1
|
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
|
2
|
Chen J, Liu S, Ruan Z, Wang K, Xi X, Mao J. Thrombotic events associated with immune checkpoint inhibitors and novel antithrombotic strategies to mitigate bleeding risk. Blood Rev 2024; 67:101220. [PMID: 38876840 DOI: 10.1016/j.blre.2024.101220] [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: 04/26/2024] [Revised: 05/23/2024] [Accepted: 06/05/2024] [Indexed: 06/16/2024]
Abstract
Although immunotherapy is expanding treatment options for cancer patients, the prognosis of advanced cancer remains poor, and these patients must contend with both cancers and cancer-related thrombotic events. In particular, immune checkpoint inhibitors are associated with an increased risk of atherosclerotic thrombotic events. Given the fundamental role of platelets in atherothrombosis, co-administration of antiplatelet agents is always indicated. Platelets are also involved in all steps of cancer progression. Classical antithrombotic drugs can cause inevitable hemorrhagic side effects due to blocking integrin β3 bidirectional signaling, which regulates simultaneously thrombosis and hemostasis. Meanwhile, many promising new targets are emerging with minimal bleeding risk and desirable anti-tumor effects. This review will focus on the issue of thrombosis during immune checkpoint inhibitor treatment and the role of platelet activation in cancer progression as well as explore the mechanisms by which novel antiplatelet therapies may exert both antithrombotic and antitumor effects without excessive bleeding risk.
Collapse
Affiliation(s)
- Jiayi Chen
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, Collaborative Innovation Center of Hematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Shuang Liu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, Collaborative Innovation Center of Hematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Zheng Ruan
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, Collaborative Innovation Center of Hematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Kankan Wang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Sino-French Research Center for Life Sciences and Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Xiaodong Xi
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, Collaborative Innovation Center of Hematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Jianhua Mao
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, Collaborative Innovation Center of Hematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| |
Collapse
|
3
|
Gupta S, Dalpati N, Rai SK, Sehrawat A, Pai V, Sarangi PP. A synthetic bioactive peptide of the C-terminal fragment of adhesion protein Fibulin7 attenuates the inflammatory functions of innate immune cells in LPS-induced systemic inflammation. Inflamm Res 2024; 73:1333-1348. [PMID: 38836870 DOI: 10.1007/s00011-024-01903-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 05/21/2024] [Accepted: 05/30/2024] [Indexed: 06/06/2024] Open
Abstract
OBJECTIVE Systemic inflammation is associated with improper localization of hyperactive neutrophils and monocytes in visceral organs. Previously, a C-terminal fragment of adhesion protein Fibulin7 (Fbln7-C) was shown to regulate innate immune functionality during inflammation. Recently, a shorter bioactive peptide of Fbln7-C, FC-10, via integrin binding was shown to reduce ocular angiogenesis. However, the role of FC-10 in regulating the neutrophils and monocyte functionality during systemic inflammatory conditions is unknown. The study sought to explore the role of FC-10 peptide on the functionality of innate immune cells during inflammation and endotoxemic mice. METHODS Neutrophils and monocytes were isolated from healthy donors and septic patient clinical samples and Cell adhesion assay was performed using a UV spectrophotometer. Gene expression studies were performed using qPCR. Protein level expression was measured using ELISA and flow cytometry. ROS assay, and activation markers analysis in vitro, and in vivo were done using flow cytometry. TREATMENT Cells were stimulated with LPS (100 ng/mL) and studied in the presence of peptides (10 μg, and 20 μg/mL) in vitro. In an in vivo study, mice were administered with LPS (36.8 mg/kg bw) and peptide (20 μg). RESULTS This study demonstrates that human neutrophils and monocytes adhere to FC-10 via integrin β1, inhibit spreading, ROS, surface activation markers (CD44, CD69), phosphorylated Src kinase, pro-inflammatory genes, and protein expression, compared to scrambled peptide in cells isolated from healthy donors and clinical sample. In line with the in vitro data, FC-10 (20 μg) administration significantly decreases innate cell infiltration at inflammatory sites, improves survival in endotoxemia animals & reduces the inflammatory properties of neutrophils and monocytes isolated from septic patients. CONCLUSION FC-10 peptide can regulate neutrophils and monocyte functions and has potential to be used as an immunomodulatory therapeutic in inflammatory diseases.
Collapse
Affiliation(s)
- Saloni Gupta
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, 247667, India
| | - Nibedita Dalpati
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, 247667, India
| | - Shubham Kumar Rai
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, 247667, India
| | - Amit Sehrawat
- All India Institute of Medical Sciences Rishikesh, Rishikesh, Uttarakhand, India
| | - Venkatesh Pai
- All India Institute of Medical Sciences Rishikesh, Rishikesh, Uttarakhand, India
| | - Pranita P Sarangi
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, 247667, India.
| |
Collapse
|
4
|
Sidibé A, Mykuliak VV, Zhang P, Hytönen VP, Wu J, Wehrle-Haller B. Acetyl-NPKY of integrin-β1 binds KINDLIN2 to control endothelial cell proliferation and junctional integrity. iScience 2024; 27:110129. [PMID: 38904068 PMCID: PMC11187247 DOI: 10.1016/j.isci.2024.110129] [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: 08/08/2023] [Revised: 02/09/2024] [Accepted: 05/24/2024] [Indexed: 06/22/2024] Open
Abstract
Integrin-dependent crosstalk between cell-matrix adhesions and cell-cell junctions is critical for controlling endothelial permeability and proliferation in cancer and inflammatory diseases but remains poorly understood. Here, we investigated how acetylation of the distal NPKY-motif of Integrin-β1 influences endothelial cell physiology and barrier function. Expression of an acetylation-mimetic β1-K794Q-GFP mutant led to the accumulation of immature cell-matrix adhesions accompanied by a transcriptomic reprograming of endothelial cells, involving genes associated with cell adhesion, proliferation, polarity, and barrier function. β1-K794Q-GFP induced constitutive MAPK signaling, junctional impairment, proliferation, and reduced contact inhibition at confluence. Structural analysis of Integrin-β1 interaction with KINDLIN2, biochemical pulldown assay, and binding energy determination by using molecular dynamics simulation showed that acetylation of K794 and the K794Q-mutant increased KINDLIN2 binding affinity to the Integrin-β1. Thus, enhanced recruitment of KINDLIN2 to Lysine-acetylated Integrin-β1 and resulting modulation of barrier function, offers new therapeutic possibilities for controlling vascular permeability and disease conditions.
Collapse
Affiliation(s)
- Adama Sidibé
- Department of Cell Physiology and Metabolism, University of Geneva, Centre Médical Universitaire, Rue Michel-Servet 1, CH-1211 Geneva, Switzerland
| | - Vasyl V. Mykuliak
- Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, FI-33520 Tampere, Finland
| | - Pingfeng Zhang
- Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, FI-33520 Tampere, Finland
| | - Vesa P. Hytönen
- Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, FI-33520 Tampere, Finland
- Fimlab Laboratories, Biokatu 4, FI-33520 Tampere, Finland
| | - Jinhua Wu
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA 19111, USA
| | - Bernhard Wehrle-Haller
- Department of Cell Physiology and Metabolism, University of Geneva, Centre Médical Universitaire, Rue Michel-Servet 1, CH-1211 Geneva, Switzerland
| |
Collapse
|
5
|
Jang I, Menon S, Indra I, Basith R, Beningo KA. Calpain Small Subunit Mediated Secretion of Galectin-3 Regulates Traction Stress. Biomedicines 2024; 12:1247. [PMID: 38927454 PMCID: PMC11200796 DOI: 10.3390/biomedicines12061247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 05/29/2024] [Accepted: 05/30/2024] [Indexed: 06/28/2024] Open
Abstract
The complex regulation of traction forces (TF) produced during cellular migration remains poorly understood. We have previously found that calpain 4 (Capn4), the small non-catalytic subunit of the calpain 1 and 2 proteases, regulates the production of TF independent of the proteolytic activity of the larger subunits. Capn4 was later found to facilitate tyrosine phosphorylation and secretion of the lectin-binding protein galectin-3 (Gal3). In this study, recombinant Gal3 (rGal3) was added to the media-enhanced TF generated by capn4-/- mouse embryonic fibroblasts (MEFs). Extracellular Gal3 also rescued defects in the distribution, morphology, and adhesive strength of focal adhesions present in capn4-/- MEF cells. Surprisingly, extracellular Gal3 does not influence mechanosensing. c-Abl kinase was found to affect Gal3 secretion and the production of TF through phosphorylation of Y107 on Gal3. Our study also suggests that Gal3-mediated regulation of TF occurs through signaling pathways triggered by β1 integrin but not by focal adhesion kinase (FAK) Y397 autophosphorylation. Our findings provide insights into the signaling mechanism by which Capn4 and secreted Gal3 regulate cell migration through the modulation of TF distinctly independent from a mechanosensing mechanism.
Collapse
Affiliation(s)
| | | | | | | | - Karen A. Beningo
- Department of Biological Sciences, Wayne State University, Detroit, MI 48202, USA; (I.J.)
| |
Collapse
|
6
|
Najafi S, Asemani Y, Majidpoor J, Mahmoudi R, Aghaei-Zarch SM, Mortezaee K. Tumor-educated platelets. Clin Chim Acta 2024; 552:117690. [PMID: 38056548 DOI: 10.1016/j.cca.2023.117690] [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: 10/15/2023] [Revised: 11/29/2023] [Accepted: 12/02/2023] [Indexed: 12/08/2023]
Abstract
Beyond traditional roles in homeostasis and coagulation, growing evidence suggests that platelets also reflect malignant transformation in cancer. Platelets are present in the tumor microenvironment where they interact with cancer cells. This interaction results in direct and indirect "education" as evident by platelet alterations in adhesion molecules, glycoproteins, nucleic acids, proteins and various receptors. Subsequently, these tumor-educated platelets (TEPs) circulate throughout the body and play pivotal roles in promotion of tumor growth and dissemination. Accordingly, platelet status can be considered a unique blood-based biomarker that can potentially predict prognosis and therapeutic success. Recently, liquid biopsies including TEPs have received much attention as safe, minimally invasive and sensitive alternatives for patient management. Herein, we provide an overview of TEPs and explore their benefits and limitations in cancer.
Collapse
Affiliation(s)
- Sajad Najafi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Yahya Asemani
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Jamal Majidpoor
- Department of Anatomy, School of Medicine, Infectious Diseases Research Center, Gonabad University of Medical Sciences, Gonabad, Iran
| | - Reza Mahmoudi
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Mohsen Aghaei-Zarch
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Keywan Mortezaee
- Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran.
| |
Collapse
|
7
|
Tan H, Ma L, Qin T, Liu K, Liu Y, Wen C, You K, Pang C, Luo H, Wei L, Shu Y, Yang X, Shen X, Zhou C. Myo6 mediates osteoclast function and is essential for joint damage in collagen-induced arthritis. Biochim Biophys Acta Mol Basis Dis 2024; 1870:166902. [PMID: 37816396 DOI: 10.1016/j.bbadis.2023.166902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 09/19/2023] [Accepted: 09/26/2023] [Indexed: 10/12/2023]
Abstract
OBJECTIVES To explore the novel function of MYO6 on Osteoclast differentiation and its joint destruction capacity in Rheumatoid arthritis mice model. METHODS We examined joint erosion in a collagen-induced arthritis (CIA) mouse model using micro-CT, with the mice having a MYO6 knockout background. Inflammatory cytokines were analyzed using an enzyme-linked immunosorbent assay (ELISA). In vitro, we investigated the osteoclastogenesis ability of bone marrow-derived macrophages isolated from MYO6-/- mice and their littermate controls, examining both morphological and functional differences. Furthermore, we explored podosome formation and endosome maturation using immunofluorescence staining. RESULTS We found that MYO6 deficiency attenuated arthritis development and bone destruction in CIA mice as well as impaired osteoclast differentiation by inhibiting NFATc1 induction. Our findings indicate that MYO6 is essential for the organization of podosomes by modulating the FAK/AKT and integrin-β3/Src pathways. MYO6 also mediates endosome transportation by regulating the expression of Rab5 and GM130. This may impact the maintenance and functionality of the ruffled border, as well as the regulation of autophagy in osteoclasts. CONCLUSION Our results demonstrated a critical function of MYO6 in osteoclast differentiation and its potential relevance in experimental arthritis.
Collapse
Affiliation(s)
- Huijing Tan
- SMU-KI United Medical Inflammatory Center, School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of Shock and Microcirculation, Southern Medical University, Guangzhou 510515, China
| | - Liqing Ma
- SMU-KI United Medical Inflammatory Center, School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of Shock and Microcirculation, Southern Medical University, Guangzhou 510515, China
| | - Tian Qin
- SMU-KI United Medical Inflammatory Center, School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of Shock and Microcirculation, Southern Medical University, Guangzhou 510515, China
| | - Kaifei Liu
- Department of Pharmacy, Jingzhou Central Hospital, Jingzhou, Hubei 434020, China
| | - Ying Liu
- School of Pharmacy, Guangzhou Xinhua University, Guangzhou 510520, China
| | - Cailing Wen
- SMU-KI United Medical Inflammatory Center, School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of Shock and Microcirculation, Southern Medical University, Guangzhou 510515, China
| | - Keyuan You
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Caixia Pang
- SMU-KI United Medical Inflammatory Center, School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of Shock and Microcirculation, Southern Medical University, Guangzhou 510515, China
| | - Hui Luo
- SMU-KI United Medical Inflammatory Center, School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of Shock and Microcirculation, Southern Medical University, Guangzhou 510515, China
| | - Linlin Wei
- SMU-KI United Medical Inflammatory Center, School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of Shock and Microcirculation, Southern Medical University, Guangzhou 510515, China
| | - Yue Shu
- SMU-KI United Medical Inflammatory Center, School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of Shock and Microcirculation, Southern Medical University, Guangzhou 510515, China
| | - Xinru Yang
- SMU-KI United Medical Inflammatory Center, School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of Shock and Microcirculation, Southern Medical University, Guangzhou 510515, China
| | - Xiaoyan Shen
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Chun Zhou
- SMU-KI United Medical Inflammatory Center, School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of Shock and Microcirculation, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
8
|
Liu K, Hao Z, Zheng H, Wang H, Zhang L, Yan M, Tuerhong R, Zhou Y, Wang Y, Pang T, Shi L. Repurposing of rilpivirine for preventing platelet β3 integrin-dependent thrombosis by targeting c-Src active autophosphorylation. Thromb Res 2023; 229:53-68. [PMID: 37413892 DOI: 10.1016/j.thromres.2023.06.031] [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: 04/17/2023] [Revised: 06/19/2023] [Accepted: 06/30/2023] [Indexed: 07/08/2023]
Abstract
BACKGROUND HIV-infected individuals are known to be at higher risk for thrombotic cardiovascular disease (CVD), which may also be differentially affected by components of anti-HIV drugs. To identify the effects of a series of FDA-approved anti-HIV drugs on platelet aggregation in humans, focusing on the novel pharmacological effects of rilpivirine (RPV), a reverse transcriptase inhibitor, on platelet function both in vitro and in vivo and the mechanisms involved. METHODS AND RESULTS In vitro studies showed that RPV was the only anti-HIV reagent that consistently and efficiently inhibited aggregation elicited by different agonists, exocytosis, morphological extension on fibrinogen, and clot retraction. Treatment of mice with RPV significantly prevented thrombus formation in FeCl3-injured mesenteric vessels, postcava with stenosis surgery, and ADP -induced pulmonary embolism models without defects in platelet viability, tail bleeding, and coagulation activities. RPV also improved cardiac performance in mice with post-ischemic reperfusion. A mechanistic study revealed that RPV preferentially attenuated fibrinogen-stimulated Tyr773 phosphorylation of β3-integrin by inhibiting Tyr419 autophosphorylation of c-Src. Molecular docking and surface plasmon resonance analyses showed that RPV can bind directly to c-Src. Further mutational analysis showed that the Phe427 residue of c-Src is critical for RPV interaction, suggesting a novel interaction site for targeting c-Src to block β3-integrin outside-in signaling. CONCLUSION These results demonstrated that RPV was able to prevent the progression of thrombotic CVDs by interrupting β3-integrin-mediated outside-in signaling via inhibiting c-Src activation without hemorrhagic side effects, highlighting RPV as a promising reagent for the prevention and therapy of thrombotic CVDs.
Collapse
Affiliation(s)
- Kui Liu
- Xiamen Key Laboratory of Cardiovascular Disease, Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, 2999 Jinshan Road, Xiamen 361000, China; State Key Laboratory of Natural Medicines, New Drug Screening Center, Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), China Pharmaceutical University, Nanjing 210009, China
| | - Zhen Hao
- Xiamen Key Laboratory of Cardiovascular Disease, Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, 2999 Jinshan Road, Xiamen 361000, China; College of Basic Medical Sciences, Dalian Medical University, No. 9 West Section, South Lv shun Road, Dalian 116044, China
| | - Hao Zheng
- College of Basic Medical Sciences, Dalian Medical University, No. 9 West Section, South Lv shun Road, Dalian 116044, China
| | - Haojie Wang
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Luying Zhang
- College of Basic Medical Sciences, Dalian Medical University, No. 9 West Section, South Lv shun Road, Dalian 116044, China
| | - Minghui Yan
- College of Basic Medical Sciences, Dalian Medical University, No. 9 West Section, South Lv shun Road, Dalian 116044, China
| | - Reyisha Tuerhong
- College of Basic Medical Sciences, Dalian Medical University, No. 9 West Section, South Lv shun Road, Dalian 116044, China
| | - Yuling Zhou
- Xiamen Key Laboratory of Cardiovascular Disease, Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, 2999 Jinshan Road, Xiamen 361000, China
| | - Yan Wang
- Xiamen Key Laboratory of Cardiovascular Disease, Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, 2999 Jinshan Road, Xiamen 361000, China.
| | - Tao Pang
- State Key Laboratory of Natural Medicines, New Drug Screening Center, Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), China Pharmaceutical University, Nanjing 210009, China.
| | - Lei Shi
- Xiamen Key Laboratory of Cardiovascular Disease, Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, 2999 Jinshan Road, Xiamen 361000, China; College of Basic Medical Sciences, Dalian Medical University, No. 9 West Section, South Lv shun Road, Dalian 116044, China.
| |
Collapse
|
9
|
Kumar S, Stainer A, Dubrulle J, Simpkins C, Cooper JA. Cas phosphorylation regulates focal adhesion assembly. eLife 2023; 12:e90234. [PMID: 37489578 PMCID: PMC10435235 DOI: 10.7554/elife.90234] [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: 06/16/2023] [Accepted: 07/19/2023] [Indexed: 07/26/2023] Open
Abstract
Integrin-mediated cell attachment rapidly induces tyrosine kinase signaling. Despite years of research, the role of this signaling in integrin activation and focal adhesion assembly is unclear. We provide evidence that the Src-family kinase (SFK) substrate Cas (Crk-associated substrate, p130Cas, BCAR1) is phosphorylated and associated with its Crk/CrkL effectors in clusters that are precursors of focal adhesions. The initial phospho-Cas clusters contain integrin β1 in its inactive, bent closed, conformation. Later, phospho-Cas and total Cas levels decrease as integrin β1 is activated and core focal adhesion proteins including vinculin, talin, kindlin, and paxillin are recruited. Cas is required for cell spreading and focal adhesion assembly in epithelial and fibroblast cells on collagen and fibronectin. Cas cluster formation requires Cas, Crk/CrkL, SFKs, and Rac1 but not vinculin. Rac1 provides positive feedback onto Cas through reactive oxygen, opposed by negative feedback from the ubiquitin proteasome system. The results suggest a two-step model for focal adhesion assembly in which clusters of phospho-Cas, effectors and inactive integrin β1 grow through positive feedback prior to integrin activation and recruitment of core focal adhesion proteins.
Collapse
Affiliation(s)
- Saurav Kumar
- Fred Hutchinson Cancer CenterSeattleUnited States
| | | | | | | | | |
Collapse
|
10
|
Kaiser R, Anjum A, Kammerer L, Loew Q, Akhalkatsi A, Rossaro D, Escaig R, Droste zu Senden A, Raude B, Lorenz M, Gold C, Pekayvaz K, Brocker T, Kranich J, Holch JW, Spiekermann K, Massberg S, Gaertner F, Nicolai L. Mechanosensing via a GpIIb/Src/14-3-3ζ axis critically regulates platelet migration in vascular inflammation. Blood 2023; 141:2973-2992. [PMID: 37018659 PMCID: PMC10646815 DOI: 10.1182/blood.2022019210] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 02/17/2023] [Accepted: 03/05/2023] [Indexed: 04/07/2023] Open
Abstract
Platelets are not only the first responders in thrombosis and hemostasis but also central players in inflammation. Compared with platelets recruited to thrombi, immune-responsive platelets use distinct effector functions including actin-related protein complex 2/3-dependent migration along adhesive substrate gradients (haptotaxis), which prevents inflammatory bleeding and contributes to host defense. How platelet migration in this context is regulated on a cellular level is incompletely understood. Here, we use time-resolved morphodynamic profiling of individual platelets to show that migration, in contrast to clot retraction, requires anisotropic myosin IIa-activity at the platelet rear which is preceded by polarized actin polymerization at the front to initiate and maintain migration. Integrin GPIIb-dependent outside-in signaling via Gα13 coordinates polarization of migrating platelets to trigger tyrosine kinase c-Src/14-3-3ζ-dependent lamellipodium formation and functions independent of soluble agonists or chemotactic signals. Inhibitors of this signaling cascade, including the clinically used ABL/c-Src inhibitor dasatinib, interfere predominantly with the migratory capacity of platelets, without major impairment of classical platelet functions. In murine inflammation models, this translates to reduced migration of platelets visualized by 4D intravital microscopy, resulting in increased inflammation-associated hemorrhage in acute lung injury. Finally, platelets isolated from patients with leukemia treated with dasatinib who are prone to clinically relevant hemorrhage exhibit prominent migration defects, whereas other platelet functions are only partially affected. In summary, we define a distinct signaling pathway essential for migration and provide novel mechanistic insights explaining dasatinib-related platelet dysfunction and bleeding.
Collapse
Affiliation(s)
- Rainer Kaiser
- Medizinische Klinik und Poliklinik I, University Hospital Ludwig-Maximilian University, Munich, Germany
- German Centre for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
| | - Afra Anjum
- Medizinische Klinik und Poliklinik I, University Hospital Ludwig-Maximilian University, Munich, Germany
- German Centre for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
| | - Lisa Kammerer
- Medizinische Klinik und Poliklinik I, University Hospital Ludwig-Maximilian University, Munich, Germany
- German Centre for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
| | - Quentin Loew
- Medizinische Klinik und Poliklinik I, University Hospital Ludwig-Maximilian University, Munich, Germany
- German Centre for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
| | - Anastassia Akhalkatsi
- Medizinische Klinik und Poliklinik I, University Hospital Ludwig-Maximilian University, Munich, Germany
- German Centre for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
| | - Dario Rossaro
- Medizinische Klinik und Poliklinik I, University Hospital Ludwig-Maximilian University, Munich, Germany
- German Centre for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
| | - Raphael Escaig
- Medizinische Klinik und Poliklinik I, University Hospital Ludwig-Maximilian University, Munich, Germany
- German Centre for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
| | - Augustin Droste zu Senden
- Medizinische Klinik und Poliklinik I, University Hospital Ludwig-Maximilian University, Munich, Germany
- German Centre for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
| | - Ben Raude
- Department of Vascular Surgery, Charité–Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Michael Lorenz
- Medizinische Klinik und Poliklinik I, University Hospital Ludwig-Maximilian University, Munich, Germany
| | - Christoph Gold
- Medizinische Klinik und Poliklinik I, University Hospital Ludwig-Maximilian University, Munich, Germany
- German Centre for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
| | - Kami Pekayvaz
- Medizinische Klinik und Poliklinik I, University Hospital Ludwig-Maximilian University, Munich, Germany
- German Centre for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
| | - Thomas Brocker
- Institute for Immunology, Biomedical Center, Medical Faculty, Ludwig-Maximilian University Munich, Munich, Germany
| | - Jan Kranich
- Institute for Immunology, Biomedical Center, Medical Faculty, Ludwig-Maximilian University Munich, Munich, Germany
| | - Julian Walter Holch
- Department of Medicine III, University Hospital, Ludwig-Maximilian University Munich, Munich, Germany
- Comprehensive Cancer Center, University Hospital, Ludwig-Maximilian University Munich, Munich, Germany
- German Cancer Consortium, Partner Site Munich and German Cancer Research Centre, Heidelberg, Germany
| | - Karsten Spiekermann
- Department of Medicine III, University Hospital, Ludwig-Maximilian University Munich, Munich, Germany
- Comprehensive Cancer Center, University Hospital, Ludwig-Maximilian University Munich, Munich, Germany
- German Cancer Consortium, Partner Site Munich and German Cancer Research Centre, Heidelberg, Germany
| | - Steffen Massberg
- Medizinische Klinik und Poliklinik I, University Hospital Ludwig-Maximilian University, Munich, Germany
- German Centre for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
| | - Florian Gaertner
- Medizinische Klinik und Poliklinik I, University Hospital Ludwig-Maximilian University, Munich, Germany
- German Centre for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
| | - Leo Nicolai
- Medizinische Klinik und Poliklinik I, University Hospital Ludwig-Maximilian University, Munich, Germany
- German Centre for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
| |
Collapse
|
11
|
Ha BH, Yigit S, Natarajan N, Morse EM, Calderwood DA, Boggon TJ. Molecular basis for integrin adhesion receptor binding to p21-activated kinase 4 (PAK4). Commun Biol 2022; 5:1257. [PMID: 36385162 PMCID: PMC9669019 DOI: 10.1038/s42003-022-04157-3] [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: 10/20/2021] [Accepted: 10/24/2022] [Indexed: 11/18/2022] Open
Abstract
Integrin adhesion receptors provide links between extracellular ligands and cytoplasmic signaling. Multiple kinases have been found to directly engage with integrin β tails, but the molecular basis for these interactions remain unknown. Here, we assess the interaction between the kinase domain of p21-activated kinase 4 (PAK4) and the cytoplasmic tail of integrin β5. We determine three crystal structures of PAK4-β5 integrin complexes and identify the PAK-binding site. This is a region in the membrane-proximal half of the β5 tail and confirmed by site-directed mutagenesis. The β5 tail engages the kinase substrate-binding groove and positions the non-phosphorylatable integrin residue Glu767 at the phosphoacceptor site. Consistent with this, integrin β5 is poorly phosphorylated by PAK4, and in keeping with its ability to occlude the substrate-binding site, weakly inhibits kinase activity. These findings demonstrate the molecular basis for β5 integrin-PAK4 interactions but suggest modifications in understanding the potential cellular role of this interaction.
Collapse
Affiliation(s)
- Byung Hak Ha
- The Department of Pharmacology, Yale University, 333 Cedar St., New Haven, CT, 06520, USA
| | - Sezin Yigit
- The Department of Pharmacology, Yale University, 333 Cedar St., New Haven, CT, 06520, USA
| | - Nalini Natarajan
- The Department of Pharmacology, Yale University, 333 Cedar St., New Haven, CT, 06520, USA
| | - Elizabeth M Morse
- The Department of Cell Biology, Yale University, 333 Cedar St., New Haven, CT, 06520, USA
| | - David A Calderwood
- The Department of Pharmacology, Yale University, 333 Cedar St., New Haven, CT, 06520, USA.
- The Department of Cell Biology, Yale University, 333 Cedar St., New Haven, CT, 06520, USA.
| | - Titus J Boggon
- The Department of Pharmacology, Yale University, 333 Cedar St., New Haven, CT, 06520, USA.
- The Department of Molecular Biophysics and Biochemistry, Yale University, 333 Cedar St., New Haven, CT, 06520, USA.
| |
Collapse
|
12
|
Promalignant effects of antiangiogenics in the tumor microenvironment. Semin Cancer Biol 2022; 86:199-206. [PMID: 35248730 DOI: 10.1016/j.semcancer.2022.03.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 03/02/2022] [Accepted: 03/02/2022] [Indexed: 02/07/2023]
Abstract
Antiangiogenic therapies are considered a promising strategy against solid tumors. Their aim is to inhibit the formation of new blood vasculature, thereby reducing the oxygen and nutrient supply to prevent further tumor growth and spreading. However, the strategy has seen limitations, as survival benefits are modest and often accompanied with increased tumor aggressiveness in form of invasion and metastasis. Antiangiogenic induced changes in the tumor microenvironment, such as hypoxia, mechanical stress or extracellular acidification can activate different receptors of tumoral and stromal cells and induce an extensive remodeling of the entire tumor microenvironment, with the overall goal to invade nearby tissues and regain access to the vasculature. In this regard, receptor tyrosine kinases have been studied intensively and especially the inhibition of c-Met has given promising results, characterized by a reduction in invasiveness and prolonged survival. Receptors that sense changes in the extracellular matrix like integrins or proteoglycans can also induce downstream signaling that stimulates the expression of remodeling factors such as new matrix components, enzymes or chemoattractants. Targeting multiple receptors and sensors of cancer cells simultaneously might represent an effective second line treatment that prevents the formation of malignant side effects.
Collapse
|
13
|
Hedna R, Kovacic H, Pagano A, Peyrot V, Robin M, Devred F, Breuzard G. Tau Protein as Therapeutic Target for Cancer? Focus on Glioblastoma. Cancers (Basel) 2022; 14:5386. [PMID: 36358803 PMCID: PMC9653627 DOI: 10.3390/cancers14215386] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 10/28/2022] [Accepted: 10/28/2022] [Indexed: 08/27/2023] Open
Abstract
Despite being extensively studied for several decades, the microtubule-associated protein Tau has not finished revealing its secrets. For long, Tau has been known for its ability to promote microtubule assembly. A less known feature of Tau is its capability to bind to cancer-related protein kinases, suggesting a possible role of Tau in modulating microtubule-independent cellular pathways that are associated with oncogenesis. With the intention of finding new therapeutic targets for cancer, it appears essential to examine the interaction of Tau with these kinases and their consequences. This review aims at collecting the literature data supporting the relationship between Tau and cancer with a particular focus on glioblastoma tumors in which the pathological significance of Tau remains largely unexplored. We will first treat this subject from a mechanistic point of view showing the pivotal role of Tau in oncogenic processes. Then, we will discuss the involvement of Tau in dysregulating critical pathways in glioblastoma. Finally, we will outline promising strategies to target Tau protein for the therapy of glioblastoma.
Collapse
Affiliation(s)
- Rayane Hedna
- Faculté des Sciences Médicales et Paramédicales, Institut de Neurophysiopathologie (INP), UMR 7051, CNRS, Aix Marseille Université, 13005 Marseille, France
| | - Hervé Kovacic
- Faculté des Sciences Médicales et Paramédicales, Institut de Neurophysiopathologie (INP), UMR 7051, CNRS, Aix Marseille Université, 13005 Marseille, France
| | - Alessandra Pagano
- Faculté des Sciences Médicales et Paramédicales, Institut de Neurophysiopathologie (INP), UMR 7051, CNRS, Aix Marseille Université, 13005 Marseille, France
| | - Vincent Peyrot
- Faculté des Sciences Médicales et Paramédicales, Institut de Neurophysiopathologie (INP), UMR 7051, CNRS, Aix Marseille Université, 13005 Marseille, France
| | - Maxime Robin
- Faculté de Pharmacie, Institut Méditerranéen de Biodiversité et Ecologie marine et continentale (IMBE), UMR 7263, CNRS, IRD 237, Aix-Marseille Université, 13005 Marseille, France
| | - François Devred
- Faculté des Sciences Médicales et Paramédicales, Institut de Neurophysiopathologie (INP), UMR 7051, CNRS, Aix Marseille Université, 13005 Marseille, France
| | - Gilles Breuzard
- Faculté des Sciences Médicales et Paramédicales, Institut de Neurophysiopathologie (INP), UMR 7051, CNRS, Aix Marseille Université, 13005 Marseille, France
| |
Collapse
|
14
|
TNS1: Emerging Insights into Its Domain Function, Biological Roles, and Tumors. BIOLOGY 2022; 11:biology11111571. [PMID: 36358270 PMCID: PMC9687257 DOI: 10.3390/biology11111571] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 10/24/2022] [Accepted: 10/25/2022] [Indexed: 01/25/2023]
Abstract
Tensins are a family of cellular-adhesion constituents that have been extensively studied. They have instrumental roles in the pathogenesis of numerous diseases. The mammalian tensin family comprises four members: tensin1 (TNS1), tensin2, tensin3, and tensin4. Among them, TNS1 has recently received attention from researchers because of its structural properties. TNS1 engages in various biological processes, such as cell adhesion, polarization, migration, invasion, proliferation, apoptosis, and mechano-transduction, by interacting with various partner proteins. Moreover, the abnormal expression of TNS1 in vivo is associated with the development of various diseases, especially tumors. Interestingly, the role of TNS1 in different tumors is still controversial. Here, we systematically summarize three aspects of TNS1: the gene structure, the biological processes underlying its action, and the dual regulatory role of TNS1 in different tumors through different mechanisms, of which we provide the first overview.
Collapse
|
15
|
Tamari S, Menju T, Toyazaki T, Miyamoto H, Chiba N, Noguchi M, Ishikawa H, Miyata R, Kayawake H, Tanaka S, Yamada Y, Yutaka Y, Nakajima D, Ohsumi A, Hamaji M, Date H. Nrf2/p‑Fyn/ABCB1 axis accompanied by p‑Fyn nuclear accumulation plays pivotal roles in vinorelbine resistance in non‑small cell lung cancer. Oncol Rep 2022; 48:171. [PMID: 35959810 DOI: 10.3892/or.2022.8386] [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: 04/12/2022] [Accepted: 07/19/2022] [Indexed: 11/05/2022] Open
Abstract
Adjuvant cisplatin‑vinorelbine is a standard therapy for stage II/III lung cancer. However, a poor survival rate of patients with lung cancer is attributed to vinorelbine resistance arising from ATP‑binding cassette (ABC) sub‑family B member 1 (ABCB1) and phosphorylated Fyn (p‑Fyn) overexpression. However, the underlying mechanisms remain unclear. NF‑E2‑related factor 2 (Nrf2) regulates the ABC family and activates the nuclear transport of Fyn. The present study evaluated the roles of the Nrf2/p‑Fyn/ABCB1 axis in vinorelbine‑resistant (VR) cells and clinical samples. To establish VR cells, H1299 cells were exposed to vinorelbine, and the intracellular reactive oxygen species (ROS) level in the H1299 cells was determined using a DCFH‑DA assay. The total and subcellular expression of Nrf2, ABCB1 and p‑Fyn in VR cells was evaluated. Immunofluorescence was used to detect the subcellular localization of p‑Fyn in VR cells. A cell viability assay was used to examine whether the sensitivity of VR cells to vinorelbine is dependent on Nrf2 activity. Immunohistochemistry was performed on 104 tissue samples from patients with lung cancer who underwent surgery followed by cisplatin‑vinorelbine treatment. The results revealed that persistent exposure to vinorelbine induced intracellular ROS formation in H1299 cells. p‑Fyn was localized in the nucleus, and ABCB1 and Nrf2 were overexpressed in VR cells. ABCB1 expression was dependent on Nrf2 downstream activation. The decreased expression of Nrf2 restored the sensitivity of VR cells to vinorelbine. In the surgical samples, Nrf2 and ABCB1 were associated with disease‑free survival, and p‑Fyn was associated with overall survival (P<0.05). On the whole, the present study demonstrates that Nrf2 upregulates ABCB1 and, accompanied by the nuclear accumulation of p‑Fyn, induces vinorelbine resistance. These findings may facilitate the development of drug resistance prevention strategies or new drug targets against non‑small cell lung cancer.
Collapse
Affiliation(s)
- Shigeyuki Tamari
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, Kyoto 606‑8507, Japan
| | - Toshi Menju
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, Kyoto 606‑8507, Japan
| | - Toshiya Toyazaki
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, Kyoto 606‑8507, Japan
| | - Hideaki Miyamoto
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, Kyoto 606‑8507, Japan
| | - Naohisa Chiba
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, Kyoto 606‑8507, Japan
| | - Misa Noguchi
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, Kyoto 606‑8507, Japan
| | - Hiroaki Ishikawa
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, Kyoto 606‑8507, Japan
| | - Ryo Miyata
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, Kyoto 606‑8507, Japan
| | - Hidenao Kayawake
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, Kyoto 606‑8507, Japan
| | - Satona Tanaka
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, Kyoto 606‑8507, Japan
| | - Yoshito Yamada
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, Kyoto 606‑8507, Japan
| | - Yojiro Yutaka
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, Kyoto 606‑8507, Japan
| | - Daisuke Nakajima
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, Kyoto 606‑8507, Japan
| | - Akihiro Ohsumi
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, Kyoto 606‑8507, Japan
| | - Masatsugu Hamaji
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, Kyoto 606‑8507, Japan
| | - Hiroshi Date
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, Kyoto 606‑8507, Japan
| |
Collapse
|
16
|
Torres-Gomez A, Fiyouzi T, Guerra-Espinosa C, Cardeñes B, Clares I, Toribio V, Reche PA, Cabañas C, Lafuente EM. Expression of the phagocytic receptors αMβ2 and αXβ2 is controlled by RIAM, VASP and Vinculin in neutrophil-differentiated HL-60 cells. Front Immunol 2022; 13:951280. [PMID: 36238292 PMCID: PMC9552961 DOI: 10.3389/fimmu.2022.951280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 08/23/2022] [Indexed: 11/29/2022] Open
Abstract
Activation of the integrin phagocytic receptors CR3 (αMβ2, CD11b/CD18) and CR4 (αXβ2, CD11c/CD18) requires Rap1 activation and RIAM function. RIAM controls integrin activation by recruiting Talin to β2 subunits, enabling the Talin-Vinculin interaction, which in term bridges integrins to the actin-cytoskeleton. RIAM also recruits VASP to phagocytic cups and facilitates VASP phosphorylation and function promoting particle internalization. Using a CRISPR-Cas9 knockout approach, we have analyzed the requirement for RIAM, VASP and Vinculin expression in neutrophilic-HL-60 cells. All knockout cells displayed abolished phagocytosis that was accompanied by a significant and specific reduction in ITGAM (αM), ITGAX (αX) and ITGB2 (β2) mRNA, as revealed by RT-qPCR. RIAM, VASP and Vinculin KOs presented reduced cellular F-actin content that correlated with αM expression, as treatment with the actin filament polymerizing and stabilizing drug jasplakinolide, partially restored αM expression. In general, the expression of αX was less responsive to jasplakinolide treatment than αM, indicating that regulatory mechanisms independent of F-actin content may be involved. The Serum Response Factor (SRF) was investigated as the potential transcription factor controlling αMβ2 expression, since its coactivator MRTF-A requires actin polymerization to induce transcription. Immunofluorescent MRTF-A localization in parental cells was primarily nuclear, while in knockouts it exhibited a diffuse cytoplasmic pattern. Localization of FHL-2 (SRF corepressor) was mainly sub-membranous in parental HL-60 cells, but in knockouts the localization was disperse in the cytoplasm and the nucleus, suggesting RIAM, VASP and Vinculin are required to maintain FHL-2 close to cytoplasmic membranes, reducing its nuclear localization and inhibiting its corepressor activity. Finally, reexpression of VASP in the VASP knockout resulted in a complete reversion of the phenotype, as knock-ins restored αM expression. Taken together, our results suggest that RIAM, VASP and Vinculin, are necessary for the correct expression of αMβ2 and αXβ2 during neutrophilic differentiation in the human promyelocytic HL-60 cell line, and strongly point to an involvement of these proteins in the acquisition of a phagocytic phenotype.
Collapse
Affiliation(s)
- Alvaro Torres-Gomez
- Department of Immunology, Ophthalmology and Otorhinolaryngology, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (i+12), Inflammatory Diseases and Immune Disorders (Lymphocyte Immunobiology Unit), Madrid, Spain
- *Correspondence: Esther M. Lafuente, ; Alvaro Torres-Gomez,
| | - Tara Fiyouzi
- Department of Immunology, Ophthalmology and Otorhinolaryngology, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (i+12), Inflammatory Diseases and Immune Disorders (Lymphocyte Immunobiology Unit), Madrid, Spain
| | - Claudia Guerra-Espinosa
- Department of Immunology, Ophthalmology and Otorhinolaryngology, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| | - Beatriz Cardeñes
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (i+12), Inflammatory Diseases and Immune Disorders (Lymphocyte Immunobiology Unit), Madrid, Spain
| | - Irene Clares
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (i+12), Inflammatory Diseases and Immune Disorders (Lymphocyte Immunobiology Unit), Madrid, Spain
| | - Víctor Toribio
- Tissue and Organ Homeostasis Program (Cell-Cell Communication and Inflammation Unit), Centre for Molecular Biology "Severo Ochoa", Consejo Superior de Investigaciones Científicas (CSIC)-Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Pedro A. Reche
- Department of Immunology, Ophthalmology and Otorhinolaryngology, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (i+12), Inflammatory Diseases and Immune Disorders (Lymphocyte Immunobiology Unit), Madrid, Spain
| | - Carlos Cabañas
- Department of Immunology, Ophthalmology and Otorhinolaryngology, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (i+12), Inflammatory Diseases and Immune Disorders (Lymphocyte Immunobiology Unit), Madrid, Spain
- Tissue and Organ Homeostasis Program (Cell-Cell Communication and Inflammation Unit), Centre for Molecular Biology "Severo Ochoa", Consejo Superior de Investigaciones Científicas (CSIC)-Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Esther M. Lafuente
- Department of Immunology, Ophthalmology and Otorhinolaryngology, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (i+12), Inflammatory Diseases and Immune Disorders (Lymphocyte Immunobiology Unit), Madrid, Spain
- *Correspondence: Esther M. Lafuente, ; Alvaro Torres-Gomez,
| |
Collapse
|
17
|
Song G, Meng F, Luo B. The β
8
integrin EGF domains support a constitutive extended conformation, and the cytoplasmic domain impairs outside‐in signaling. J Cell Physiol 2022; 237:4251-4261. [DOI: 10.1002/jcp.30871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 07/28/2022] [Accepted: 08/27/2022] [Indexed: 11/06/2022]
Affiliation(s)
- Guannan Song
- Department of Biological Sciences Louisiana State University Baton Rouge Louisiana USA
| | - Fei Meng
- Department of Biological Sciences Louisiana State University Baton Rouge Louisiana USA
| | - Bing‐Hao Luo
- Department of Biological Sciences Louisiana State University Baton Rouge Louisiana USA
| |
Collapse
|
18
|
Zhang RM, Tiedemann K, Muthu ML, Dinesh NEH, Komarova S, Ramkhelawon B, Reinhardt DP. Fibrillin-1-regulated miR-122 has a critical role in thoracic aortic aneurysm formation. Cell Mol Life Sci 2022; 79:314. [PMID: 35606547 PMCID: PMC11072253 DOI: 10.1007/s00018-022-04337-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 04/08/2022] [Accepted: 04/28/2022] [Indexed: 11/30/2022]
Abstract
Thoracic aortic aneurysms (TAA) in Marfan syndrome, caused by fibrillin-1 mutations, are characterized by elevated cytokines and fragmentated elastic laminae in the aortic wall. This study explored whether and how specific fibrillin-1-regulated miRNAs mediate inflammatory cytokine expression and elastic laminae degradation in TAA. miRNA expression profiling at early and late TAA stages using a severe Marfan mouse model (Fbn1mgR/mgR) revealed a spectrum of differentially regulated miRNAs. Bioinformatic analyses predicted the involvement of these miRNAs in inflammatory and extracellular matrix-related pathways. We demonstrate that upregulation of pro-inflammatory cytokines and matrix metalloproteinases is a common characteristic of mouse and human TAA tissues. miR-122, the most downregulated miRNA in the aortae of 10-week-old Fbn1mgR/mgR mice, post-transcriptionally upregulated CCL2, IL-1β and MMP12. Similar data were obtained at 70 weeks of age using Fbn1C1041G/+ mice. Deficient fibrillin-1-smooth muscle cell interaction suppressed miR-122 levels. The marker for tissue hypoxia HIF-1α was upregulated in the aortic wall of Fbn1mgR/mgR mice, and miR-122 was reduced under hypoxic conditions in cell and organ cultures. Reduced miR-122 was partially rescued by HIF-1α inhibitors, digoxin and 2-methoxyestradiol in aortic smooth muscle cells. Digoxin-treated Fbn1mgR/mgR mice demonstrated elevated miR-122 and suppressed CCL2 and MMP12 levels in the ascending aortae, with reduced elastin fragmentation and aortic dilation. In summary, this study demonstrates that miR-122 in the aortic wall inhibits inflammatory responses and matrix remodeling, which is suppressed by deficient fibrillin-1-cell interaction and hypoxia in TAA.
Collapse
Affiliation(s)
- Rong-Mo Zhang
- Faculty of Medicine and Health Sciences, McGill University, Montreal, Canada
| | - Kerstin Tiedemann
- Shriners Hospital for Children-Canada, Montreal, Canada
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, Canada
| | - Muthu L Muthu
- Faculty of Medicine and Health Sciences, McGill University, Montreal, Canada
| | - Neha E H Dinesh
- Faculty of Medicine and Health Sciences, McGill University, Montreal, Canada
| | - Svetlana Komarova
- Shriners Hospital for Children-Canada, Montreal, Canada
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, Canada
| | - Bhama Ramkhelawon
- Department of Surgery, New York University School of Medicine, New York, USA
| | - Dieter P Reinhardt
- Faculty of Medicine and Health Sciences, McGill University, Montreal, Canada.
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, Canada.
- Department of Anatomy and Cell Biology, McGill University, 3640 University Street, Montreal, QC, H3A 0C7, Canada.
| |
Collapse
|
19
|
Gahmberg CG, Grönholm M, Madhavan S. Regulation of Dynamic Cell Adhesion by Integrin-Integrin Crosstalk. Cells 2022; 11:cells11101685. [PMID: 35626722 PMCID: PMC9140058 DOI: 10.3390/cells11101685] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 05/11/2022] [Accepted: 05/17/2022] [Indexed: 02/07/2023] Open
Abstract
Most cells express several integrins. The integrins are able to respond to various cellular functions and needs by modifying their own activation state, but in addition by their ability to regulate each other by activation or inhibition. This crosstalk or transdominant regulation is strictly controlled. The mechanisms resulting in integrin crosstalk are incompletely understood, but they often involve intracellular signalling routes also used by other cell surface receptors. Several studies show that the integrin cytoplasmic tails bind to a number of cytoskeletal and adaptor molecules in a regulated manner. Recent work has shown that phosphorylations of integrins and key intracellular molecules are of pivotal importance in integrin-cytoplasmic interactions, and these in turn affect integrin activity and crosstalk. The integrin β-chains play a central role in regulating crosstalk. In addition to Integrin-integrin crosstalk, crosstalk may also occur between integrins and related receptors, including other adhesion receptors, growth factor and SARS-CoV-2 receptors.
Collapse
Affiliation(s)
- Carl G. Gahmberg
- Molecular and Integrative Biosciences Research Program, Faculty of Biological and Environmental Sciences, University of Helsinki, Viikinkaari 9 C, 00014 Helsinki, Finland; (M.G.); (S.M.)
- Correspondence: ; Tel.: +358-50-539-9439
| | - Mikaela Grönholm
- Molecular and Integrative Biosciences Research Program, Faculty of Biological and Environmental Sciences, University of Helsinki, Viikinkaari 9 C, 00014 Helsinki, Finland; (M.G.); (S.M.)
- Drug Research Program, Faculty of Pharmacy, University of Helsinki, Viikinkaari 9 C, 00014 Helsinki, Finland
| | - Sudarrshan Madhavan
- Molecular and Integrative Biosciences Research Program, Faculty of Biological and Environmental Sciences, University of Helsinki, Viikinkaari 9 C, 00014 Helsinki, Finland; (M.G.); (S.M.)
| |
Collapse
|
20
|
Lin J, Dong H, Wen Y, Zhuang X, Li S. Surface Free Energy of Titanium Disks Enhances Osteoblast Activity by Affecting the Conformation of Adsorbed Fibronectin. FRONTIERS IN MATERIALS 2022; 9. [DOI: 10.3389/fmats.2022.840813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/28/2023]
Abstract
This study evaluated the influence of surface free energy (SFE) of titanium disks on the adsorption and conformation of fibronectin (FN) and the biological behavior of osteoblasts cultured on the FN-treated modified surfaces. High [H]-SFE titanium disks were irradiated by a 30 W UV light, while low (L)-SFE titanium disks received no treatment. The surface characteristics of the titanium disks were examined using scanning electron microscope, optical surface profilometer, X-ray photoelectron spectroscopy, and contact angle measurements. Adsorbed FN on different groups was investigated using attenuated total reflection-Fourier transform infrared spectroscopy. MG-63 cells were cultured on FN-treated titanium disks to evaluate the in vitro bioactivity. The experiment showed H-SFE titanium disks adsorbed more FN and acquired more ß-turn content than L-SFE group. MG-63 cells cultured on FN-treated H-SFE titanium disks showed better osteogenic responses, including adhesion, proliferation, alkaline phosphatase activity and mineralization than that on FN-treated L-SFE titanium disks. Compared to L-SFE titanium disks, integrin-β1, integrin-α5 and Rac-1 mRNA levels were significantly higher in MG-63 cells on FN-treated H-SFE after 3 h of culture. These findings suggest that the higher SFE of H-SFE compared to L-SFE titanium disks induced changes in the conformation of adsorbed FN that enhanced the osteogenic activity of MG-63 cells.
Collapse
|
21
|
Daday C, de Buhr S, Mercadante D, Gräter F. Mechanical force can enhance c-Src kinase activity by impairing autoinhibition. Biophys J 2022; 121:684-691. [PMID: 35120901 PMCID: PMC8943751 DOI: 10.1016/j.bpj.2022.01.028] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 12/21/2021] [Accepted: 01/28/2022] [Indexed: 11/25/2022] Open
Abstract
Cellular mechanosensing is pivotal for virtually all biological processes, and many molecular mechano-sensors and their way of function are being uncovered. In this work, we suggest that c-Src kinase acts as a direct mechano-sensor. c-Src is responsible for, among others, cell proliferation, and shows increased activity in stretched cells. In its native state, c-Src has little basal activity, because its kinase domain binds to an SH2 and SH3 domain. However, it is known that c-Src can bind to p130Cas, through which force can be transmitted to the membrane. Using molecular dynamics simulations, we show that force acting between the membrane-bound N-terminus of the SH3 domain and p130Cas induces partial SH3 unfolding, thereby impeding rebinding of the kinase domain onto SH2/SH3 and effectively enhancing kinase activity. Forces involved in this process are slightly lower or similar to the forces required to pull out c-Src from the membrane through the myristoyl linker, and key interactions involved in this anchoring are salt bridges between negative lipids and nearby basic residues in c-Src. Thus, c-Src appears to be a candidate for an intriguing mechanosensing mechanism of impaired kinase inhibition, which can be potentially tuned by membrane composition and other environmental factors.
Collapse
Affiliation(s)
- Csaba Daday
- Heidelberg Institute for Theoretical Studies, Heidelberg, Germany
| | - Svenja de Buhr
- Heidelberg Institute for Theoretical Studies, Heidelberg, Germany
| | | | - Frauke Gräter
- Heidelberg Institute for Theoretical Studies, Heidelberg, Germany; Interdisciplinary Center for Scientific Computing (IWR), Heidelberg University, Mathematikon, Heidelberg, Germany.
| |
Collapse
|
22
|
Mao J, Zhu K, Long Z, Zhang H, Xiao B, Xi W, Wang Y, Huang J, Liu J, Shi X, Jiang H, Lu T, Wen Y, Zhang N, Meng Q, Zhou H, Ruan Z, Wang J, Luo C, Xi X. Targeting the RT loop of Src SH3 in Platelets Prevents Thrombosis without Compromising Hemostasis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2103228. [PMID: 35023301 PMCID: PMC8895158 DOI: 10.1002/advs.202103228] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 11/30/2021] [Indexed: 05/05/2023]
Abstract
Conventional antiplatelet agents indiscriminately inhibit both thrombosis and hemostasis, and the increased bleeding risk thus hampers their use at more aggressive dosages to achieve adequate effect. Blocking integrin αIIbβ3 outside-in signaling by separating the β3/Src interaction, yet to be proven in vivo, may nonetheless resolve this dilemma. Identification of a specific druggable target for this strategy remains a fundamental challenge as Src SH3 is known to be responsible for binding to not only integrin β3 but also the proteins containing the PXXP motif. In vitro and in vivo mutational analyses show that the residues, especially E97, in the RT loop of Src SH3 are critical for interacting with β3. DCDBS84, a small molecule resulting from structure-based virtual screening, is structurally validated to be directed toward the projected target. It specifically disrupts β3/Src interaction without affecting canonical PXXP binding and thus inhibits the outside-in signaling-regulated platelet functions. Treatment of mice with DCDBS84 causes a profound inhibition of thrombosis, equivalent to that induced by extremely high doses of αIIbβ3 antagonist, but does not compromise primary hemostasis. Specific targets are revealed for a preferential inhibition of thrombosis that may lead to new classes of potent antithrombotics without hemorrhagic side effects.
Collapse
Affiliation(s)
- Jianhua Mao
- State Key Laboratory of Medical GenomicsShanghai Institute of HematologyCollaborative Innovation Center of HematologyRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025China
| | - Kongkai Zhu
- Drug Discovery and Design Centerthe Center for Chemical BiologyState Key Laboratory of Drug ResearchShanghai Institute of Materia MedicaChinese Academy of SciencesUniversity of Chinese Academy of SciencesShanghai201203China
| | - Zhangbiao Long
- State Key Laboratory of Medical GenomicsShanghai Institute of HematologyCollaborative Innovation Center of HematologyRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025China
| | - Huimin Zhang
- Drug Discovery and Design Centerthe Center for Chemical BiologyState Key Laboratory of Drug ResearchShanghai Institute of Materia MedicaChinese Academy of SciencesUniversity of Chinese Academy of SciencesShanghai201203China
- School of Life Science and TechnologyShanghai Tech UniversityShanghai201210China
| | - Bing Xiao
- State Key Laboratory of Medical GenomicsShanghai Institute of HematologyCollaborative Innovation Center of HematologyRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025China
| | - Wenda Xi
- Shanghai Institute of HypertensionRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025China
| | - Yun Wang
- State Key Laboratory of Medical GenomicsShanghai Institute of HematologyCollaborative Innovation Center of HematologyRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025China
| | - Jiansong Huang
- State Key Laboratory of Medical GenomicsShanghai Institute of HematologyCollaborative Innovation Center of HematologyRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025China
| | - Jingqiu Liu
- Drug Discovery and Design Centerthe Center for Chemical BiologyState Key Laboratory of Drug ResearchShanghai Institute of Materia MedicaChinese Academy of SciencesUniversity of Chinese Academy of SciencesShanghai201203China
| | - Xiaofeng Shi
- State Key Laboratory of Medical GenomicsShanghai Institute of HematologyCollaborative Innovation Center of HematologyRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025China
| | - Hao Jiang
- Drug Discovery and Design Centerthe Center for Chemical BiologyState Key Laboratory of Drug ResearchShanghai Institute of Materia MedicaChinese Academy of SciencesUniversity of Chinese Academy of SciencesShanghai201203China
| | - Tian Lu
- Drug Discovery and Design Centerthe Center for Chemical BiologyState Key Laboratory of Drug ResearchShanghai Institute of Materia MedicaChinese Academy of SciencesUniversity of Chinese Academy of SciencesShanghai201203China
| | - Yi Wen
- Drug Discovery and Design Centerthe Center for Chemical BiologyState Key Laboratory of Drug ResearchShanghai Institute of Materia MedicaChinese Academy of SciencesUniversity of Chinese Academy of SciencesShanghai201203China
| | - Naixia Zhang
- Drug Discovery and Design Centerthe Center for Chemical BiologyState Key Laboratory of Drug ResearchShanghai Institute of Materia MedicaChinese Academy of SciencesUniversity of Chinese Academy of SciencesShanghai201203China
| | - Qian Meng
- Drug Discovery and Design Centerthe Center for Chemical BiologyState Key Laboratory of Drug ResearchShanghai Institute of Materia MedicaChinese Academy of SciencesUniversity of Chinese Academy of SciencesShanghai201203China
| | - Hu Zhou
- Drug Discovery and Design Centerthe Center for Chemical BiologyState Key Laboratory of Drug ResearchShanghai Institute of Materia MedicaChinese Academy of SciencesUniversity of Chinese Academy of SciencesShanghai201203China
| | - Zheng Ruan
- State Key Laboratory of Medical GenomicsShanghai Institute of HematologyCollaborative Innovation Center of HematologyRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025China
| | - Jin Wang
- State Key Laboratory of Medical GenomicsShanghai Institute of HematologyCollaborative Innovation Center of HematologyRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025China
| | - Cheng Luo
- Drug Discovery and Design Centerthe Center for Chemical BiologyState Key Laboratory of Drug ResearchShanghai Institute of Materia MedicaChinese Academy of SciencesUniversity of Chinese Academy of SciencesShanghai201203China
- School of Life Science and TechnologyShanghai Tech UniversityShanghai201210China
- School of Pharmaceutical Science and TechnologyHangzhou Institute for Advanced StudyUCASHangzhou310024China
| | - Xiaodong Xi
- State Key Laboratory of Medical GenomicsShanghai Institute of HematologyCollaborative Innovation Center of HematologyRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025China
| |
Collapse
|
23
|
Moretti L, Stalfort J, Barker TH, Abebayehu D. The interplay of fibroblasts, the extracellular matrix, and inflammation in scar formation. J Biol Chem 2022; 298:101530. [PMID: 34953859 PMCID: PMC8784641 DOI: 10.1016/j.jbc.2021.101530] [Citation(s) in RCA: 135] [Impact Index Per Article: 67.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 12/08/2021] [Indexed: 02/06/2023] Open
Abstract
Various forms of fibrosis, comprising tissue thickening and scarring, are involved in 40% of deaths across the world. Since the discovery of scarless functional healing in fetuses prior to a certain stage of development, scientists have attempted to replicate scarless wound healing in adults with little success. While the extracellular matrix (ECM), fibroblasts, and inflammatory mediators have been historically investigated as separate branches of biology, it has become increasingly necessary to consider them as parts of a complex and tightly regulated system that becomes dysregulated in fibrosis. With this new paradigm, revisiting fetal scarless wound healing provides a unique opportunity to better understand how this highly regulated system operates mechanistically. In the following review, we navigate the four stages of wound healing (hemostasis, inflammation, repair, and remodeling) against the backdrop of adult versus fetal wound healing, while also exploring the relationships between the ECM, effector cells, and signaling molecules. We conclude by singling out recent findings that offer promising leads to alter the dynamics between the ECM, fibroblasts, and inflammation to promote scarless healing. One factor that promises to be significant is fibroblast heterogeneity and how certain fibroblast subpopulations might be predisposed to scarless healing. Altogether, reconsidering fetal wound healing by examining the interplay of the various factors contributing to fibrosis provides new research directions that will hopefully help us better understand and address fibroproliferative diseases, such as idiopathic pulmonary fibrosis, liver cirrhosis, systemic sclerosis, progressive kidney disease, and cardiovascular fibrosis.
Collapse
Affiliation(s)
- Leandro Moretti
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
| | - Jack Stalfort
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
| | - Thomas Harrison Barker
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
| | - Daniel Abebayehu
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA.
| |
Collapse
|
24
|
Sheikh A, Alhakamy NA, Md S, Kesharwani P. Recent Progress of RGD Modified Liposomes as Multistage Rocket Against Cancer. Front Pharmacol 2022; 12:803304. [PMID: 35145405 PMCID: PMC8822168 DOI: 10.3389/fphar.2021.803304] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 12/22/2021] [Indexed: 12/24/2022] Open
Abstract
Cancer is a life-threatening disease, contributing approximately 9.4 million deaths worldwide. To address this challenge, scientific researchers have investigated molecules that could act as speed-breakers for cancer. As an abiotic drug delivery system, liposomes can hold both hydrophilic and lipophilic drugs, which promote a controlled release, accumulate in the tumor microenvironment, and achieve elongated half-life with an enhanced safety profile. To further improve the safety and impair the off-target effect, the surface of liposomes could be modified in a way that is easily identified by cancer cells, promotes uptake, and facilitates angiogenesis. Integrins are overexpressed on cancer cells, which upon activation promote downstream cell signaling and eventually activate specific pathways, promoting cell growth, proliferation, and migration. RGD peptides are easily recognized by integrin over expressed cells. Just like a multistage rocket, ligand anchored liposomes can be selectively recognized by target cells, accumulate at the specific site, and finally, release the drug in a specific and desired way. This review highlights the role of integrin in cancer development, so gain more insights into the phenomenon of tumor initiation and survival. Since RGD is recognized by the integrin family, the fate of RGD has been demonstrated after its binding with the acceptor’s family. The role of RGD based liposomes in targeting various cancer cells is also highlighted in the paper.
Collapse
Affiliation(s)
- Afsana Sheikh
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Nabil A. Alhakamy
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
- Center of Excellence for Drug Research and Pharmaceutical Industries, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Shadab Md
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
- Center of Excellence for Drug Research and Pharmaceutical Industries, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
- *Correspondence: Prashant Kesharwani,
| |
Collapse
|
25
|
Day RN, Day KH, Pavalko FM. Direct visualization by FRET-FLIM of a putative mechanosome complex involving Src, Pyk2 and MBD2 in living MLO-Y4 cells. PLoS One 2021; 16:e0261660. [PMID: 34941939 PMCID: PMC8699642 DOI: 10.1371/journal.pone.0261660] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 12/08/2021] [Indexed: 01/12/2023] Open
Abstract
Earlier, we proposed the “mechanosome” concept as a testable model for understanding how mechanical stimuli detected by cell surface adhesion molecules are transmitted to modulate gene expression inside cells. Here, for the first time we document a putative mechanosome involving Src, Pyk2 and MBD2 in MLO-Y4 osteocytes with high spatial resolution using FRET-FLIM. Src-Pyk2 complexes were concentrated at the periphery of focal adhesions and the peri-nuclear region. Pyk2-MBD2 complexes were located primarily in the nucleus and peri-nuclear region. Lifetime measurements indicated that Src and MBD2 did not interact directly. Finally, mechanical stimulation by fluid flow induced apparent accumulation of Src-Pyk2 protein complexes in the peri-nuclear/nuclear region, consistent with the proposed behavior of a mechanosome in response to a mechanical stimulus.
Collapse
Affiliation(s)
- Richard N. Day
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Kathleen H. Day
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Fredrick M. Pavalko
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
- * E-mail:
| |
Collapse
|
26
|
Mechanosensing and the Hippo Pathway in Microglia: A Potential Link to Alzheimer's Disease Pathogenesis? Cells 2021; 10:cells10113144. [PMID: 34831369 PMCID: PMC8622675 DOI: 10.3390/cells10113144] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 10/27/2021] [Accepted: 10/29/2021] [Indexed: 01/01/2023] Open
Abstract
The activation of microglia, the inflammatory cells of the central nervous system (CNS), has been linked to the pathogenesis of Alzheimer’s disease and other neurodegenerative diseases. How microglia sense the changing brain environment, in order to respond appropriately, is still being elucidated. Microglia are able to sense and respond to the mechanical properties of their microenvironment, and the physical and molecular pathways underlying this mechanosensing/mechanotransduction in microglia have recently been investigated. The Hippo pathway functions through mechanosensing and subsequent protein kinase cascades, and is critical for neuronal development and many other cellular processes. In this review, we examine evidence for the potential involvement of Hippo pathway components specifically in microglia in the pathogenesis of Alzheimer’s disease. We suggest that the Hippo pathway is worth investigating as a mechanosensing pathway in microglia, and could be one potential therapeutic target pathway for preventing microglial-induced neurodegeneration in AD.
Collapse
|
27
|
Epigenetic induction of lipocalin 2 expression drives acquired resistance to 5-fluorouracil in colorectal cancer through integrin β3/SRC pathway. Oncogene 2021; 40:6369-6380. [PMID: 34588619 DOI: 10.1038/s41388-021-02029-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 09/03/2021] [Accepted: 09/17/2021] [Indexed: 12/24/2022]
Abstract
The therapeutic efficacy of 5-fluorouracil (5-FU) is often reduced by the development of drug resistance. We observed significant upregulation of lipocalin 2 (LCN2) expression in a newly established 5-FU-resistant colorectal cancer (CRC) cell line. In this study, we demonstrated that 5-FU-treated CRC cells developed resistance through LCN2 upregulation caused by LCN2 promoter demethylation and that feedback between LCN2 and NF-κB further amplified LCN2 expression. High LCN2 expression was associated with poor prognosis in CRC patients. LCN2 attenuated the cytotoxicity of 5-FU by activating the SRC/AKT/ERK-mediated antiapoptotic program. Mechanistically, the LCN2-integrin β3 interaction enhanced integrin β3 stability, thus recruiting SRC to the cytomembrane for autoactivation, leading to downstream AKT/ERK cascade activation. Targeting LCN2 or SRC compromised the growth of CRC cells with LCN2-induced 5-FU resistance. Our findings demonstrate a novel mechanism of acquired resistance to 5-FU, suggesting that LCN2 can be used as a biomarker and/or therapeutic target for advanced CRC.
Collapse
|
28
|
Abstract
Effective regulation of immune-cell activation is critical for ensuring that the immune response, and inflammation generated for the purpose of pathogen elimination, are limited in space and time to minimize tissue damage. Autoimmune disease can occur when immunoreceptor signaling is dysregulated, leading to unrestrained inflammation and organ damage. Conversely, tumors can coopt the tissue healing and immunosuppressive functions of hematopoietic cells to promote metastasis and evade therapy. The Src-family kinase Lyn is an essential regulator of immunoreceptor signaling, initiating both proinflammatory and suppressive signaling pathways in myeloid immune cells (eg, neutrophils, dendritic cells, monocytes, macrophages) and in B lymphocytes. Defects in Lyn signaling are implicated in autoimmune disease, but mechanisms by which Lyn, expressed along with a battery of other Src-family kinases, may uniquely direct both positive and negative signaling remain incompletely defined. This review describes our current understanding of the activating and inhibitory contributions of Lyn to immunoreceptor signaling and how these processes contribute to myeloid and B-cell function. We also highlight recent work suggesting that the 2 proteins generated by alternative splicing of lyn, LynA and LynB, differentially regulate both immune and cancer-cell signaling. These principles may also extend to other Lyn-expressing cells, such as neuronal and endocrine cells. Unraveling the common and cell-specific aspects of Lyn function could lead to new approaches to therapeutically target dysregulated pathways in pathologies ranging from autoimmune and neurogenerative disease to cancer.
Collapse
Affiliation(s)
- Ben F Brian
- Department of Pharmacology, University of Minnesota, Minneapolis, MN, USA
- Current Affiliation: Current affiliation for B.F.B.: Division of Immunology & Pathogenesis, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Tanya S Freedman
- Department of Pharmacology, University of Minnesota, Minneapolis, MN, USA
- Center for Immunology, University of Minnesota, Minneapolis, MN, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
- Center for Autoimmune Diseases Research, University of Minnesota, Minneapolis, MN, USA
- Correspondence: Tanya S. Freedman, PhD, University of Minnesota Twin Cities Campus: University of Minnesota, 6-120 Jackson Hall, 321 Church St. S.E., Minneapolis, MN 55455, USA. E-mail:
| |
Collapse
|
29
|
Schreuder WH, van der Wal JE, de Lange J, van den Berg H. Multiple versus solitary giant cell lesions of the jaw: Similar or distinct entities? Bone 2021; 149:115935. [PMID: 33771761 DOI: 10.1016/j.bone.2021.115935] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 02/27/2021] [Accepted: 03/17/2021] [Indexed: 02/06/2023]
Abstract
The majority of giant cell lesions of the jaw present as a solitary focus of disease in bones of the maxillofacial skeleton. Less frequently they occur as multifocal lesions. This raises the clinical dilemma if these should be considered distinct entities and therefore each need a specific therapeutic approach. Solitary giant cell lesions of the jaw present with a great diversity of symptoms. Recent molecular analysis revealed that these are associated with somatic gain-of-function mutations in KRAS, FGFR1 or TRPV4 in a large component of the mononuclear stromal cells which all act on the RAS/MAPK pathway. For multifocal lesions, a small group of neoplastic multifocal giant cell lesions of the jaw remain after ruling out hyperparathyroidism. Strikingly, most of these patients are diagnosed with jaw lesions before the age of 20 years, thus before the completion of dental and jaw development. These multifocal lesions are often accompanied by a diagnosis or strong clinical suspicion of a syndrome. Many of the frequently reported syndromes belong to the so-called RASopathies, with germline or mosaic mutations leading to downstream upregulation of the RAS/MAPK pathway. The other frequently reported syndrome is cherubism, with gain-of-function mutations in the SH3BP2 gene leading through assumed and unknown signaling to an autoinflammatory bone disorder with hyperactive osteoclasts and defective osteoblastogenesis. Based on this extensive literature review, a RAS/MAPK pathway activation is hypothesized in all giant cell lesions of the jaw. The different interaction between and contribution of deregulated signaling in individual cell lineages and crosstalk with other pathways among the different germline- and non-germline-based alterations causing giant cell lesions of the jaw can be explanatory for the characteristic clinical features. As such, this might also aid in the understanding of the age-dependent symptomatology of syndrome associated giant cell lesions of the jaw; hopefully guiding ideal timing when installing treatment strategies in the future.
Collapse
Affiliation(s)
- Willem H Schreuder
- Department of Oral and Maxillofacial Surgery, Amsterdam UMC and Academic Center for Dentistry Amsterdam, University of Amsterdam, Amsterdam, the Netherlands; Department of Head and Neck Surgery and Oncology, Antoni van Leeuwenhoek / Netherlands Cancer Institute, Amsterdam, the Netherlands.
| | - Jacqueline E van der Wal
- Department of Pathology, Antoni van Leeuwenhoek / Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Jan de Lange
- Department of Oral and Maxillofacial Surgery, Amsterdam UMC and Academic Center for Dentistry Amsterdam, University of Amsterdam, Amsterdam, the Netherlands
| | - Henk van den Berg
- Department of Pediatrics / Oncology, Amsterdam UMC, University of Amsterdam, Emma Children's Hospital, Amsterdam, the Netherlands
| |
Collapse
|
30
|
Zhang RM, Zeyer KA, Odenthal N, Zhang Y, Reinhardt DP. The fibrillin-1 RGD motif posttranscriptionally regulates ERK1/2 signaling and fibroblast proliferation via miR-1208. FASEB J 2021; 35:e21598. [PMID: 33871068 DOI: 10.1096/fj.202100282r] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 03/28/2021] [Accepted: 03/31/2021] [Indexed: 12/17/2022]
Abstract
Fibrillin-1 is an extracellular matrix protein which contains one conserved RGD integrin-binding motif. It constitutes the backbone of microfibrils in many tissues, and mutations in fibrillin-1 cause various connective tissue disorders. Although it is well established that fibrillin-1 interacts with several RGD-dependent integrins, very little is known about the associated intracellular signaling pathways. Recent published evidence identified a subset of miRNAs regulated by fibrillin-1 RGD-cell adhesion, with miR-1208 among the most downregulated. The present study shows that the downregulated miR-1208 controls fibroblast proliferation. Inhibitor experiments revealed that fibrillin-1 RGD suppressed miR-1208 expression via c-Src kinase and the downstream JNK signaling. Bioinformatic prediction and experimental target sequence validation demonstrated four miR-1208 binding sites on the ERK2 mRNA and one on the MEK1 mRNA. ERK2 and MEK1 are critical proliferation-promoting kinases. Decreased miR-1208 levels elevated the total and phosphorylated ERK1/2 and MEK1/2 protein levels and the phosphorylated to total ERK1/2 ratio. Together, the data demonstrate a novel outside-in signaling mechanism explaining how fibrillin-1 RGD-cell binding regulates fibroblast proliferation.
Collapse
Affiliation(s)
- Rong-Mo Zhang
- Faculty of Medicine and Health Sciences, Department of Anatomy and Cell Biology, McGill University, Montreal, Canada
| | - Karina A Zeyer
- Faculty of Medicine and Health Sciences, Department of Anatomy and Cell Biology, McGill University, Montreal, Canada
| | - Nadine Odenthal
- Department of Natural Science, University of Lübeck, Lübeck, Germany
| | - Yiyun Zhang
- Faculty of Medicine and Health Sciences, Department of Anatomy and Cell Biology, McGill University, Montreal, Canada
| | - Dieter P Reinhardt
- Faculty of Medicine and Health Sciences, Department of Anatomy and Cell Biology, McGill University, Montreal, Canada.,Faculty of Dentistry, McGill University, Montreal, Canada
| |
Collapse
|
31
|
Kim TI, Kim YJ, Kim K. Extract of Seaweed Codium fragile Inhibits Integrin αIIbβ3-Induced Outside-in Signaling and Arterial Thrombosis. Front Pharmacol 2021; 12:685948. [PMID: 34276375 PMCID: PMC8283197 DOI: 10.3389/fphar.2021.685948] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 06/11/2021] [Indexed: 11/22/2022] Open
Abstract
Seaweeds are thought to be promising candidates for functional foods and to help prevent thrombotic and related cardiovascular diseases. Codium fragile (Suringer) Hariot has been traditionally used as a culinary ingredient, and it possesses a range of biological activities, including the inhibition of platelet function. However, the mechanism of this inhibition is unclear. The aim of this study was to examine the inhibitory effect of C. fragile in platelet function. The antiplatelet activity of C. fragile on agonist-activated platelet aggregation, granule secretion, calcium mobilization, platelet spreading, and clot retraction was assessed. The phosphorylation of c-Src, Syk, PLCγ2, and several proteins involving in the αIIbβ3 integrin outside-in signaling pathway were also studied in thrombin and CRP-stimulated platelets. The antithrombotic effect was investigated in mice using ferric chloride-induced arterial thrombus formation in vivo. Transection tail bleeding time was used to evaluate whether C. fragile inhibited primary hemostasis. The main components and contents of C. fragile ethanol extract were confirmed by GC-MS analysis. C. fragile significantly impaired agonist-induced platelet aggregation granule secretion, calcium mobilization, platelet spreading, and clot retraction. Biochemical analysis revealed that C. fragile inhibited the agonist-induced activation of c-Src, Syk, and PLCγ2, as well as the phosphorylation of PI3K, AKT, and mitogen-activated protein kinases (MAPKs). The inhibitory effect of C. fragile resulted from an inhibition of platelet αIIbβ3 integrin outside-in signal transduction during cell activation. Oral administration of C. fragile efficiently blocked FeCl3-induced arterial thrombus formation in vivo without prolonging bleeding time. GC-MS analysis revealed that phytol was the main constituent and the total content of isomers was 160.8 mg/kg. Our results demonstrated that C. fragile suppresses not only the inside-out signaling of αIIbβ3 integrin but also outside-in signal transmission. Therefore, C. fragile could be an effective antiplatelet therapeutic candidate.
Collapse
Affiliation(s)
- Tae In Kim
- Korean Medicine-Application Center, Korea Institute of Oriental Medicine, Daegu, South Korea
| | - Yeon-Ji Kim
- Korean Medicine-Application Center, Korea Institute of Oriental Medicine, Daegu, South Korea
| | - Kyungho Kim
- Korean Medicine-Application Center, Korea Institute of Oriental Medicine, Daegu, South Korea
| |
Collapse
|
32
|
Dzobo K. Integrins Within the Tumor Microenvironment: Biological Functions, Importance for Molecular Targeting, and Cancer Therapeutics Innovation. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2021; 25:417-430. [PMID: 34191612 DOI: 10.1089/omi.2021.0069] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Many cellular functions important for solid tumor initiation and progression are mediated by members of the integrin family, a diverse family of cell attachment receptors. With recent studies emphasizing the role of the tumor microenvironment (TME) in tumor initiation and progression, it is not surprising that considerable attention is being paid to integrins. Several integrin antagonists are under clinical trials, with many demonstrating promising activity in patients with different cancers. A deeper knowledge of the functions of integrins within the TME is still required and might lead to better inhibitors being discovered. Integrin expression is commonly dysregulated in many tumors with integrins playing key roles in signaling as well as promotion of tumor cell invasion and migration. Integrins also play a major role in adhesion of circulating tumor cells to new sites and the resulting formation of secondary tumors. Furthermore, integrins have demonstrated the ability to promoting stem cell-like properties in tumor cells as well as drug resistance. Anti-integrin therapies rely heavily on the doses or concentrations used as these determine whether the drugs act as antagonists or as integrin agonists. This expert review offers the latest synthesis in terms of the current knowledge of integrins functions within the TME and as potential molecular targets for cancer therapeutics innovation.
Collapse
Affiliation(s)
- Kevin Dzobo
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town Component, Cape Town, South Africa.,Division of Medical Biochemistry and Institute of Infectious Disease and Molecular Medicine, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
33
|
Sakai Y, Tsunekawa M, Ohta K, Shimizu T, Pastuhov S, Hanafusa H, Hisamoto N, Matsumoto K. The Integrin Signaling Network Promotes Axon Regeneration via the Src-Ephexin-RhoA GTPase Signaling Axis. J Neurosci 2021; 41:4754-4767. [PMID: 33963050 PMCID: PMC8260174 DOI: 10.1523/jneurosci.2456-20.2021] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 04/05/2021] [Accepted: 04/07/2021] [Indexed: 12/12/2022] Open
Abstract
Axon regeneration is an evolutionarily conserved process essential for restoring the function of damaged neurons. In Caenorhabditis elegans hermaphrodites, initiation of axon regeneration is regulated by the RhoA GTPase-ROCK (Rho-associated coiled-coil kinase)-regulatory nonmuscle myosin light-chain phosphorylation signaling pathway. However, the upstream mechanism that activates the RhoA pathway remains unknown. Here, we show that axon injury activates TLN-1/talin via the cAMP-Epac (exchange protein directly activated by cAMP)-Rap GTPase cascade and that TLN-1 induces multiple downstream events, one of which is integrin inside-out activation, leading to the activation of the RhoA-ROCK signaling pathway. We found that the nonreceptor tyrosine kinase Src, a key mediator of integrin signaling, activates the Rho guanine nucleotide exchange factor EPHX-1/ephexin by phosphorylating the Tyr-568 residue in the autoinhibitory domain. Our results suggest that the C. elegans integrin signaling network regulates axon regeneration via the Src-RhoGEF-RhoA axis.SIGNIFICANCE STATEMENT The ability of axons to regenerate after injury is governed by cell-intrinsic regeneration pathways. We have previously demonstrated that the Caenorhabditis elegans RhoA GTPase-ROCK (Rho-associated coiled-coil kinase) pathway promotes axon regeneration by inducing MLC-4 phosphorylation. In this study, we found that axon injury activates TLN-1/talin through the cAMP-Epac (exchange protein directly activated by cAMP)-Rap GTPase cascade, leading to integrin inside-out activation, which promotes axonal regeneration by activating the RhoA signaling pathway. In this pathway, SRC-1/Src acts downstream of integrin activation and subsequently activates EPHX-1/ephexin RhoGEF by phosphorylating the Tyr-568 residue in the autoinhibitory domain. Our results suggest that the C. elegans integrin signaling network regulates axon regeneration via the Src-RhoGEF-RhoA axis.
Collapse
Affiliation(s)
- Yoshiki Sakai
- Division of Biological Science, Graduate School of Science, Nagoya University, Nagoya 464-8602, Japan
| | - Mayuka Tsunekawa
- Division of Biological Science, Graduate School of Science, Nagoya University, Nagoya 464-8602, Japan
| | - Kohei Ohta
- Division of Biological Science, Graduate School of Science, Nagoya University, Nagoya 464-8602, Japan
| | - Tatsuhiro Shimizu
- Division of Biological Science, Graduate School of Science, Nagoya University, Nagoya 464-8602, Japan
| | - Strahil Pastuhov
- Division of Biological Science, Graduate School of Science, Nagoya University, Nagoya 464-8602, Japan
| | - Hiroshi Hanafusa
- Division of Biological Science, Graduate School of Science, Nagoya University, Nagoya 464-8602, Japan
| | - Naoki Hisamoto
- Division of Biological Science, Graduate School of Science, Nagoya University, Nagoya 464-8602, Japan
| | - Kunihiro Matsumoto
- Division of Biological Science, Graduate School of Science, Nagoya University, Nagoya 464-8602, Japan
| |
Collapse
|
34
|
Xu Z, Liang Y, Delaney MK, Zhang Y, Kim K, Li J, Bai Y, Cho J, Ushio-Fukai M, Cheng N, Du X. Shear and Integrin Outside-In Signaling Activate NADPH-Oxidase 2 to Promote Platelet Activation. Arterioscler Thromb Vasc Biol 2021; 41:1638-1653. [PMID: 33691478 PMCID: PMC8057529 DOI: 10.1161/atvbaha.120.315773] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 03/01/2021] [Indexed: 11/18/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Zheng Xu
- Department of Pharmacology, University of Illinois at Chicago (Z.X., Y.L., M.K.D., Y.Z., K.K., J.L., Y.B., J.C., M.U.-F., N.C., X.D.)
| | - Ying Liang
- Department of Pharmacology, University of Illinois at Chicago (Z.X., Y.L., M.K.D., Y.Z., K.K., J.L., Y.B., J.C., M.U.-F., N.C., X.D.)
| | - M. Keegan Delaney
- Department of Pharmacology, University of Illinois at Chicago (Z.X., Y.L., M.K.D., Y.Z., K.K., J.L., Y.B., J.C., M.U.-F., N.C., X.D.)
- Dupage Medical Technology, Inc (M.K.D.)
| | - Yaping Zhang
- Department of Pharmacology, University of Illinois at Chicago (Z.X., Y.L., M.K.D., Y.Z., K.K., J.L., Y.B., J.C., M.U.-F., N.C., X.D.)
| | - Kyungho Kim
- Department of Pharmacology, University of Illinois at Chicago (Z.X., Y.L., M.K.D., Y.Z., K.K., J.L., Y.B., J.C., M.U.-F., N.C., X.D.)
- Korean Medicine-Application Center, Korea Institute of Oriental Medicine, Daegu (K.K.)
| | - Jing Li
- Department of Pharmacology, University of Illinois at Chicago (Z.X., Y.L., M.K.D., Y.Z., K.K., J.L., Y.B., J.C., M.U.-F., N.C., X.D.)
| | - Yanyan Bai
- Department of Pharmacology, University of Illinois at Chicago (Z.X., Y.L., M.K.D., Y.Z., K.K., J.L., Y.B., J.C., M.U.-F., N.C., X.D.)
| | - Jaehyung Cho
- Department of Pharmacology, University of Illinois at Chicago (Z.X., Y.L., M.K.D., Y.Z., K.K., J.L., Y.B., J.C., M.U.-F., N.C., X.D.)
| | - Masuko Ushio-Fukai
- Department of Pharmacology, University of Illinois at Chicago (Z.X., Y.L., M.K.D., Y.Z., K.K., J.L., Y.B., J.C., M.U.-F., N.C., X.D.)
- Department of Medicine (Cardiology), Vascular Biology Center, Medical College of Georgia at Augusta University (M.U.-F.)
| | - Ni Cheng
- Department of Pharmacology, University of Illinois at Chicago (Z.X., Y.L., M.K.D., Y.Z., K.K., J.L., Y.B., J.C., M.U.-F., N.C., X.D.)
| | - Xiaoping Du
- Department of Pharmacology, University of Illinois at Chicago (Z.X., Y.L., M.K.D., Y.Z., K.K., J.L., Y.B., J.C., M.U.-F., N.C., X.D.)
| |
Collapse
|
35
|
Ludwig BS, Kessler H, Kossatz S, Reuning U. RGD-Binding Integrins Revisited: How Recently Discovered Functions and Novel Synthetic Ligands (Re-)Shape an Ever-Evolving Field. Cancers (Basel) 2021; 13:1711. [PMID: 33916607 PMCID: PMC8038522 DOI: 10.3390/cancers13071711] [Citation(s) in RCA: 92] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/22/2021] [Accepted: 03/29/2021] [Indexed: 12/19/2022] Open
Abstract
Integrins have been extensively investigated as therapeutic targets over the last decades, which has been inspired by their multiple functions in cancer progression, metastasis, and angiogenesis as well as a continuously expanding number of other diseases, e.g., sepsis, fibrosis, and viral infections, possibly also Severe Acute Respiratory Syndrome Coronavirus (SARS-CoV-2). Although integrin-targeted (cancer) therapy trials did not meet the high expectations yet, integrins are still valid and promising targets due to their elevated expression and surface accessibility on diseased cells. Thus, for the future successful clinical translation of integrin-targeted compounds, revisited and innovative treatment strategies have to be explored based on accumulated knowledge of integrin biology. For this, refined approaches are demanded aiming at alternative and improved preclinical models, optimized selectivity and pharmacological properties of integrin ligands, as well as more sophisticated treatment protocols considering dose fine-tuning of compounds. Moreover, integrin ligands exert high accuracy in disease monitoring as diagnostic molecular imaging tools, enabling patient selection for individualized integrin-targeted therapy. The present review comprehensively analyzes the state-of-the-art knowledge on the roles of RGD-binding integrin subtypes in cancer and non-cancerous diseases and outlines the latest achievements in the design and development of synthetic ligands and their application in biomedical, translational, and molecular imaging approaches. Indeed, substantial progress has already been made, including advanced ligand designs, numerous elaborated pre-clinical and first-in-human studies, while the discovery of novel applications for integrin ligands remains to be explored.
Collapse
Affiliation(s)
- Beatrice S. Ludwig
- Department of Nuclear Medicine, University Hospital Klinikum Rechts der Isar and Central Institute for Translational Cancer Research (TranslaTUM), Technical University Munich, 81675 Munich, Germany;
| | - Horst Kessler
- Department of Chemistry, Institute for Advanced Study, Technical University Munich, 85748 Garching, Germany;
| | - Susanne Kossatz
- Department of Nuclear Medicine, University Hospital Klinikum Rechts der Isar and Central Institute for Translational Cancer Research (TranslaTUM), Technical University Munich, 81675 Munich, Germany;
- Department of Chemistry, Institute for Advanced Study, Technical University Munich, 85748 Garching, Germany;
| | - Ute Reuning
- Clinical Research Unit, Department of Obstetrics and Gynecology, University Hospital Klinikum Rechts der Isar, Technical University Munich, 81675 Munich, Germany
| |
Collapse
|
36
|
Lakk M, Križaj D. TRPV4-Rho signaling drives cytoskeletal and focal adhesion remodeling in trabecular meshwork cells. Am J Physiol Cell Physiol 2021; 320:C1013-C1030. [PMID: 33788628 PMCID: PMC8285634 DOI: 10.1152/ajpcell.00599.2020] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Intraocular pressure (IOP) is dynamically regulated by the trabecular meshwork (TM), a mechanosensitive tissue that protects the eye from injury through dynamic regulation of aqueous humor flow. TM compensates for mechanical stress impelled by chronic IOP elevations through increased actin polymerization, tissue stiffness, and contractility. This process has been associated with open angle glaucoma; however, the mechanisms that link mechanical stress to pathological cytoskeletal remodeling downstream from the mechanotransducers remain poorly understood. We used fluorescence imaging and biochemical analyses to investigate cytoskeletal and focal adhesion remodeling in human TM cells stimulated with physiological strains. Mechanical stretch promoted F-actin polymerization, increased the number and size of focal adhesions, and stimulated the activation of the Rho-associated protein kinase (ROCK). Stretch-induced activation of the small GTPase Ras homolog family member A (RhoA), and tyrosine phosphorylations of focal adhesion proteins paxillin, focal adhesion kinase (FAK), vinculin, and zyxin were time dependently inhibited by ROCK inhibitor trans-4-[(1R)-1-aminoethyl]-N-4-pyridinylcyclohexanecarboxamide dihydrochloride (Y-27632), and by HC-067047, an antagonist of transient receptor potential vanilloid 4 (TRPV4) channels. Both TRPV4 and ROCK activation were required for zyxin translocation and increase in the number/size of focal adhesions in stretched cells. Y-27632 blocked actin polymerization without affecting calcium influx induced by membrane stretch and the TRPV4 agonist GSK1016790A. These results reveal that mechanical tuning of TM cells requires parallel activation of TRPV4, integrins, and ROCK, with chronic stress leading to sustained remodeling of the cytoskeleton and focal complexes.
Collapse
Affiliation(s)
- Monika Lakk
- Department of Ophthalmology and Visual Sciences, University of Utah, Salt Lake City, Utah
| | - David Križaj
- Department of Ophthalmology and Visual Sciences, University of Utah, Salt Lake City, Utah.,Department of Bioengineering, University of Utah, Salt Lake City, Utah.,Department of Neurobiology, University of Utah, Salt Lake City, Utah
| |
Collapse
|
37
|
Senis YA, Nagy Z, Mori J, Lane S, Lane P. Platelet Src family kinases: A tale of reversible phosphorylation. Res Pract Thromb Haemost 2021; 5:376-389. [PMID: 33870023 PMCID: PMC8035799 DOI: 10.1002/rth2.12495] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 01/17/2021] [Accepted: 01/19/2021] [Indexed: 11/07/2022] Open
Abstract
Sarcoma (Src) family kinases (SFKs) have occupied a central place in platelet research for over 40 years. Discovered by virologists and oncologists as the proto proto-oncogene, Src tyrosine kinase spurred a phenomenal burst of research on reversible tyrosine phosphorylation and signal transduction. For a time, platelets were adopted as the model of choice for studying the biological functions of Src, owing to their ease of isolation, high Src expression, and lack of a nucleus, only to be abandoned due to challenges of culturing and manipulating using common molecular biology-based techniques. For platelet biologists, SFKs have remained an important area of investigation, initiating and amplifying signals from all major adhesion, activation, and inhibitory receptors, including the integrin αIIbβ3, the collagen receptor complex glycoprotein VI-Fc receptor γ-chain, the G protein-coupled ADP receptor P2Y12 and the inhibitory receptors platelet endothelial cell adhesion molecule-1 and G6b-B. The vital roles of SFKs in platelets is highlighted by the severe phenotypes of null and gain-of-function mutations in SFKs in mice and humans, and effects of pharmacologic inhibitors on platelet activation, thrombosis, and hemostasis. The recent description of critical regulators of SFKs in platelets, namely, C-terminal Src kinase (Csk), Csk homologous kinase (Chk), the receptor-type protein-tyrosine phosphatase receptor type J (PTPRJ) helps explain some of the bleeding side effects of tyrosine kinase inhibitors and are novel therapeutic targets for regulating the thrombotic and hemostatic capacity of platelets. Recent findings from Chk, Csk, and PTPRJ knockout mouse models highlighted that SFKs are able to autoinhibit by phosphorylating their C-terminal tyrosine residues, providing fundamental insights into SFK autoregulation.
Collapse
Affiliation(s)
- Yotis A Senis
- Unité Mixte de Recherche-S 1255 Fédération de Médecine Translationnelle de Strasbourg Université de Strasbourg Institut National de la Santé et de la Recherche Médicale Etablissement Français du Sang Grand Est Strasbourg France
| | - Zoltan Nagy
- Institute of Experimental Biomedicine University Hospital and Rudolf Virchow Center University of Würzburg Würzburg Germany
| | - Jun Mori
- Research and Development Align Technology Inc. Yokohama Japan
| | - Sophia Lane
- Illustration and Design, ScEYEnce Studios Elkins Park PA USA
| | - Patrick Lane
- Illustration and Design, ScEYEnce Studios Elkins Park PA USA
| |
Collapse
|
38
|
The 14-3-3ζ-c-Src-integrin-β3 complex is vital for platelet activation. Blood 2021; 136:974-988. [PMID: 32584951 DOI: 10.1182/blood.2019002314] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 04/27/2020] [Indexed: 12/14/2022] Open
Abstract
Several adaptor molecules bind to cytoplasmic tails of β-integrins and facilitate bidirectional signaling, which is critical in thrombosis and hemostasis. Interfering with integrin-adaptor interactions spatially or temporally to inhibit thrombosis without affecting hemostasis is an attractive strategy for the development of safe antithrombotic drugs. We show for the first time that the 14-3-3ζ-c-Src-integrin-β3 complex is formed during platelet activation. 14-3-3ζ-c-Src interaction is mediated by the -PIRLGLALNFSVFYYE- fragment (PE16) on the 14-3-3ζ and SH2-domain on c-Src, whereas the 14-3-3ζ-integrin-β3 interaction is mediated by the -ESKVFYLKMKGDYYRYL- fragment (EL17) on the 14-3-3ζ and -KEATSTF- fragment (KF7) on the β3-integrin cytoplasmic tail. The EL17-motif inhibitor, or KF7 peptide, interferes with the formation of the 14-3-3ζ-c-Src-integrin-β3 complex and selectively inhibits β3 outside-in signaling without affecting the integrin-fibrinogen interaction, which suppresses thrombosis without causing significant bleeding. This study characterized a previously unidentified 14-3-3ζ-c-Src-integrin-β3 complex in platelets and provided a novel strategy for the development of safe and effective antithrombotic treatments.
Collapse
|
39
|
Rudd CE. How the Discovery of the CD4/CD8-p56 lck Complexes Changed Immunology and Immunotherapy. Front Cell Dev Biol 2021; 9:626095. [PMID: 33791292 PMCID: PMC8005572 DOI: 10.3389/fcell.2021.626095] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 01/11/2021] [Indexed: 12/22/2022] Open
Abstract
The past 25 years have seen enormous progress in uncovering the receptors and signaling mechanisms on T-cells that activate their various effecter functions. Until the late 1980s, most studies on T-cells had focused on the influx of calcium and the levels of cAMP/GMP in T-cells. My laboratory then uncovered the interaction of CD4 and CD8 co-receptors with the protein-tyrosine kinase p56lck which are now widely accepted as the initiators of the tyrosine phosphorylation cascade leading to T-cell activation. The finding explained how immune recognition receptors expressed by many immune cells, which lack intrinsic catalytic activity, can transduce activation signals via non-covalent association with non-receptor tyrosine kinases. The discovery also established the concept that a protein tyrosine phosphorylation cascade operated in T-cells. In this vein, we and others then showed that the CD4- and CD8-p56lck complexes phosphorylate the TCR complexes which led to the identification of other protein-tyrosine kinases such as ZAP-70 and an array of substrates that are now central to studies in T-cell immunity. Other receptors such as B-cell receptor, Fc receptors and others were also subsequently found to use src kinases to control cell growth. In T-cells, p56lck driven phosphorylation targets include co-receptors such as CD28 and CTLA-4 and immune cell-specific adaptor proteins such as LAT and SLP-76 which act to integrate signals proximal to surface receptors. CD4/CD8-p56lck regulated events in T-cells include intracellular calcium mobilization, integrin activation and the induction of transcription factors for gene expression. Lastly, the identification of the targets of p56lck in the TCR and CD28 provided the framework for the development of chimeric antigen receptor (CAR) therapy in the treatment of cancer. In this review, I outline a history of the development of events that led to the development of the "TCR signaling paradigm" and its implications to immunology and immunotherapy.
Collapse
Affiliation(s)
- Christopher E. Rudd
- Division of Immunology-Oncology, Centre de Recherche Hôpital Maisonneuve-Rosemont (CR-HMR), Montreal, QC, Canada
- Department of Microbiology, Infection and Immunology, Faculty of Medicine, Universite de Montreal, Montreal, QC, Canada
- Division of Experimental Medicine, Department of Medicine, McGill University Health Center, McGill University, Montreal, QC, Canada
| |
Collapse
|
40
|
Trevisan B, Morsi A, Aleman J, Rodriguez M, Shields J, Meares D, Farland AM, Doering CB, Spencer HT, Atala A, Skardal A, Porada CD, Almeida-Porada G. Effects of Shear Stress on Production of FVIII and vWF in a Cell-Based Therapeutic for Hemophilia A. Front Bioeng Biotechnol 2021; 9:639070. [PMID: 33732691 PMCID: PMC7957060 DOI: 10.3389/fbioe.2021.639070] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 01/25/2021] [Indexed: 01/05/2023] Open
Abstract
Microfluidic technology enables recapitulation of organ-level physiology to answer pertinent questions regarding biological systems that otherwise would remain unanswered. We have previously reported on the development of a novel product consisting of human placental cells (PLC) engineered to overexpress a therapeutic factor VIII (FVIII) transgene, mcoET3 (PLC-mcoET3), to treat Hemophilia A (HA). Here, microfluidic devices were manufactured to model the physiological shear stress in liver sinusoids, where infused PLC-mcoET3 are thought to lodge after administration, to help us predict the therapeutic outcome of this novel biological strategy. In addition to the therapeutic transgene, PLC-mcoET3 also constitutively produce endogenous FVIII and von Willebrand factor (vWF), which plays a critical role in FVIII function, immunogenicity, stability, and clearance. While vWF is known to respond to flow by changing conformation, whether and how shear stress affects the production and secretion of vWF and FVIII has not been explored. We demonstrated that exposure of PLC-mcoET3 to physiological levels of shear stress present within the liver sinusoids significantly reduced mRNA levels and secreted FVIII and vWF when compared to static conditions. In contrast, mRNA for the vector-encoded mcoET3 was unaltered by flow. To determine the mechanism responsible for the observed decrease in FVIII and vWF mRNA, PCR arrays were performed to evaluate expression of genes involved in shear mechanosensing pathways. We found that flow conditions led to a significant increase in KLF2, which induces miRNAs that negatively regulate expression of FVIII and vWF, providing a mechanistic explanation for the reduced expression of these proteins in PLC under conditions of flow. In conclusion, microfluidic technology allowed us to unmask novel pathways by which endogenous FVIII and vWF are affected by shear stress, while demonstrating that expression of the therapeutic mcoET3 gene will be maintained in the gene-modified PLCs upon transplantation, irrespective of whether they engraft within sites that expose them to conditions of shear stress.
Collapse
Affiliation(s)
- Brady Trevisan
- Fetal Research and Therapy Program, Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Alshaimaa Morsi
- Fetal Research and Therapy Program, Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, United States.,Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Julio Aleman
- Fetal Research and Therapy Program, Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Martin Rodriguez
- Fetal Research and Therapy Program, Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Jordan Shields
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta and Department of Pediatrics, Emory University, Atlanta, GA, United States
| | - Diane Meares
- Department of Medicine, Section on Hematology and Oncology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Andrew M Farland
- Department of Medicine, Section on Hematology and Oncology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Christopher B Doering
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta and Department of Pediatrics, Emory University, Atlanta, GA, United States
| | - H Trent Spencer
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta and Department of Pediatrics, Emory University, Atlanta, GA, United States
| | - Anthony Atala
- Fetal Research and Therapy Program, Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Aleks Skardal
- Fetal Research and Therapy Program, Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Christopher D Porada
- Fetal Research and Therapy Program, Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Graça Almeida-Porada
- Fetal Research and Therapy Program, Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, United States
| |
Collapse
|
41
|
Gao WJ, Liu JX, Xie Y, Luo P, Liu ZQ, Liu L, Zhou H. Suppression of macrophage migration by down-regulating Src/FAK/P130Cas activation contributed to the anti-inflammatory activity of sinomenine. Pharmacol Res 2021; 167:105513. [PMID: 33617975 DOI: 10.1016/j.phrs.2021.105513] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 02/18/2021] [Accepted: 02/18/2021] [Indexed: 01/01/2023]
Abstract
A large number of macrophages in inflamed sites not only amplify the severity of inflammatory responses but also contribute to the deleterious progression of many chronic inflammatory diseases, autoimmune diseases and cancers. Macrophage migration is a prerequisite for their entry into inflammatory sites and their participation of macrophages in the pathologic processes. Inhibition of macrophage migration is therefore a potential anti-inflammatory mechanism. Moreover, alleviation of inflammation also prevents the macrophages infiltration. Sinomenine (SIN) is an alkaloid derived from the Chinese medicinal plant Sinomenium acutum. It has multiple pharmacological effects, including anti-inflammation, immunosuppression, and anti-arthritis. However, its anti-inflammatory molecular mechanisms and effect on macrophage migration are not fully understood. The purpose of this research was to investigate the pharmacological effects and the molecular mechanism of SIN on macrophage migration in vivo and in vitro as well as to elucidate its anti-inflammatory mechanisms associated with macrophage migration. Our results showed that SIN reduced the number of RAW264.7 cells migrating into inflammatory paws and blocked lipopolysaccharide (LPS)-induced RAW264.7 cells and bone marrow-derived macrophages (BMDMs) migration in vitro. Furthermore, SIN attenuated the 3D mesenchymal migration of BMDMs. The absence of macrophage migration after circulatory and periphery macrophages depletion led to a reduction in the severity of inflammatory response. In macrophages depleted (macrophages-/-) mice, as inflammatory severity decreased, RAW264.7 cells migration was suppressed. A non-obvious effect of SIN on the inflammatory response was found in macrophages-/- mice, while the inhibitory effect of SIN on RAW264.7 cells migration was still observed. Furthermore, the migration of RAW264.7 cells pre-treated with SIN was suppressed in normal mice. Finally, Src/focal adhesion kinase (FAK)/P130Cas axis activation, which supports macrophages mesenchymal migration, and iNOS expression, NO production, integrin αV and in integrin β3 expressions, which promote Src/FAK/P130Cas activation, were down-regulated by SIN. However, SIN had no obvious effect on the expression of the monocyte chemoattractant protein-1 (MCP-1), which is an important chemokine for macrophage migration. These results indicated that SIN significantly inhibited macrophage mesenchymal migration by down-regulating on Src/FAK/P130Cas axis activation. There was a mutual regulatory correlation between the inflammatory response and macrophage migration, and the effects of SIN on macrophage migration were involved in its anti-inflammatory activity.
Collapse
Affiliation(s)
- Wan-Jiao Gao
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, PR China
| | - Jian-Xin Liu
- School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua City, Hunan Province, PR China
| | - Yie Xie
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, PR China
| | - Pei Luo
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, PR China
| | - Zhong-Qiu Liu
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, Guangzhou University of Chinese Medicine, Guangzhou, PR China
| | - Liang Liu
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, PR China; Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, Guangzhou University of Chinese Medicine, Guangzhou, PR China.
| | - Hua Zhou
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, PR China; Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, Guangzhou University of Chinese Medicine, Guangzhou, PR China.
| |
Collapse
|
42
|
Bouti P, Webbers SDS, Fagerholm SC, Alon R, Moser M, Matlung HL, Kuijpers TW. β2 Integrin Signaling Cascade in Neutrophils: More Than a Single Function. Front Immunol 2021; 11:619925. [PMID: 33679708 PMCID: PMC7930317 DOI: 10.3389/fimmu.2020.619925] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 12/31/2020] [Indexed: 12/13/2022] Open
Abstract
Neutrophils are the most prevalent leukocytes in the human body. They have a pivotal role in the innate immune response against invading bacterial and fungal pathogens, while recent emerging evidence also demonstrates their role in cancer progression and anti-tumor responses. The efficient execution of many neutrophil effector responses requires the presence of β2 integrins, in particular CD11a/CD18 or CD11b/CD18 heterodimers. Although extensively studied at the molecular level, the exact signaling cascades downstream of β2 integrins still remain to be fully elucidated. In this review, we focus mainly on inside-out and outside-in signaling of these two β2 integrin members expressed on neutrophils and describe differences between various neutrophil stimuli with respect to integrin activation, integrin ligand binding, and the pertinent differences between mouse and human studies. Last, we discuss how integrin signaling studies could be used to explore the therapeutic potential of targeting β2 integrins and the intracellular signaling cascade in neutrophils in several, among other, inflammatory conditions in which neutrophil activity should be dampened to mitigate disease.
Collapse
Affiliation(s)
- Panagiota Bouti
- Sanquin Research and Landsteiner Laboratory, Department of Blood Cell Research, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Steven D S Webbers
- Sanquin Research and Landsteiner Laboratory, Department of Blood Cell Research, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, Netherlands.,Department of Pediatric Immunology, Rheumatology and Infectious Disease, Amsterdam University Medical Center (AUMC), Emma Children's Hospital, University of Amsterdam, Amsterdam, Netherlands
| | - Susanna C Fagerholm
- Research Program of Molecular and Integrative Biosciences, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | - Ronen Alon
- Department of Immunology, The Weizmann Institute of Science, Rehovot, Israel
| | - Markus Moser
- Institute of Experimental Hematology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Hanke L Matlung
- Sanquin Research and Landsteiner Laboratory, Department of Blood Cell Research, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Taco W Kuijpers
- Sanquin Research and Landsteiner Laboratory, Department of Blood Cell Research, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, Netherlands.,Department of Pediatric Immunology, Rheumatology and Infectious Disease, Amsterdam University Medical Center (AUMC), Emma Children's Hospital, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
43
|
Understanding the role of integrins in breast cancer invasion, metastasis, angiogenesis, and drug resistance. Oncogene 2021; 40:1043-1063. [PMID: 33420366 DOI: 10.1038/s41388-020-01588-2] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 11/11/2020] [Accepted: 11/26/2020] [Indexed: 12/13/2022]
Abstract
Integrins are cell adhesion receptors, which are typically transmembrane glycoproteins that connect to the extracellular matrix (ECM). The function of integrins regulated by biochemical events within the cells. Understanding the mechanisms of cell growth by integrins is important in elucidating their effects on tumor progression. One of the major events in integrin signaling is integrin binding to extracellular ligands. Another event is distant signaling that gathers chemical signals from outside of the cell and transmit the signals upon cell adhesion to the inside of the cell. In normal breast tissue, integrins function as checkpoints to monitor effects on cell proliferation, while in cancer tissue these functions altered. The combination of tumor microenvironment and its associated components determines the cell fate. Hypoxia can increase the expression of several integrins. The exosomal integrins promote the growth of metastatic cells. Expression of certain integrins is associated with increased metastasis and decreased prognosis in cancers. In addition, integrin-binding proteins promote invasion and metastasis in breast cancer. Targeting specific integrins and integrin-binding proteins may provide new therapeutic approaches for breast cancer therapies. This review will examine the current knowledge of integrins' role in breast cancer.
Collapse
|
44
|
Su B, Wu J. Phosphorylation of RIAM Activates Its Adaptor Function in Mediating Integrin Signaling. JOURNAL OF CELLULAR SIGNALING 2021; 2:103-110. [PMID: 35128538 PMCID: PMC8813058 DOI: 10.33696/signaling.2.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Integrins are cellular receptors that regulate cell adhesion and many other cellular functions. Integrins can be activated via an "inside-out pathway" that is promoted by RAP1 GTPase. RAP1-GTP-Interacting Adaptor Molecular (RIAM) mediates integrin activation by linking RAP1 GTPase to talin, an integrin activator. RIAM's function in integrin signaling is tightly regulated. In this commentary, we review recent studies of the molecular mechanisms underlying RIAM autoinhibition via both intramolecular interaction and oligomer assembly, and the phosphorylation-dependent activation of RIAM.
Collapse
Affiliation(s)
| | - Jinhua Wu
- Correspondence should be addressed to Jinhua Wu;
| |
Collapse
|
45
|
Deak D, Gorcea-Andronic N, Sas V, Teodorescu P, Constantinescu C, Iluta S, Pasca S, Hotea I, Turcas C, Moisoiu V, Zimta AA, Galdean S, Steinheber J, Rus I, Rauch S, Richlitzki C, Munteanu R, Jurj A, Petrushev B, Selicean C, Marian M, Soritau O, Andries A, Roman A, Dima D, Tanase A, Sigurjonsson O, Tomuleasa C. A narrative review of central nervous system involvement in acute leukemias. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:68. [PMID: 33553361 PMCID: PMC7859772 DOI: 10.21037/atm-20-3140] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Acute leukemias (both myeloid and lymphoblastic) are a group of diseases for which each year more successful therapies are implemented. However, in a subset of cases the overall survival (OS) is still exceptionally low due to the infiltration of leukemic cells in the central nervous system (CNS) and the subsequent formation of brain tumors. The CNS involvement is more common in acute lymphocytic leukemia (ALL), than in adult acute myeloid leukemia (AML), although the rates for the second case might be underestimated. The main reasons for CNS invasion are related to the expression of specific adhesion molecules (VLA-4, ICAM-1, VCAM, L-selectin, PECAM-1, CD18, LFA-1, CD58, CD44, CXCL12) by a subpopulation of leukemic cells, called “sticky cells” which have the ability to interact and adhere to endothelial cells. Moreover, the microenvironment becomes hypoxic and together with secretion of VEGF-A by ALL or AML cells the permeability of vasculature in the bone marrow increases, coupled with the disruption of blood brain barrier. There is a single subpopulation of leukemia cells, called leukemia stem cells (LSCs) that is able to resist in the new microenvironment due to its high adaptability. The LCSs enter into the arachnoid, migrate, and intensively proliferate in cerebrospinal fluid (CSF) and consequently infiltrate perivascular spaces and brain parenchyma. Moreover, the CNS is an immune privileged site that also protects leukemic cells from chemotherapy. CD56/NCAM is the most important surface molecule often overexpressed by leukemic stem cells that offers them the ability to infiltrate in the CNS. Although asymptomatic or with unspecific symptoms, CNS leukemia should be assessed in both AML/ALL patients, through a combination of flow cytometry and cytological analysis of CSF. Intrathecal therapy (ITT) is a preventive measure for CNS involvement in AML and ALL, still much research is needed in finding the appropriate target that would dramatically lower CNS involvement in acute leukemia.
Collapse
Affiliation(s)
- Dalma Deak
- Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj-Napoca, Romania.,Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Nicolae Gorcea-Andronic
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Valentina Sas
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania.,Department of Pediatrics, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Patric Teodorescu
- Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj-Napoca, Romania.,Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Catalin Constantinescu
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania.,Intensive Care Unit, Ion Chiricuta Clinical Cancer Center, Cluj-Napoca, Romania
| | - Sabina Iluta
- Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj-Napoca, Romania.,Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Sergiu Pasca
- Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj-Napoca, Romania.,Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Ionut Hotea
- Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj-Napoca, Romania.,Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Cristina Turcas
- Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj-Napoca, Romania.,Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Vlad Moisoiu
- Department of Neurosurgery, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Alina-Andreea Zimta
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Simona Galdean
- Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj-Napoca, Romania
| | - Jakob Steinheber
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Ioana Rus
- Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj-Napoca, Romania
| | - Sebastian Rauch
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Cedric Richlitzki
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Raluca Munteanu
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Ancuta Jurj
- Research Center for Functional Genomics and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Bobe Petrushev
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Cristina Selicean
- Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj-Napoca, Romania
| | - Mirela Marian
- Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj-Napoca, Romania
| | - Olga Soritau
- Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj-Napoca, Romania
| | - Alexandra Andries
- Department of Radiology, Ion Chiricuta Clinical Cancer Center, Cluj-Napoca, Romania
| | - Andrei Roman
- Department of Radiology, Ion Chiricuta Clinical Cancer Center, Cluj-Napoca, Romania.,Department of Radiology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Delia Dima
- Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj-Napoca, Romania
| | - Alina Tanase
- Department of Stem Cell Transplantation, Fundeni Clinical Institute, Bucharest, Romania
| | | | - Ciprian Tomuleasa
- Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj-Napoca, Romania.,Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania.,Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| |
Collapse
|
46
|
Koudelková L, Brábek J, Rosel D. Src kinase: Key effector in mechanosignalling. Int J Biochem Cell Biol 2020; 131:105908. [PMID: 33359015 DOI: 10.1016/j.biocel.2020.105908] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 12/15/2020] [Accepted: 12/15/2020] [Indexed: 02/07/2023]
Abstract
Cells have developed a unique set of molecular mechanisms that allows them to probe mechanical properties of the surrounding environment. These systems are based on deformable primary mechanosensors coupled to tension transmitting proteins and enzymes generating biochemical signals. This modular setup enables to transform a mechanical load into more versatile biochemical information. Src kinase appears to be one of the central components of the mechanotransduction network mediating force-induced signalling across multiple cellular contexts. In tight cooperation with primary sensors and the cytoskeleton, Src functions as an effector molecule necessary for transformation of mechanical stimuli into biochemical outputs executing cellular response and adaptation to mechanical cues.
Collapse
Affiliation(s)
- Lenka Koudelková
- Department of Cell Biology, Charles University, 12800, Prague, Czech Republic; Biotechnology and Biomedicine Centre of the Academy of Sciences and Charles University (BIOCEV), 25250, Vestec, Czech Republic
| | - Jan Brábek
- Department of Cell Biology, Charles University, 12800, Prague, Czech Republic; Biotechnology and Biomedicine Centre of the Academy of Sciences and Charles University (BIOCEV), 25250, Vestec, Czech Republic
| | - Daniel Rosel
- Department of Cell Biology, Charles University, 12800, Prague, Czech Republic; Biotechnology and Biomedicine Centre of the Academy of Sciences and Charles University (BIOCEV), 25250, Vestec, Czech Republic.
| |
Collapse
|
47
|
Dolinschek R, Hingerl J, Benge A, Zafiu C, Schüren E, Ehmoser EK, Lössner D, Reuning U. Constitutive activation of integrin αvβ3 contributes to anoikis resistance of ovarian cancer cells. Mol Oncol 2020; 15:503-522. [PMID: 33155399 PMCID: PMC7858284 DOI: 10.1002/1878-0261.12845] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 09/30/2020] [Accepted: 10/21/2020] [Indexed: 02/01/2023] Open
Abstract
Epithelial ovarian cancer involves the shedding of single tumor cells or spheroids from the primary tumor into ascites, followed by their survival, and transit to the sites of metastatic colonization within the peritoneal cavity. During their flotation, anchorage-dependent epithelial-type tumor cells gain anoikis resistance, implicating integrins, including αvß3. In this study, we explored anoikis escape, cisplatin resistance, and prosurvival signaling as a function of the αvß3 transmembrane conformational activation state in cells suspended in ascites. A high-affinity and constitutively signaling-competent αvß3 variant, which harbored unclasped transmembrane domains, was found to confer delayed anoikis onset, enhanced cisplatin resistance, and reduced cell proliferation in ascites or 3D-hydrogels, involving p27kip upregulation. Moreover, it promoted EGF-R expression and activation, prosurvival signaling, implicating FAK, src, and PKB/Akt. This led to the induction of the anti-apoptotic factors Bcl-2 and survivin suppressing caspase activation, compared to a signaling-incapable αvß3 variant displaying firmly associated transmembrane domains. Dissecting the mechanistic players for αvß3-dependent survival and peritoneal metastasis of ascitic ovarian cancer spheroids is of paramount importance to target their anchorage independence by reversing anoikis resistance and blocking αvß3-triggered prosurvival signaling.
Collapse
Affiliation(s)
- Romana Dolinschek
- Department for Obstetrics & Gynecology, Clinical Research Unit, Technische Universität München, Germany
| | - Julia Hingerl
- Department for Obstetrics & Gynecology, Clinical Research Unit, Technische Universität München, Germany
| | - Anke Benge
- Department for Obstetrics & Gynecology, Clinical Research Unit, Technische Universität München, Germany
| | - Christian Zafiu
- Department of Water, Atmosphere, and Environment, University for Natural Resources and Applied Life Sciences (BOKU), Vienna, Austria
| | - Elisabeth Schüren
- Department for Obstetrics & Gynecology, Clinical Research Unit, Technische Universität München, Germany
| | - Eva-Kathrin Ehmoser
- Department for Nanobiotechnology, Institute for Synthetic Bioarchitectures, University for Natural Resources and Applied Life Sciences (BOKU), Vienna, Austria
| | - Daniela Lössner
- Faculties of Engineering and Medicine, Nursing & Health Sciences, Monash University, Melbourne, Vic., Australia
| | - Ute Reuning
- Department for Obstetrics & Gynecology, Clinical Research Unit, Technische Universität München, Germany
| |
Collapse
|
48
|
De Kock L, Freson K. The (Patho)Biology of SRC Kinase in Platelets and Megakaryocytes. ACTA ACUST UNITED AC 2020; 56:medicina56120633. [PMID: 33255186 PMCID: PMC7759910 DOI: 10.3390/medicina56120633] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 11/17/2020] [Accepted: 11/21/2020] [Indexed: 02/07/2023]
Abstract
Proto-oncogene tyrosine-protein kinase SRC (SRC), as other members of the SRC family kinases (SFK), plays an important role in regulating signal transduction by different cell surface receptors after changes in the cellular environment. Here, we reviewed the role of SRC in platelets and megakaryocytes (MK). In platelets, inactive closed SRC is coupled to the β subunit of integrin αIIbβ3 while upon fibrinogen binding during platelet activation, αIIbβ3-mediated outside-in signaling is initiated by activation of SRC. Active open SRC now further stimulates many downstream effectors via tyrosine phosphorylation of enzymes, adaptors, and especially cytoskeletal components. Functional platelet studies using SRC knockout mice or broad spectrum SFK inhibitors pointed out that SRC mediates their spreading on fibrinogen. On the other hand, an activating pathological SRC missense variant E527K in humans that causes bleeding inhibits collagen-induced platelet activation while stimulating platelet spreading. The role of SRC in megakaryopoiesis is much less studied. SRC knockout mice have a normal platelet count though studies with SFK inhibitors point out that SRC could interfere with MK polyploidization and proplatelet formation but these inhibitors are not specific. Patients with the SRC E527K variant have thrombocytopenia due to hyperactive SRC that inhibits proplatelet formation after increased spreading of MK on fibrinogen and enhanced formation of podosomes. Studies in humans have contributed significantly to our understanding of SRC signaling in platelets and MK.
Collapse
|
49
|
Liu L, He F, Yu Y, Wang Y. Application of FRET Biosensors in Mechanobiology and Mechanopharmacological Screening. Front Bioeng Biotechnol 2020; 8:595497. [PMID: 33240867 PMCID: PMC7680962 DOI: 10.3389/fbioe.2020.595497] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Accepted: 10/19/2020] [Indexed: 12/15/2022] Open
Abstract
Extensive studies have shown that cells can sense and modulate the biomechanical properties of the ECM within their resident microenvironment. Thus, targeting the mechanotransduction signaling pathways provides a promising way for disease intervention. However, how cells perceive these mechanical cues of the microenvironment and transduce them into biochemical signals remains to be answered. Förster or fluorescence resonance energy transfer (FRET) based biosensors are a powerful tool that can be used in live-cell mechanotransduction imaging and mechanopharmacological drug screening. In this review, we will first introduce FRET principle and FRET biosensors, and then, recent advances on the integration of FRET biosensors and mechanobiology in normal and pathophysiological conditions will be discussed. Furthermore, we will summarize the current applications and limitations of FRET biosensors in high-throughput drug screening and the future improvement of FRET biosensors. In summary, FRET biosensors have provided a powerful tool for mechanobiology studies to advance our understanding of how cells and matrices interact, and the mechanopharmacological screening for disease intervention.
Collapse
Affiliation(s)
| | | | | | - Yingxiao Wang
- Department of Bioengineering, Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA, United States
| |
Collapse
|
50
|
Masuzaki R, Ray KC, Roland J, Zent R, Lee YA, Karp SJ. Integrin β1 Establishes Liver Microstructure and Modulates Transforming Growth Factor β during Liver Development and Regeneration. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 191:309-319. [PMID: 33159885 DOI: 10.1016/j.ajpath.2020.10.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 10/07/2020] [Accepted: 10/13/2020] [Indexed: 01/16/2023]
Abstract
A unique and complex microstructure underlies the diverse functions of the liver. Breakdown of this organization, as occurs in fibrosis and cirrhosis, impairs liver function and leads to disease. The role of integrin β1 was examined both in establishing liver microstructure and recreating it after injury. Embryonic deletion of integrin β1 in the liver disrupts the normal development of hepatocyte polarity, specification of cell-cell junctions, and canalicular formation. This in turn leads to the expression of transforming growth factor β (TGF-β) and widespread fibrosis. Targeted deletion of integrin β1 in adult hepatocytes prevents recreation of normal hepatocyte architecture after liver injury, with resultant fibrosis. In vitro, integrin β1 is essential for canalicular formation and is needed to prevent stellate cell activation by modulating TGF-β. Taken together, these findings identify integrin β1 as a key determinant of liver architecture with a critical role as a regulator of TGF-β secretion. These results suggest that disrupting the hepatocyte-extracellular matrix interaction is sufficient to drive fibrosis.
Collapse
Affiliation(s)
- Ryota Masuzaki
- Section of Surgical Sciences, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Kevin C Ray
- Section of Surgical Sciences, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Joseph Roland
- Section of Surgical Sciences, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Roy Zent
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee; Department of Medicine, Nashville Veterans Affairs Hospital, Nashville, Tennessee
| | - Youngmin A Lee
- Section of Surgical Sciences, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Seth J Karp
- Section of Surgical Sciences, Vanderbilt University School of Medicine, Nashville, Tennessee.
| |
Collapse
|