1
|
Murthi P, Overton E, Brennecke SP, Keogh RJ. Happy feet: the key roles of podosomes and invadopodia in trophoblast invasion at the maternal-fetal interface. Front Endocrinol (Lausanne) 2025; 16:1576732. [PMID: 40519515 PMCID: PMC12163045 DOI: 10.3389/fendo.2025.1576732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Accepted: 05/06/2025] [Indexed: 06/18/2025] Open
Abstract
Cells move by forming specialized projections or invasive feet known as podosomes in normal invasive cells and invadopodia in transformed and cancer cells. An understanding of invasive projections of trophoblasts at the maternal-fetal interface and their formation is important for developing novel therapies for pregnancy complications where invasion is abnormal, in instances where over- or under-invasion of cells manifests as serious pregnancy pathologies such as accreta or preeclampsia. Podosomes and invadopodia have distinctive morphological and molecular features that are used to distinguish them from each other. Despite this, there is still debate and uncertainty around how to definitively classify them. Analyses of novel models of cell invasion have demonstrated the existence of hybrid structures that are neither true podosomes nor invadopodia but which display features of both. This raises the question as to whether the classification of invasive structures needs redefining.
Collapse
Affiliation(s)
- Padma Murthi
- Department of Maternal Fetal Medicine, Pregnancy Research Centre, Royal Women’s Hospital, Parkville, Australia
- Department of Obstetrics, Gynaecology and Newborn Health, University of Melbourne, Parkville, Australia
| | - Emily Overton
- Department of Maternal Fetal Medicine, Pregnancy Research Centre, Royal Women’s Hospital, Parkville, Australia
- Department of Obstetrics, Gynaecology and Newborn Health, University of Melbourne, Parkville, Australia
| | - Shaun P. Brennecke
- Department of Maternal Fetal Medicine, Pregnancy Research Centre, Royal Women’s Hospital, Parkville, Australia
- Department of Obstetrics, Gynaecology and Newborn Health, University of Melbourne, Parkville, Australia
| | - Rosemary J. Keogh
- Department of Maternal Fetal Medicine, Pregnancy Research Centre, Royal Women’s Hospital, Parkville, Australia
- Department of Obstetrics, Gynaecology and Newborn Health, University of Melbourne, Parkville, Australia
| |
Collapse
|
2
|
Noureddine M, Mikolajek H, Morgan NV, Denning C, Loughna S, Gehmlich K, Mohammed F. Structural and functional insights into α-actinin isoforms and their implications in cardiovascular disease. J Gen Physiol 2025; 157:e202413684. [PMID: 39918740 PMCID: PMC11804879 DOI: 10.1085/jgp.202413684] [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: 10/07/2024] [Revised: 12/11/2024] [Accepted: 01/13/2025] [Indexed: 02/09/2025] Open
Abstract
α-actinin (ACTN) is a pivotal member of the actin-binding protein family, crucial for the anchoring and organization of actin filaments within the cytoskeleton. Four isoforms of α-actinin exist: two non-muscle isoforms (ACTN1 and ACTN4) primarily associated with actin stress fibers and focal adhesions, and two muscle-specific isoforms (ACTN2 and ACTN3) localized to the Z-disk of the striated muscle. Although these isoforms share structural similarities, they exhibit distinct functional characteristics that reflect their specialized roles in various tissues. Genetic variants in α-actinin isoforms have been implicated in a range of pathologies, including cardiomyopathies, thrombocytopenia, and non-cardiovascular diseases, such as nephropathy. However, the precise impact of these genetic variants on the α-actinin structure and their contribution to disease pathogenesis remains poorly understood. This review provides a comprehensive overview of the structural and functional attributes of the four α-actinin isoforms, emphasizing their roles in actin crosslinking and sarcomere stabilization. Furthermore, we present detailed structural modeling of select ACTN1 and ACTN2 variants to elucidate mechanisms underlying disease pathogenesis, with a particular focus on macrothrombocytopenia and hypertrophic cardiomyopathy. By advancing our understanding of α-actinin's role in both normal cellular function and disease states, this review lays the groundwork for future research and the development of targeted therapeutic interventions.
Collapse
Affiliation(s)
- Maya Noureddine
- Department of Cardiovascular Sciences, School of Medical Sciences, College of Medicine and Health University of Birmingham, Birmingham, UK
| | - Halina Mikolajek
- Diamond Light Source Ltd., Harwell Science and Innovation Campus, Didcot, UK
| | - Neil V. Morgan
- Department of Cardiovascular Sciences, School of Medical Sciences, College of Medicine and Health University of Birmingham, Birmingham, UK
| | - Chris Denning
- Biodiscovery Institute, University of Nottingham, Nottingham, UK
| | - Siobhan Loughna
- School of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, UK
| | - Katja Gehmlich
- Department of Cardiovascular Sciences, School of Medical Sciences, College of Medicine and Health University of Birmingham, Birmingham, UK
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Oxford, UK
| | - Fiyaz Mohammed
- Department of Immunology and Immunotherapy, School of Infection, Inflammation and Immunology, College of Medicine and Health, University of Birmingham, Birmingham, UK
| |
Collapse
|
3
|
Carlos AJ, Yang D, Thomas DM, Huang S, Harter KI, Moellering RE. Family-Wide Photoproximity Profiling of Integrin Protein Social Networks in Cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.18.613588. [PMID: 39345550 PMCID: PMC11429684 DOI: 10.1101/2024.09.18.613588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Integrin family transmembrane receptors mediate dynamic interactions between cells and their extracellular microenvironment. The heterogeneous interaction partners of integrins directly regulate cell adhesion, motility, proliferation, and intracellular signaling. Despite the recognized importance of protein-protein interactions and the formation of signaling hubs around integrins, the ability to detect and quantify these dynamic binding partners with high spatial and temporal resolution remains challenging. Here, we developed an integrin-family-directed quantitative photoproximity protein interaction (PhotoPPI) profiling method to detect and quantify native integrin-centered protein social networks on live cells and tissues without the need for genetic manipulation, antibodies, or non-physiologic cell culture conditions. We drafted quantitative maps of integrin-centered protein social networks, highlighting conserved and unique binding partners between different cell types and cellular microenvironments. Comparison of integrin social networks in cancer cell lines of diverse tissue of origin and disease state identified specific AND-gate binding partners involved cell migration, microenvironmental interactions and proliferation that serve as markers of tumor cell metastatic state. Finally, we identified unique combinations - or barcodes - of integrin-proximal proteins on the surface of pre- and post-metastatic triple negative breast cancer (TNBC) cells whose expression strongly correlate with both positive and negative disease progression and outcomes in TNBC patients. Taken together, these data provide the first family-wide high-resolution maps of native protein interactors on live cells and identify dynamic integrin-centered social networks as potential AND-gate markers of cell identity, microenvironmental context and disease state.
Collapse
Affiliation(s)
- Anthony J. Carlos
- Department of Chemistry, The University of Chicago. Chicago, IL, 60637, USA
| | - Dongbo Yang
- Department of Chemistry, The University of Chicago. Chicago, IL, 60637, USA
| | - Deborah M. Thomas
- Department of Chemistry, The University of Chicago. Chicago, IL, 60637, USA
| | - Shuyuan Huang
- Department of Chemistry, The University of Chicago. Chicago, IL, 60637, USA
| | - Keira I. Harter
- Department of Chemistry, The University of Chicago. Chicago, IL, 60637, USA
| | | |
Collapse
|
4
|
Cui L, Lu Y, Zheng J, Guo B, Zhao X. ACTN1 promotes HNSCC tumorigenesis and cisplatin resistance by enhancing MYH9-dependent degradation of GSK-3β and integrin β1-mediated phosphorylation of FAK. J Exp Clin Cancer Res 2023; 42:335. [PMID: 38057867 DOI: 10.1186/s13046-023-02904-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 11/15/2023] [Indexed: 12/08/2023] Open
Abstract
BACKGROUND Head and neck squamous cell carcinoma (HNSCC) is one of the most common malignant tumors globally. Understanding the molecular basis of tumor progression and drug resistance can offer innovative strategies to enhance clinical outcomes for HNSCC patients. METHODS The cytoskeletal remodeling genes associated with cisplatin resistance were screened using a PCR array. The role of alpha-actinin 1 (ACTN1) in modulating cisplatin resistance and tumorigenesis in HNSCC was evaluated both in vitro and in vivo. Co-immunoprecipitation (Co-IP), IP-mass spectrometry (MS), western blotting, dual-luciferase assay, and bioinformatics analysis were performed to elucidate the underlying mechanisms involved. RESULTS Our study identifies ACTN1 as a crucial contributor to cisplatin resistance and tumorigenesis in HNSCC, as evidenced across cellular, animal, and patient-derived xenograft models. From a clinical perspective, overexpression of ACTN1 significantly correlates with a suboptimal response to neoadjuvant chemotherapy and reduced overall survival in HNSCC patients. Mechanistically, ACTN1 predominantly activates β-catenin-mediated signaling by promoting the interaction between myosin heavy chain 9 (MYH9) and GSK-3β, leading to the ubiquitin-dependent degradation of GSK-3β. ACTN1 also interacts with integrin β1, subsequently activating the FAK/PI3K/AKT pathway, providing an additional avenue for the activation of β-catenin signaling. Our study also unveils that the β-catenin/c-Myc axis transcriptionally regulates ACTN1, thereby creating a positive feedback loop promoting HNSCC tumorigenesis and drug resistance. CONCLUSIONS These insights underscore the novel mechanisms that highlight ACTN1's pivotal role in driving HNSCC progression and resistance to chemotherapy, suggesting ACTN1 as a promising therapeutic target in HNSCC management.
Collapse
Affiliation(s)
- Li Cui
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, Guangdong, China.
| | - Ye Lu
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, Guangdong, China
| | - Jiarong Zheng
- Department of Dentistry, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Bing Guo
- Department of Dentistry, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Xinyuan Zhao
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, Guangdong, China.
| |
Collapse
|
5
|
Jain K, Lim KYE, Sheetz MP, Kanchanawong P, Changede R. Intrinsic self-organization of integrin nanoclusters within focal adhesions is required for cellular mechanotransduction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.20.567975. [PMID: 38045378 PMCID: PMC10690202 DOI: 10.1101/2023.11.20.567975] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Upon interaction with the extracellular matrix, the integrin receptors form nanoclusters as a first biochemical response to ligand binding. Here, we uncover a critical biodesign principle where these nanoclusters are spatially self-organized, facilitating effective mechanotransduction. Mouse Embryonic Fibroblasts (MEFs) with integrin β3 nanoclusters organized themselves with an intercluster distance of ∼550 nm on uniformly coated fibronectin substrates, leading to larger focal adhesions. We determined that this spatial organization was driven by cell-intrinsic factors since there was no pre-existing pattern on the substrates. Altering this spatial organization using cyclo-RGD functionalized Titanium nanodiscs (of 100 nm, corroborating to the integrin nanocluster size) spaced at intervals of 300 nm (almost half), 600 nm (normal) or 1000 nm (almost double) resulted in abrogation in mechanotransduction, indicating that a new parameter i.e., an optimal intercluster distance is necessary for downstream function. Overexpression of α-actinin, which induces a kink in the integrin tail, disrupted the establishment of the optimal intercluster distance, while simultaneous co-overexpression of talin head with α-actinin rescued it, indicating a concentration-dependent competition, and that cytoplasmic activation of integrin by talin head is required for the optimal intercluster organization. Additionally, talin head-mediated recruitment of FHOD1 that facilitates local actin polymerization at nanoclusters, and actomyosin contractility were also crucial for establishing the optimal intercluster distance and a robust mechanotransduction response. These findings demonstrate that cell-intrinsic machinery plays a vital role in organizing integrin receptor nanoclusters within focal adhesions, encoding essential information for downstream mechanotransduction signalling.
Collapse
|
6
|
Putra VDL, Kilian KA, Knothe Tate ML. Biomechanical, biophysical and biochemical modulators of cytoskeletal remodelling and emergent stem cell lineage commitment. Commun Biol 2023; 6:75. [PMID: 36658332 PMCID: PMC9852586 DOI: 10.1038/s42003-022-04320-w] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 11/30/2022] [Indexed: 01/20/2023] Open
Abstract
Across complex, multi-time and -length scale biological systems, redundancy confers robustness and resilience, enabling adaptation and increasing survival under dynamic environmental conditions; this review addresses ubiquitous effects of cytoskeletal remodelling, triggered by biomechanical, biophysical and biochemical cues, on stem cell mechanoadaptation and emergent lineage commitment. The cytoskeleton provides an adaptive structural scaffold to the cell, regulating the emergence of stem cell structure-function relationships during tissue neogenesis, both in prenatal development as well as postnatal healing. Identification and mapping of the mechanical cues conducive to cytoskeletal remodelling and cell adaptation may help to establish environmental contexts that can be used prospectively as translational design specifications to target tissue neogenesis for regenerative medicine. In this review, we summarize findings on cytoskeletal remodelling in the context of tissue neogenesis during early development and postnatal healing, and its relevance in guiding lineage commitment for targeted tissue regeneration. We highlight how cytoskeleton-targeting chemical agents modulate stem cell differentiation and govern responses to mechanical cues in stem cells' emerging form and function. We further review methods for spatiotemporal visualization and measurement of cytoskeletal remodelling, as well as its effects on the mechanical properties of cells, as a function of adaptation. Research in these areas may facilitate translation of stem cells' own healing potential and improve the design of materials, therapies, and devices for regenerative medicine.
Collapse
Affiliation(s)
- Vina D L Putra
- School of Chemistry and School of Materials Science & Engineering, University of New South Wales, Sydney, NSW, Australia
| | - Kristopher A Kilian
- School of Chemistry and School of Materials Science & Engineering, University of New South Wales, Sydney, NSW, Australia.
| | - Melissa L Knothe Tate
- Blue Mountains World Interdisciplinary Innovation Institute (bmwi³), Blue Mountains, NSW, Australia.
| |
Collapse
|
7
|
Tseng CC, Zheng RH, Lin TW, Chou CC, Shih YC, Liang SW, Lee HH. α-Actinin-4 recruits Shp2 into focal adhesions to potentiate ROCK2 activation in podocytes. Life Sci Alliance 2022; 5:5/11/e202201557. [PMID: 36096674 PMCID: PMC9468603 DOI: 10.26508/lsa.202201557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 08/20/2022] [Accepted: 08/22/2022] [Indexed: 11/24/2022] Open
Abstract
α-Actinin-4 is crucial in the regulation of Shp2 FA targeting to enhance ROCK2-mediated actomyosin contractility and thereby strengthening cell adhesion and cytoskeletal architecture in podocytes. Cell–matrix adhesions are mainly provided by integrin-mediated focal adhesions (FAs). We previously found that Shp2 is essential for FA maturation by promoting ROCK2 activation at FAs. In this study, we further delineated the role of α-actinin-4 in the FA recruitment and activation of Shp2. We used the conditional immortalized mouse podocytes to examine the role of α-actinin-4 in the regulation of Shp2 and ROCK2 signaling. After the induction of podocyte differentiation, Shp2 and ROCK2 were strongly activated, concomitant with the formation of matured FAs, stress fibers, and interdigitating intracellular junctions in a ROCK-dependent manner. Gene knockout of α-actinin-4 abolished the Shp2 activation and subsequently reduced matured FAs in podocytes. We also demonstrated that gene knockout of ROCK2 impaired the generation of contractility and interdigitating intercellular junctions. Our results reveal the role of α-actinin-4 in the recruitment of Shp2 at FAs to potentiate ROCK2 activation for the maintenance of cellular contractility and cytoskeletal architecture in the cultured podocytes.
Collapse
Affiliation(s)
- Chien-Chun Tseng
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Ru-Hsuan Zheng
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Ting-Wei Lin
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chih-Chiang Chou
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yu-Chia Shih
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Shao-Wei Liang
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Hsiao-Hui Lee
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan .,Center for Intelligent Drug Systems and Smart Bio-Devices (IDS2B), National Yang Ming Chiao Tung University, Taipei, Taiwan
| |
Collapse
|
8
|
Wen L, Lyu Q, Ley K, Goult BT. Structural Basis of β2 Integrin Inside—Out Activation. Cells 2022; 11:cells11193039. [PMID: 36231001 PMCID: PMC9564206 DOI: 10.3390/cells11193039] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 09/19/2022] [Accepted: 09/27/2022] [Indexed: 11/16/2022] Open
Abstract
β2 integrins are expressed on all leukocytes. Precise regulation of the β2 integrin is critical for leukocyte adhesion and trafficking. In neutrophils, β2 integrins participate in slow rolling. When activated by inside–out signaling, fully activated β2 integrins mediate rapid leukocyte arrest and adhesion. The two activation pathways, starting with selectin ligand engagement and chemokine receptor ligation, respectively, converge on phosphoinositide 3-kinase, talin-1, kindlin-3 and Rap1. Here, we focus on recent structural insights into autoinhibited talin-1 and autoinhibited trimeric kindlin-3. When activated, both talin-1 and kindlin-3 can bind the β2 cytoplasmic tail at separate but adjacent sites. We discuss possible pathways for talin-1 and kindlin-3 activation, recruitment to the plasma membrane, and their role in integrin activation. We propose new models of the final steps of integrin activation involving the complex of talin-1, kindlin-3, integrin and the plasma membrane.
Collapse
Affiliation(s)
- Lai Wen
- Department of Pharmacology, Center for Molecular and Cellular Signaling in the Cardiovascular System, Reno School of Medicine, University of Nevada, Reno, NV 89577, USA
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Qingkang Lyu
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
- Immunology Center of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Klaus Ley
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
- Immunology Center of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Benjamin T. Goult
- School of Biosciences, University of Kent, Canterbury CT2 7NJ, UK
- Correspondence: ; Tel.: +44-(0)1227-816-142
| |
Collapse
|
9
|
Integrin Regulators in Neutrophils. Cells 2022; 11:cells11132025. [PMID: 35805108 PMCID: PMC9266208 DOI: 10.3390/cells11132025] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 06/17/2022] [Accepted: 06/22/2022] [Indexed: 02/01/2023] Open
Abstract
Neutrophils are the most abundant leukocytes in humans and are critical for innate immunity and inflammation. Integrins are critical for neutrophil functions, especially for their recruitment to sites of inflammation or infections. Integrin conformational changes during activation have been heavily investigated but are still not fully understood. Many regulators, such as talin, Rap1-interacting adaptor molecule (RIAM), Rap1, and kindlin, are critical for integrin activation and might be potential targets for integrin-regulating drugs in treating inflammatory diseases. In this review, we outline integrin activation regulators in neutrophils with a focus on the above critical regulators, as well as newly discovered modulators that are involved in integrin activation.
Collapse
|
10
|
O’Sullivan LR, Cahill MR, Young PW. The Importance of Alpha-Actinin Proteins in Platelet Formation and Function, and Their Causative Role in Congenital Macrothrombocytopenia. Int J Mol Sci 2021; 22:9363. [PMID: 34502272 PMCID: PMC8431150 DOI: 10.3390/ijms22179363] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 08/25/2021] [Accepted: 08/26/2021] [Indexed: 12/04/2022] Open
Abstract
The actin cytoskeleton plays a central role in platelet formation and function. Alpha-actinins (actinins) are actin filament crosslinking proteins that are prominently expressed in platelets and have been studied in relation to their role in platelet activation since the 1970s. However, within the past decade, several groups have described mutations in ACTN1/actinin-1 that cause congenital macrothrombocytopenia (CMTP)-accounting for approximately 5% of all cases of this condition. These findings are suggestive of potentially novel functions for actinins in platelet formation from megakaryocytes in the bone marrow and/or platelet maturation in circulation. Here, we review some recent insights into the well-known functions of actinins in platelet activation before considering possible roles for actinins in platelet formation that could explain their association with CMTP. We describe what is known about the consequences of CMTP-linked mutations on actinin-1 function at a molecular and cellular level and speculate how these changes might lead to the alterations in platelet count and morphology observed in CMTP patients. Finally, we outline some unanswered questions in this area and how they might be addressed in future studies.
Collapse
Affiliation(s)
- Leanne R. O’Sullivan
- School of Biochemistry & Cell Biology, University College Cork, T12 XF62 Cork, Ireland;
| | - Mary R. Cahill
- Department of Haematology and CancerResearch@UCC, Cork University Hospital, University College Cork, T12 XF62 Cork, Ireland;
| | - Paul W. Young
- School of Biochemistry & Cell Biology, University College Cork, T12 XF62 Cork, Ireland;
| |
Collapse
|
11
|
Dias C, Nita E, Faktor J, Tynan AC, Hernychova L, Vojtesek B, Nylandsted J, Hupp TR, Kunath T, Ball KL. CHIP-dependent regulation of the actin cytoskeleton is linked to neuronal cell membrane integrity. iScience 2021; 24:102878. [PMID: 34401662 PMCID: PMC8350547 DOI: 10.1016/j.isci.2021.102878] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 04/13/2021] [Accepted: 07/15/2021] [Indexed: 12/12/2022] Open
Abstract
CHIP is an E3-ubiquitin ligase that contributes to healthy aging and has been characterized as neuroprotective. To elucidate dominant CHIP-dependent changes in protein steady-state levels in a patient-derived human neuronal model, CHIP function was ablated using gene-editing and an unbiased proteomic analysis conducted to compare knock-out and wild-type isogenic induced pluripotent stem cell (iPSC)-derived cortical neurons. Rather than a broad effect on protein homeostasis, loss of CHIP function impacted on a focused cohort of proteins from actin cytoskeleton signaling and membrane integrity networks. In support of the proteomics, CHIP knockout cells had enhanced sensitivity to induced membrane damage. We conclude that the major readout of CHIP function in cortical neurons derived from iPSC of a patient with elevate α-synuclein, Parkinson's disease and dementia, is the modulation of substrates involved in maintaining cellular "health". Thus, regulation of the actin cytoskeletal and membrane integrity likely contributes to the neuroprotective function(s) of CHIP.
Collapse
Affiliation(s)
- Catarina Dias
- Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XU, UK
- Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, The University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Erisa Nita
- Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Jakub Faktor
- Research Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, 656 53 Brno, Czech Republic
- University of Gdansk, International Centre for Cancer Vaccine Science, 80-822 Gdansk, Poland
| | - Ailish C. Tynan
- Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Lenka Hernychova
- Research Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, 656 53 Brno, Czech Republic
| | - Borivoj Vojtesek
- Research Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, 656 53 Brno, Czech Republic
| | - Jesper Nylandsted
- Membrane Integrity Group, Danish Cancer Society Research Center, Strandboulevarden 49, 2100, Copenhagen, Denmark
| | - Ted R. Hupp
- Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XU, UK
- University of Gdansk, International Centre for Cancer Vaccine Science, 80-822 Gdansk, Poland
| | - Tilo Kunath
- Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, The University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Kathryn L. Ball
- Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XU, UK
| |
Collapse
|
12
|
Cardin GB, Bernard M, Rodier F, Christopoulos A. DCBLD1 is associated with the integrin signaling pathway and has prognostic value in non-small cell lung and invasive breast carcinoma. Sci Rep 2021; 11:12753. [PMID: 34140574 PMCID: PMC8211811 DOI: 10.1038/s41598-021-92090-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 06/02/2021] [Indexed: 01/27/2023] Open
Abstract
Germline single nucleotide polymorphisms in the promoter region of the DCBLD1 gene are associated with non-smoking cases of both non-small cell lung carcinoma (NSCLC) and human papillomavirus-negative head and neck cancer. However the clinical relevance and function of DCBLD1 remain unclear. This multicenter retrospective study was designed to evaluate the prognostic value and function of DCBLD1 in the four main solid cancers: NSCLC, invasive breast carcinoma, colorectal adenocarcinoma and prostate adenocarcinoma. We included the following cohorts: GSE81089 NSCLC, METABRIC invasive breast carcinoma, GSE14333 colorectal adenocarcinoma, GSE70770 prostate adenocarcinoma and The Cancer Genome Atlas (TCGA) Firehose Legacy cohorts of all four cancers. DCBLD1 gene expression was associated with a worse overall survival in multivariate analyses for both NSCLC cohorts (TCGA: P = 0.03 and GSE81089: P = 0.04) and both invasive breast carcinoma cohorts (TCGA: P = 0.02 and METABRIC: P < 0.001). Patients with high DCBLD1 expression showed an upregulation of the integrin signaling pathway in comparison to those with low DCBLD1 expression in the TCGA NSCLC cohort (FDR = 5.16 × 10-14) and TCGA invasive breast carcinoma cohort (FDR = 1.94 × 10-05).
Collapse
Affiliation(s)
- Guillaume B Cardin
- Centre de recherche du Centre hospitalier de l'université de Montréal, Montreal, QC, Canada.,Institut du cancer de Montréal, 900 Saint-Denis, Montreal, QC, H2X 0A9, Canada
| | - Monique Bernard
- Centre de recherche du Centre hospitalier de l'université de Montréal, Montreal, QC, Canada.,Institut du cancer de Montréal, 900 Saint-Denis, Montreal, QC, H2X 0A9, Canada
| | - Francis Rodier
- Centre de recherche du Centre hospitalier de l'université de Montréal, Montreal, QC, Canada.,Institut du cancer de Montréal, 900 Saint-Denis, Montreal, QC, H2X 0A9, Canada.,Département de radiologie, radio-oncologie et médecine nucléaire, Université de Montréal, Montreal, QC, Canada
| | - Apostolos Christopoulos
- Centre de recherche du Centre hospitalier de l'université de Montréal, Montreal, QC, Canada. .,Institut du cancer de Montréal, 900 Saint-Denis, Montreal, QC, H2X 0A9, Canada. .,Otolaryngology-Head and Neck Surgery Service, Centre hospitalier de l'Université de Montréal, Montreal, QC, Canada.
| |
Collapse
|
13
|
Wubshet NH, Bashirzadeh Y, Liu AP. Fascin-induced actin protrusions are suppressed by dendritic networks in giant unilamellar vesicles. Mol Biol Cell 2021; 32:1634-1640. [PMID: 34133215 PMCID: PMC8684724 DOI: 10.1091/mbc.e21-02-0080] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
The interactions between actin networks and cell membrane are immensely important for eukaryotic cell functions including cell shape changes, motility, polarity establishment, and adhesion. Actin-binding proteins are known to compete and cooperate using a finite amount of actin monomers to form distinct actin networks. How actin-bundling protein fascin and actin-branching protein Arp2/3 complex compete to remodel membranes is not entirely clear. To investigate fascin- and Arp2/3-mediated actin network remodeling, we applied a reconstitution approach encapsulating bundled and dendritic actin networks inside giant unilamellar vesicles (GUVs). Independently reconstituted, membrane-bound Arp2/3 nucleation forms an actin cortex in GUVs, whereas fascin mediates formation of actin bundles that protrude out of GUVs. Coencapsulating both fascin and Arp2/3 complex leads to polarized dendritic aggregates and significantly reduces membrane protrusions, irrespective of whether the dendritic network is membrane bound or not. However, reducing Arp2/3 complex while increasing fascin restores membrane protrusion. Such changes in network assembly and the subsequent interplay with membrane can be attributed to competition between fascin and Arp2/3 complex to utilize a finite pool of actin.
Collapse
Affiliation(s)
- Nadab H Wubshet
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, 48109
| | - Yashar Bashirzadeh
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, 48109
| | - Allen P Liu
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, 48109.,Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109.,Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, MI, 48109.,Department of Biophysics, University of Michigan, Ann Arbor, MI, 48109
| |
Collapse
|
14
|
Skripchenko A, Gelderman MP, Vostal JG. P38 mitogen activated protein kinase inhibitor improves platelet in vitro parameters and in vivo survival in a SCID mouse model of transfusion for platelets stored at cold or temperature cycled conditions for 14 days. PLoS One 2021; 16:e0250120. [PMID: 33974660 PMCID: PMC8112650 DOI: 10.1371/journal.pone.0250120] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 03/30/2021] [Indexed: 11/26/2022] Open
Abstract
Platelets for transfusion are stored at room temperature (20-24°C) up to 7 days but decline in biochemical and morphological parameters during storage and can support bacterial proliferation. This decline is reduced with p38MAPK inhibitor, VX-702. Storage of platelets in the cold (4-6°C) can reduce bacterial proliferation but platelets get activated and have reduced circulation when transfused. Thermocycling (cold storage with brief periodic warm ups) reduces some of the effects of cold storage. We evaluated in vitro properties and in vivo circulation in SCID mouse model of human platelet transfusion of platelets stored in cold or thermocycled for 14 days with and without VX-702. Apheresis platelet units (N = 15) were each aliquoted into five storage bags and stored under different conditions: room temperature; cold temperature; thermocycled temperature; cold temperature with VX-702; thermocycled temperature with VX-702. Platelet in vitro parameters were evaluated at 1, 7 and 14 days. On day 14, platelets were infused into SCID mice to assess their retention in circulation by flow cytometry. VX-702 reduced negative platelet parameters associated with cold and thermocycled storage such as an increase in expression of activation markers CD62, CD63 and of phosphatidylserine (marker of apoptosis measured by Annexin binding) and lowered the rise in lactate (marker of increase in anaerobic metabolism). However, VX-702 did not inhibit agonist-induced platelet aggregation indicating that it does not interfere with platelet hemostatic function. In vivo, VX-702 improved initial recovery and area under the curve in circulation of human platelets infused into a mouse model that has been previously validated against a human platelet infusion clinical trial. In conclusion, inhibition of p38MAPK during 14-days platelet storage in cold or thermocycling conditions improved in vitro platelet parameters and platelet circulation in the mouse model indicating that VX-702 may improve cell physiology and clinical performance of human platelets stored in cold conditions.
Collapse
Affiliation(s)
- Andrey Skripchenko
- Division of Blood Components and Devices, Laboratory of Cellular Hematology, Office of Blood Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Monique P. Gelderman
- Division of Blood Components and Devices, Laboratory of Cellular Hematology, Office of Blood Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Jaroslav G. Vostal
- Division of Blood Components and Devices, Laboratory of Cellular Hematology, Office of Blood Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, United States of America
| |
Collapse
|
15
|
Choi E, Kim D, Kang D, Yang GH, Jung B, Yeo M, Park MJ, An S, Lee K, Kim JS, Kim JC, Jeong W, Yoo HH, Jeon H. 3D-printed gelatin methacrylate (GelMA)/silanated silica scaffold assisted by two-stage cooling system for hard tissue regeneration. Regen Biomater 2021; 8:rbab001. [PMID: 33738115 PMCID: PMC7955716 DOI: 10.1093/rb/rbab001] [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: 09/23/2020] [Revised: 12/23/2020] [Accepted: 01/01/2021] [Indexed: 12/12/2022] Open
Abstract
Among many biomaterials, gelatin methacrylate (GelMA), a photocurable protein, has been widely used in 3D bioprinting process owing to its excellent cellular responses, biocompatibility and biodegradability. However, GelMA still shows a low processability due to the severe temperature dependence of viscosity. To overcome this obstacle, we propose a two-stage temperature control system to effectively control the viscosity of GelMA. To optimize the process conditions, we evaluated the temperature of the cooling system (jacket and stage). Using the established system, three GelMA scaffolds were fabricated in which different concentrations (0, 3 and 10 wt%) of silanated silica particles were embedded. To evaluate the performances of the prepared scaffolds suitable for hard tissue regeneration, we analyzed the physical (viscoelasticity, surface roughness, compressive modulus and wettability) and biological (human mesenchymal stem cells growth, western blotting and osteogenic differentiation) properties. Consequently, the composite scaffold with greater silica contents (10 wt%) showed enhanced physical and biological performances including mechanical strength, cell initial attachment, cell proliferation and osteogenic differentiation compared with those of the controls. Our results indicate that the GelMA/silanated silica composite scaffold can be potentially used for hard tissue regeneration.
Collapse
Affiliation(s)
- Eunjeong Choi
- Research Institute of Additive Manufacturing and Regenerative Medicine, Baobab Healthcare Inc, 55 Hanyangdaehak-Ro, Ansan, Gyeonggi-do 15588, South Korea
| | - Dongyun Kim
- Department of Mechanical Engineering, Korea Polytechnic University, Sangidaehak-ro, Siheung, Gyeonggi-do 15073, South Korea
| | - Donggu Kang
- Research Institute of Additive Manufacturing and Regenerative Medicine, Baobab Healthcare Inc, 55 Hanyangdaehak-Ro, Ansan, Gyeonggi-do 15588, South Korea
| | - Gi Hoon Yang
- Research Institute of Additive Manufacturing and Regenerative Medicine, Baobab Healthcare Inc, 55 Hanyangdaehak-Ro, Ansan, Gyeonggi-do 15588, South Korea
| | - Bongsu Jung
- Medical Device Development Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), 80, Cheombok-ro, Dong-gu, Daegu 41061, South Korea
| | - MyungGu Yeo
- Medical Device Development Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), 80, Cheombok-ro, Dong-gu, Daegu 41061, South Korea
| | - Min-Jeong Park
- Medical Device Development Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), 80, Cheombok-ro, Dong-gu, Daegu 41061, South Korea
| | - SangHyun An
- Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), 80 Cheombok-ro, Dong-gu, Daegu 41061, South Korea
| | - KyoungHo Lee
- Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), 80 Cheombok-ro, Dong-gu, Daegu 41061, South Korea
| | - Jun Sik Kim
- Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), 80 Cheombok-ro, Dong-gu, Daegu 41061, South Korea
| | - Jong Chul Kim
- Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), 80 Cheombok-ro, Dong-gu, Daegu 41061, South Korea
| | - Woonhyeok Jeong
- Department of Plastic and Reconstructive Surgery, Dongsan Medical Center, Keimyung University College of Medicine, 1035 Dalgubeol-daero, Dalseo-gu, Daegu 42601, South Korea
| | - Hye Hyun Yoo
- Institute of Pharmaceutical Science and Technology, College of Pharmacy, Hanyang University, 55 Hanyangdaehak-Ro, Ansan, Gyeonggi-Do 15588, South Korea
| | - Hojun Jeon
- Research Institute of Additive Manufacturing and Regenerative Medicine, Baobab Healthcare Inc, 55 Hanyangdaehak-Ro, Ansan, Gyeonggi-do 15588, South Korea
| |
Collapse
|
16
|
Choi E, Bae S, Kim D, Yang GH, Lee K, You HJ, Kang HJ, Gwak SJ, An S, Jeon H. Characterization and intracellular mechanism of electrospun poly (ε-caprolactone) (PCL) fibers incorporated with bone-dECM powder as a potential membrane for guided bone regeneration. J IND ENG CHEM 2021. [DOI: 10.1016/j.jiec.2020.11.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
17
|
Katoh K. Regulation of Fibroblast Cell Polarity by Src Tyrosine Kinase. Biomedicines 2021; 9:biomedicines9020135. [PMID: 33535441 PMCID: PMC7912711 DOI: 10.3390/biomedicines9020135] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 01/22/2021] [Accepted: 01/26/2021] [Indexed: 11/20/2022] Open
Abstract
Src protein tyrosine kinases (SFKs) are a family of nonreceptor tyrosine kinases that are localized beneath the plasma membrane and are activated during cell adhesion, migration, and elongation. Due to their involvement in the activation of signal transduction cascades, SFKs have been suggested to play important roles in the determination of cell polarity during cell extension and elongation. However, the mechanism underlying Src-mediated polarity formation remains unclear. The present study was performed to investigate the mechanisms underlying Src-induced cell polarity formation and cell elongation using Src knockout fibroblasts (SYFs) together with an inhibitor of Src. Normal and Src knockout fibroblasts were also transfected with a wild-type c-Src, dominant negative c-Src, or constitutively active c-Src gene to analyze the changes in cell morphology. SYF cells cultured on a glass substrate elongated symmetrically into spindle-shaped cells, with the formation of focal adhesions at both ends of the cells. When normal fibroblasts were treated with Src Inhibitor No. 5, a selective inhibitor of Src tyrosine kinases, they elongated into symmetrical spindle-shaped cells, similar to SYF cells. These results suggest that cell polarity during extension and elongation may be regulated by SFKs and that the expression and regulation of Src are important for the formation of polarity during cell elongation.
Collapse
Affiliation(s)
- Kazuo Katoh
- Laboratory of Human Anatomy and Cell Biology, Faculty of Health Sciences, Tsukuba University of Technology, Tsukuba-city, Ibaraki 305-8521, Japan
| |
Collapse
|
18
|
Boujemaa-Paterski R, Martins B, Eibauer M, Beales CT, Geiger B, Medalia O. Talin-activated vinculin interacts with branched actin networks to initiate bundles. eLife 2020; 9:e53990. [PMID: 33185186 PMCID: PMC7682986 DOI: 10.7554/elife.53990] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 11/12/2020] [Indexed: 12/18/2022] Open
Abstract
Vinculin plays a fundamental role in integrin-mediated cell adhesion. Activated by talin, it interacts with diverse adhesome components, enabling mechanical coupling between the actin cytoskeleton and the extracellular matrix. Here we studied the interactions of activated full-length vinculin with actin and the way it regulates the organization and dynamics of the Arp2/3 complex-mediated branched actin network. Through a combination of surface patterning and light microscopy experiments we show that vinculin can bundle dendritic actin networks through rapid binding and filament crosslinking. We show that vinculin promotes stable but flexible actin bundles having a mixed-polarity organization, as confirmed by cryo-electron tomography. Adhesion-like synthetic design of vinculin activation by surface-bound talin revealed that clustered vinculin can initiate and immobilize bundles from mobile Arp2/3-branched networks. Our results provide a molecular basis for coordinate actin bundle formation at nascent adhesions.
Collapse
Affiliation(s)
- Rajaa Boujemaa-Paterski
- Department of Biochemistry, University of ZurichZurichSwitzerland
- Université Grenoble AlpesGrenobleFrance
| | - Bruno Martins
- Department of Biochemistry, University of ZurichZurichSwitzerland
| | - Matthias Eibauer
- Department of Biochemistry, University of ZurichZurichSwitzerland
| | - Charlie T Beales
- Department of Biochemistry, University of ZurichZurichSwitzerland
| | - Benjamin Geiger
- Department of Immunology, Weizmann Institute of ScienceRehovotIsrael
| | - Ohad Medalia
- Department of Biochemistry, University of ZurichZurichSwitzerland
| |
Collapse
|
19
|
Taha-Salaime L, Lebedev G, Abo-Nassar J, Marzouk S, Inbar M, Ghanim M, Aly R. Activity of Ajuga iva Extracts Against the African Cotton Leafworm Spodoptera littoralis. INSECTS 2020; 11:insects11110726. [PMID: 33114086 PMCID: PMC7690827 DOI: 10.3390/insects11110726] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 10/21/2020] [Accepted: 10/21/2020] [Indexed: 12/24/2022]
Abstract
Control of the crop pest African cotton leafworm, Spodoptera littoralis (Boisduval), by chemical insecticides has led to serious resistance problems. Ajuga plants contain phytoecdysteroids (arthropod steroid hormone analogs regulating metamorphosis) and clerodanes (diterpenoids exhibiting antifeedant activity). We analyzed these compounds in leaf extracts of the Israeli Ajuga iva L. by liquid chromatography time-of-flight mass spectrometry (LC-TOF-MS) and thin-layer chromatography (TLC), and their efficiency at reducing S.littoralis fitness. First and third instars of S. littoralis were fed castor bean leaves (Ricinus communis) smeared with an aqueous suspension of dried methanolic crude extract of A. iva phytoecdysteroids and clerodanes. Mortality, larval weight gain, relative growth rate and survival were compared to feeding on control leaves. We used '4',6-diamidino-2-phenylindole (DAPI, a fluorescent stain) and phalloidin staining to localize A. iva crude leaf extract activity in the insect gut. Ajuga iva crude leaf extract (50, 100 and 250 µg/µL) significantly increased mortality of first-instar S. littoralis (36%, 70%, and 87%, respectively) compared to controls (6%). Third-instar larval weight gain decreased significantly (by 52%, 44% and 30%, respectively), as did relative growth rate (-0.05 g/g per day compared to the relevant controls), ultimately resulting in few survivors. Crude leaf extract (250 µg/µL) reduced gut size, with relocation of nuclei and abnormal actin-filament organization. Ajug iva extract has potential for alternative, environmentally safe insect-pest control.
Collapse
Affiliation(s)
- Leena Taha-Salaime
- Department of Evolutionary and Environmental Biology, The Faculty of Natural Science, University of Haifa, Haifa 3498838, Israel; (L.T.-S.); (M.I.)
- Department of Plant Pathology and Weeds Research, Newe Ya’ar Research Center, Agricultural Research Organization, Ramat Yishay 30095, Israel; (J.A.-N.); (S.M.)
| | - Galina Lebedev
- Department of Entomology, Agricultural Research Organization, The Volcani Center, Rishon LeTsiyon 7528809, Israel; (G.L.); (M.G.)
| | - Jackline Abo-Nassar
- Department of Plant Pathology and Weeds Research, Newe Ya’ar Research Center, Agricultural Research Organization, Ramat Yishay 30095, Israel; (J.A.-N.); (S.M.)
| | - Sally Marzouk
- Department of Plant Pathology and Weeds Research, Newe Ya’ar Research Center, Agricultural Research Organization, Ramat Yishay 30095, Israel; (J.A.-N.); (S.M.)
| | - Moshe Inbar
- Department of Evolutionary and Environmental Biology, The Faculty of Natural Science, University of Haifa, Haifa 3498838, Israel; (L.T.-S.); (M.I.)
| | - Murad Ghanim
- Department of Entomology, Agricultural Research Organization, The Volcani Center, Rishon LeTsiyon 7528809, Israel; (G.L.); (M.G.)
| | - Radi Aly
- Department of Plant Pathology and Weeds Research, Newe Ya’ar Research Center, Agricultural Research Organization, Ramat Yishay 30095, Israel; (J.A.-N.); (S.M.)
- Correspondence:
| |
Collapse
|
20
|
Sbai O, Soussi R, Bole A, Khrestchatisky M, Esclapez M, Ferhat L. The actin binding protein α-actinin-2 expression is associated with dendritic spine plasticity and migrating granule cells in the rat dentate gyrus following pilocarpine-induced seizures. Exp Neurol 2020; 335:113512. [PMID: 33098872 DOI: 10.1016/j.expneurol.2020.113512] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 10/08/2020] [Accepted: 10/19/2020] [Indexed: 12/24/2022]
Abstract
α-actinin-2 (α-actn-2) is an F-actin-crosslinking protein, localized in dendritic spines. In vitro studies suggested that it is involved in spinogenesis, morphogenesis, actin organization, cell migration and anchoring of the NR1 subunit of the N-methyl-D-aspartate (NMDA) receptors in dendritic spines. However, little is known regarding its function in vivo. We examined the levels of α-actn-2 expression within the dentate gyrus (DG) during the development of chronic limbic seizures (epileptogenesis) induced by pilocarpine in rats. In this model, plasticity of the DG glutamatergic granule cells including spine loss, spinogenesis, morphogenesis, neo-synaptogenesis, aberrant migration, and alterations of NMDA receptors have been well characterized. We showed that α-actn-2 immunolabeling was reduced in the inner molecular layer at 1-2 weeks post-status epilepticus (SE), when granule cell spinogenesis and morphogenesis occur. This low level persisted at the chronic stage when new functional synapses are established. This decreased of α-actn-2 protein is concomitant with the recovery of drebrin A (DA), another actin-binding protein, at the chronic stage. Indeed, we demonstrated in cultured cells that in contrast to DA, α-actn-2 did not protect F-actin destabilization and DA inhibited α-actn-2 binding to F-actin. Such alteration could affect the anchoring of NR1 in dendritic spines. Furthermore, we showed that the expression of α-actn-2 and NR1 are co-down-regulated in membrane fractions of pilocarpine animals at chronic stage. Last, we showed that α-actn-2 is expressed in migrating newly born granule cells observed within the hilus of pilocarpine-treated rats. Altogether, our results suggest that α-actn-2 is not critical for the structural integrity and stabilization of granule cell dendritic spines. Instead, its expression is regulated when spinogenesis and morphogenesis occur and within migrating granule cells. Our data also suggest that the balance between α-actn-2 and DA expression levels may modulate NR1 anchoring within dendritic spines.
Collapse
Affiliation(s)
- Oualid Sbai
- Aix-Marseille Univ, CNRS, INP, Inst Neurophysiopathol, Marseille, France
| | - Rabia Soussi
- Aix-Marseille Univ, CNRS, INP, Inst Neurophysiopathol, Marseille, France
| | - Angélique Bole
- Aix-Marseille Univ, CNRS, INP, Inst Neurophysiopathol, Marseille, France
| | | | - Monique Esclapez
- Aix-Marseille Univ, INSERM, INS, Inst Neurosci Syst, Marseille, France
| | - Lotfi Ferhat
- Aix-Marseille Univ, CNRS, INP, Inst Neurophysiopathol, Marseille, France.
| |
Collapse
|
21
|
Abstract
Integrins are heterodimeric cell surface receptors ensuring the mechanical connection between cells and the extracellular matrix. In addition to the anchorage of cells to the extracellular matrix, these receptors have critical functions in intracellular signaling, but are also taking center stage in many physiological and pathological conditions. In this review, we provide some historical, structural, and physiological notes so that the diverse functions of these receptors can be appreciated and put into the context of the emerging field of mechanobiology. We propose that the exciting journey of the exploration of these receptors will continue for at least another new generation of researchers.
Collapse
Affiliation(s)
- Michael Bachmann
- Department of Cell Physiology and Metabolism, University of Geneva, Centre Médical Universitaire , Geneva , Switzerland ; and Faculty of Medicine and Health Technology, Tampere University, and Fimlab Laboratories , Tampere , Finland
| | - Sampo Kukkurainen
- Department of Cell Physiology and Metabolism, University of Geneva, Centre Médical Universitaire , Geneva , Switzerland ; and Faculty of Medicine and Health Technology, Tampere University, and Fimlab Laboratories , Tampere , Finland
| | - Vesa P Hytönen
- Department of Cell Physiology and Metabolism, University of Geneva, Centre Médical Universitaire , Geneva , Switzerland ; and Faculty of Medicine and Health Technology, Tampere University, and Fimlab Laboratories , Tampere , Finland
| | - Bernhard Wehrle-Haller
- Department of Cell Physiology and Metabolism, University of Geneva, Centre Médical Universitaire , Geneva , Switzerland ; and Faculty of Medicine and Health Technology, Tampere University, and Fimlab Laboratories , Tampere , Finland
| |
Collapse
|
22
|
Bildyug N. Extracellular Matrix in Regulation of Contractile System in Cardiomyocytes. Int J Mol Sci 2019; 20:E5054. [PMID: 31614676 PMCID: PMC6834325 DOI: 10.3390/ijms20205054] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 10/07/2019] [Accepted: 10/09/2019] [Indexed: 12/16/2022] Open
Abstract
The contractile apparatus of cardiomyocytes is considered to be a stable system. However, it undergoes strong rearrangements during heart development as cells progress from their non-muscle precursors. Long-term culturing of mature cardiomyocytes is also accompanied by the reorganization of their contractile apparatus with the conversion of typical myofibrils into structures of non-muscle type. Processes of heart development as well as cell adaptation to culture conditions in cardiomyocytes both involve extracellular matrix changes, which appear to be crucial for the maturation of contractile apparatus. The aim of this review is to analyze the role of extracellular matrix in the regulation of contractile system dynamics in cardiomyocytes. Here, the remodeling of actin contractile structures and the expression of actin isoforms in cardiomyocytes during differentiation and adaptation to the culture system are described along with the extracellular matrix alterations. The data supporting the regulation of actin dynamics by extracellular matrix are highlighted and the possible mechanisms of such regulation are discussed.
Collapse
Affiliation(s)
- Natalya Bildyug
- Institute of Cytology, Russian Academy of Sciences, St-Petersburg 194064, Russia.
| |
Collapse
|
23
|
Hemidesmosomes and Focal Adhesions Treadmill as Separate but Linked Entities during Keratinocyte Migration. J Invest Dermatol 2019; 139:1876-1888.e4. [PMID: 30951704 DOI: 10.1016/j.jid.2019.03.1139] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 03/18/2019] [Accepted: 03/19/2019] [Indexed: 01/25/2023]
Abstract
Hemidesmosomes anchor the epidermal keratin filament cytoskeleton to the extracellular matrix. They are crucial for the mechanical integrity of skin. Their role in keratinocyte migration, however, remains unclear. Examining migrating primary human keratinocytes, we find that hemidesmosomes cluster as ordered arrays consisting of multiple chevrons that are flanked by actin-associated focal adhesions. These hemidesmosomal arrays with intercalated focal adhesions extend from the cell rear to the cell front. New hemidesmosomal chevrons form subsequent to focal adhesion assembly at the cell's leading front, whereas chevrons and associated focal adhesions disassemble at the cell rear in reverse order. The bulk of the hemidesmosome-focal adhesion composite, however, remains attached to the substratum during cell translocation. Similar hemidesmosome-focal adhesion patterns emerge on X-shaped fibronectin-coated micropatterns, during cell spreading and in leader cells during collective cell migration. We further find that hemidesmosomes and focal adhesions affect each other's distribution. We propose that both junctions are separate but linked entities, which treadmill coordinately to support efficient directed cell migration and cooperate to coordinate the dynamic interplay between the keratin and actin cytoskeleton.
Collapse
|
24
|
Lornage X, Romero NB, Grosgogeat CA, Malfatti E, Donkervoort S, Marchetti MM, Neuhaus SB, Foley AR, Labasse C, Schneider R, Carlier RY, Chao KR, Medne L, Deleuze JF, Orlikowski D, Bönnemann CG, Gupta VA, Fardeau M, Böhm J, Laporte J. ACTN2 mutations cause "Multiple structured Core Disease" (MsCD). Acta Neuropathol 2019; 137:501-519. [PMID: 30701273 DOI: 10.1007/s00401-019-01963-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 01/17/2019] [Accepted: 01/18/2019] [Indexed: 12/11/2022]
Abstract
The identification of genes implicated in myopathies is essential for diagnosis and for revealing novel therapeutic targets. Here we characterize a novel subclass of congenital myopathy at the morphological, molecular, and functional level. Through exome sequencing, we identified de novo ACTN2 mutations, a missense and a deletion, in two unrelated patients presenting with progressive early-onset muscle weakness and respiratory involvement. Morphological and ultrastructural analyses of muscle biopsies revealed a distinctive pattern with the presence of muscle fibers containing small structured cores and jagged Z-lines. Deeper analysis of the missense mutation revealed mutant alpha-actinin-2 properly localized to the Z-line in differentiating myotubes and its level was not altered in muscle biopsy. Modelling of the disease in zebrafish and mice by exogenous expression of mutated alpha-actinin-2 recapitulated the abnormal muscle function and structure seen in the patients. Motor deficits were noted in zebrafish, and muscle force was impaired in isolated muscles from AAV-transduced mice. In both models, sarcomeric disorganization was evident, while expression of wild-type alpha-actinin-2 did not result in muscle anomalies. The murine muscles injected with mutant ACTN2 displayed cores and Z-line defects. Dominant ACTN2 mutations were previously associated with cardiomyopathies, and our data demonstrate that specific mutations in the well-known Z-line regulator alpha-actinin-2 can cause a skeletal muscle disorder.
Collapse
Affiliation(s)
- Xavière Lornage
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), 1, rue Laurent Fries, BP 10142, 67404, Illkirch, France
- INSERM U1258, 67404, Illkirch, France
- CNRS, UMR7104, 67404, Illkirch, France
- Université de Strasbourg, 67404, Illkirch, France
| | - Norma B Romero
- Université Sorbonne, UPMC Univ Paris 06, INSERM UMRS974, CNRS FRE3617, Center for Research in Myology, GH Pitié-Salpêtrière, 47 Boulevard de l'hôpital, 75013, Paris, France
- Centre de référence de Pathologie Neuromusculaire Paris-Est, Institut de Myologie, GHU Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris, 75013, Paris, France
- Neuromuscular Morphology Unit, Myology Institute, GHU Pitié-Salpêtrière, 75013, Paris, France
| | - Claire A Grosgogeat
- Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Edoardo Malfatti
- Centre de référence de Pathologie Neuromusculaire Paris-Est, Institut de Myologie, GHU Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris, 75013, Paris, France
- Neuromuscular Morphology Unit, Myology Institute, GHU Pitié-Salpêtrière, 75013, Paris, France
- Neurology Department, Raymond-Poincaré teaching hospital, Centre de référence des maladies neuromusculaires Nord/Est/Ile-de-France, AP-HP, 92380, Garches, France
| | - Sandra Donkervoort
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Michael M Marchetti
- Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Sarah B Neuhaus
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA
| | - A Reghan Foley
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Clémence Labasse
- Centre de référence de Pathologie Neuromusculaire Paris-Est, Institut de Myologie, GHU Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris, 75013, Paris, France
- Neuromuscular Morphology Unit, Myology Institute, GHU Pitié-Salpêtrière, 75013, Paris, France
| | - Raphaël Schneider
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), 1, rue Laurent Fries, BP 10142, 67404, Illkirch, France
- INSERM U1258, 67404, Illkirch, France
- CNRS, UMR7104, 67404, Illkirch, France
- Université de Strasbourg, 67404, Illkirch, France
| | - Robert Y Carlier
- Neurolocomotor Division, Department of Radiology, Raymond Poincare Hospital, University Hospitals Paris-Ile-de-France West, Public Hospital Network of Paris, 92380, Garches, France
- Versailles Saint-Quentin-en-Yvelines University, 78000, Versailles, France
| | - Katherine R Chao
- Center for Mendelian Genomics, Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Boston, MA, 02115, USA
| | - Livija Medne
- Division of Human Genetics, The Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Jean-François Deleuze
- Centre National de Recherche en Génomique Humaine (CNRGH), Institut de biologie François Jacob, CEA, 91000, Evry, France
| | - David Orlikowski
- CIC 1429, INSERM, AP-HP, Hôpital Raymond Poincaré, 92380, Garches, France
| | - Carsten G Bönnemann
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Vandana A Gupta
- Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Michel Fardeau
- Université Sorbonne, UPMC Univ Paris 06, INSERM UMRS974, CNRS FRE3617, Center for Research in Myology, GH Pitié-Salpêtrière, 47 Boulevard de l'hôpital, 75013, Paris, France
- Centre de référence de Pathologie Neuromusculaire Paris-Est, Institut de Myologie, GHU Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris, 75013, Paris, France
- Neuromuscular Morphology Unit, Myology Institute, GHU Pitié-Salpêtrière, 75013, Paris, France
| | - Johann Böhm
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), 1, rue Laurent Fries, BP 10142, 67404, Illkirch, France
- INSERM U1258, 67404, Illkirch, France
- CNRS, UMR7104, 67404, Illkirch, France
- Université de Strasbourg, 67404, Illkirch, France
| | - Jocelyn Laporte
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), 1, rue Laurent Fries, BP 10142, 67404, Illkirch, France.
- INSERM U1258, 67404, Illkirch, France.
- CNRS, UMR7104, 67404, Illkirch, France.
- Université de Strasbourg, 67404, Illkirch, France.
| |
Collapse
|
25
|
Tousley A, Iuliano M, Weisman E, Sapp E, Richardson H, Vodicka P, Alexander J, Aronin N, DiFiglia M, Kegel-Gleason KB. Huntingtin associates with the actin cytoskeleton and α-actinin isoforms to influence stimulus dependent morphology changes. PLoS One 2019; 14:e0212337. [PMID: 30768638 PMCID: PMC6377189 DOI: 10.1371/journal.pone.0212337] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 01/31/2019] [Indexed: 01/09/2023] Open
Abstract
One response of cells to growth factor stimulus involves changes in morphology driven by the actin cytoskeleton and actin associated proteins which regulate functions such as cell adhesion, motility and in neurons, synaptic plasticity. Previous studies suggest that Huntingtin may be involved in regulating morphology however, there has been limited evidence linking endogenous Huntingtin localization or function with cytoplasmic actin in cells. We found that depletion of Huntingtin in human fibroblasts reduced adhesion and altered morphology and these phenotypes were made worse with growth factor stimulation, whereas the presence of the Huntington's Disease mutation inhibited growth factor induced changes in morphology and increased numbers of vinculin-positive focal adhesions. Huntingtin immunoreactivity localized to actin stress fibers, vinculin-positive adhesion contacts and membrane ruffles in fibroblasts. Interactome data from others has shown that Huntingtin can associate with α-actinin isoforms which bind actin filaments. Mapping studies using a cDNA encoding α-actinin-2 showed that it interacts within Huntingtin aa 399-969. Double-label immunofluorescence showed Huntingtin and α-actinin-1 co-localized to stress fibers, membrane ruffles and lamellar protrusions in fibroblasts. Proximity ligation assays confirmed a close molecular interaction between Huntingtin and α-actinin-1 in human fibroblasts and neurons. Huntingtin silencing with siRNA in fibroblasts blocked the recruitment of α-actinin-1 to membrane foci. These studies support the idea that Huntingtin is involved in regulating adhesion and actin dependent functions including those involving α-actinin.
Collapse
Affiliation(s)
- Adelaide Tousley
- Laboratory of Cellular Neurobiology, Department of Neurology, Massachusetts General Hospital, Charlestown, Massachusetts, United States of America
| | - Maria Iuliano
- Laboratory of Cellular Neurobiology, Department of Neurology, Massachusetts General Hospital, Charlestown, Massachusetts, United States of America
| | - Elizabeth Weisman
- Laboratory of Cellular Neurobiology, Department of Neurology, Massachusetts General Hospital, Charlestown, Massachusetts, United States of America
| | - Ellen Sapp
- Laboratory of Cellular Neurobiology, Department of Neurology, Massachusetts General Hospital, Charlestown, Massachusetts, United States of America
| | - Heather Richardson
- Laboratory of Cellular Neurobiology, Department of Neurology, Massachusetts General Hospital, Charlestown, Massachusetts, United States of America
| | - Petr Vodicka
- Laboratory of Cellular Neurobiology, Department of Neurology, Massachusetts General Hospital, Charlestown, Massachusetts, United States of America
| | - Jonathan Alexander
- Laboratory of Cellular Neurobiology, Department of Neurology, Massachusetts General Hospital, Charlestown, Massachusetts, United States of America
| | - Neil Aronin
- Department of Medicine and Cell Biology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Marian DiFiglia
- Laboratory of Cellular Neurobiology, Department of Neurology, Massachusetts General Hospital, Charlestown, Massachusetts, United States of America
| | - Kimberly B. Kegel-Gleason
- Laboratory of Cellular Neurobiology, Department of Neurology, Massachusetts General Hospital, Charlestown, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
26
|
|
27
|
Tseng HY, Samarelli AV, Kammerer P, Scholze S, Ziegler T, Immler R, Zent R, Sperandio M, Sanders CR, Fässler R, Böttcher RT. LCP1 preferentially binds clasped αMβ2 integrin and attenuates leukocyte adhesion under flow. J Cell Sci 2018; 131:jcs.218214. [PMID: 30333137 DOI: 10.1242/jcs.218214] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 09/07/2018] [Indexed: 12/13/2022] Open
Abstract
Integrins are α/β heterodimers that interconvert between inactive and active states. In the active state the α/β cytoplasmic domains recruit integrin-activating proteins and separate the transmembrane and cytoplasmic (TMcyto) domains (unclasped TMcyto). Conversely, in the inactive state the α/β TMcyto domains bind integrin-inactivating proteins, resulting in the association of the TMcyto domains (clasped TMcyto). Here, we report the isolation of integrin cytoplasmic tail interactors using either lipid bicelle-incorporated integrin TMcyto domains (α5, αM, αIIb, β1, β2 and β3 integrin TMcyto) or a clasped, lipid bicelle-incorporated αMβ2 TMcyto. Among the proteins found to preferentially bind clasped rather than the isolated αM and β2 subunits was L-plastin (LCP1, also known as plastin-2), which binds to and maintains the inactive state of αMβ2 integrin in vivo and thereby regulates leukocyte adhesion to integrin ligands under flow. Our findings offer a global view on cytoplasmic proteins interacting with different integrins and provide evidence for the existence of conformation-specific integrin interactors.
Collapse
Affiliation(s)
- Hui-Yuan Tseng
- Department of Molecular Medicine, Max Planck Institute for Biochemistry, 82152 Martinsried, Germany
| | - Anna V Samarelli
- Department of Molecular Medicine, Max Planck Institute for Biochemistry, 82152 Martinsried, Germany
| | - Patricia Kammerer
- Department of Molecular Medicine, Max Planck Institute for Biochemistry, 82152 Martinsried, Germany
| | - Sarah Scholze
- Department of Molecular Medicine, Max Planck Institute for Biochemistry, 82152 Martinsried, Germany
| | - Tilman Ziegler
- Department of Molecular Medicine, Max Planck Institute for Biochemistry, 82152 Martinsried, Germany
| | - Roland Immler
- Walter Brendel Center for Experimental Medicine, Ludwig-Maximilians-University, 81377 Munich, Germany
| | - Roy Zent
- Division of Nephrology, Department of Medicine, Vanderbilt University, Nashville, 37232 Tennessee, USA.,Department of Medicine, Veterans Affairs Medical Center, Nashville, 37232 Tennessee, USA
| | - Markus Sperandio
- Walter Brendel Center for Experimental Medicine, Ludwig-Maximilians-University, 81377 Munich, Germany
| | - Charles R Sanders
- Department of Biochemistry, Center for Structural Biology, and Institute of Chemical Biology, Vanderbilt University School of Medicine, Nashville, 37232 Tennessee, USA
| | - Reinhard Fässler
- Department of Molecular Medicine, Max Planck Institute for Biochemistry, 82152 Martinsried, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, 80802 Munich, Germany
| | - Ralph T Böttcher
- Department of Molecular Medicine, Max Planck Institute for Biochemistry, 82152 Martinsried, Germany .,DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, 80802 Munich, Germany
| |
Collapse
|
28
|
Houweling PJ, Papadimitriou ID, Seto JT, Pérez LM, Coso JD, North KN, Lucia A, Eynon N. Is evolutionary loss our gain? The role of
ACTN3
p.Arg577Ter (R577X) genotype in athletic performance, ageing, and disease. Hum Mutat 2018; 39:1774-1787. [DOI: 10.1002/humu.23663] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2018] [Revised: 09/20/2018] [Accepted: 09/26/2018] [Indexed: 01/13/2023]
Affiliation(s)
- Peter J. Houweling
- Murdoch Children's Research Institute Melbourne, Victoria Australia
- Department of Paediatrics University of Melbourne The Royal Children's Hospital Melbourne, Victoria Australia
| | | | - Jane T. Seto
- Murdoch Children's Research Institute Melbourne, Victoria Australia
- Department of Paediatrics University of Melbourne The Royal Children's Hospital Melbourne, Victoria Australia
| | - Laura M. Pérez
- Universidad Europea de Madrid (Faculty of Sport Sciences) Madrid Spain
- Instituto de Investigación Hospital 12 de Octubre Madrid Spain
| | - Juan Del Coso
- Exercise Physiology Laboratory Camilo José Cela University Madrid Spain
| | - Kathryn N. North
- Murdoch Children's Research Institute Melbourne, Victoria Australia
- Department of Paediatrics University of Melbourne The Royal Children's Hospital Melbourne, Victoria Australia
| | - Alejandro Lucia
- Universidad Europea de Madrid (Faculty of Sport Sciences) Madrid Spain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable Madrid Spain
| | - Nir Eynon
- Institute for Health and Sport (iHeS) Victoria University Victoria Australia
| |
Collapse
|
29
|
Kemp JP, Brieher WM. The actin filament bundling protein α-actinin-4 actually suppresses actin stress fibers by permitting actin turnover. J Biol Chem 2018; 293:14520-14533. [PMID: 30049798 DOI: 10.1074/jbc.ra118.004345] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 07/21/2018] [Indexed: 01/07/2023] Open
Abstract
Cells organize actin filaments into contractile bundles known as stress fibers that resist mechanical stress, increase cell adhesion, remodel the extracellular matrix, and maintain tissue integrity. α-actinin is an actin filament bundling protein that is thought to be essential for stress fiber formation and stability. However, previous studies have also suggested that α-actinin might disrupt fibers, making the true function of this biomolecule unclear. Here we use fluorescence imaging to show that kidney epithelial cells depleted of α-actinin-4 via shRNA or CRISPR/Cas9, or expressing a disruptive mutant make more massive stress fibers that are less dynamic than those in WT cells, leading to defects in cell motility and wound healing. The increase in stress fiber mass and stability can be explained, in part, by increased loading of the filament component tropomyosin onto stress fibers in the absence of α-actinin, as monitored via immunofluorescence. We show using imaging and cosedimentation that α-actinin and tropomyosin compete for binding to F-actin and that tropomyosin shields actin filaments from cofilin-mediated disassembly in vitro and in cells. Perturbing tropomyosin in cells lacking α-actinin-4 results in a complete loss of stress fibers. Our results with α-actinin-4 on stress fiber organization are the opposite of what might have been predicted from previous in vitro biochemistry and further highlight how the complex interactions of multiple proteins competing for filament binding lead to unexpected functions for actin-binding proteins in cells.
Collapse
Affiliation(s)
| | - William M Brieher
- From the Department of Cell and Developmental Biology, University of Illinois, Urbana-Champaign, Urbana, Illinois 61801
| |
Collapse
|
30
|
Li M, Wu X, Guo X, Bao P, Ding X, Chu M, Liang C, Yan P. Comparative iTRAQ proteomics revealed proteins associated with horn development in yak. Proteome Sci 2018; 16:14. [PMID: 30061793 PMCID: PMC6056918 DOI: 10.1186/s12953-018-0141-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 05/31/2018] [Indexed: 01/15/2023] Open
Abstract
Background The practice of dehorning yak raises animal safety concerns, which have been addressed by selective breeding to obtain genetically hornless yak. The POLLED locus in yak has been studied extensively; however, little is known regarding the proteins that regulate horn bud development. Methods A differential proteomic analysis was performed to compare the skin from the horn bud region of polled yak fetuses and the horn bud tissue of horned yak fetuses using isobaric tags for relative and absolute quantitation (iTRAQ) technology coupled with 2D LC-MS/MS. Results One hundred differentially abundant proteins (DAPs) were identified. Of these, 29 were up-regulated and 71 were down-regulated in skin from the horn bud region of polled fetuses when compared to the horn bud tissue of horned fetuses. Bioinformatics analyses showed that the up-regulated DAPs were mainly associated with metabolic activities, while the down-regulated DAPs were significantly enriched in cell adhesion and cell movement activities. Conclusions We concluded that some important proteins were associated with cell adhesion, cell motility, keratinocyte differentiation, cytoskeleton organization, osteoblast differentiation, and fatty acid metabolism during horn bud development. These results advance our understanding of the molecular mechanisms underlying horn development.
Collapse
Affiliation(s)
- Mingna Li
- Key Laboratory for Yak Breeding Engineering of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, 730050 People's Republic of China
| | - Xiaoyun Wu
- Key Laboratory for Yak Breeding Engineering of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, 730050 People's Republic of China
| | - Xian Guo
- Key Laboratory for Yak Breeding Engineering of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, 730050 People's Republic of China
| | - Pengjia Bao
- Key Laboratory for Yak Breeding Engineering of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, 730050 People's Republic of China
| | - Xuezhi Ding
- Key Laboratory for Yak Breeding Engineering of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, 730050 People's Republic of China
| | - Min Chu
- Key Laboratory for Yak Breeding Engineering of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, 730050 People's Republic of China
| | - Chunnian Liang
- Key Laboratory for Yak Breeding Engineering of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, 730050 People's Republic of China
| | - Ping Yan
- Key Laboratory for Yak Breeding Engineering of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, 730050 People's Republic of China
| |
Collapse
|
31
|
Abstract
This overview article for the Comprehensive Physiology collection is focused on detailing platelets, how platelets respond to various stimuli, how platelets interact with their external biochemical environment, and the role of platelets in physiological and pathological processes. Specifically, we will discuss the four major functions of platelets: activation, adhesion, aggregation, and inflammation. We will extend this discussion to include various mechanisms that can induce these functional changes and a discussion of some of the salient receptors that are responsible for platelets interacting with their external environment. We will finish with a discussion of how platelets interact with their vascular environment, with a special focus on interactions with the extracellular matrix and endothelial cells, and finally how platelets can aid and possibly initiate the progression of various vascular diseases. Throughout this overview, we will highlight both the historical investigations into the role of platelets in health and disease as well as some of the more current work. Overall, the authors aim for the readers to gain an appreciation for the complexity of platelet functions and the multifaceted role of platelets in the vascular system. © 2017 American Physiological Society. Compr Physiol 8:1117-1156, 2018.
Collapse
Affiliation(s)
- David A Rubenstein
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, New York, USA
| | - Wei Yin
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, New York, USA
| |
Collapse
|
32
|
Hsu CP, Moghadaszadeh B, Hartwig JH, Beggs AH. Sarcomeric and nonmuscle α-actinin isoforms exhibit differential dynamics at skeletal muscle Z-lines. Cytoskeleton (Hoboken) 2018; 75:213-228. [PMID: 29518289 PMCID: PMC5943145 DOI: 10.1002/cm.21442] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 03/02/2018] [Accepted: 03/05/2018] [Indexed: 01/12/2023]
Abstract
The α-actinin proteins are a highly conserved family of actin crosslinkers that mediate interactions between several cytoskeletal and sarcomeric proteins. Nonsarcomeric α-actinin-1 and α-actinin-4 crosslink actin filaments in the cytoskeleton, while sarcomeric α-actinin-2 and α-actinin-3 serve a crucial role in anchoring actin filaments to the muscle Z-line. To assess the difference in turnover dynamics and structure/function properties between the α-actinin isoforms at the sarcomeric Z-line, we used Fluorescence Recovery After Photobleaching (FRAP) in primary myofiber cultures. We found that the recovery kinetics of these proteins followed three distinct patterns: α-actinin-2/α-actinin-3 had the slowest turn over, α-actinin-1 recovered to an intermediate degree, and α-actinin-4 had the fastest recovery. Interestingly, the isoforms' patterns of recovery were reversed at adhesion plaques in fibroblasts. This disparity suggests that the different α-actinin isoforms have unique association kinetics in myofibers and that nonmuscle isoform interactions are more dynamic at the sarcomeric Z-line. Protein domain-specific investigations using α-actinin-2/4 chimeric proteins showed that differential dynamics between sarcomeric and nonmuscle isoforms are regulated by cooperative interactions between the N-terminal actin-binding domain, the spectrin-like linker region and the C-terminal calmodulin-like EF hand domain. Together, these findings demonstrate that α-actinin isoforms are unique in binding dynamics at the Z-line and suggest differentially evolved interactive and Z-line association capabilities of each functional domain.
Collapse
Affiliation(s)
- Cynthia P Hsu
- Division of Genetics and Genomics, The Manton Center for Orphan Disease Research, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Behzad Moghadaszadeh
- Division of Genetics and Genomics, The Manton Center for Orphan Disease Research, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - John H Hartwig
- Translational Medicine Division, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Alan H Beggs
- Division of Genetics and Genomics, The Manton Center for Orphan Disease Research, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
33
|
Pan L, Zhao Y, Farouk MH, Bao N, Wang T, Qin G. Integrins Were Involved in Soybean Agglutinin Induced Cell Apoptosis in IPEC-J2. Int J Mol Sci 2018; 19:E587. [PMID: 29462933 PMCID: PMC5855809 DOI: 10.3390/ijms19020587] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Revised: 02/02/2018] [Accepted: 02/09/2018] [Indexed: 12/30/2022] Open
Abstract
Soybean agglutinin (SBA), is a non-fiber carbohydrate related protein and a major anti-nutritional factor. Integrins, transmembrane glycoproteins, are involved in many biological processes. Although recent work suggested that integrins are involved in SBA-induced cell-cycle alterations, no comprehensive study has reported whether integrins are involved in SBA-induced cell apoptosis (SCA) in IPEC-J2. The relationship between SBA and integrins are still unclear. We aimed to elucidate the effects of SBA on IPEC-J2 cell proliferation and cell apoptosis; to study the roles of integrins in IPEC-J2 normal cell apoptosis (NCA) and SCA; and to illustrate the relationship and connection type between SBA and integrins. Thus, IPEC-J2 cells were treated with SBA at the levels of 0, 0.125, 0.25, 0.5, 1.0 or 2.0 mg/mL to determine cell proliferation and cell apoptosis. The cells were divided into control, SBA treated groups, integrin inhibitor groups, and SBA + integrin inhibitor groups to determine the integrin function in SCA. The results showed that SBA significantly (p < 0.05) lowered cell proliferation and induced cell apoptosis in IPEC-J2 (p < 0.05). Inhibition of any integrin type induced the cell apoptosis (p < 0.05) and these integrins were involved in SCA (p < 0.05). Even SBA had no physical connection with integrins, an association was detected between SBA and α-actinin-2 ACTN2 (integrin-binding protein). Additionally, SBA reduced the mRNA expression of integrins by down regulating the gene expression level of ACTN2. We concluded an evidence for the anti-nutritional mechanism of SBA by ACTN2 with integrins. Further trials are needed to prove whether ACTN2 is the only protein for connecting SBA with integrin.
Collapse
Affiliation(s)
- Li Pan
- Key Laboratory of Animal Production, Product Quality and Security, Ministry of Education, Key Laboratory of Animal Nutrition and Feed Science, Jilin Province, College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, China.
| | - Yuan Zhao
- Key Laboratory of Animal Production, Product Quality and Security, Ministry of Education, Key Laboratory of Animal Nutrition and Feed Science, Jilin Province, College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, China.
| | - Mohamed Hamdy Farouk
- Key Laboratory of Animal Production, Product Quality and Security, Ministry of Education, Key Laboratory of Animal Nutrition and Feed Science, Jilin Province, College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, China.
- Department of Animal Production, Faculty of Agriculture, Al-Azhar University, Nasr City, Cairo 11884, Egypt.
| | - Nan Bao
- Key Laboratory of Animal Production, Product Quality and Security, Ministry of Education, Key Laboratory of Animal Nutrition and Feed Science, Jilin Province, College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, China.
| | - Tao Wang
- Key Laboratory of Animal Production, Product Quality and Security, Ministry of Education, Key Laboratory of Animal Nutrition and Feed Science, Jilin Province, College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, China.
| | - Guixin Qin
- Key Laboratory of Animal Production, Product Quality and Security, Ministry of Education, Key Laboratory of Animal Nutrition and Feed Science, Jilin Province, College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, China.
| |
Collapse
|
34
|
Urner S, Kelly-Goss M, Peirce SM, Lammert E. Mechanotransduction in Blood and Lymphatic Vascular Development and Disease. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2017; 81:155-208. [PMID: 29310798 DOI: 10.1016/bs.apha.2017.08.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The blood and lymphatic vasculatures are hierarchical networks of vessels, which constantly transport fluids and, therefore, are exposed to a variety of mechanical forces. Considering the role of mechanotransduction is key for fully understanding how these vascular systems develop, function, and how vascular pathologies evolve. During embryonic development, for example, initiation of blood flow is essential for early vascular remodeling, and increased interstitial fluid pressure as well as initiation of lymph flow is needed for proper development and maturation of the lymphatic vasculature. In this review, we introduce specific mechanical forces that affect both the blood and lymphatic vasculatures, including longitudinal and circumferential stretch, as well as shear stress. In addition, we provide an overview of the role of mechanotransduction during atherosclerosis and secondary lymphedema, which both trigger tissue fibrosis.
Collapse
Affiliation(s)
- Sofia Urner
- Institute of Metabolic Physiology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Molly Kelly-Goss
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, United States
| | - Shayn M Peirce
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, United States
| | - Eckhard Lammert
- Institute of Metabolic Physiology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany; Institute for Beta Cell Biology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany.
| |
Collapse
|
35
|
Jansen K, Atherton P, Ballestrem C. Mechanotransduction at the cell-matrix interface. Semin Cell Dev Biol 2017; 71:75-83. [DOI: 10.1016/j.semcdb.2017.07.027] [Citation(s) in RCA: 182] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 07/19/2017] [Accepted: 07/19/2017] [Indexed: 01/09/2023]
|
36
|
Burridge K. Focal adhesions: a personal perspective on a half century of progress. FEBS J 2017; 284:3355-3361. [PMID: 28796323 DOI: 10.1111/febs.14195] [Citation(s) in RCA: 186] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 08/07/2017] [Indexed: 12/28/2022]
Abstract
Focal adhesions (FAs) are specialized sites within the cell where clustered integrin receptors interact with the extracellular matrix on the outside of cells and with the actin cytoskeleton on the inside. They provide strong adhesion to the matrix and transmit mechanical tension generated within cells across the plasma membrane to the external environment. Additionally, they act as scaffolds for many signaling pathways triggered by integrin engagement or mechanical force exerted on cells. Here I describe my personal perspective on FA research which I have witnessed since the initial discovery and description of FAs as electron dense regions of the ventral plasma nearly half a century ago.
Collapse
Affiliation(s)
- Keith Burridge
- Department of Cell Biology and Physiology, Lineberger Comprehensive Cancer Center, McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
37
|
Xu Q, Huff LP, Fujii M, Griendling KK. Redox regulation of the actin cytoskeleton and its role in the vascular system. Free Radic Biol Med 2017; 109:84-107. [PMID: 28285002 PMCID: PMC5497502 DOI: 10.1016/j.freeradbiomed.2017.03.004] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 02/17/2017] [Accepted: 03/06/2017] [Indexed: 12/17/2022]
Abstract
The actin cytoskeleton is critical for form and function of vascular cells, serving mechanical, organizational and signaling roles. Because many cytoskeletal proteins are sensitive to reactive oxygen species, redox regulation has emerged as a pivotal modulator of the actin cytoskeleton and its associated proteins. Here, we summarize work implicating oxidants in altering actin cytoskeletal proteins and focus on how these alterations affect cell migration, proliferation and contraction of vascular cells. Finally, we discuss the role of oxidative modification of the actin cytoskeleton in vivo and highlight its importance for vascular diseases.
Collapse
Affiliation(s)
- Qian Xu
- Division of Cardiology, Department of Medicine, Emory University, 101 Woodruff Circle, 308a WMB, Atlanta, GA 30322, United States; Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Lauren P Huff
- Division of Cardiology, Department of Medicine, Emory University, 101 Woodruff Circle, 308a WMB, Atlanta, GA 30322, United States
| | - Masakazu Fujii
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Japan
| | - Kathy K Griendling
- Division of Cardiology, Department of Medicine, Emory University, 101 Woodruff Circle, 308a WMB, Atlanta, GA 30322, United States.
| |
Collapse
|
38
|
Bachir AI, Horwitz AR, Nelson WJ, Bianchini JM. Actin-Based Adhesion Modules Mediate Cell Interactions with the Extracellular Matrix and Neighboring Cells. Cold Spring Harb Perspect Biol 2017; 9:9/7/a023234. [PMID: 28679638 DOI: 10.1101/cshperspect.a023234] [Citation(s) in RCA: 114] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Cell adhesions link cells to the extracellular matrix (ECM) and to each other and depend on interactions with the actin cytoskeleton. Both cell-ECM and cell-cell adhesion sites contain discrete, yet overlapping, functional modules. These modules establish physical associations with the actin cytoskeleton, locally modulate actin organization and dynamics, and trigger intracellular signaling pathways. Interplay between these modules generates distinct actin architectures that underlie different stages, types, and functions of cell-ECM and cell-cell adhesions. Actomyosin contractility is required to generate mature, stable adhesions, as well as to sense and translate the mechanical properties of the cellular environment into changes in cell organization and behavior. Here, we review the organization and function of different adhesion modules and how they interact with the actin cytoskeleton. We highlight the molecular mechanisms of mechanotransduction in adhesions and how adhesion molecules mediate cross talk between cell-ECM and cell-cell adhesion sites.
Collapse
Affiliation(s)
- Alexia I Bachir
- Protein and Cell Analysis, Biosciences Division, Thermo Fisher Scientific, Eugene, Oregon 97402
| | - Alan Rick Horwitz
- Protein and Cell Analysis, Biosciences Division, Thermo Fisher Scientific, Eugene, Oregon 97402
| | - W James Nelson
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia 22903
| | - Julie M Bianchini
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia 22903
| |
Collapse
|
39
|
Strength of Neisseria meningitidis binding to endothelial cells requires highly-ordered CD147/β 2-adrenoceptor clusters assembled by alpha-actinin-4. Nat Commun 2017; 8:15764. [PMID: 28569760 PMCID: PMC5461506 DOI: 10.1038/ncomms15764] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 04/26/2017] [Indexed: 12/24/2022] Open
Abstract
Neisseria meningitidis (meningococcus) is an invasive bacterial pathogen that colonizes human vessels, causing thrombotic lesions and meningitis. Establishment of tight interactions with endothelial cells is crucial for meningococci to resist haemodynamic forces. Two endothelial receptors, CD147 and the β2-adrenergic receptor (β2AR), are sequentially engaged by meningococci to adhere and promote signalling events leading to vascular colonization, but their spatiotemporal coordination is unknown. Here we report that CD147 and β2AR form constitutive hetero-oligomeric complexes. The scaffolding protein α-actinin-4 directly binds to the cytosolic tail of CD147 and governs the assembly of CD147–β2AR complexes in highly ordered clusters at bacterial adhesion sites. This multimolecular assembly process increases the binding strength of meningococci to endothelial cells under shear stress, and creates molecular platforms for the elongation of membrane protrusions surrounding adherent bacteria. Thus, the specific organization of cellular receptors has major impacts on host–pathogen interaction. Neisseria meningitidis bacteria bind to host proteins CD147 and β2-adrenergic receptor on the surface of endothelial cells. Here, Maïssa et al. show that the two proteins interact with each other forming clusters that increase the binding strength of the bacteria to endothelial cells.
Collapse
|
40
|
Proteoglycans, ion channels and cell-matrix adhesion. Biochem J 2017; 474:1965-1979. [PMID: 28546458 DOI: 10.1042/bcj20160747] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 04/10/2017] [Accepted: 04/12/2017] [Indexed: 01/09/2023]
Abstract
Cell surface proteoglycans comprise a transmembrane or membrane-associated core protein to which one or more glycosaminoglycan chains are covalently attached. They are ubiquitous receptors on nearly all animal cell surfaces. In mammals, the cell surface proteoglycans include the six glypicans, CD44, NG2 (CSPG4), neuropilin-1 and four syndecans. A single syndecan is present in invertebrates such as nematodes and insects. Uniquely, syndecans are receptors for many classes of proteins that can bind to the heparan sulphate chains present on syndecan core proteins. These range from cytokines, chemokines, growth factors and morphogens to enzymes and extracellular matrix (ECM) glycoproteins and collagens. Extracellular interactions with other receptors, such as some integrins, are mediated by the core protein. This places syndecans at the nexus of many cellular responses to extracellular cues in development, maintenance, repair and disease. The cytoplasmic domains of syndecans, while having no intrinsic kinase activity, can nevertheless signal through binding proteins. All syndecans appear to be connected to the actin cytoskeleton and can therefore contribute to cell adhesion, notably to the ECM and migration. Recent data now suggest that syndecans can regulate stretch-activated ion channels. The structure and function of the syndecans and the ion channels are reviewed here, along with an analysis of ion channel functions in cell-matrix adhesion. This area sheds new light on the syndecans, not least since evidence suggests that this is an evolutionarily conserved relationship that is also potentially important in the progression of some common diseases where syndecans are implicated.
Collapse
|
41
|
Wei Z, Shaikh ZA. Cadmium stimulates metastasis-associated phenotype in triple-negative breast cancer cells through integrin and β-catenin signaling. Toxicol Appl Pharmacol 2017; 328:70-80. [PMID: 28527916 DOI: 10.1016/j.taap.2017.05.017] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 04/21/2017] [Accepted: 05/16/2017] [Indexed: 12/15/2022]
Abstract
Cadmium (Cd) is a carcinogenic heavy metal which is implicated in breast cancer development. While the mechanisms of Cd-induced breast cancer initiation and promotion have been studied, the molecular processes involved in breast cancer progression remain to be investigated. The purpose of the present study was to evaluate the influence of Cd on metastasis-associated phenotypes, such as cell adhesion, migration, and invasion in triple-negative breast cancer cells. Treatment of MDA-MB-231 cells with 1μM Cd increased cell spreading and cell migration. This was associated with the activation of integrin β1, FAK, Src, and Rac1. Treatment with Cd also inhibited GSK3β activity and induced T-cell factor/lymphoid enhancer factor (TCF/LEF) transcription, indicating the involvement of β-catenin signaling. Furthermore, treatment with 3μM Cd for 4weeks increased the expression of β-catenin and enhanced TCF/LEF-mediated transcription. Furthermore, enhanced expressions of integrins α5 and β1, paxillin, and vimentin indicated that prolonged Cd treatment reorganized the cytoskeleton, which aided malignancy, as evidenced by enhanced matrix metalloprotease 2/9 (MMP2/9) secretion and cell invasion. Prolonged Cd treatment also caused an increase in cell growth. Together, these results indicate that Cd alters key signaling processes involved in the regulation of cytoskeleton to enhance cancer cell migration, invasion, adhesion, and proliferation.
Collapse
Affiliation(s)
- Zhengxi Wei
- Center for Molecular Toxicology, Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI 02881, USA
| | - Zahir A Shaikh
- Center for Molecular Toxicology, Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI 02881, USA.
| |
Collapse
|
42
|
Aguilar-Cuenca R, Llorente-Gonzalez C, Vicente C, Vicente-Manzanares M. Microfilament-coordinated adhesion dynamics drives single cell migration and shapes whole tissues. F1000Res 2017; 6:160. [PMID: 28299195 PMCID: PMC5321130 DOI: 10.12688/f1000research.10356.1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/13/2017] [Indexed: 12/26/2022] Open
Abstract
Cell adhesion to the substratum and/or other cells is a crucial step of cell migration. While essential in the case of solitary migrating cells (for example, immune cells), it becomes particularly important in collective cell migration, in which cells maintain contact with their neighbors while moving directionally. Adhesive coordination is paramount in physiological contexts (for example, during organogenesis) but also in pathology (for example, tumor metastasis). In this review, we address the need for a coordinated regulation of cell-cell and cell-matrix adhesions during collective cell migration. We emphasize the role of the actin cytoskeleton as an intracellular integrator of cadherin- and integrin-based adhesions and the emerging role of mechanics in the maintenance, reinforcement, and turnover of adhesive contacts. Recent advances in understanding the mechanical regulation of several components of cadherin and integrin adhesions allow us to revisit the adhesive clutch hypothesis that controls the degree of adhesive engagement during protrusion. Finally, we provide a brief overview of the major impact of these discoveries when using more physiological three-dimensional models of single and collective cell migration.
Collapse
Affiliation(s)
- Rocio Aguilar-Cuenca
- Universidad Autonoma de Madrid School of Medicine, Madrid, Spain; Instituto de Investigacion Sanitaria Hospital Universitario de la Princesa, Madrid, Spain
| | - Clara Llorente-Gonzalez
- Universidad Autonoma de Madrid School of Medicine, Madrid, Spain; Instituto de Investigacion Sanitaria Hospital Universitario de la Princesa, Madrid, Spain
| | - Carlos Vicente
- Team of Cell Interactions in Plant Symbiosis, Faculty of Biology, Complutense University, Madrid, Spain
| | - Miguel Vicente-Manzanares
- Universidad Autonoma de Madrid School of Medicine, Madrid, Spain; Team of Cell Interactions in Plant Symbiosis, Faculty of Biology, Complutense University, Madrid, Spain
| |
Collapse
|
43
|
Zhang Y, Cao G, Zhu L, Chen F, Zar MS, Wang S, Hu X, Wei Y, Xue R, Gong C. Integrin beta and receptor for activated protein kinase C are involved in the cell entry of Bombyx mori cypovirus. Appl Microbiol Biotechnol 2017; 101:3703-3716. [PMID: 28175946 DOI: 10.1007/s00253-017-8158-z] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Revised: 01/23/2017] [Accepted: 01/25/2017] [Indexed: 12/22/2022]
Abstract
Receptor-mediated endocytosis using a β1 integrin-dependent internalization was considered as the primary mechanism for the initiation of mammalian reovirus infection. Bombyx mori cypovirus (BmCPV) is a member of Reoviridae family which mainly infects the midgut epithelium of silkworm; the cell entry of BmCPV is poorly explored. In this study, co-immunoprecipitation (Co-IP), virus overlay protein binding assay (VOPBA), and BmCPV-protein interaction on the polyvinylidene difluoride membrane (BmCPV-PI-PVDF) methods were employed to screen the interacting proteins of BmCPV, and several proteins including integrin beta and receptor for activated protein kinase C (RACK1) were identified as the candidate interacting proteins for establishing the infection of BmCPV. The infectivity of BmCPV was investigated in vivo and in vitro by RNA interference (RNAi) and antibody blocking methods, and the results showed that the infectivity of BmCPV was significantly reduced by either small interfering RNA-mediated silencing of integrin beta and RACK1 or antibody blocking of integrin beta and RACK1. The expression level of integrin beta or RACK1 is not the highest in the silkworm midgut which is a principal target tissue of BmCPV, suggesting that the molecules other than integrin beta or RACK1 might play a key role in determining the tissue tropism of BmCPV infection. The establishment of BmCPV infection depends on other factors, and these factors interacted with integrin beta and RACK1 to form receptor complex for the cell entry of BmCPV.
Collapse
Affiliation(s)
- Yiling Zhang
- School of Biology and Basic Medical Sciences, Soochow University, Suzhou, 215123, Jiangsu, People's Republic of China
- Suzhou Municipal Key Laboratory of Molecular Diagnostics and Therapeutics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, 215163, People's Republic of China
| | - Guangli Cao
- School of Biology and Basic Medical Sciences, Soochow University, Suzhou, 215123, Jiangsu, People's Republic of China
- National Engineering Laboratory for Modern Silk, Soochow University, Postal address: No. 199 Ren'ai Road, Suzhou Industrial Park, Suzhou, 215123, Jiangsu, People's Republic of China
| | - Liyuan Zhu
- School of Biology and Basic Medical Sciences, Soochow University, Suzhou, 215123, Jiangsu, People's Republic of China
| | - Fei Chen
- School of Biology and Basic Medical Sciences, Soochow University, Suzhou, 215123, Jiangsu, People's Republic of China
| | - Mian Sahib Zar
- School of Biology and Basic Medical Sciences, Soochow University, Suzhou, 215123, Jiangsu, People's Republic of China
| | - Simei Wang
- School of Biology and Basic Medical Sciences, Soochow University, Suzhou, 215123, Jiangsu, People's Republic of China
- Department of Hematology, The First Affiliated Hospital of Gannan Medical College, Ganzhou, 341000, Jiangxi, People's Republic of China
| | - Xiaolong Hu
- School of Biology and Basic Medical Sciences, Soochow University, Suzhou, 215123, Jiangsu, People's Republic of China
- National Engineering Laboratory for Modern Silk, Soochow University, Postal address: No. 199 Ren'ai Road, Suzhou Industrial Park, Suzhou, 215123, Jiangsu, People's Republic of China
| | - Yuhong Wei
- School of Biology and Basic Medical Sciences, Soochow University, Suzhou, 215123, Jiangsu, People's Republic of China
| | - Renyu Xue
- School of Biology and Basic Medical Sciences, Soochow University, Suzhou, 215123, Jiangsu, People's Republic of China
- National Engineering Laboratory for Modern Silk, Soochow University, Postal address: No. 199 Ren'ai Road, Suzhou Industrial Park, Suzhou, 215123, Jiangsu, People's Republic of China
| | - Chengliang Gong
- School of Biology and Basic Medical Sciences, Soochow University, Suzhou, 215123, Jiangsu, People's Republic of China.
- National Engineering Laboratory for Modern Silk, Soochow University, Postal address: No. 199 Ren'ai Road, Suzhou Industrial Park, Suzhou, 215123, Jiangsu, People's Republic of China.
| |
Collapse
|
44
|
Neuronal migration is mediated by inositol hexakisphosphate kinase 1 via α-actinin and focal adhesion kinase. Proc Natl Acad Sci U S A 2017; 114:2036-2041. [PMID: 28154132 DOI: 10.1073/pnas.1700165114] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Inositol hexakisphosphate kinase 1 (IP6K1), which generates 5-diphosphoinositol pentakisphosphate (5-IP7), physiologically mediates numerous functions. We report that IP6K1 deletion leads to brain malformation and abnormalities of neuronal migration. IP6K1 physiologically associates with α-actinin and localizes to focal adhesions. IP6K1 deletion disrupts α-actinin's intracellular localization and function. The IP6K1 deleted cells display substantial decreases of stress fiber formation and impaired cell migration and spreading. Regulation of α-actinin by IP6K1 requires its kinase activity. Deletion of IP6K1 abolishes α-actinin tyrosine phosphorylation, which is known to be regulated by focal adhesion kinase (FAK). FAK phosphorylation is substantially decreased in IP6K1 deleted cells. 5-IP7, a product of IP6K1, promotes FAK autophosphorylation. Pharmacologic inhibition of IP6K by TNP [N2-(m-Trifluorobenzyl), N6-(p-nitrobenzyl)purine] recapitulates the phenotype of IP6K1 deletion. These findings establish that IP6K1 physiologically regulates neuronal migration by binding to α-actinin and influencing phosphorylation of both FAK and α-actinin through its product 5-IP7.
Collapse
|
45
|
Extracellular matrix-mediated cellular communication in the heart. J Mol Cell Cardiol 2016; 91:228-37. [PMID: 26778458 DOI: 10.1016/j.yjmcc.2016.01.011] [Citation(s) in RCA: 112] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 01/10/2016] [Accepted: 01/11/2016] [Indexed: 01/13/2023]
Abstract
The extracellular matrix (ECM) is a complex and dynamic scaffold that maintains tissue structure and dynamics. However, the view of the ECM as an inert architectural support has been increasingly challenged. The ECM is a vibrant meshwork, a crucial organizer of cellular microenvironments. It plays a direct role in cellular interactions regulating cell growth, survival, spreading, proliferation, differentiation and migration through the intricate relationship among cellular and acellular tissue components. This complex interrelationship preserves cardiac function during homeostasis; however it is also responsible for pathologic remodeling following myocardial injury. Therefore, enhancing our understanding of this cross-talk may provide mechanistic insights into the pathogenesis of heart failure and suggest new approaches to novel, targeted pharmacologic therapies. This review explores the implications of ECM-cell interactions in myocardial cell behavior and cardiac function at baseline and following myocardial injury.
Collapse
|
46
|
Horton ER, Byron A, Askari JA, Ng DHJ, Millon-Frémillon A, Robertson J, Koper EJ, Paul NR, Warwood S, Knight D, Humphries JD, Humphries MJ. Definition of a consensus integrin adhesome and its dynamics during adhesion complex assembly and disassembly. Nat Cell Biol 2015; 17:1577-1587. [PMID: 26479319 PMCID: PMC4663675 DOI: 10.1038/ncb3257] [Citation(s) in RCA: 404] [Impact Index Per Article: 40.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 09/18/2015] [Indexed: 12/14/2022]
Abstract
Integrin receptor activation initiates the formation of integrin adhesion complexes (IACs) at the cell membrane that transduce adhesion-dependent signals to control a multitude of cellular functions. Proteomic analyses of isolated IACs have revealed an unanticipated molecular complexity; however, a global view of the consensus composition and dynamics of IACs is lacking. Here, we have integrated several IAC proteomes and generated a 2,412-protein integrin adhesome. Analysis of this data set reveals the functional diversity of proteins in IACs and establishes a consensus adhesome of 60 proteins. The consensus adhesome is likely to represent a core cell adhesion machinery, centred around four axes comprising ILK-PINCH-kindlin, FAK-paxillin, talin-vinculin and α-actinin-zyxin-VASP, and includes underappreciated IAC components such as Rsu-1 and caldesmon. Proteomic quantification of IAC assembly and disassembly detailed the compositional dynamics of the core cell adhesion machinery. The definition of this consensus view of integrin adhesome components provides a resource for the research community.
Collapse
Affiliation(s)
- Edward R. Horton
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, UK
| | - Adam Byron
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, UK
- Edinburgh Cancer Research UK Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XR, UK
| | - Janet A. Askari
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, UK
| | - Daniel H. J. Ng
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, UK
| | - Angélique Millon-Frémillon
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, UK
| | - Joseph Robertson
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, UK
| | - Ewa J. Koper
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, UK
| | - Nikki R. Paul
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, UK
| | - Stacey Warwood
- Biological Mass Spectrometry Core Facility, Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, UK
| | - David Knight
- Biological Mass Spectrometry Core Facility, Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, UK
| | - Jonathan D. Humphries
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, UK
| | - Martin J. Humphries
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, UK
| |
Collapse
|
47
|
Qi L, Yu Y, Chi X, Xu W, Lu D, Song Y, Zhang Y, Zhang H. Kindlin-2 interacts with α-actinin-2 and β1 integrin to maintain the integrity of the Z-disc in cardiac muscles. FEBS Lett 2015; 589:2155-62. [PMID: 26143257 DOI: 10.1016/j.febslet.2015.06.022] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Revised: 06/18/2015] [Accepted: 06/21/2015] [Indexed: 01/06/2023]
Abstract
Kindlin-2, as an integrin-interacting protein, was known to be required for the maintenance of cardiac structure and function in zebrafish. However, the mechanism remains unclear. We found that Kindlin-2 interacts and colocalizes with α-actinin-2 at the Z-disc of mouse cardiac muscles and there Kindlin-2 also interacts with β1 integrin. Knockdown of Kindlin-2 influences the association of β1 integrin with α-actinin-2 and disrupts the structure of the Z-disc and leads to cardiac dysfunction. Our data indicated that Kindlin-2 is a novel α-actinin-2-interacting protein and plays an important role in the regulation of cardiac structure and function.
Collapse
Affiliation(s)
- Lihua Qi
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, and State Key Laboratory of Natural and Biomimetic Drugs, Peking University Health Science Center, Beijing 100191, China; Department of Anatomy, Histology and Embryology, Peking University Health Science Center, Beijing 100191, China
| | - Yu Yu
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, and State Key Laboratory of Natural and Biomimetic Drugs, Peking University Health Science Center, Beijing 100191, China; Department of Anatomy, Histology and Embryology, Peking University Health Science Center, Beijing 100191, China
| | - Xiaochun Chi
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, and State Key Laboratory of Natural and Biomimetic Drugs, Peking University Health Science Center, Beijing 100191, China; Department of Anatomy, Histology and Embryology, Peking University Health Science Center, Beijing 100191, China
| | - Weizhi Xu
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, and State Key Laboratory of Natural and Biomimetic Drugs, Peking University Health Science Center, Beijing 100191, China; Department of Anatomy, Histology and Embryology, Peking University Health Science Center, Beijing 100191, China
| | - Danyu Lu
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, and State Key Laboratory of Natural and Biomimetic Drugs, Peking University Health Science Center, Beijing 100191, China; Department of Anatomy, Histology and Embryology, Peking University Health Science Center, Beijing 100191, China
| | - Yao Song
- Institute for Cardiovascular Research, Peking University Health Science Center, Beijing 100191, China
| | - Youyi Zhang
- Institute for Cardiovascular Research, Peking University Health Science Center, Beijing 100191, China
| | - Hongquan Zhang
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, and State Key Laboratory of Natural and Biomimetic Drugs, Peking University Health Science Center, Beijing 100191, China; Department of Anatomy, Histology and Embryology, Peking University Health Science Center, Beijing 100191, China.
| |
Collapse
|
48
|
Integration of actin dynamics and cell adhesion by a three-dimensional, mechanosensitive molecular clutch. Nat Cell Biol 2015; 17:955-63. [PMID: 26121555 DOI: 10.1038/ncb3191] [Citation(s) in RCA: 380] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Accepted: 05/15/2015] [Indexed: 12/12/2022]
Abstract
During cell migration, the forces generated in the actin cytoskeleton are transmitted across transmembrane receptors to the extracellular matrix or other cells through a series of mechanosensitive, regulable protein-protein interactions termed the molecular clutch. In integrin-based focal adhesions, the proteins forming this linkage are organized into a conserved three-dimensional nano-architecture. Here we discuss how the physical interactions between the actin cytoskeleton and focal-adhesion-associated molecules mediate force transmission from the molecular clutch to the extracellular matrix.
Collapse
|
49
|
Rooj AK, Liu Z, McNicholas CM, Fuller CM. Physical and functional interactions between a glioma cation channel and integrin-β1 require α-actinin. Am J Physiol Cell Physiol 2015; 309:C308-19. [PMID: 26108662 DOI: 10.1152/ajpcell.00036.2015] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Accepted: 06/17/2015] [Indexed: 11/22/2022]
Abstract
Major plasma membrane components of the tumor cell, ion channels, and integrins play crucial roles in metastasis. Glioma cells express an amiloride-sensitive nonselective cation channel composed of acid-sensing ion channel (ASIC)-1 and epithelial Na(+) channel (ENaC) α- and γ-subunits. Inhibition of this channel is associated with reduced cell migration and proliferation. Using the ASIC-1 subunit as a reporter for the channel complex, we found a physical and functional interaction between this channel and integrin-β1. Short hairpin RNA knockdown of integrin-β1 attenuated the amiloride-sensitive current, which was due to loss of surface expression of ASIC-1. In contrast, upregulation of membrane expression of integrin-β1 increased the surface expression of ASIC-1. The link between the amiloride-sensitive channel and integrin-β1 was mediated by α-actinin. Downregulation of α-actinin-1 or -4 attenuated the amiloride-sensitive current. Mutation of the putative binding site for α-actinin on the COOH terminus of ASIC-1 reduced the membrane localization of ASIC-1 and also resulted in attenuation of the amiloride-sensitive current. Our data suggest a novel interaction between the amiloride-sensitive glioma cation channel and integrin-β1, mediated by α-actinin. This interaction may form a mechanism by which channel activity can regulate glioma cell proliferation and migration.
Collapse
Affiliation(s)
- Arun K Rooj
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Zhiyong Liu
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Carmel M McNicholas
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Catherine M Fuller
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
50
|
α-Actinin-4 enhances colorectal cancer cell invasion by suppressing focal adhesion maturation. PLoS One 2015; 10:e0120616. [PMID: 25860875 PMCID: PMC4393021 DOI: 10.1371/journal.pone.0120616] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2014] [Accepted: 01/24/2015] [Indexed: 01/09/2023] Open
Abstract
α-Actinins (ACTNs) are known to crosslink actin filaments at focal adhesions in migrating cells. Among the four isoforms of mammalian ACTNs, ACTN1 and ACTN4 are ubiquitously expressed. Recently, ACTN4 was reported to enhance cancer cell motility, invasion, and metastasis. However, the mechanism by which ACTN4 drives these malignant phenotypes remains unclear. Here, we show that ACTN4, but not ACTN1, induces the formation of immature focal adhesions in DLD-1 cells, leading to the rapid turnover of focal adhesions. Interestingly, zyxin (ZYX) assembly to focal adhesions was markedly decreased in ACTN4-expressing DLD-1 cells, while the recruitment of paxillin (PAX) occurred normally. On the other hand, in ACTN1-expressing DLD-1 cells, PAX and ZYX were normally recruited to focal adhesions, suggesting that ACTN4 specifically impairs focal adhesion maturation by inhibiting the recruitment of ZYX to focal complexes. Using purified recombinant proteins, we found that ZYX binding to ACTN4 was defective under conditions where ZYX binding to ACTN1 was observed. Furthermore, Matrigel invasion of SW480 cells that express high endogenous levels of ACTN4 protein was inhibited by ectopic expression of ACTN1. Altogether, our results suggest that ZYX defective binding to ACTN4, which occupies focal adhesions instead of ACTN1, induces the formation of immature focal adhesions, resulting in the enhancement of cell motility and invasion.
Collapse
|