adhesion, activation, and mechano-sensing during hemostasis and thrombosis. Disulfide bonds are the covalent links between the sulfur atoms of two cysteine residues and their role in the functioning of αIIbβ3 has been a topic of investigation for over two decades. The advent of differential cysteine alkylation using isotopic alkylators and mass spectrometry has led to the identification of multiple partially disulfide-bonded states of αIIbβ3 that are constitutively produced by megakaryocytes and reside in the platelet surface membrane, and an allosteric disulfide that is cleaved in the mature receptor to control function. One of the disulfide-bonded integrin states has reduced capacity due to particular clustering, internalisation, and recycling dynamics and lower avidity for fibrinogen, suggesting that other states may also have specific properties. Cleavage of an allosteric disulfide bond in the activated integrin uncouples the receptor from its ligand, and it is likely that other allosteric disulfides are yet to be identified. This review presents the current knowledge of the role of specific disulfide bonds in the regulation of αIIbβ3 integrin and perspectives on the future.
Collapse
Affiliation(s)
- Aster E Pijning
- School of Life Sciences, University of Technology Sydney and Centenary Institute, University of Sydney, Sydney, NSW, Australia
| | - Philip J Hogg
- School of Life Sciences, University of Technology Sydney and Centenary Institute, University of Sydney, Sydney, NSW, Australia.
| |
Collapse
|
3
|
Zhu S, Liu J, Xu K, Xu F, Jiang Y, Dai L, Pei T, Zhu Y, Liu D, Zhang X, Xu J, Yang J, Pan Z, Tao J, Hou Z. Comparative transcriptomic analyses of macrophages infected with Toxoplasma gondii strains of different virulence provide molecular insights into the response of macrophage in phagocytosis and polarization to infection. Mol Immunol 2025; 183:259-273. [PMID: 40414092 DOI: 10.1016/j.molimm.2025.05.003] [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: 12/16/2024] [Revised: 03/07/2025] [Accepted: 05/05/2025] [Indexed: 05/27/2025]
Abstract
Macrophages are essential for the proliferation and spread of Toxoplasma gondii. Modulating macrophage activation to improve the inflammatory environment is an effective approach for disease treatment. However, the molecular mechanism through which T. gondii alters macrophage function remain unknown. Based on transcriptomic data analysis of various macrophage types infected with T. gondii, current research revealed differences in the regulation of macrophage functions among strains with different virulence: RH was primarily involved in cell cycle regulation, ME49 was associated with cAMP signaling, and CEP mainly participated in ion channel activity. All three T. gondii strains were involved in regulating immune response activation, including leukocyte adhesion and the MAPK signaling pathway. Nineteen shared DEGs associated with macrophage phagocytosis or polarization were identified through the GeneCards database, and PPI analysis confirmed Il6 as the hub gene in the regulatory network. In vivo and in vitro experiments showed that the YZ-1 strain significantly regulated the expressions of eight DEGs (Il6, Rel, Cd83, Myc, Adora2b, Egr2, Gja1 and Nr4a2), and promoted macrophage phagocytic activity and induced M1 polarization, confirming a significant correlation with Il6. This study revealed the dissimilarities and commonalities in macrophage function regulated by T. gondii strains of different virulence, and identified key molecules involved in the regulation of macrophage phagocytosis and polarization during T. gondii infection. This is crucial for identifying potential drug targets against T. gondii and provides a new perspective on the etiopathogenesis and therapeutic approaches for toxoplasmosis.
Collapse
Affiliation(s)
- Shifan Zhu
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou 225009, PR China; International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, PR China; Jiangsu Key Laboratory of Zoonosis, Yangzhou 225009, PR China
| | - Jiantao Liu
- YEBIO Bioengineering Co., Ltd of QINGDAO, Qingdao 266113, PR China
| | - Kangzhi Xu
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou 225009, PR China; International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, PR China; Jiangsu Key Laboratory of Zoonosis, Yangzhou 225009, PR China
| | - Fan Xu
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou 225009, PR China; International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, PR China; Jiangsu Key Laboratory of Zoonosis, Yangzhou 225009, PR China
| | - Yuwei Jiang
- Lingkou Town Animal Epidemic Prevention Station, Danyang 212353, PR China
| | - Linwei Dai
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou 225009, PR China; International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, PR China; Jiangsu Key Laboratory of Zoonosis, Yangzhou 225009, PR China
| | - Tianxu Pei
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou 225009, PR China; International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, PR China; Jiangsu Key Laboratory of Zoonosis, Yangzhou 225009, PR China
| | - Yuyang Zhu
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou 225009, PR China; International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, PR China; Jiangsu Key Laboratory of Zoonosis, Yangzhou 225009, PR China
| | - Dandan Liu
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou 225009, PR China; International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, PR China; Jiangsu Key Laboratory of Zoonosis, Yangzhou 225009, PR China
| | - Xinjun Zhang
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou 225009, PR China; International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, PR China; Jiangsu Key Laboratory of Zoonosis, Yangzhou 225009, PR China
| | - Jinjun Xu
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou 225009, PR China; International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, PR China; Jiangsu Key Laboratory of Zoonosis, Yangzhou 225009, PR China
| | - Jin Yang
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225000, PR China.
| | - Zhiming Pan
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou 225009, PR China; Jiangsu Key Laboratory of Zoonosis, Yangzhou 225009, PR China
| | - Jianping Tao
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou 225009, PR China; International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, PR China; Jiangsu Key Laboratory of Zoonosis, Yangzhou 225009, PR China.
| | - Zhaofeng Hou
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou 225009, PR China; International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, PR China; Jiangsu Key Laboratory of Zoonosis, Yangzhou 225009, PR China.
| |
Collapse
|
4
|
Kulkarni S, Tebar F, Rentero C, Zhao M, Sáez P. Competing signaling pathways controls electrotaxis. iScience 2025; 28:112329. [PMID: 40292314 PMCID: PMC12032939 DOI: 10.1016/j.isci.2025.112329] [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: 08/08/2024] [Revised: 10/01/2024] [Accepted: 03/28/2025] [Indexed: 04/30/2025] Open
Abstract
Understanding how cells follow exogenous cues is a key question for biology, medicine, and bioengineering. Growing evidence shows that electric fields represent a precise and programmable method to control cell migration. Most data suggest that the polarization of membrane proteins and the following downstream signaling are central to electrotaxis. Unfortunately, how these multiple mechanisms coordinate with the motile machinery of the cell is still poorly understood. Here, we develop a mechanistic model that explains electrotaxis across different cell types. Using the zebrafish proteome, we identify membrane proteins directly related to migration signaling pathways that polarize anodally and cathodally. Further, we show that the simultaneous and asymmetric distribution of these membrane receptors establish multiple cooperative and competing stimuli for directing the anodal and cathodal migration of the cell. Using electric fields, we enhance, cancel, or switch directed cell migration, with clear implications in promoting tissue regeneration or arresting tumor progression.
Collapse
Affiliation(s)
- S. Kulkarni
- Laboratori de Càlcul Numèric (LaCàN), ETS de Ingeniería de Caminos, Canales y Puertos, Universitat Politècnica de Catalunya, Barcelona, Spain
| | - F. Tebar
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain
- Cell Compartments and Signaling Group, Fundació de Recerca Clínic Barcelona - Institut d’Investigacions Biomèdiques August Pi i Sunyer (FRCB-IDIBAPS), 08036 Barcelona, Spain
| | - C. Rentero
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain
- Cell Compartments and Signaling Group, Fundació de Recerca Clínic Barcelona - Institut d’Investigacions Biomèdiques August Pi i Sunyer (FRCB-IDIBAPS), 08036 Barcelona, Spain
| | - M. Zhao
- Department of Ophthalmology & Vision Science, School of Medicine, University of California, Davis, Sacramento, CA, USA
| | - P. Sáez
- Laboratori de Càlcul Numèric (LaCàN), ETS de Ingeniería de Caminos, Canales y Puertos, Universitat Politècnica de Catalunya, Barcelona, Spain
- IMTech (Institute of Mathematics), Universitat Politècnica de Catalunya-BarcelonaTech., 08034 Barcelona, Spain
| |
Collapse
|
5
|
Cheah K, Chu P, Schmidt G, Scarlata S. Imaging methods to monitor and quantify cell differentiation. Front Cell Dev Biol 2025; 13:1584858. [PMID: 40433548 PMCID: PMC12106324 DOI: 10.3389/fcell.2025.1584858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Accepted: 04/17/2025] [Indexed: 05/29/2025] Open
Abstract
The transition of a cell from a stem to a differentiated state involves an interrelated and complex series of events. These events include dynamic changes in cellular nucleic acid and protein content that are mediated by both intrinsic and extrinsic factors which ultimately lead to differentiation into specific lineage. Quantifying the parameters associated with differentiation and their changes under different conditions would not only allow for a better understanding of this process but also would enable the development of approaches that control differentiation. Here, we describe processes associated with the differentiation of two types of cultured cells, neurons and fibroblasts, and the tools to follow changes in real time. Specifically, we discuss methods to the identify cell lineage, changes in morphology, shifts in specific mRNA and miRNA levels as well as the changes in protein localization, interactions and assemblies that accompany differentiation.
Collapse
Affiliation(s)
| | | | | | - Suzanne Scarlata
- Department of Chemistry and Biochemistry, Worcester Polytechnic Institute, Worcester, MA, United States
| |
Collapse
|
6
|
Salem Y, Yacov N, Kafri P, Propheta-Meiran O, Karni A, Maharshak N, Furer V, Elkayam O, Mendel I. MOSPD2 regulates the activation state of αLβ2 integrin to control monocyte migration: applicability for treatment of chronic inflammatory diseases. Immunol Res 2025; 73:78. [PMID: 40312574 PMCID: PMC12045827 DOI: 10.1007/s12026-025-09633-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Accepted: 04/21/2025] [Indexed: 05/03/2025]
Abstract
Monocytes are innate immune cells that drive the chronicity of various inflammatory diseases. Monocyte migration to inflamed tissues involves multiple steps of interaction with the vascular endothelium and the extracellular matrix (ECM), a process mediated through conformational transitions in cell surface integrins. We previously described motile sperm domain-containing protein 2 (MOSPD2) as a surface protein expressed on myeloid cells that is essential for the migration of monocytes and a key regulator of inflammation. Investigating MOSPD2's mechanism of action, we assessed whether it plays a role in regulating integrin activation and monocyte adhesion. Data show that silencing of MOSPD2 expression in the THP-1 monocytic cell line significantly increased cell adhesion to various ECM molecules. Employing IW-601, a humanized anti-human MOSDP2 monoclonal antibody, on primary human monocytes increased adhesion to ECM molecules as well as to adhesion molecules. At the molecular level, silencing of MOSPD2 or blocking MOSPD2 using IW-601 led to a transition in integrin αLβ2 (CD11a/CD18, LFA-1) conformation into an active high-affinity binding form and to the induction of adhesion-associated signaling pathways. Co-immunoprecipitation experiments showed that MOSPD2 binds integrin-β2 (CD18), but not integrin-β1 (CD29). Our results reveal a novel mechanism controlling monocyte migration, in which MOSPD2 acts as an adhesion checkpoint that governs the balance between monocyte adhesion and release. By demonstrating the inhibitory effect of IW-601 on the migration of primary monocytes isolated from patients with chronic inflammatory diseases, we provide proof of concept for translating MOSPD2's mechanism into a potential treatment for inflammatory diseases, further supported by in vivo data in models of RA and IBD.
Collapse
Affiliation(s)
- Yaniv Salem
- ImmuneWalk Therapeutics, SVP Research, 20, Hamagshimim St, 4934829, Petach-Tikva, Israel
- ImmuneWalk Therapeutics, 1 Blue Hill Plaza, Pearl River, NY, 10965, USA
- , Current Address: 77, Shtern Yair St., 5560706, Kiryat-Ono, Israel
| | - Niva Yacov
- ImmuneWalk Therapeutics, SVP Research, 20, Hamagshimim St, 4934829, Petach-Tikva, Israel
- ImmuneWalk Therapeutics, 1 Blue Hill Plaza, Pearl River, NY, 10965, USA
| | - Pinhas Kafri
- ImmuneWalk Therapeutics, SVP Research, 20, Hamagshimim St, 4934829, Petach-Tikva, Israel
- ImmuneWalk Therapeutics, 1 Blue Hill Plaza, Pearl River, NY, 10965, USA
- Current Address: Teva Pharmaceuticals, 12 Hatrufa St, 4250483, Netanya, Israel
| | - Oshrat Propheta-Meiran
- ImmuneWalk Therapeutics, SVP Research, 20, Hamagshimim St, 4934829, Petach-Tikva, Israel
- ImmuneWalk Therapeutics, 1 Blue Hill Plaza, Pearl River, NY, 10965, USA
| | - Arnon Karni
- Neuroimmunology and Multiple Sclerosis Unit of the Department of Neurology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
- Department of Neurology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Nitsan Maharshak
- Department of Gastroenterology and Liver Diseases, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Victoria Furer
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Department of Rheumatology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Ori Elkayam
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Department of Rheumatology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Itzhak Mendel
- ImmuneWalk Therapeutics, SVP Research, 20, Hamagshimim St, 4934829, Petach-Tikva, Israel.
- ImmuneWalk Therapeutics, 1 Blue Hill Plaza, Pearl River, NY, 10965, USA.
| |
Collapse
|
7
|
Perner F, Pahl HL, Zeiser R, Heidel FH. Malignant JAK-signaling: at the interface of inflammation and malignant transformation. Leukemia 2025; 39:1011-1030. [PMID: 40140631 PMCID: PMC12055591 DOI: 10.1038/s41375-025-02569-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 02/21/2025] [Accepted: 03/13/2025] [Indexed: 03/28/2025]
Abstract
The JAK pathway is central to mammalian cell communication, characterized by rapid responses, receptor versatility, and fine-tuned regulation. It involves Janus kinases (JAK1, JAK2, JAK3, TYK2), which are activated when natural ligands bind to receptors, leading to autophosphorylation and activation of STAT transcription factors [1, 2]. JAK-dependent signaling plays a pivotal role in coordinating cell communication networks across a broad spectrum of biological systems including development, immune responses, cell growth, and differentiation. JAKs are frequently mutated in the aging hematopoietic system [3, 4] and in hematopoietic cancers [5]. Thus, dysregulation of the pathway results in various diseases, including cancers and immune disorders. The binding of extracellular ligands to class I and II cytokine receptors initiates a critical signaling cascade through the activation of Janus kinases (JAKs). Upon ligand engagement, JAKs become activated and phosphorylate specific tyrosine residues on the receptor, creating docking sites for signal transducer and activator of transcription (STAT) proteins. Subsequent JAK-mediated phosphorylation of STATs enables their dimerization and nuclear translocation, where they function as transcription factors to modulate gene expression. Under physiological conditions, JAK-signaling is a tightly regulated mechanism that governs cellular responses to external cues, such as cytokines and growth factors, ensuring homeostasis and maintaining the functional integrity of tissues and organs. Highly defined regulation of JAK-signaling is essential for balancing cellular responses to inflammatory stimuli and growth signals, thus safeguarding tissue health. In contrast, dysregulated JAK-signaling results in chronic inflammation and unrestrained cellular proliferation associated with various diseases. Understanding the qualitative and quantitative differences at the interface of physiologic JAK-signaling and its aberrant activation in disease is crucial for the development of targeted therapies that precisely tune this pathway to target pathologic activation patterns while leaving homeostatic processes largely unaffected. Consequently, pharmaceutical research has targeted this pathway for drug development leading to the approval of several substances with different selectivity profiles towards individual JAKs. Yet, the precise impact of inhibitor selectivity and the complex interplay of different functional modules within normal and malignant cells remains incompletely understood. In this review, we summarize the current knowledge on JAK-signaling in health and disease and highlight recent advances and future directions in the field.
Collapse
Affiliation(s)
- Florian Perner
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School (MHH), Hannover, Germany
| | - Heike L Pahl
- Department of Medicine I, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Robert Zeiser
- Department of Medicine I, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Florian H Heidel
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School (MHH), Hannover, Germany.
- Leibniz-Institute on Aging, Fritz-Lipmann-Institute (FLI), Jena, Germany.
- Cellular Therapy Center (CTC), Hannover Medical School (MHH), Hannover, Germany.
| |
Collapse
|
8
|
Hong H, Kim D, Jung H, Kim S, Min S, Kim C, Kim K, Rha H, Kang H. Biomimetic Dynamics of Nanoscale Groove and Ridge Topography for Stem Cell Regulation. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025:e2419416. [PMID: 40285558 DOI: 10.1002/adma.202419416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 04/10/2025] [Indexed: 04/29/2025]
Abstract
Native extracellular matrix exhibits multiscale groove and ridge structures that continuously change, such as collagen fibril-based nanogrooves in bone tissue, and regulate cellular responses. However, dynamic switching between groove and ridge nanostructures at the molecular level has not been demonstrated. Herein, materials capable of dynamic groove-ridge switching at tens-of-nanometers scale are developed by flexibly conjugating RGD-magnetically activatable nanoridges (MANs) to non-magnetic nanogrooves with independently tuned widths comparable to the sizes of integrin-presenting filopodia by modulating hydrophobicity in bicontinuous microemulsion, allowing for cyclic modulation of RGD accessibility and cellular adhesion. Nanogrooves with medium width restrict RGD accessibility in the "groove" state in which the RGD-MANs are buried, which is reversed by magnetically raising them to protrude and form the "ridge" state that fully exposes the RGDs. This reversibly stimulates integrin recruitment, focal adhesion complex assembly, mechanotransduction, and differentiation of stem cells in vivo. This is the first demonstration of molecular-level groove and ridge nanostructures that exhibit unprecedented switchability between groove and ridge nanostructures. Versatile tuning of the width, height, pitch, and shape of intricate nanogroove structures with remote manipulability can enlighten the understanding of molecular-scale cell-ligand interactions for stem cell engineering-based treatment of aging, injuries, and stress-related diseases.
Collapse
Affiliation(s)
- Hyunsik Hong
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Dahee Kim
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Hwapyung Jung
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Seongyeol 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
| | - Chowon Kim
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Kanghyeon Kim
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Hyunji Rha
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Heemin Kang
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
- College of Medicine, Korea University, Seoul, 02841, Republic of Korea
| |
Collapse
|
9
|
Zhevlakova I, Liu H, Dudiki T, Gao D, Yakubenko V, Tkachenko S, Cherepanova O, Podrez EA, Byzova TV. Mechanisms and consequences of myeloid adhesome dysfunction in atherogenesis. Cardiovasc Res 2025; 121:62-76. [PMID: 39393814 PMCID: PMC11999018 DOI: 10.1093/cvr/cvae223] [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: 04/10/2024] [Revised: 07/24/2024] [Accepted: 08/23/2024] [Indexed: 10/13/2024] Open
Abstract
AIMS In the context of atherosclerosis, macrophages exposed to oxidized low-density lipoproteins (oxLDLs) exhibit cellular abnormalities, specifically in adhesome functions, yet the mechanisms and implications of these adhesive dysfunctions remain largely unexplored. METHODS AND RESULTS This study reveals a significant depletion of Kindlin3 (K3) or Fermt3, an essential component of the adhesome regulating integrin functions, in macrophages located within atherosclerotic plaques in vivo and following oxLDL exposure in vitro. To examine the effects of K3 deficiency, the study utilized hyperlipidaemic bone marrow chimeras devoid of myeloid Kindlin3 expression. The absence of myeloid K3 increased atherosclerotic plaque burden in the aortas in vivo and enhanced lipid accumulation and lipoprotein uptake in macrophages from Kindlin3-null chimeric mice in vitro. Importantly, re-expression of K3 in macrophages ameliorated these abnormalities. RNA sequencing of bone marrow-derived macrophages (BMDM) from K3-deficient mice revealed extensive deregulation in adhesion-related pathways, echoing changes observed in wild-type cells treated with oxLDL. Notably, there was an increase in Olr1 expression [encoding the lectin-like oxidized LDL receptor-1 (LOX1)], a gene implicated in atherogenesis. The disrupted K3-integrin axis in macrophages led to a significant elevation in the LOX1 receptor, contributing to increased oxLDL uptake and foam cell formation. Inhibition of LOX1 normalized lipid uptake in Kindlin3-null macrophages. A similar proatherogenic phenotype, marked by increased macrophage LOX1 expression and foam cell formation, was observed in myeloid-specific Itgβ1-deficient mice but not in Itgβ2-deficient mice, underscoring the critical role of K3/Itgβ1 interaction. CONCLUSION This study shows that the loss of Kindlin3 in macrophages upon exposure to oxLDL leads to adhesome dysfunction in atherosclerosis and reveals the pivotal role of Kindlin3 in macrophage function and its contribution to the progression of atherosclerosis, providing valuable insights into the molecular mechanisms that could be targeted for therapeutic interventions.
Collapse
Affiliation(s)
- Irina Zhevlakova
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Ave, Cleveland, OH 44195, USA
| | - Huan Liu
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Ave, Cleveland, OH 44195, USA
| | - Tejasvi Dudiki
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Ave, Cleveland, OH 44195, USA
| | - Detao Gao
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Ave, Cleveland, OH 44195, USA
| | - Valentin Yakubenko
- Department of Biomedical Sciences, Center of Excellence for Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37684, USA
| | - Svyatoslav Tkachenko
- Department of Genetics and Genome Sciences, Case Western Reserve University, 2109 Adelbert Rd Building, Cleveland, OH 44106, USA
| | - Olga Cherepanova
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Ave, Cleveland, OH 44195, USA
| | - Eugene A Podrez
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Ave, Cleveland, OH 44195, USA
| | - Tatiana V Byzova
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Ave, Cleveland, OH 44195, USA
| |
Collapse
|
10
|
Hsu CP, Hordeichyk A, Aretz J, Fässler R, Bausch AR. Synergistic effect of PIP 2 and PIP 3 on membrane-induced phase separation of integrin complexes. Biophys J 2025:S0006-3495(25)00238-3. [PMID: 40235120 DOI: 10.1016/j.bpj.2025.04.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 02/17/2025] [Accepted: 04/10/2025] [Indexed: 04/17/2025] Open
Abstract
The assembly of integrin adhesion complexes at the inner leaflet of the plasma membrane regulates cell adhesion to the extracellular matrix. The multivalent protein interactions within the complexes and with the cell membrane display characteristics of membrane-associated biomolecular condensates driven by liquid-liquid phase separation. The composition of lipids and the distribution of the cell membrane are crucial for forming integrin adhesion complexes. Here, we report that phosphatidylinositol 4,5-bisphosphate (PIP2) and phosphatidylinositol (3,4,5)-trisphosphate (PIP3) in the model membrane synergistically regulate the formation of membrane-induced integrin adhesion condensates, which consist of β1 tails, kindlin, talin, paxillin, and focal adhesion kinase. We show that the preferential bindings of kindlin to PIP3 and talin to PIP2 enhance their recruitment to the membrane, which in turn increases the probability of membrane-associated phase separation. Our results indicate that modulating the intricate balance of membrane composition is a strategy to localize integrin adhesion complexes and optimize their density on lipid membranes.
Collapse
Affiliation(s)
- Chiao-Peng Hsu
- Heinz Nixdorf Chair for Cellular Biophysics, Center for Functional Protein Assemblies, Center for Organoid Systems, Department of Bioscience, Technical University of Munich, Technical University of Munich School of Natural Sciences, Garching, Germany
| | - Arsenii Hordeichyk
- Heinz Nixdorf Chair for Cellular Biophysics, Center for Functional Protein Assemblies, Center for Organoid Systems, Department of Bioscience, Technical University of Munich, Technical University of Munich School of Natural Sciences, Garching, Germany; Max Planck School Matter to Life, Heidelberg, Germany
| | - Jonas Aretz
- Department of Molecular Medicine, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Reinhard Fässler
- Department of Molecular Medicine, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Andreas R Bausch
- Heinz Nixdorf Chair for Cellular Biophysics, Center for Functional Protein Assemblies, Center for Organoid Systems, Department of Bioscience, Technical University of Munich, Technical University of Munich School of Natural Sciences, Garching, Germany; Max Planck School Matter to Life, Heidelberg, Germany.
| |
Collapse
|
11
|
Bagheri L, Javanbakht M, Malekian S, Ghahderijani BH, Taghipour S, Tanha FD, Ranjkesh M, Cegolon L, Zhao S. Antifibrotic therapeutic strategies in systemic sclerosis: Critical role of the Wnt/β-catenin and TGF-β signal transduction pathways as potential targets. Eur J Pharmacol 2025; 999:177607. [PMID: 40209848 DOI: 10.1016/j.ejphar.2025.177607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 03/25/2025] [Accepted: 04/07/2025] [Indexed: 04/12/2025]
Abstract
Systemic sclerosis (SSc) is a prototypic fibrosing disorder characterized by widespread fibrosis and immune dysregulation. Current evidence highlights the intricate cross-talk between the canonical Wnt/β-catenin signaling pathway and transforming growth factor-beta (TGF-β) signaling, both of which play fundamental roles in the pathogenesis of fibrosis. This review aims to elucidate the central role of the Wnt/β-catenin-TGF-β pathway and TGF-β signal transduction pathway in fibrotic diseases, focusing on SSc. We summarized evidence from cellular biology studies, animal model investigations and clinical observations to provide a comprehensive view of the mechanisms causing pathological fibrosis. In addition, we explore the possibilities of antifibrotic therapeutic strategies against Wnt/β-catenin-TGF-β signaling to counteract fibrosis, delineating approaches for treatment of SSc patients by targeting these interconnected signaling pathways.
Collapse
Affiliation(s)
- Leyla Bagheri
- Department of Internal Medicine, Shahid Modarres Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Mohammad Javanbakht
- Nephrology and Urology Research Center, Clinical Science Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Sheida Malekian
- Department of Internal Medicine, Shahid Modarres Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Sadra Taghipour
- Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Fatemeh Davari Tanha
- Department of Infertility, Yas Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Luca Cegolon
- Department of Medical, Surgical & Health Sciences, University of Trieste, 34128, Trieste, Italy; Public Health Unit, University Health Agency Giuliano-Isontina (ASUGI), 34148, Trieste, Italy
| | - Shi Zhao
- School of Public Health, Tianjin Medical University, Tianjin, 300070, China
| |
Collapse
|
12
|
Shi S, Gao J, Zhang Y, Zhan M, Tan Z, Wang P, Fu J, Liu J. Inflammation and platelet hyperresponsiveness in coronary artery disease and the influence of Talin-1/αIIbβ3-mediated bidirectional signaling pathway. Front Pharmacol 2025; 16:1535182. [PMID: 40183091 PMCID: PMC11965607 DOI: 10.3389/fphar.2025.1535182] [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: 12/10/2024] [Accepted: 02/24/2025] [Indexed: 04/05/2025] Open
Abstract
Background While platelet hyperreactivity constitutes an independent risk factor for major adverse cardiovascular events (MACEs) in coronary artery disease, its molecular underpinnings remain poorly characterized. Recent advances in transcriptomic profiling have revealed potential associations with specific RNA signatures. Through systematic bioinformatics analysis of differential gene expression patterns and pathway activation in CHD patients, this study aims to elucidate key molecular regulators of platelet hyperactivity, establishing a theoretical framework for developing precision therapeutic strategies to mitigate post-CHD complications. Methods This randomized controlled study included 16 CHD patients and 16 healthy controls. Inflammation markers, platelet aggregation function, and CD62p levels were assessed using flow cytometry. Mitochondrial morphology and organelles were observed using scanning electron microscopy and transmission electron microscopy. Genes related to symptom alteration between CHD patients and healthy controls were identified using the criteria of p < 0.05. The molecular correlations of these genes were analyzed using a comprehensive perspective that included Gene Ontology (GO) biological process and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses. Western blot and correlation analyses were also conducted to validate the expression and diagnostic value of the DEGs. Results CHD patients exhibited alterations in platelet organelles ultrastructure, heightened platelet activation and aggregation, and disturbance of the inflammatory equilibrium. RNA sequencing demonstrated distinct changes in the gene expression profiles of circulating platelets from CHD patients. The increase in platelet activation and aggregation could be partially associated with the upregulation of the Talin-1 and αIIbβ3 proteins expression. Conclusion Abnormal transcription and platelet activation occur after CHD onset, and upregulation of the Talin-1/αIIbβ3-mediated bidirectional signaling pathway are the primary pathological features. Clinical Trial Registration https://www.chictr.org.cn/, identifier ChiCTR2100041998.
Collapse
Affiliation(s)
- Shengnan Shi
- Beijing Key Laboratory of Pharmacology of Chinese Materia Medica, Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jiaming Gao
- Beijing Key Laboratory of Pharmacology of Chinese Materia Medica, Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yehao Zhang
- Beijing Key Laboratory of Pharmacology of Chinese Materia Medica, Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Min Zhan
- Department of Encephalopathy, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhanfei Tan
- Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Peili Wang
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jianhua Fu
- Beijing Key Laboratory of Pharmacology of Chinese Materia Medica, Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jianxun Liu
- Beijing Key Laboratory of Pharmacology of Chinese Materia Medica, Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
13
|
Angeli S, Neophytou C, Kalli M, Stylianopoulos T, Mpekris F. The mechanopathology of the tumor microenvironment: detection techniques, molecular mechanisms and therapeutic opportunities. Front Cell Dev Biol 2025; 13:1564626. [PMID: 40171226 PMCID: PMC11958720 DOI: 10.3389/fcell.2025.1564626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 02/27/2025] [Indexed: 04/03/2025] Open
Abstract
The mechanical properties of the tumor microenvironment (TME) undergo significant changes during tumor growth, primarily driven by alterations in extracellular (ECM) stiffness and tumor viscoelasticity. These mechanical changes not only promote tumor progression but also hinder therapeutic efficacy by impairing drug delivery and activating mechanotransduction pathways that regulate crucial cellular processes such as migration, proliferation, and resistance to therapy. In this review, we examine the mechanisms through which tumor cells sense and transmit mechanical signals to maintain homeostasis in the biomechanically altered TME. We explore current computational modelling strategies for mechanotransduction pathways, highlighting the need for developing models that incorporate additional components of the mechanosignaling machinery. Furthermore, we review available methods for measuring the mechanical properties of tumors in clinical settings and strategies aiming at restoring the TME and blocking deregulated mechanotransduction pathways. Finally, we propose that proper characterization and a deeper understanding of the mechanical landscape of the TME, both at the tissue and cellular levels, are essential for developing therapeutic strategies that account for the influence of mechanical forces on treatment efficacy.
Collapse
Affiliation(s)
| | | | | | | | - Fotios Mpekris
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| |
Collapse
|
14
|
Sun Y, Hamlin AJ, Schwarzbauer JE. Fibronectin matrix assembly at a glance. J Cell Sci 2025; 138:jcs263834. [PMID: 40130407 PMCID: PMC12050093 DOI: 10.1242/jcs.263834] [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: 03/26/2025] Open
Abstract
The organization and mechanics of extracellular matrix (ECM) protein polymers determine tissue structure and function. Secreted ECM components are assembled into polymers via a cell-mediated process. The specific mechanisms that cells use for assembly are crucial for generating tissue-appropriate matrices. Fibronectin (FN) is a ubiquitous and abundant ECM protein that is assembled into a fibrillar matrix by a receptor-mediated process, and the FN matrix provides a foundation for incorporation of many other proteins into the ECM. In this Cell Science at a Glance article and the accompanying poster, we describe the domain organization of FN and the events that initiate and propagate a stable insoluble network of FN fibrils. We also discuss intracellular pathways that regulate FN assembly and the impact of changes in assembly on disease progression.
Collapse
Affiliation(s)
- Yu Sun
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Aaron J. Hamlin
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | | |
Collapse
|
15
|
Zhao H, Tu X. The potential key genes within focal adhesion that regulate mesenchymal stem cells osteogenesis or adipogenesis in microgravity related disuse osteoporosis: an integrated analysis. Front Endocrinol (Lausanne) 2025; 16:1469400. [PMID: 40130165 PMCID: PMC11930814 DOI: 10.3389/fendo.2025.1469400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 02/14/2025] [Indexed: 03/26/2025] Open
Abstract
This study aimed to identify key genes related to focal adhesions (FA) and cells involved in osteoblast (OS) and adipocyte (AD) differentiation in osteoporosis. A mouse model of disuse osteoporosis was made by hindlimbs unloading (HLU)/Tail - suspension. Micro - CT and histological analysis were done, and differentially expressed genes (DEGs) from GSE100930 were analyzed. Soft clustering on GSE80614 OS/AD samples found FA - related candidate genes. protein-protein interaction (PPI) network and cytoHubba's Degree algorithm identified key FA - genes, validated by quantitative polymerase chain reaction (qPCR). Key OS/AD - associated cells were identified by single - cell analysis. The mouse model showed decreased bone density, microstructure damage, increased marrow adiposity, and altered gene expression. Key FA - related genes for osteogenesis (ITGB3, LAMC1, COL6A3, ITGA8, PDGFRB) and adipogenesis (ITGB3, ITGA4, LAMB1, ITGA8, LAMA4) were found and validated. Key cells (chondrocyte, adipocyte, and osteoblast progenitors) are involved in specific pathways, with osteoblast progenitors having stronger interactions. Pseudotime analysis implies differentiation from chondrocyte progenitors to adipocyte, then osteoblast progenitors. This study provides new insights for disuse osteoporosis research.
Collapse
Affiliation(s)
| | - Xiaolin Tu
- Laboratory of Skeletal Development and Regeneration, Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| |
Collapse
|
16
|
Liu Z, Zhang X, Ben T, Li M, Jin Y, Wang T, Song Y. Focal adhesion in the tumour metastasis: from molecular mechanisms to therapeutic targets. Biomark Res 2025; 13:38. [PMID: 40045379 PMCID: PMC11884212 DOI: 10.1186/s40364-025-00745-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 02/11/2025] [Indexed: 03/09/2025] Open
Abstract
The tumour microenvironment is the "hotbed" of tumour cells, providing abundant extracellular support for growth and metastasis. However, the tumour microenvironment is not static and is constantly remodelled by a variety of cellular components, including tumour cells, through mechanical, biological and chemical means to promote metastasis. Focal adhesion plays an important role in cell-extracellular matrix adhesion. An in-depth exploration of the role of focal adhesion in tumour metastasis, especially their contribution at the biomechanical level, is an important direction of current research. In this review, we first summarize the assembly of focal adhesions and explore their kinetics in tumour cells. Then, we describe in detail the role of focal adhesion in various stages of tumour metastasis, especially its key functions in cell migration, invasion, and matrix remodelling. Finally, we describe the anti-tumour strategies targeting focal adhesion and the current progress in the development of some inhibitors against focal adhesion proteins. In this paper, we summarize for the first time that focal adhesion play a positive feedback role in pro-tumour metastatic matrix remodelling by summarizing the five processes of focal adhesion assembly in a multidimensional way. It is beneficial for researchers to have a deeper understanding of the role of focal adhesion in the biological behaviour of tumour metastasis and the potential of focal adhesion as a therapeutic target, providing new ideas for the prevention and treatment of metastases.
Collapse
Affiliation(s)
- Zonghao Liu
- Department of Radiotherapy, Cancer Hospital of China Medical University, No.44 Xiaoheyan Road, Dadong District, Shenyang, Liaoning Province, 110042, P. R. China
- The First Clinical College, China Medical University, Shenyang, Liaoning Province, 110122, P. R. China
| | - Xiaofang Zhang
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, Liaoning, 110001, China
| | - Tianru Ben
- The First Clinical College, China Medical University, Shenyang, Liaoning Province, 110122, P. R. China
| | - Mo Li
- Department of Breast Surgery, Liaoning Cancer Hospital and Institute, No.44 Xiaoheyan Road, Dadong District, Shenyang, Liaoning Province, 110042, P. R. China
| | - Yi Jin
- Department of Breast Surgery, Liaoning Cancer Hospital and Institute, No.44 Xiaoheyan Road, Dadong District, Shenyang, Liaoning Province, 110042, P. R. China
| | - Tianlu Wang
- Department of Radiotherapy, Cancer Hospital of China Medical University, No.44 Xiaoheyan Road, Dadong District, Shenyang, Liaoning Province, 110042, P. R. China.
- Department of Radiotherapy, Cancer Hospital of Dalian University of Technology, Shenyang, Liaoning Province, 110042, People's Republic of China.
- Faculty of Medicine, Dalian University of Technology, Dalian, Liaoning Province, 116024, P. R. China.
| | - Yingqiu Song
- Department of Radiotherapy, Cancer Hospital of China Medical University, No.44 Xiaoheyan Road, Dadong District, Shenyang, Liaoning Province, 110042, P. R. China.
- Department of Radiotherapy, Liaoning Cancer Hospital & Institute, No.44 Xiaoheyan Road, Dadong District, Shenyang, Liaoning Province, 110042, P. R. China.
| |
Collapse
|
17
|
Reuning U, D'Amore VM, Hodivala-Dilke K, Marinelli L, Kessler H. Importance of integrin transmembrane helical interactions for antagonistic versus agonistic ligand behavior: Consequences for medical applications. Bioorg Chem 2025; 156:108193. [PMID: 39842299 DOI: 10.1016/j.bioorg.2025.108193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 11/27/2024] [Accepted: 01/17/2025] [Indexed: 01/24/2025]
Abstract
Integrins are well-characterized receptors involved in cell adhesion and signaling. With six approved drugs, they are recognized as valuable therapeutic targets. Here, we explore potential activation mechanisms that may clarify the agonist versus antagonist behavior of integrin ligands. The reorganization of the transmembrane domain (TMD) in the integrin receptor, forming homooligomers within focal adhesions, could be key to the understanding of the agonistic properties of integrin ligands at substoichiometric concentrations. This has significant implications for medical applications. While we focus on the RGD peptide-recognizing integrin subfamily, we propose that these mechanistic insights may also apply to other integrin subtypes. For application of integrin ligands in medicine it is essential to consider this mechanism and its consequences for affinity and bioavailability.
Collapse
Affiliation(s)
- Ute Reuning
- TUM University Hospital, Klinikum Rechts der Isar, School of Medicine and Health, Technical University of Munich, Department of Gynecology and Obstetrics, Clinical Research Unit, Ismaninger Strasse 22, 81675 Munich, Germany.
| | - Vincenzo Maria D'Amore
- University of Naples Federico II, UNINA-Department of Pharmacy, C.so Umberto I, 40, 80138 Naples, Italy.
| | - Kairbaan Hodivala-Dilke
- Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, United Kingdom.
| | - Luciana Marinelli
- University of Naples Federico II, UNINA-Department of Pharmacy, C.so Umberto I, 40, 80138 Naples, Italy.
| | - Horst Kessler
- Institute for Advanced Study, Department of Chemistry, School of Natural Sciences and Bavarian NMR Center (BNMRZ), Technical University Munich, Ernst-Otto-Fischer-Str. 2, 85748 Garching, Germany.
| |
Collapse
|
18
|
Bock F, Li S, Pozzi A, Zent R. Integrins in the kidney - beyond the matrix. Nat Rev Nephrol 2025; 21:157-174. [PMID: 39643697 DOI: 10.1038/s41581-024-00906-1] [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] [Accepted: 10/21/2024] [Indexed: 12/09/2024]
Abstract
The development and proper functioning of the kidney is dependent on the interaction of kidney cells with the surrounding extracellular matrix (ECM). These interactions are mediated by heterodimeric membrane-bound receptors called integrins, which bind to the ECM via their extracellular domain and via their cytoplasmic tail to intracellular adaptor proteins, to assemble large macromolecular adhesion complexes. These interactions enable integrins to control cellular functions such as intracellular signalling and organization of the actin cytoskeleton and are therefore crucial to organ function. The different nephron segments and the collecting duct system have unique morphologies, functions and ECM environments and are thus equipped with unique sets of integrins with distinct specificities for the ECM with which they interact. These cell-type-specific functions are facilitated by specific intracellular integrin binding proteins, which are critical in determining the integrin activation status, ligand-binding affinity and the type of ECM signals that are relayed to the intracellular structures. The spatiotemporal expression of integrins and their specific interactions with binding partners underlie the proper development, function and repair processes of the kidney. This Review summarizes our current understanding of how integrins, their binding partners and the actin cytoskeleton regulate kidney development, physiology and pathology.
Collapse
Affiliation(s)
- Fabian Bock
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA.
- Department of Veterans Affairs Hospital, Tennessee Valley Healthcare System, Nashville, Tennessee, USA.
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA.
| | - Shensen Li
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Ambra Pozzi
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Veterans Affairs Hospital, Tennessee Valley Healthcare System, Nashville, Tennessee, USA
- Department of Physiology and Molecular Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Roy Zent
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA.
- Department of Veterans Affairs Hospital, Tennessee Valley Healthcare System, Nashville, Tennessee, USA.
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA.
| |
Collapse
|
19
|
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
|
20
|
Baster Z, Russell L, Rajfur Z. A Review of Talin- and Integrin-Dependent Molecular Mechanisms in Cancer Invasion and Metastasis. Int J Mol Sci 2025; 26:1798. [PMID: 40076426 PMCID: PMC11899650 DOI: 10.3390/ijms26051798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 02/04/2025] [Accepted: 02/06/2025] [Indexed: 03/14/2025] Open
Abstract
Cancer is the second most common cause of death in the world, representing one of the main economic burdens in health care and research. The effort of research has mainly focused on limiting the growth of a localized tumor, but most recently, there has been more attention focused on restricting the spreading of the cancer via invasion and metastasis. The signaling pathways behind these two processes share many molecules with physiological pathways regulating cell adhesion and migration, and, moreover, adhesion and migration processes themselves underlie tumor potential for invasion. In this work, we reviewed the latest literature about cancer development and invasion and their regulation by cell migration- and adhesion-related proteins, with a specific focus on talins and integrins. We also summarized the most recent developments and approaches to anti-cancer therapies, concentrating on cell migration-related therapies.
Collapse
Affiliation(s)
- Zbigniew Baster
- Institute of Physics, Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, 30-348 Kraków, Poland
- Laboratory for Cell and Tissue Engineering, Department of Biomedical Engineering, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands
| | - Lindsay Russell
- Undergraduate Program, Barnard College of Columbia University, New York, NY 10027, USA;
| | - Zenon Rajfur
- Institute of Physics, Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, 30-348 Kraków, Poland
- Jagiellonian Center of Biomedical Imaging, Jagiellonian University, 30-348 Kraków, Poland
| |
Collapse
|
21
|
Negrescu AM, Ionascu I, Necula MG, Tudor N, Kamaleev M, Zarnescu O, Mazare A, Schmuki P, Cimpean A. Lateral Spacing of TiO 2 Nanotube Coatings Modulates In Vivo Early New Bone Formation. Biomimetics (Basel) 2025; 10:81. [PMID: 39997104 PMCID: PMC11853438 DOI: 10.3390/biomimetics10020081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 01/23/2025] [Accepted: 01/24/2025] [Indexed: 02/26/2025] Open
Abstract
Due to the bio-inert nature of titanium (Ti) and subsequent accompanying chronic inflammatory response, an implant's stability and function can be significantly affected, which is why various surface modifications have been employed, including the deposition of titanium oxide (TiO2) nanotubes (TNTs) onto the native surface through the anodic oxidation method. While the influence of nanotube diameter on cell behaviour and osteogenesis is very well documented, information regarding the effects of nanotube lateral spacing on the in vivo new bone formation process is insufficient and hard to find. Considering this, the present study's aim was to evaluate the mechanical properties and the osteogenic ability of two types of TNTs-based pins with different lateral spacing, e.g., 25 nm (TNTs) and 92 nm (spTNTs). The mechanical properties of the TNT-coated implants were characterised from a morphological point of view (tube diameter, spacing, and tube length) using scanning electron microscopy (SEM). In addition, the chemical composition of the implants was evaluated using X-ray photoelectron spectroscopy, while surface roughness and topography were characterised using atomic force microscopy (AFM). Finally, the implants' hardness and elastic modulus were investigated using nanoindentation measurements. The in vivo new bone formation was histologically evaluated (haematoxylin and eosin-HE staining) at 6 and 30 days post-implantation in a rat model. Mechanical characterisation revealed that the two morphologies presented a similar chemical composition and mechanical strength, but, in terms of surface roughness, the spTNTs exhibited a higher average roughness. The microscopic examination at 1 month post-implantation revealed that spTNTs pins (57.21 ± 34.93) were capable of promoting early new bone tissue formation to a greater extent than the TNTs-coated implants (24.37 ± 6.5), with a difference in the average thickness of the newly formed bone tissue of ~32.84 µm, thus highlighting the importance of this parameter when designing future dental/orthopaedic implants.
Collapse
Affiliation(s)
- Andreea Mariana Negrescu
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, 91-95 Spl. Independentei, 050657 Bucharest, Romania; (A.M.N.); (M.G.N.); (O.Z.)
- Research Institute of the University of Bucharest (ICUB), University of Bucharest, 050657 Bucharest, Romania
| | - Iuliana Ionascu
- Faculty of Veterinary Medicine, University of Agronomic Sciences and Veterinary Medicine, 105 Spl. Independentei, 050097 Bucharest, Romania; (I.I.); (N.T.)
| | - Madalina Georgiana Necula
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, 91-95 Spl. Independentei, 050657 Bucharest, Romania; (A.M.N.); (M.G.N.); (O.Z.)
| | - Niculae Tudor
- Faculty of Veterinary Medicine, University of Agronomic Sciences and Veterinary Medicine, 105 Spl. Independentei, 050097 Bucharest, Romania; (I.I.); (N.T.)
| | - Maksim Kamaleev
- Department of Materials Science and Engineering, Chair of General Materials Properties, Friedrich-Alexander-University of Erlangen-Nürnberg, Martensstraße 5, 91058 Erlangen, Germany;
| | - Otilia Zarnescu
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, 91-95 Spl. Independentei, 050657 Bucharest, Romania; (A.M.N.); (M.G.N.); (O.Z.)
| | - Anca Mazare
- Department of Materials Science and Engineering, WW4-LKO, Friedrich-Alexander-University of Erlangen-Nürnberg, Martensstrasse 7, 91058 Erlangen, Germany;
| | - Patrik Schmuki
- Department of Materials Science and Engineering, WW4-LKO, Friedrich-Alexander-University of Erlangen-Nürnberg, Martensstrasse 7, 91058 Erlangen, Germany;
- Regional Centre of Advanced Technologies and Materials, Šlechtitelů 27, 78371 Olomouc, Czech Republic
| | - Anisoara Cimpean
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, 91-95 Spl. Independentei, 050657 Bucharest, Romania; (A.M.N.); (M.G.N.); (O.Z.)
| |
Collapse
|
22
|
Zhu Y, Chen J, Chen C, Tang R, Xu J, Shi S, Yu X. Deciphering mechanical cues in the microenvironment: from non-malignant settings to tumor progression. Biomark Res 2025; 13:11. [PMID: 39849659 PMCID: PMC11755887 DOI: 10.1186/s40364-025-00727-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 01/05/2025] [Indexed: 01/25/2025] Open
Abstract
The tumor microenvironment functions as a dynamic and intricate ecosystem, comprising a diverse array of cellular and non-cellular components that precisely orchestrate pivotal tumor behaviors, including invasion, metastasis, and drug resistance. While unraveling the intricate interplay between the tumor microenvironment and tumor behaviors represents a tremendous challenge, recent research illuminates a crucial biological phenomenon known as cellular mechanotransduction. Within the microenvironment, mechanical cues like tensile stress, shear stress, and stiffness play a pivotal role by activating mechanosensitive effectors such as PIEZO proteins, integrins, and Yes-associated protein. This activation initiates cascades of intrinsic signaling pathways, effectively linking the physical properties of tissues to their physiological and pathophysiological processes like morphogenesis, regeneration, and immunity. This mechanistic insight offers a novel perspective on how the mechanical cues within the tumor microenvironment impact tumor behaviors. While the intricacies of the mechanical tumor microenvironment are yet to be fully elucidated, it exhibits distinct physical attributes from non-malignant tissues, including elevated solid stresses, interstitial hypertension, augmented matrix stiffness, and enhanced viscoelasticity. These traits exert notable influences on tumor progression and treatment responses, enriching our comprehension of the multifaceted nature of the microenvironment. Through this innovative review, we aim to provide a new lens to decipher the mechanical attributes within the tumor microenvironment from non-malignant contexts, broadening our knowledge on how these factors promote or inhibit tumor behaviors, and thus offering valuable insights to identify potential targets for anti-tumor strategies.
Collapse
Affiliation(s)
- Yicheng Zhu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Shanghai Key Laboratory of Precision Medicine for Pancreatic Cancer, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Jiaoshun Chen
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Shanghai Key Laboratory of Precision Medicine for Pancreatic Cancer, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Chen Chen
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Shanghai Key Laboratory of Precision Medicine for Pancreatic Cancer, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Rong Tang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Shanghai Key Laboratory of Precision Medicine for Pancreatic Cancer, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Jin Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Shanghai Key Laboratory of Precision Medicine for Pancreatic Cancer, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Si Shi
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China.
- Shanghai Key Laboratory of Precision Medicine for Pancreatic Cancer, Shanghai, 200032, China.
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China.
| | - Xianjun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China.
- Shanghai Key Laboratory of Precision Medicine for Pancreatic Cancer, Shanghai, 200032, China.
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
23
|
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
|
24
|
Cui L, Zhao S, Teng HL, Yang B, Liu Q, Qin A. Integrins identified as potential prognostic markers in osteosarcoma through multi-omics and multi-dataset analysis. NPJ Precis Oncol 2025; 9:19. [PMID: 39825088 PMCID: PMC11742673 DOI: 10.1038/s41698-024-00794-5] [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: 06/27/2024] [Accepted: 12/19/2024] [Indexed: 01/20/2025] Open
Abstract
Osteosarcoma represents 20% of primary malignant bone tumors globally. Assessing its prognosis is challenging due to the complex roles of integrins in tumor development and metastasis. This study utilized 209,268 osteosarcoma cells from the GEO database to identify integrin-associated genes using advanced analysis methods. A novel machine learning framework combining 10 algorithms was developed to construct an Integrin-related Signature (IRS), which demonstrated robust predictive power across multiple datasets. The IRS's utility in predicting overall survival was confirmed using data from The Cancer Genome Atlas, underscoring its potential in personalized cancer management.
Collapse
Affiliation(s)
- Lei Cui
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning, Guangxi, China
| | - Shuai Zhao
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning, Guangxi, China
| | - Hai Long Teng
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning, Guangxi, China
| | - Biao Yang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangxi Medical University, Nanning, China
| | - Qian Liu
- Guangxi Key Laboratory of Regenerative Medicine, Orthopaedic Department, The First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi, China.
| | - An Qin
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning, Guangxi, China.
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
25
|
Alonso-Matilla R, Provenzano PP, Odde DJ. Physical principles and mechanisms of cell migration. NPJ BIOLOGICAL PHYSICS AND MECHANICS 2025; 2:2. [PMID: 39829952 PMCID: PMC11738987 DOI: 10.1038/s44341-024-00008-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Accepted: 11/19/2024] [Indexed: 01/22/2025]
Abstract
Cell migration is critical in processes such as developmental biology, wound healing, immune response, and cancer invasion/metastasis. Understanding its regulation is essential for developing targeted therapies in regenerative medicine, cancer treatment and immune modulation. This review examines cell migration mechanisms, highlighting fundamental physical principles, key molecular components, and cellular behaviors, identifying existing gaps in current knowledge, and suggesting potential directions for future research.
Collapse
Affiliation(s)
- Roberto Alonso-Matilla
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN USA
- University of Minnesota Center for Multiparametric Imaging of Tumor Immune Microenvironments, Minneapolis, MN USA
| | - Paolo P. Provenzano
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN USA
- University of Minnesota Center for Multiparametric Imaging of Tumor Immune Microenvironments, Minneapolis, MN USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN USA
- Department of Hematology, Oncology, and Transplantation, University of Minnesota, Minneapolis, MN USA
- Stem Cell Institute, University of Minnesota, Minneapolis, MN USA
| | - David J. Odde
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN USA
- University of Minnesota Center for Multiparametric Imaging of Tumor Immune Microenvironments, Minneapolis, MN USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN USA
| |
Collapse
|
26
|
Wang X, Yu S, Xie L, Xiang M, Ma H. The role of the extracellular matrix in cardiac regeneration. Heliyon 2025; 11:e41157. [PMID: 39834404 PMCID: PMC11745795 DOI: 10.1016/j.heliyon.2024.e41157] [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: 07/08/2024] [Revised: 11/16/2024] [Accepted: 12/11/2024] [Indexed: 01/22/2025] Open
Abstract
The extracellular matrix (ECM) is a complex and dynamic three-dimensional network that functions as an architectural scaffold to maintain cardiac homeostasis. Important biochemical and mechanical signals associated with cell‒cell communication are provided via the reciprocal interaction between cells and the ECM. By converting mechanical cues into biochemical signals, the ECM regulates many cell processes, including migration, adhesion, growth, differentiation, proliferation, and apoptosis. Moreover, the ECM facilitates the replacement of dead cells and preserves the structural integrity of the heart, making it essential in conditions such as myocardial infarction and other pathological states. When excessive ECM deposition or abnormal production of ECM components occurs, the heart undergoes fibrosis, leading to cardiac dysfunction and heart failure. However, emerging evidence suggests that the ECM may contribute to heart regeneration following cardiac injury. The present review offers a complete overview of the existing information and novel discoveries regarding the involvement of the ECM in heart regeneration from both mechanical and biochemical perspectives. Understanding the ECM and its involvement in mechanotransduction holds significant potential for advancing therapeutic approaches in heart repair and regeneration.
Collapse
Affiliation(s)
- Xiying Wang
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China
- Department of Gastroenterology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, China
| | - Shuo Yu
- Department of Anesthesiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Lan Xie
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China
- Department of Rheumatology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang, China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, China
| | - Meixiang Xiang
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, China
| | - Hong Ma
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, China
| |
Collapse
|
27
|
Ranga V, Dakal TC, Maurya PK, Johnson MS, Sharma NK, Kumar A. Role of RGD-binding Integrins in ovarian cancer progression, metastasis and response to therapy. Integr Biol (Camb) 2025; 17:zyaf003. [PMID: 39916547 DOI: 10.1093/intbio/zyaf003] [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: 05/08/2024] [Revised: 09/12/2024] [Accepted: 01/29/2025] [Indexed: 05/08/2025]
Abstract
Integrins are transmembrane receptors that play a crucial role in cell adhesion and signaling by connecting the extracellular environment to the intracellular cytoskeleton. After binding with specific ligands in the extracellular matrix (ECM), integrins undergo conformational changes that transmit signals across the cell membrane. The integrin-mediated bidirectional signaling triggers various cellular responses, such as changes in cell shape, migration and proliferation. Irregular integrin expression and activity are closely linked to tumor initiation, angiogenesis, cell motility, invasion, and metastasis. Thus, understanding the intricate regulatory mechanism is essential for slowing cancer progression and preventing carcinogenesis. Among the four classes of integrins, the arginine-glycine-aspartic acid (RGD)-binding integrins stand out as the most crucial integrin receptor subfamily in cancer and its metastasis. Dysregulation of almost all RGD-binding integrins promotes ECM degradation in ovarian cancer, resulting in ovarian carcinoma progression and resistance to therapy. Preclinical studies have demonstrated that targeting these integrins with therapeutic antibodies and ligands, such as RGD-containing peptides and their derivatives, can enhance the precision of these therapeutic agents in treating ovarian cancer. Therefore, the development of novel therapeutic agents is essential for treating ovarian cancer. This review mainly discusses genes and their importance across different ovarian cancer subtypes, the involvement of RGD motif-containing ECM proteins in integrin-mediated signaling in ovarian carcinoma, ongoing, completed, partially completed, and unsuccessful clinical trials of therapeutic agents, as well as existing limitations and challenges, advancements made so far, potential strategies, and directions for future research in the field. Insight Box Integrin-mediated signaling regulates cell migration, proliferation and differentiation. Dysregulated integrin expression and activity promote tumor growth and dissemination. Thus, a proper understanding of this complex regulatory mechanism is essential to delay cancer progression and prevent carcinogenesis. Notably, integrins binding to RGD motifs play an important role in tumor initiation, evolution, and metastasis. Preclinical studies have demonstrated that therapeutic agents, such as antibodies and small molecules with RGD motifs, target RGD-binding integrins and disrupt their interactions with the ECM, thereby inhibiting ovarian cancer proliferation and migration. Altogether, this review highlights the potential of RGD-binding integrins in providing new insights into the progression and metastasis of ovarian cancer and how these integrins have been utilized to develop effective treatment plans.
Collapse
Affiliation(s)
- Vipin Ranga
- DBT-North East Centre for Agricultural Biotechnology (DBT-NECAB), Assam Agricultural University, Agriculture University Road, Jorhat, Assam 785013, India
| | - Tikam Chand Dakal
- Genome and Computational Biology Laboratory, Department of Biotechnology, Mohanlal Sukhadia University, University Road, Udaipur, Rajasthan 313001, India
| | - Pawan Kumar Maurya
- Department of Biochemistry, Central University of Haryana, Central University of Haryana Road, Mahendergarh, Haryana 123031, India
| | - Mark S Johnson
- Structural Bioinformatics Laboratory and InFLAMES Research Flagship Center, Biochemistry, Faculty of Science and Engineering, Åbo Akademi University, Tykistökatu 6A, Turku 20520, Finland
| | - Narendra Kumar Sharma
- Department of Bioscience and Biotechnology, Banasthali Vidyapith, Vanasthali Road, Tonk, Rajasthan 304022, India
| | - Abhishek Kumar
- Manipal Academy of Higher Education (MAHE), Tiger Circle Road, Manipal, Karnataka 576104, India
- Institute of Bioinformatics, Discoverer Building, International Technology Park, Whitefield, Bangalore, Karnataka 560006, India
| |
Collapse
|
28
|
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
|
29
|
Hansen CE, Hollaus D, Kamermans A, de Vries HE. Tension at the gate: sensing mechanical forces at the blood-brain barrier in health and disease. J Neuroinflammation 2024; 21:325. [PMID: 39696463 PMCID: PMC11657007 DOI: 10.1186/s12974-024-03321-2] [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: 10/07/2024] [Accepted: 12/07/2024] [Indexed: 12/20/2024] Open
Abstract
Microvascular brain endothelial cells tightly limit the entry of blood components and peripheral cells into the brain by forming the blood-brain barrier (BBB). The BBB is regulated by a cascade of mechanical and chemical signals including shear stress and elasticity of the adjacent endothelial basement membrane (BM). During physiological aging, but especially in neurological diseases including multiple sclerosis (MS), stroke, small vessel disease, and Alzheimer's disease (AD), the BBB is exposed to inflammation, rigidity changes of the BM, and disturbed cerebral blood flow (CBF). These altered forces lead to increased vascular permeability, reduced endothelial reactivity to vasoactive mediators, and promote leukocyte transmigration. Whereas the molecular players involved in leukocyte infiltration have been described in detail, the importance of mechanical signalling throughout this process has only recently been recognized. Here, we review relevant features of mechanical forces acting on the BBB under healthy and pathological conditions, as well as the endothelial mechanosensory elements detecting and responding to altered forces. We demonstrate the underlying complexity by focussing on the family of transient receptor potential (TRP) ion channels. A better understanding of these processes will provide insights into the pathogenesis of several neurological disorders and new potential leads for treatment.
Collapse
Affiliation(s)
- Cathrin E Hansen
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands
- MS Center Amsterdam, Amsterdam UMC Location VU Medical Center, Amsterdam, The Netherlands
| | - David Hollaus
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
| | - Alwin Kamermans
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands
| | - Helga E de Vries
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands.
- Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands.
- MS Center Amsterdam, Amsterdam UMC Location VU Medical Center, Amsterdam, The Netherlands.
| |
Collapse
|
30
|
Pal K. Unravelling molecular mechanobiology using DNA-based fluorogenic tension sensors. J Mater Chem B 2024; 13:37-53. [PMID: 39564891 DOI: 10.1039/d4tb01858c] [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: 11/21/2024]
Abstract
Investigations of the biological system have revealed many principles that govern regular life processes. Recently, the analysis of tiny mechanical forces associated with many biological processes revealed their significance in understanding biological functions. Consequently, this piqued the interest of researchers, and a series of technologies have been developed to understand biomechanical cues at the molecular level. Notable techniques include single-molecule force spectroscopy, traction force microscopy, and molecular tension sensors. Well-defined double-stranded DNA structures could possess programmable mechanical characteristics, and hence, they have become one of the central molecules in molecular tension sensor technology. With the advancement of DNA technology, DNA or nucleic acid-based robust tension sensors offer the possibility of understanding mechanobiology in the bulk to single-molecule level range with desired spatiotemporal resolution. This review presents a comprehensive account of molecular tension sensors with a special emphasis on DNA-based fluorogenic tension sensors. Along with a detailed discussion on irreversible and reversible DNA-based tension sensors and their application in super-resolution microscopy, a discussion on biomolecules associated with cellular mechanotransduction and key findings in the field are included. This review ends with an elaborate discussion on the current challenges and future prospects of molecular tension sensors.
Collapse
Affiliation(s)
- Kaushik Pal
- Biophysical Chemistry Laboratory, Department of Chemistry, Indian Institute of Technology Tirupati, Yerpedu, Tirupati, AP-517619, India.
| |
Collapse
|
31
|
Peterson RJ, Reed RC, Zamecnik CR, Sallam MA, Finbloom JA, Martinez FJ, Levy JM, Moonwiriyakit A, Desai TA, Koval M. Apical integrins as a switchable target to regulate the epithelial barrier. J Cell Sci 2024; 137:jcs263580. [PMID: 39552289 PMCID: PMC11795292 DOI: 10.1242/jcs.263580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 11/12/2024] [Indexed: 11/19/2024] Open
Abstract
Tight junctions regulate epithelial barrier function and have been shown to be influenced by multiple classes of proteins. Apical integrins have been identified as potential regulators of epithelial barrier function; however, only indirect approaches have been used to measure integrin regulation of the epithelial barrier. Here, we used polymeric nanowires conjugated with anti-integrin β1 antibodies to specifically target apically localized integrins in either their closed or open conformation. Barrier regulation by apical integrins was found to be conformation specific. Nanowires targeting integrins in the closed conformation increased epithelial permeability and caused zonula occludens-1 (ZO-1, also known as TJP1) to change from a linear to a ruffled morphology. Claudin-2 and claudin-4 colocalized with ZO-1 and were also ruffled; however, claudin-1 and claudin-7 remained linear. Ruffling was dependent on myosin light chain kinases (MLCKs) and Rho kinases (ROCKs). Conversely, targeting integrins in the open conformation decreased epithelial permeability and made junctions more linearized. Anti-integrin β1 nanowires differentially affected actin and talin (analyzed using pan-talin antibodies), depending on whether they contained activating or inhibitory antibodies. Thus, apical integrins can act as a conformation-sensitive switch that regulates epithelial barrier function.
Collapse
Affiliation(s)
- Raven J. Peterson
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Ryan C. Reed
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Colin R. Zamecnik
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA 94158, USA
| | - Marwa A. Sallam
- School of Engineering, Brown University, Providence, RI 02912, USA
| | - Joel A. Finbloom
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA 94158, USA
| | - Francisco J. Martinez
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Joshua M. Levy
- Department of Otolaryngology, Emory University School of Medicine, Atlanta, GA 30322, USA
- National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892, USA
| | - Aekkacha Moonwiriyakit
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bang Phli, Samut Prakan, 10540, Thailand
| | - Tejal A. Desai
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA 94158, USA
- School of Engineering, Brown University, Providence, RI 02912, USA
| | - Michael Koval
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
32
|
Ma N, Wu F, Liu J, Wu Z, Wang L, Li B, Liu Y, Dong X, Hu J, Fang X, Zhang H, Ai D, Zhou J, Wang X. Kindlin-2 Phase Separation in Response to Flow Controls Vascular Stability. Circ Res 2024; 135:1141-1160. [PMID: 39492718 DOI: 10.1161/circresaha.124.324773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 10/11/2024] [Accepted: 10/17/2024] [Indexed: 11/05/2024]
Abstract
BACKGROUND Atheroprotective shear stress preserves endothelial barrier function, while atheroprone shear stress enhances endothelial permeability. Yet, the underlying mechanisms through which distinct flow patterns regulate EC integrity remain to be clarified. This study aimed to investigate the involvement of Kindlin-2, a key component of focal adhesion and endothelial adherens junctions crucial for regulating endothelial cell (EC) integrity and vascular stability. METHODS Mouse models of atherosclerosis in EC-specific Kindlin-2 knockout mice (Kindlin-2iΔEC) were used to study the role of Kindlin-2 in atherogenesis. Pulsatile shear (12±4 dynes/cm2) or oscillatory shear (0.5±4 dynes/cm2) were applied to culture ECs. Live-cell imaging, fluorescence recovery after photobleaching assay, and OptoDroplet assay were used to study the liquid-liquid phase separation (LLPS) of Kindlin-2. Co-immunoprecipitation, mutagenesis, proximity ligation assay, and transendothelial electrical resistance assay were used to explore the underlying mechanism of flow-regulated Kindlin-2 function. RESULTS We found that Kindlin-2 localization is altered under different flow patterns. Kindlin-2iΔEC mice showed heightened vascular permeability. Kindlin-2iΔEC were bred onto ApoE-/- mice to generate Kindlin-2iΔEC; ApoE-/- mice, which displayed a significant increase in atherosclerosis lesions. In vitro data showed that in ECs, Kindlin-2 underwent LLPS, a critical process for proper focal adhesion assembly, maturation, and junction formation. Mass spectrometry analysis revealed that oscillatory shear increased arginine methylation of Kindlin-2, catalyzed by PRMT5 (protein arginine methyltransferase 5). Functionally, arginine hypermethylation inhibits Kindlin-2 LLPS, impairing focal adhesion assembly and junction maturation. Notably, we identified R290 of Kindlin-2 as a crucial residue for LLPS and a key site for arginine methylation. Finally, pharmacologically inhibiting arginine methylation reduces EC activation and plaque formation. CONCLUSIONS Collectively, our study elucidates that mechanical force induces arginine methylation of Kindlin-2, thereby regulating vascular stability through its impact on Kindlin-2 LLPS. Targeting Kindlin-2 arginine methylation emerges as a promising hemodynamic-based strategy for treating vascular disorders and atherosclerosis.
Collapse
Affiliation(s)
- Nina Ma
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, State Key Laboratory of Experimental Hematology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics (N.M., F.W., Z.W., L.W., Y.L., X.D., X.W.), School of Basic Medical Sciences, Tianjin Medical University, China
- Department of Ophthalmology, Laboratory of Molecular Ophthalmology and Tianjin Key Laboratory of Ocular Trauma, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Medical University General Hospital, China (N.M., F.W., Z.W., L.W., Y.L., X.D., X.W.)
| | - Fangfang Wu
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, State Key Laboratory of Experimental Hematology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics (N.M., F.W., Z.W., L.W., Y.L., X.D., X.W.), School of Basic Medical Sciences, Tianjin Medical University, China
- Department of Ophthalmology, Laboratory of Molecular Ophthalmology and Tianjin Key Laboratory of Ocular Trauma, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Medical University General Hospital, China (N.M., F.W., Z.W., L.W., Y.L., X.D., X.W.)
| | - Jiayu Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, National Health Commission Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University, Beijing, China (J.L., J.Z.)
| | - Ziru Wu
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, State Key Laboratory of Experimental Hematology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics (N.M., F.W., Z.W., L.W., Y.L., X.D., X.W.), School of Basic Medical Sciences, Tianjin Medical University, China
- Department of Ophthalmology, Laboratory of Molecular Ophthalmology and Tianjin Key Laboratory of Ocular Trauma, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Medical University General Hospital, China (N.M., F.W., Z.W., L.W., Y.L., X.D., X.W.)
| | - Lu Wang
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, State Key Laboratory of Experimental Hematology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics (N.M., F.W., Z.W., L.W., Y.L., X.D., X.W.), School of Basic Medical Sciences, Tianjin Medical University, China
- Department of Ophthalmology, Laboratory of Molecular Ophthalmology and Tianjin Key Laboratory of Ocular Trauma, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Medical University General Hospital, China (N.M., F.W., Z.W., L.W., Y.L., X.D., X.W.)
| | - Bochuan Li
- Department of Physiology and Pathophysiology (B.L., D.A.), School of Basic Medical Sciences, Tianjin Medical University, China
| | - Yuming Liu
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, State Key Laboratory of Experimental Hematology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics (N.M., F.W., Z.W., L.W., Y.L., X.D., X.W.), School of Basic Medical Sciences, Tianjin Medical University, China
- Department of Ophthalmology, Laboratory of Molecular Ophthalmology and Tianjin Key Laboratory of Ocular Trauma, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Medical University General Hospital, China (N.M., F.W., Z.W., L.W., Y.L., X.D., X.W.)
| | - Xue Dong
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, State Key Laboratory of Experimental Hematology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics (N.M., F.W., Z.W., L.W., Y.L., X.D., X.W.), School of Basic Medical Sciences, Tianjin Medical University, China
- Department of Ophthalmology, Laboratory of Molecular Ophthalmology and Tianjin Key Laboratory of Ocular Trauma, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Medical University General Hospital, China (N.M., F.W., Z.W., L.W., Y.L., X.D., X.W.)
| | - Junhao Hu
- Laboratory of Vascular Biology and Organ Homeostasis, Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, China (J.H.)
| | - Xi Fang
- Department of Medicine, University of California San Diego, La Jolla (X.F.)
| | - Heng Zhang
- Department of Biochemistry and Molecular Biology (H.Z.), School of Basic Medical Sciences, Tianjin Medical University, China
| | - Ding Ai
- Department of Physiology and Pathophysiology (B.L., D.A.), School of Basic Medical Sciences, Tianjin Medical University, China
| | - Jing Zhou
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, National Health Commission Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University, Beijing, China (J.L., J.Z.)
| | - Xiaohong Wang
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, State Key Laboratory of Experimental Hematology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics (N.M., F.W., Z.W., L.W., Y.L., X.D., X.W.), School of Basic Medical Sciences, Tianjin Medical University, China
- Department of Ophthalmology, Laboratory of Molecular Ophthalmology and Tianjin Key Laboratory of Ocular Trauma, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Medical University General Hospital, China (N.M., F.W., Z.W., L.W., Y.L., X.D., X.W.)
| |
Collapse
|
33
|
Zhang F, Xia Y, Su J, Quan F, Zhou H, Li Q, Feng Q, Lin C, Wang D, Jiang Z. Neutrophil diversity and function in health and disease. Signal Transduct Target Ther 2024; 9:343. [PMID: 39638788 PMCID: PMC11627463 DOI: 10.1038/s41392-024-02049-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: 05/17/2024] [Revised: 09/21/2024] [Accepted: 10/31/2024] [Indexed: 12/07/2024] Open
Abstract
Neutrophils, the most abundant type of granulocyte, are widely recognized as one of the pivotal contributors to the acute inflammatory response. Initially, neutrophils were considered the mobile infantry of the innate immune system, tasked with the immediate response to invading pathogens. However, recent studies have demonstrated that neutrophils are versatile cells, capable of regulating various biological processes and impacting both human health and disease. Cytokines and other active mediators regulate the functional activity of neutrophils by activating multiple receptors on these cells, thereby initiating downstream signal transduction pathways. Dysfunctions in neutrophils and disruptions in neutrophil homeostasis have been implicated in the pathogenesis of numerous diseases, including cancer and inflammatory disorders, often due to aberrant intracellular signaling. This review provides a comprehensive synthesis of neutrophil biological functions, integrating recent advancements in this field. Moreover, it examines the biological roles of receptors on neutrophils and downstream signaling pathways involved in the regulation of neutrophil activity. The pathophysiology of neutrophils in numerous human diseases and emerging therapeutic approaches targeting them are also elaborated. This review also addresses the current limitations within the field of neutrophil research, highlighting critical gaps in knowledge that warrant further investigation. In summary, this review seeks to establish a comprehensive and multidimensional model of neutrophil regulation, providing new perspectives for potential clinical applications and further research.
Collapse
Affiliation(s)
- Fengyuan Zhang
- Department of Hand and Foot Surgery, Orthopedics Center, The First Hospital of Jilin University, Changchun, People's Republic of China
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China
| | - Yidan Xia
- Department of Hand and Foot Surgery, Orthopedics Center, The First Hospital of Jilin University, Changchun, People's Republic of China
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China
| | - Jiayang Su
- Department of Hand and Foot Surgery, Orthopedics Center, The First Hospital of Jilin University, Changchun, People's Republic of China
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China
| | - Fushi Quan
- Laboratory Animal Center, College of Animal Science, Jilin University, Changchun, China
| | - Hengzong Zhou
- Laboratory Animal Center, College of Animal Science, Jilin University, Changchun, China
| | - Qirong Li
- Laboratory Animal Center, College of Animal Science, Jilin University, Changchun, China
| | - Qiang Feng
- Laboratory Animal Center, College of Animal Science, Jilin University, Changchun, China
| | - Chao Lin
- School of Grain Science and Technology, Jilin Business and Technology College, Changchun, China
| | - Dongxu Wang
- Laboratory Animal Center, College of Animal Science, Jilin University, Changchun, China.
| | - Ziping Jiang
- Department of Hand and Foot Surgery, Orthopedics Center, The First Hospital of Jilin University, Changchun, People's Republic of China.
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
34
|
Li S, Chen A, Gui J, Zhou H, Zhu L, Mi Y. TLN1: an oncogene associated with tumorigenesis and progression. Discov Oncol 2024; 15:716. [PMID: 39589610 PMCID: PMC11599537 DOI: 10.1007/s12672-024-01593-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 11/15/2024] [Indexed: 11/27/2024] Open
Abstract
Talin-1 (TLN1), encoded by the TLN1 gene, is a focal adhesion-related protein capable of binding various proteins in the cytoskeleton. It is also expressed at high levels in many cancers wherein it influences cellular adhesion and the activation of integrins. TLN1 is also capable of promoting tumor cell invasivity, proliferation, and metastatic progression, in addition to being a relevant biomarker and therapeutic target in certain cancers. The present review offers a comprehensive overview of current knowledge regarding TLN1 with respect to its structural properties, functions, and role in tumor development.
Collapse
Affiliation(s)
- Sixin Li
- Department of Urology, Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Wuxi, 214122, Jiangsu, China
- Wuxi School of Medicine, Jiangnan University, 1800 Lihudadao, Wuxi, 214122, Jiangsu, China
| | - Anjie Chen
- Department of Urology, Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Wuxi, 214122, Jiangsu, China
- Wuxi School of Medicine, Jiangnan University, 1800 Lihudadao, Wuxi, 214122, Jiangsu, China
| | - Jiandong Gui
- Department of Urology, Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Wuxi, 214122, Jiangsu, China
- Wuxi School of Medicine, Jiangnan University, 1800 Lihudadao, Wuxi, 214122, Jiangsu, China
| | - Hangsheng Zhou
- Department of Urology, Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Wuxi, 214122, Jiangsu, China
- Wuxi School of Medicine, Jiangnan University, 1800 Lihudadao, Wuxi, 214122, Jiangsu, China
| | - Lijie Zhu
- Department of Urology, Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Wuxi, 214122, Jiangsu, China.
| | - Yuanyuan Mi
- Department of Urology, Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Wuxi, 214122, Jiangsu, China.
| |
Collapse
|
35
|
Bogdanović B, Fagret D, Ghezzi C, Montemagno C. Integrin Targeting and Beyond: Enhancing Cancer Treatment with Dual-Targeting RGD (Arginine-Glycine-Aspartate) Strategies. Pharmaceuticals (Basel) 2024; 17:1556. [PMID: 39598465 PMCID: PMC11597078 DOI: 10.3390/ph17111556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 10/11/2024] [Accepted: 10/18/2024] [Indexed: 11/29/2024] Open
Abstract
Integrins, an important superfamily of cell adhesion receptors, play an essential role in cancer progression, metastasis, and angiogenesis, establishing them as prime targets for both diagnostic and therapeutic applications. Despite their significant potential, integrin-targeted therapies have faced substantial challenges in clinical trials, including variable efficacy and unmet high expectations. Nevertheless, the consistent expression of integrins on tumor and stromal cells underscores their ongoing relevance and potential. Traditional RGD-based imaging and therapeutic agents have faced limitations, such as inconsistent target expression and rapid systemic clearance, which have reduced their effectiveness. To overcome these challenges, recent research has focused on advancing RGD-based strategies and exploring innovative solutions. This review offers a thorough analysis of the latest developments in the RGD-integrin field, with a particular focus on addressing previous limitations. It delves into new dual-targeting approaches and cutting-edge RGD-based agents designed to improve both tumor diagnosis and therapeutic outcomes. By examining these advancements, this review illuminates new pathways for enhancing the specificity and efficacy of integrin-targeted therapies, paving the way for more effective cancer diagnosis and treatment strategies.
Collapse
Affiliation(s)
- Bojana Bogdanović
- INSERM, CHU Grenoble Alpes, Laboratory of Bioclinical Radiopharmaceutics, University Grenoble Alpes, 38000 Grenoble, France; (B.B.); (D.F.); (C.G.)
| | - Daniel Fagret
- INSERM, CHU Grenoble Alpes, Laboratory of Bioclinical Radiopharmaceutics, University Grenoble Alpes, 38000 Grenoble, France; (B.B.); (D.F.); (C.G.)
| | - Catherine Ghezzi
- INSERM, CHU Grenoble Alpes, Laboratory of Bioclinical Radiopharmaceutics, University Grenoble Alpes, 38000 Grenoble, France; (B.B.); (D.F.); (C.G.)
| | | |
Collapse
|
36
|
Xia T, Pan Z, Wan H, Li Y, Mao G, Zhao J, Zhang F, Pan S. Mechanisms of mechanical stimulation in the development of respiratory system diseases. Am J Physiol Lung Cell Mol Physiol 2024; 327:L724-L739. [PMID: 39316681 DOI: 10.1152/ajplung.00122.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 09/06/2024] [Accepted: 09/11/2024] [Indexed: 09/26/2024] Open
Abstract
During respiration, mechanical stress can initiate biological responses that impact the respiratory system. Mechanical stress plays a crucial role in the development of the respiratory system. However, pathological mechanical stress can impact the onset and progression of respiratory diseases by influencing the extracellular matrix and cell transduction processes. In this article, we explore the mechanisms by which mechanical forces communicate with and influence cells. We outline the basic knowledge of respiratory mechanics, elucidating the important role of mechanical stimulation in influencing respiratory system development and differentiation from a microscopic perspective. We also explore the potential mechanisms of mechanical transduction in the pathogenesis and development of respiratory diseases such as asthma, lung injury, pulmonary fibrosis, and lung cancer. Finally, we look forward to new research directions in cellular mechanotransduction, aiming to provide fresh insights for future therapeutic research on respiratory diseases.
Collapse
Affiliation(s)
- Tian Xia
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
- Institute of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Ziyin Pan
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University, Shanghai, People's Republic of China
| | - Haoxin Wan
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
- Institute of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Yongsen Li
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
- Institute of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Guocai Mao
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
- Institute of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Jun Zhao
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
- Institute of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Fangbiao Zhang
- Department of Cardiothoracic Surgery, Lishui Municipal Central Hospital, Lishui, People's Republic of China
| | - Shu Pan
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
- Institute of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| |
Collapse
|
37
|
Qin L, Li H, Lu H, Chen J, Wang H, Liao E. Tandem Mass Tag-based proteomic analysis of protein changes in superchilled crayfish (Procambarus clarkii) presoaked with carrageenan oligosaccharides. Food Chem 2024; 457:140126. [PMID: 38936119 DOI: 10.1016/j.foodchem.2024.140126] [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: 11/14/2023] [Revised: 06/10/2024] [Accepted: 06/13/2024] [Indexed: 06/29/2024]
Abstract
To assess the effectiveness of carrageenan oligosaccharides (COs) in enhancing superchilling storage of crayfish, the physicochemical features of muscle and protein abundance in the refrigerated sample (RS), superchilled sample (SS) and COs soaked superchilled sample (CS) were evaluated. Microstructural and SDS-PAGE analyses suggested that CS exhibited fewer pores, with a microstructure and protein subunits distribution more similar to RS. Tandem Mass Tags quantitative proteomic analysis revealed 66 up-regulated differentially abundant proteins (DAPs) in the CS vs. SS batch, including myosin light chain 2, neural cadherin, integrin beta, lectin-like protein, toll-1, reticulon-1, and moesin/ezrin/radixin homolog 1, which facilitate cells adhesion and maintain membrane/cytoskeleton integrity. Eukaryotic Clusters of Orthologous Groups results confirmed that COs treatment increased the stability of crayfish myofibrillar proteins by up-regulating DAPs, which were concentrated in functional categories such as "posttranslation modification, protein turnover, chaperones", "signal transduction mechanisms", "energy production and conversion", and "cytoskeleton".
Collapse
Affiliation(s)
- Lerong Qin
- College of Food Science and Engineering, Wuhan Polytechnic University, Wuhan 430023, China
| | - Han Li
- College of Food Science and Engineering, Wuhan Polytechnic University, Wuhan 430023, China
| | - Hongyan Lu
- College of Food Science and Engineering, Wuhan Polytechnic University, Wuhan 430023, China; Hubei Key Laboratory for Processing and Transformation of Agricultural Products (Wuhan Polytechnic University), Wuhan 430023, China
| | - Jiwang Chen
- College of Food Science and Engineering, Wuhan Polytechnic University, Wuhan 430023, China; Hubei Key Laboratory for Processing and Transformation of Agricultural Products (Wuhan Polytechnic University), Wuhan 430023, China; National Research & Development Branch Center for Crayfish Processing (Qianjiang), Qianjiang 433100, China
| | - Haibin Wang
- College of Food Science and Engineering, Wuhan Polytechnic University, Wuhan 430023, China; Hubei Key Laboratory for Processing and Transformation of Agricultural Products (Wuhan Polytechnic University), Wuhan 430023, China; National Research & Development Branch Center for Crayfish Processing (Qianjiang), Qianjiang 433100, China
| | - E Liao
- College of Food Science and Engineering, Wuhan Polytechnic University, Wuhan 430023, China; Hubei Key Laboratory for Processing and Transformation of Agricultural Products (Wuhan Polytechnic University), Wuhan 430023, China; National Research & Development Branch Center for Crayfish Processing (Qianjiang), Qianjiang 433100, China.
| |
Collapse
|
38
|
Mierke CT. Mechanosensory entities and functionality of endothelial cells. Front Cell Dev Biol 2024; 12:1446452. [PMID: 39507419 PMCID: PMC11538060 DOI: 10.3389/fcell.2024.1446452] [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: 06/09/2024] [Accepted: 10/04/2024] [Indexed: 11/08/2024] Open
Abstract
The endothelial cells of the blood circulation are exposed to hemodynamic forces, such as cyclic strain, hydrostatic forces, and shear stress caused by the blood fluid's frictional force. Endothelial cells perceive mechanical forces via mechanosensors and thus elicit physiological reactions such as alterations in vessel width. The mechanosensors considered comprise ion channels, structures linked to the plasma membrane, cytoskeletal spectrin scaffold, mechanoreceptors, and junctional proteins. This review focuses on endothelial mechanosensors and how they alter the vascular functions of endothelial cells. The current state of knowledge on the dysregulation of endothelial mechanosensitivity in disease is briefly presented. The interplay in mechanical perception between endothelial cells and vascular smooth muscle cells is briefly outlined. Finally, future research avenues are highlighted, which are necessary to overcome existing limitations.
Collapse
|
39
|
Ahmadi Jazi S, Tajik F, Rezagholizadeh F, Taha SR, Shariat Zadeh M, Bouzari B, Madjd Z. Higher Expression of Talin-1 is Associated With Less Aggressive Tumor Behavior in Pancreatic Cancer. Appl Immunohistochem Mol Morphol 2024; 32:425-435. [PMID: 39258796 DOI: 10.1097/pai.0000000000001220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 07/16/2024] [Indexed: 09/12/2024]
Abstract
Talin-1 is one of the major scaffold proteins in focal adhesions playing a vital role in cell migration, metastasis, and cancer progression. Although studies regarding the importance of Talin-1 in cancer have rapidly developed, its prognostic and diagnostic value still remain unsatisfying in pancreatic cancer (PC). Therefore, the present study aims to investigate the expression, clinical significance, as well as the prognostic and diagnostic value of Talin-1 in different types of PC. Bioinformatic analysis was applied to determine the clinical importance and biological role of Talin-1 expression in PC tumors and the normal adjacent samples. The expression patterns, clinical significance, prognosis, and diagnosis value of Talin-1 were evaluated in tissue microarrays (TMAs) of 190 PC samples including 170 pancreatic ductal adenocarcinoma (PDAC), and 20 pancreatic neuroendocrine tumors (PNET), along with 24 adjacent normal tissues using immunohistochemistry (IHC). The results indicated that the expression of Talin-1 was upregulated in tumor cells compared with adjacent normal tissues. A statistically significant association was observed between the higher cytoplasmic expression of Talin-1 and lower histologic grade ( P <0.001) in PDAC samples. Further, our findings indicated an inverse significant correlation between cytoplasmic expression of Talin-1 and recurrence ( P =0.014) in PNET samples. No significant association was observed between the cytoplasmic expression of Talin-1 and survival outcomes as well as diagnostic accuracy. In conclusion, our observations demonstrated that a higher cytoplasmic level of Talin-1 protein was significantly associated with less aggressive tumor behaviors in PC samples. Nevertheless, further investigations are required to explore the prognostic plus diagnostic value, and mechanism of action of Talin-1 in pancreatic cancer.
Collapse
Affiliation(s)
- Samira Ahmadi Jazi
- Department of Pathology, School of Medicine, Iran University of Medical Sciences
| | - Fatemeh Tajik
- Oncopathology Research Center, Iran University of Medical Sciences
- Department of Surgery, University of California, Irvine Medical Center, Orange, CA
| | - Fereshteh Rezagholizadeh
- Oncopathology Research Center, Iran University of Medical Sciences
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Seyed Reza Taha
- Oncopathology Research Center, Iran University of Medical Sciences
| | | | - Behnaz Bouzari
- Department of Pathology, School of Medicine, Iran University of Medical Sciences
| | - Zahra Madjd
- Department of Pathology, School of Medicine, Iran University of Medical Sciences
- Oncopathology Research Center, Iran University of Medical Sciences
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
40
|
Mariasoosai C, Bose S, Natesan S. Structural insights into the molecular recognition of integrin αVβ3 by RGD-containing ligands: The role of the specificity-determining loop (SDL). BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.23.614545. [PMID: 39386435 PMCID: PMC11463590 DOI: 10.1101/2024.09.23.614545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Integrin αVβ3 is a prominent member of the "RGD-recognizing" integrin family of cell surface receptors. αVβ3 binds to various extracellular matrix (ECM) proteins and oxysterols such as 25-hydroxycholesterol, is implicated in several diseases, including cancer metastasis, lung fibrosis, inflammation, and autoimmune diseases, and is pursued as a valuable therapeutic target. Despite enormous efforts to seek a pure antagonist, to date, no single drug candidate has successfully reached clinics due to associated partial agonism and toxicity issues. Developing effective and safe inhibitors require a thorough understanding of the molecular interactions and structural changes related to the receptor's activation and inhibition mechanisms. This study offers a comprehensive residue-residue contact and network analyses of the ligand-binding β-propeller βI domains (headpiece) based on all available experimental structures of integrin αVβ3 in unliganded, agonist-, antagonist-, and antibody-bound states. The analyses reveal many critical interactions that were not reported before and show that specific orientation and interactions of residues from the specificity-determining loop (SDL) are critical in molecular recognition and regulation. Also, the network analysis reveals that residues from the nearby allosteric site (site II) connect to the primary RGD-binding site via SDL, which likely acts as an interface between the two sites. Our results provide valuable insights into molecular interactions, structural changes, distinct features of the active and inactive headpiece conformations, the role of SDL in ligand recognition, and SDL-mediated allostery. Thus, the insights from this study may facilitate the designing of pure antagonists or site II-mediated allosteric modulators to integrin αVβ3 to treat various diseases.
Collapse
Affiliation(s)
- Charles Mariasoosai
- College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA 99202, USA
| | - Santanu Bose
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA
| | - Senthil Natesan
- College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA 99202, USA
| |
Collapse
|
41
|
Cui Y, Rolova T, Fagerholm SC. The role of integrins in brain health and neurodegenerative diseases. Eur J Cell Biol 2024; 103:151441. [PMID: 39002282 DOI: 10.1016/j.ejcb.2024.151441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 06/28/2024] [Accepted: 07/02/2024] [Indexed: 07/15/2024] Open
Abstract
Integrins are heterodimeric membrane proteins expressed on the surface of most cells. They mediate adhesion and signaling processes relevant for a wealth of physiological processes, including nervous system development and function. Interestingly, integrins are also recognized therapeutic targets for inflammatory diseases, such as multiple sclerosis. Here, we discuss the role of integrins in brain development and function, as well as in neurodegenerative diseases affecting the brain (Alzheimer's disease, multiple sclerosis, stroke). Furthermore, we discuss therapeutic targeting of these adhesion receptors in inflammatory diseases of the brain.
Collapse
Affiliation(s)
- Yunhao Cui
- Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki 00790, Finland
| | - Taisia Rolova
- Neuroscience Center, HiLIFE, University of Helsinki, Helsinki 00290, Finland
| | - Susanna C Fagerholm
- Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki 00790, Finland.
| |
Collapse
|
42
|
Baade T, Michaelis M, Prestel A, Paone C, Klishin N, Herbinger M, Scheinost L, Nedielkov R, Hauck CR, Möller HM. A flexible loop in the paxillin LIM3 domain mediates its direct binding to integrin β subunits. PLoS Biol 2024; 22:e3002757. [PMID: 39231388 PMCID: PMC11374337 DOI: 10.1371/journal.pbio.3002757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 07/17/2024] [Indexed: 09/06/2024] Open
Abstract
Integrins are fundamental for cell adhesion and the formation of focal adhesions (FA). Accordingly, these receptors guide embryonic development, tissue maintenance, and haemostasis but are also involved in cancer invasion and metastasis. A detailed understanding of the molecular interactions that drive integrin activation, FA assembly, and downstream signalling cascades is critical. Here, we reveal a direct association of paxillin, a marker protein of FA sites, with the cytoplasmic tails of the integrin β1 and β3 subunits. The binding interface resides in paxillin's LIM3 domain, where based on the NMR structure and functional analyses, a flexible, 7-amino acid loop engages the unstructured part of the integrin cytoplasmic tail. Genetic manipulation of the involved residues in either paxillin or integrin β3 compromises cell adhesion and motility of murine fibroblasts. This direct interaction between paxillin and the integrin cytoplasmic domain identifies an alternative, kindlin-independent mode of integrin outside-in signalling particularly important for integrin β3 function.
Collapse
Affiliation(s)
- Timo Baade
- Lehrstuhl Zellbiologie, Universität Konstanz, Konstanz, Germany
- Konstanz Research School Chemical Biology, Universität Konstanz, Konstanz, Germany
| | - Marcus Michaelis
- Analytische Chemie, Universität Potsdam, Potsdam, Germany
- DFG Research Training Group 2473 "Bioactive Peptides"
| | | | - Christoph Paone
- Lehrstuhl Zellbiologie, Universität Konstanz, Konstanz, Germany
- Konstanz Research School Chemical Biology, Universität Konstanz, Konstanz, Germany
| | - Nikolai Klishin
- Analytische Chemie, Universität Potsdam, Potsdam, Germany
- DFG Research Training Group 2473 "Bioactive Peptides"
| | | | - Laura Scheinost
- Lehrstuhl Zellbiologie, Universität Konstanz, Konstanz, Germany
| | | | - Christof R Hauck
- Lehrstuhl Zellbiologie, Universität Konstanz, Konstanz, Germany
- Konstanz Research School Chemical Biology, Universität Konstanz, Konstanz, Germany
| | - Heiko M Möller
- Analytische Chemie, Universität Potsdam, Potsdam, Germany
- DFG Research Training Group 2473 "Bioactive Peptides"
| |
Collapse
|
43
|
Wang X, Baster Z, Azizi L, Li L, Rajfur Z, Hytönen VP, Huang C. Talin2 binds to non-muscle myosin IIa and regulates cell attachment and fibronectin secretion. Sci Rep 2024; 14:20175. [PMID: 39215026 PMCID: PMC11364542 DOI: 10.1038/s41598-024-70866-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024] Open
Abstract
Talin2 is localized to large focal adhesions and is indispensable for traction force generation, invadopodium formation, cell invasion as well as metastasis. Talin2 has a higher affinity toward β-integrin tails than talin1. Moreover, disruption of the talin2-β-integrin interaction inhibits traction force generation, invadopodium formation and cell invasion, indicating that a strong talin2-β-integrin interaction is required for talin2 to fulfill these functions. Nevertheless, the role of talin2 in mediation of these processes remains unknown. Here we show that talin2 binds to the N-terminus of non-muscle myosin IIA (NMIIA) through its F3 subdomain. Moreover, talin2 co-localizes with NMIIA at cell edges as well as at some cytoplasmic spots. Talin2 also co-localizes with cortactin, an invadopodium marker. Furthermore, overexpression of NMIIA promoted the talin2 head binding to the β1-integrin tail, whereas knockdown of NMIIA reduced fibronectin and matrix metalloproteinase secretion as well as inhibited cell attachment on fibronectin-coated substrates. These results suggest that talin2 binds to NMIIA to control the secretion of extracellular matrix proteins and this interaction modulates cell adhesion.
Collapse
Affiliation(s)
- Xiaochuan Wang
- The Second Hospital of Shandong University, Jinan, 250033, Shandong, China.
| | - Zbigniew Baster
- Markey Cancer Center, University of Kentucky, Lexington, KY, 40506, USA
- Institute of Physics, Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, 30-348, Kraków, Poland
| | - Latifeh Azizi
- Faculty of Medicine and Health Technology, Tampere University, 33520, Tampere, Finland
- Fimlab Laboratories, Tampere, Finland
| | - Liqing Li
- Markey Cancer Center, University of Kentucky, Lexington, KY, 40506, USA
| | - Zenon Rajfur
- Institute of Physics, Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, 30-348, Kraków, Poland
- Jagiellonian Center of Biomedical Imaging, Jagiellonian University, 30-348, Kraków, Poland
| | - Vesa P Hytönen
- Faculty of Medicine and Health Technology, Tampere University, 33520, Tampere, Finland.
- Fimlab Laboratories, Tampere, Finland.
| | - Cai Huang
- Markey Cancer Center, University of Kentucky, Lexington, KY, 40506, USA.
- Doer Biologics Inc, 2nd Floor, Building 3, Hexiang Science and Technology Center, Medicine Port Town, Qiantang District, Hangzhou, Zhejiang Province, China.
| |
Collapse
|
44
|
Tavasoli M, McMaster CR. Defects in integrin complex formation promote CHKB-mediated muscular dystrophy. Life Sci Alliance 2024; 7:e202301956. [PMID: 38749543 PMCID: PMC11096732 DOI: 10.26508/lsa.202301956] [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: 01/27/2023] [Revised: 05/03/2024] [Accepted: 05/07/2024] [Indexed: 05/18/2024] Open
Abstract
Phosphatidylcholine (PC) is the major membrane phospholipid in most eukaryotic cells. Bi-allelic loss of function variants in CHKB, encoding the first step in the synthesis of PC, is the cause of a rostrocaudal muscular dystrophy in both humans and mice. Loss of sarcolemma integrity is a hallmark of muscular dystrophies; however, how this occurs in the absence of choline kinase function is not known. We determine that in Chkb -/- mice there is a failure of the α7β1 integrin complex that is specific to affected muscle. We observed that in Chkb -/- hindlimb muscles there is a decrease in sarcolemma association/abundance of the PI(4,5)P2 binding integrin complex proteins vinculin, and α-actinin, and a decrease in actin association with the sarcolemma. In cells, pharmacological inhibition of choline kinase activity results in internalization of a fluorescent PI(4,5)P2 reporter from discrete plasma membrane clusters at the cell surface membrane to cytosol, this corresponds with a decreased vinculin localization at plasma membrane focal adhesions that was rescued by overexpression of CHKB.
Collapse
Affiliation(s)
- Mahtab Tavasoli
- Department of Pharmacology, Dalhousie University, Halifax, Canada
| | | |
Collapse
|
45
|
Younesi FS, Miller AE, Barker TH, Rossi FMV, Hinz B. Fibroblast and myofibroblast activation in normal tissue repair and fibrosis. Nat Rev Mol Cell Biol 2024; 25:617-638. [PMID: 38589640 DOI: 10.1038/s41580-024-00716-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/21/2024] [Indexed: 04/10/2024]
Abstract
The term 'fibroblast' often serves as a catch-all for a diverse array of mesenchymal cells, including perivascular cells, stromal progenitor cells and bona fide fibroblasts. Although phenotypically similar, these subpopulations are functionally distinct, maintaining tissue integrity and serving as local progenitor reservoirs. In response to tissue injury, these cells undergo a dynamic fibroblast-myofibroblast transition, marked by extracellular matrix secretion and contraction of actomyosin-based stress fibres. Importantly, whereas transient activation into myofibroblasts aids in tissue repair, persistent activation triggers pathological fibrosis. In this Review, we discuss the roles of mechanical cues, such as tissue stiffness and strain, alongside cell signalling pathways and extracellular matrix ligands in modulating myofibroblast activation and survival. We also highlight the role of epigenetic modifications and myofibroblast memory in physiological and pathological processes. Finally, we discuss potential strategies for therapeutically interfering with these factors and the associated signal transduction pathways to improve the outcome of dysregulated healing.
Collapse
Affiliation(s)
- Fereshteh Sadat Younesi
- Keenan Research Institute for Biomedical Science of the St. Michael's Hospital, Toronto, Ontario, Canada
- Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - Andrew E Miller
- Department of Biomedical Engineering, School of Engineering and Applied Science, University of Virginia, Charlottesville, VA, USA
| | - Thomas H Barker
- Department of Biomedical Engineering, School of Engineering and Applied Science, University of Virginia, Charlottesville, VA, USA
| | - Fabio M V Rossi
- School of Biomedical Engineering and Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Boris Hinz
- Keenan Research Institute for Biomedical Science of the St. Michael's Hospital, Toronto, Ontario, Canada.
- Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
46
|
Shi H, Song J, Gao L, Shan X, Panicker SR, Yao L, McDaniel M, Zhou M, McGee S, Zhong H, Griffin CT, Xia L, Shao B. Deletion of Talin1 in Myeloid Cells Facilitates Atherosclerosis in Mice. Arterioscler Thromb Vasc Biol 2024; 44:1799-1812. [PMID: 38899470 DOI: 10.1161/atvbaha.123.319677] [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: 06/01/2023] [Accepted: 04/23/2024] [Indexed: 06/21/2024]
Abstract
BACKGROUND Integrin-regulated monocyte recruitment and cellular responses of monocyte-derived macrophages are critical for the pathogenesis of atherosclerosis. In the canonical model, talin1 controls ligand binding to integrins, a prerequisite for integrins to mediate leukocyte recruitment and induce immune responses. However, the role of talin1 in the development of atherosclerosis has not been studied. Our study investigated how talin1 in myeloid cells regulates the progression of atherosclerosis. METHODS On an Apoe-/- background, myeloid talin1-deficient mice and the control mice were fed with a high-fat diet for 8 or 12 weeks to induce atherosclerosis. The atherosclerosis development in the aorta and monocyte recruitment into atherosclerotic lesions were analyzed. RESULTS Myeloid talin1 deletion facilitated the formation of atherosclerotic lesions and macrophage deposition in lesions. Talin1 deletion abolished integrin β2-mediated adhesion of monocytes but did not impair integrin α4β1-dependent cell adhesion in a flow adhesion assay. Strikingly, talin1 deletion did not prevent Mn2+- or chemokine-induced activation of integrin α4β1 to the high-affinity state for ligands. In an in vivo competitive homing assay, monocyte infiltration into inflamed tissues was prohibited by antibodies to integrin α4β1 but was not affected by talin1 deletion or antibodies to integrin β2. Furthermore, quantitative polymerase chain reaction and ELISA (enzyme-linked immunosorbent assay) analysis showed that macrophages produced cytokines to promote inflammation and the proliferation of smooth muscle cells. Ligand binding to integrin β3 inhibited cytokine generation in macrophages, although talin1 deletion abolished the negative effects of integrin β3. CONCLUSIONS Integrin α4β1 controls monocyte recruitment during atherosclerosis. Talin1 is dispensable for integrin α4β1 activation to the high-affinity state and integrin α4β1-mediated monocyte recruitment. Yet, talin1 is required for integrin β3 to inhibit the production of inflammatory cytokines in macrophages. Thus, intact monocyte recruitment and elevated inflammatory responses cause enhanced atherosclerosis in talin1-deficient mice. Our study provides novel insights into the roles of myeloid talin1 and integrins in the progression of atherosclerosis.
Collapse
Affiliation(s)
- Huiping Shi
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation (H.S., J.S., L.G., X.S., S.R.P., L.Y., M.M., M.Z., S.M., C.T.G., L.X., B.S.)
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center (H.S., L.X.)
| | - Jianhua Song
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation (H.S., J.S., L.G., X.S., S.R.P., L.Y., M.M., M.Z., S.M., C.T.G., L.X., B.S.)
| | - Liang Gao
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation (H.S., J.S., L.G., X.S., S.R.P., L.Y., M.M., M.Z., S.M., C.T.G., L.X., B.S.)
| | - Xindi Shan
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation (H.S., J.S., L.G., X.S., S.R.P., L.Y., M.M., M.Z., S.M., C.T.G., L.X., B.S.)
| | - Sumith R Panicker
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation (H.S., J.S., L.G., X.S., S.R.P., L.Y., M.M., M.Z., S.M., C.T.G., L.X., B.S.)
| | - Longbiao Yao
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation (H.S., J.S., L.G., X.S., S.R.P., L.Y., M.M., M.Z., S.M., C.T.G., L.X., B.S.)
| | - Michael McDaniel
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation (H.S., J.S., L.G., X.S., S.R.P., L.Y., M.M., M.Z., S.M., C.T.G., L.X., B.S.)
| | - Meixiang Zhou
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation (H.S., J.S., L.G., X.S., S.R.P., L.Y., M.M., M.Z., S.M., C.T.G., L.X., B.S.)
| | - Samuel McGee
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation (H.S., J.S., L.G., X.S., S.R.P., L.Y., M.M., M.Z., S.M., C.T.G., L.X., B.S.)
| | - Hui Zhong
- Lindsley F. Kimball Research Institute, New York Blood Center (H.Z., B.S.)
| | - Courtney T Griffin
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation (H.S., J.S., L.G., X.S., S.R.P., L.Y., M.M., M.Z., S.M., C.T.G., L.X., B.S.)
| | - Lijun Xia
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation (H.S., J.S., L.G., X.S., S.R.P., L.Y., M.M., M.Z., S.M., C.T.G., L.X., B.S.)
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center (H.S., L.X.)
| | - Bojing Shao
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation (H.S., J.S., L.G., X.S., S.R.P., L.Y., M.M., M.Z., S.M., C.T.G., L.X., B.S.)
- Lindsley F. Kimball Research Institute, New York Blood Center (H.Z., B.S.)
| |
Collapse
|
47
|
Miao MZ, Lee JS, Yamada KM, Loeser RF. Integrin signalling in joint development, homeostasis and osteoarthritis. Nat Rev Rheumatol 2024; 20:492-509. [PMID: 39014254 PMCID: PMC11886400 DOI: 10.1038/s41584-024-01130-8] [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] [Accepted: 05/24/2024] [Indexed: 07/18/2024]
Abstract
Integrins are key regulators of cell-matrix interactions during joint development and joint tissue homeostasis, as well as in the development of osteoarthritis (OA). The signalling cascades initiated by the interactions of integrins with a complex network of extracellular matrix (ECM) components and intracellular adaptor proteins orchestrate cellular responses necessary for maintaining joint tissue integrity. Dysregulated integrin signalling, triggered by matrix degradation products such as matrikines, disrupts this delicate balance, tipping the scales towards an environment conducive to OA pathogenesis. The interplay between integrin signalling and growth factor pathways further underscores the multifaceted nature of OA. Moreover, emerging insights into the role of endocytic trafficking in regulating integrin signalling add a new layer of complexity to the understanding of OA development. To harness the therapeutic potential of targeting integrins for mitigation of OA, comprehensive understanding of their molecular mechanisms across joint tissues is imperative. Ultimately, deciphering the complexities of integrin signalling will advance the ability to treat OA and alleviate its global burden.
Collapse
Affiliation(s)
- Michael Z Miao
- Cell Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
- Craniofacial Anomalies and Regeneration Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
- Division of Rheumatology, Allergy, and Immunology and the Thurston Arthritis Research Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Janice S Lee
- Craniofacial Anomalies and Regeneration Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
- Office of the Clinical Director, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - Kenneth M Yamada
- Cell Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA.
| | - Richard F Loeser
- Division of Rheumatology, Allergy, and Immunology and the Thurston Arthritis Research Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
48
|
Li Y, Wang S, Zhang Y, Liu Z, Zheng Y, Zhang K, Chen S, Lv X, Huang M, Pan X, Zheng Y, Yuan M, Ge G, Zeng YA, Lin C, Chen J. Ca 2+ transients on the T cell surface trigger rapid integrin activation in a timescale of seconds. Nat Commun 2024; 15:6131. [PMID: 39033133 PMCID: PMC11271479 DOI: 10.1038/s41467-024-50464-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Accepted: 07/12/2024] [Indexed: 07/23/2024] Open
Abstract
One question in lymphocyte homing is how integrins are rapidly activated to enable immediate arrest of fast rolling lymphocytes upon encountering chemokines at target vascular beds given the slow chemokine-induced integrin inside-out activation. Herein we demonstrate that chemokine CCL25-triggered Ca2+ influx induces T cell membrane-proximal external Ca2+ concentration ([Ca2+]ex) drop in 6 s from physiological concentration 1.2 mM to 0.3 mM, a critical extracellular Ca2+ threshold for inducing αLβ2 activation, triggering rapid αLβ2 activation and T cell arrest before occurrence of αLβ2 inside-out activation. Talin knockdown inhibits the slow inside-out activation of αLβ2 but not [Ca2+]ex drop-triggered αLβ2 quick activation. Blocking Ca2+ influx significantly suppresses T cell rolling-to-arrest transition and homing to skin lesions in a mouse psoriasis model, thus alleviating skin inflammation. [Ca2+]ex decrease-triggered rapid integrin activation bridges the gap between initial chemokine stimulation and slow integrin inside-out activation, ensuring immediate lymphocyte arrest and subsequent diapedesis on the right location.
Collapse
Affiliation(s)
- Yue Li
- State Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - ShiHui Wang
- State Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - YouHua Zhang
- Department of Pathology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - ZhaoYuan Liu
- State Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - YunZhe Zheng
- State Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Kun Zhang
- State Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - ShiYang Chen
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China
| | - XiaoYing Lv
- Fundamental Research Center, Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - MengWen Huang
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China
| | - XingChao Pan
- State Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - YaJuan Zheng
- State Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - MengYa Yuan
- State Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - GaoXiang Ge
- State Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China
| | - Yi Arial Zeng
- State Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China
| | - ChangDong Lin
- Fundamental Research Center, Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), School of Life Sciences and Technology, Tongji University, Shanghai, China.
- Frontier Science Center for Stem Cell Research, Tongji University, Shanghai, China.
| | - JianFeng Chen
- State Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China.
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China.
| |
Collapse
|
49
|
García-Chamé M, Wadhwani P, Pfeifer J, Schepers U, Niemeyer CM, Domínguez CM. A Versatile Microfluidic Platform for Extravasation Studies Based on DNA Origami-Cell Interactions. Angew Chem Int Ed Engl 2024; 63:e202318805. [PMID: 38687094 DOI: 10.1002/anie.202318805] [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: 12/07/2023] [Revised: 04/12/2024] [Accepted: 04/24/2024] [Indexed: 05/02/2024]
Abstract
The adhesion of circulating tumor cells (CTCs) to the endothelial lumen and their extravasation to surrounding tissues are crucial in the seeding of metastases and remain the most complex events of the metastatic cascade to study. Integrins expressed on CTCs are major regulators of the extravasation process. This knowledge is primarily derived from animal models and biomimetic systems based on artificial endothelial layers, but these methods have ethical or technical limitations. We present a versatile microfluidic device to study cancer cell extravasation that mimics the endothelial barrier by using a porous membrane functionalized with DNA origami nanostructures (DONs) that display nanoscale patterns of adhesion peptides to circulating cancer cells. The device simulates physiological flow conditions and allows direct visualization of cell transmigration through microchannel pores using 3D confocal imaging. Using this system, we studied integrin-specific adhesion in the absence of other adhesive events. Specifically, we show that the transmigration ability of the metastatic cancer cell line MDA-MB-231 is influenced by the type, distance, and density of adhesion peptides present on the DONs. Furthermore, studies with mixed ligand systems indicate that integrins binding to RGD (arginine-glycine-aspartic acid) and IDS (isoleucine-aspartic acid-serine) did not synergistically enhance the extravasation process of MDA-MB-231 cells.
Collapse
Affiliation(s)
- Miguel García-Chamé
- Karlsruhe Institute of Technology (KIT), Institute for Biological Interfaces 1 (IBG 1), Hermann-von-Helmholtz-Platz, 76344, Eggenstein-Leopoldshafen, Germany
| | - Parvesh Wadhwani
- Karlsruhe Institute of Technology (KIT), Institute for Biological Interfaces 2 (IBG 2), Hermann-von-Helmholtz-Platz, 76344, Eggenstein-Leopoldshafen, Germany
| | - Juliana Pfeifer
- Karlsruhe Institute of Technology (KIT), Institute of Functional Interfaces (IFG), Hermann-von-Helmholtz-Platz, 76344, Eggenstein-Leopoldshafen, Germany
| | - Ute Schepers
- Karlsruhe Institute of Technology (KIT), Institute of Functional Interfaces (IFG), Hermann-von-Helmholtz-Platz, 76344, Eggenstein-Leopoldshafen, Germany
| | - Christof M Niemeyer
- Karlsruhe Institute of Technology (KIT), Institute for Biological Interfaces 1 (IBG 1), Hermann-von-Helmholtz-Platz, 76344, Eggenstein-Leopoldshafen, Germany
| | - Carmen M Domínguez
- Karlsruhe Institute of Technology (KIT), Institute for Biological Interfaces 1 (IBG 1), Hermann-von-Helmholtz-Platz, 76344, Eggenstein-Leopoldshafen, Germany
| |
Collapse
|
50
|
Zhang W, Hou Y, Yin S, Miao Q, Lee K, Zhou X, Wang Y. Advanced gene nanocarriers/scaffolds in nonviral-mediated delivery system for tissue regeneration and repair. J Nanobiotechnology 2024; 22:376. [PMID: 38926780 PMCID: PMC11200991 DOI: 10.1186/s12951-024-02580-8] [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: 03/09/2024] [Accepted: 05/24/2024] [Indexed: 06/28/2024] Open
Abstract
Tissue regeneration technology has been rapidly developed and widely applied in tissue engineering and repair. Compared with traditional approaches like surgical treatment, the rising gene therapy is able to have a durable effect on tissue regeneration, such as impaired bone regeneration, articular cartilage repair and cancer-resected tissue repair. Gene therapy can also facilitate the production of in situ therapeutic factors, thus minimizing the diffusion or loss of gene complexes and enabling spatiotemporally controlled release of gene products for tissue regeneration. Among different gene delivery vectors and supportive gene-activated matrices, advanced gene/drug nanocarriers attract exceptional attraction due to their tunable physiochemical properties, as well as excellent adaptive performance in gene therapy for tissue regeneration, such as bone, cartilage, blood vessel, nerve and cancer-resected tissue repair. This paper reviews the recent advances on nonviral-mediated gene delivery systems with an emphasis on the important role of advanced nanocarriers in gene therapy and tissue regeneration.
Collapse
Affiliation(s)
- Wanheng Zhang
- Institute of Geriatrics, School of Medicine, Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), Shanghai University, Shanghai, 200444, China
- Department of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Yan Hou
- Institute of Geriatrics, School of Medicine, Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), Shanghai University, Shanghai, 200444, China
- Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education), Shanghai University, Shanghai, 200444, China
| | - Shiyi Yin
- Department of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Qi Miao
- Department of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Kyubae Lee
- Department of Biomedical Materials, Konyang University, Daejeon, 35365, Republic of Korea
| | - Xiaojian Zhou
- Department of Pediatrics, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200080, China.
| | - Yongtao Wang
- Institute of Geriatrics, School of Medicine, Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), Shanghai University, Shanghai, 200444, China.
- Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education), Shanghai University, Shanghai, 200444, China.
| |
Collapse
|