1
|
Camp D, Venkatesh B, Solianova V, Varela L, Goult BT, Tanentzapf G. The actin binding sites of talin have both distinct and complementary roles in cell-ECM adhesion. PLoS Genet 2024; 20:e1011224. [PMID: 38662776 PMCID: PMC11075885 DOI: 10.1371/journal.pgen.1011224] [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: 09/22/2023] [Revised: 05/07/2024] [Accepted: 03/12/2024] [Indexed: 05/08/2024] Open
Abstract
Cell adhesion requires linkage of transmembrane receptors to the cytoskeleton through intermediary linker proteins. Integrin-based adhesion to the extracellular matrix (ECM) involves large adhesion complexes that contain multiple cytoskeletal adapters that connect to the actin cytoskeleton. Many of these adapters, including the essential cytoskeletal linker Talin, have been shown to contain multiple actin-binding sites (ABSs) within a single protein. To investigate the possible role of having such a variety of ways of linking integrins to the cytoskeleton, we generated mutations in multiple actin binding sites in Drosophila talin. Using this approach, we have been able to show that different actin-binding sites in talin have both unique and complementary roles in integrin-mediated adhesion. Specifically, mutations in either the C-terminal ABS3 or the centrally located ABS2 result in lethality showing that they have unique and non-redundant function in some contexts. On the other hand, flies simultaneously expressing both the ABS2 and ABS3 mutants exhibit a milder phenotype than either mutant by itself, suggesting overlap in function in other contexts. Detailed phenotypic analysis of ABS mutants elucidated the unique roles of the talin ABSs during embryonic development as well as provided support for the hypothesis that talin acts as a dimer in in vivo contexts. Overall, our work highlights how the ability of adhesion complexes to link to the cytoskeleton in multiple ways provides redundancy, and consequently robustness, but also allows a capacity for functional specialization.
Collapse
Affiliation(s)
- Darius Camp
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Bhavya Venkatesh
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Veronika Solianova
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Lorena Varela
- School of Biosciences, University of Kent, Canterbury, Kent, United Kingdom
| | - Benjamin T. Goult
- School of Biosciences, University of Kent, Canterbury, Kent, United Kingdom
- Department of Biochemistry, Cell & Systems Biology, Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Crown Street, Liverpool L69 7ZB, United Kingdom
| | - Guy Tanentzapf
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
2
|
Sacco JL, Vaneman ZT, Gomez EW. Extracellular matrix viscoelasticity regulates TGFβ1-induced epithelial-mesenchymal transition and apoptosis via integrin linked kinase. J Cell Physiol 2024; 239:e31165. [PMID: 38149820 DOI: 10.1002/jcp.31165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 10/06/2023] [Accepted: 11/17/2023] [Indexed: 12/28/2023]
Abstract
Transforming growth factor (TGF)-β1 is a multifunctional cytokine that plays important roles in health and disease. Previous studies have revealed that TGFβ1 activation, signaling, and downstream cell responses including epithelial-mesenchymal transition (EMT) and apoptosis are regulated by the elasticity or stiffness of the extracellular matrix. However, tissues within the body are not purely elastic, rather they are viscoelastic. How matrix viscoelasticity impacts cell fate decisions downstream of TGFβ1 remains unknown. Here, we synthesized polyacrylamide hydrogels that mimic the viscoelastic properties of breast tumor tissue. We found that increasing matrix viscous dissipation reduces TGFβ1-induced cell spreading, F-actin stress fiber formation, and EMT-associated gene expression changes, and promotes TGFβ1-induced apoptosis in mammary epithelial cells. Furthermore, TGFβ1-induced expression of integrin linked kinase (ILK) and colocalization of ILK with vinculin at cell adhesions is attenuated in mammary epithelial cells cultured on viscoelastic substrata in comparison to cells cultured on nearly elastic substrata. Overexpression of ILK promotes TGFβ1-induced EMT and reduces apoptosis in cells cultured on viscoelastic substrata, suggesting that ILK plays an important role in regulating cell fate downstream of TGFβ1 in response to matrix viscoelasticity.
Collapse
Affiliation(s)
- Jessica L Sacco
- Department of Chemical Engineering, The Pennsylvania State University, University Park, Pennsylvania, USA
| | - Zachary T Vaneman
- Department of Chemical Engineering, The Pennsylvania State University, University Park, Pennsylvania, USA
| | - Esther W Gomez
- Department of Chemical Engineering, The Pennsylvania State University, University Park, Pennsylvania, USA
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, Pennsylvania, USA
| |
Collapse
|
3
|
Karkali K, Pastor-Pareja JC, Martin-Blanco E. JNK signaling and integrins cooperate to maintain cell adhesion during epithelial fusion in Drosophila. Front Cell Dev Biol 2024; 11:1034484. [PMID: 38264353 PMCID: PMC10803605 DOI: 10.3389/fcell.2023.1034484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 12/13/2023] [Indexed: 01/25/2024] Open
Abstract
The fusion of epithelial sheets is an essential and conserved morphogenetic event that requires the maintenance of tissue continuity. This is secured by membrane-bound or diffusible signals that instruct the epithelial cells, in a coordinated fashion, to change shapes and adhesive properties and when, how and where to move. Here we show that during Dorsal Closure (DC) in Drosophila, the Jun kinase (JNK) signaling pathway modulates integrins expression and ensures tissue endurance. An excess of JNK activity, as an outcome of a failure in the negative feedback implemented by the dual-specificity phosphatase Puckered (Puc), promotes the loss of integrins [the ß-subunit Myospheroid (Mys)] and amnioserosa detachment. Likewise, integrins signal back to the pathway to regulate the duration and strength of JNK activity. Mys is necessary for the regulation of JNK activity levels and in its absence, puc expression is downregulated and JNK activity increases.
Collapse
Affiliation(s)
- Katerina Karkali
- Instituto de Biologia Molecular de Barcelona, Consejo Superior de Investigaciones Científicas (IBMB-CSIC), Barcelona, Spain
| | - Jose Carlos Pastor-Pareja
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas (IN-CSIC), Alicante, Spain
| | - Enrique Martin-Blanco
- Instituto de Biologia Molecular de Barcelona, Consejo Superior de Investigaciones Científicas (IBMB-CSIC), Barcelona, Spain
| |
Collapse
|
4
|
Ready DF, Chang HC. Interommatidial cells build a tensile collagen network during Drosophila retinal morphogenesis. Curr Biol 2023; 33:2223-2234.e3. [PMID: 37209679 PMCID: PMC10247444 DOI: 10.1016/j.cub.2023.04.066] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 03/27/2023] [Accepted: 04/27/2023] [Indexed: 05/22/2023]
Abstract
Drosophila compound eye morphogenesis transforms a simple epithelium into an approximate hollow hemisphere comprised of ∼700 ommatidia, packed as tapering hexagonal prisms between a rigid external array of cuticular lenses and a parallel, rigid internal floor, the fenestrated membrane (FM). Critical to vision, photosensory rhabdomeres are sprung between these two surfaces, grading their length and shape accurately across the eye and aligning them to the optical axis. Using fluorescently tagged collagen and laminin, we show that that the FM assembles sequentially, emerging in the larval eye disc in the wake of the morphogenetic furrow as the original collagen-containing basement membrane (BM) separates from the epithelial floor and is replaced by a new, laminin-rich BM, which advances around axon bundles of newly differentiated photoreceptors as they exit the retina, forming fenestrae in this new, laminin-rich BM. In mid-pupal development, the interommatidial cells (IOCs) autonomously deposit collagen at fenestrae, forming rigid, tension-resisting grommets. In turn, stress fibers assemble in the IOC basal endfeet, where they contact grommets at anchorages mediated by integrin linked kinase (ILK). The hexagonal network of IOC endfeet tiling the retinal floor couples nearest-neighbor grommets into a supracellular tri-axial tension network. Late in pupal development, IOC stress fiber contraction folds pliable BM into a hexagonal grid of collagen-stiffened ridges, concomitantly decreasing the area of convex FM and applying essential morphogenetic longitudinal tension to rapidly growing rhabdomeres. Together, our results reveal an orderly program of sequential assembly and activation of a supramolecular tensile network that governs Drosophila retinal morphogenesis.
Collapse
Affiliation(s)
- Donald F Ready
- Department of Biological Sciences, Purdue University, 915 West State Street, West Lafayette, IN 47907-2054, USA
| | - Henry C Chang
- Department of Biological Sciences, Purdue University, 915 West State Street, West Lafayette, IN 47907-2054, USA.
| |
Collapse
|
5
|
Wang Y, Zhang H, Wu S, Wan W, Kang X, Gao B, Shi H, Zhao S, Niu L, Zou R. Substrate Stiffness Regulates the Proliferation and Apoptosis of Periodontal Ligament Cells through Integrin-Linked Kinase ILK. ACS Biomater Sci Eng 2023; 9:662-670. [PMID: 36732940 DOI: 10.1021/acsbiomaterials.2c00925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The hallmark of orthodontic tooth movement (OTM) is time-consuming during clinical treatments. The acceleration of OTM through modulating proliferation and apoptosis of periodontal ligament cells (PDLCs) possesses the potential application in clinical treatments. Here, we established an in vitro model with a graded increase in substrate stiffness to investigate the underlying mechanism of proliferation and apoptosis of PDLCs. The role of the integrin-linked kinase (ILK) in response to substrate stiffness was investigated by the depletion model of PDLCs. We found that the proliferation and apoptosis of PDLCs show a stiffness-dependent property with stiffer substrates favoring increased bias at the transcript level. Depleting integrin-linked kinase diluted the correlation between PDLCs behaviors and substrate stiffness. Our results suggest that ILK plays a significant role in modulating PDLC proliferation and apoptosis and can serve as a potential target for accelerating OTM.
Collapse
Affiliation(s)
- Yijie Wang
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an 710004, China.,Clinical Research Center of Shaanxi Province for Dental and Maxillofacial Diseases, College of Stomatology, Xi'an Jiaotong University, Xi'an 710004, China.,College of Stomatology, Xi'an Jiaotong University, Xi'an 710004, China
| | - Hui Zhang
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an 710004, China.,Clinical Research Center of Shaanxi Province for Dental and Maxillofacial Diseases, College of Stomatology, Xi'an Jiaotong University, Xi'an 710004, China.,College of Stomatology, Xi'an Jiaotong University, Xi'an 710004, China.,Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, China
| | - Shiyang Wu
- College of Stomatology, Xi'an Jiaotong University, Xi'an 710004, China
| | - Wanting Wan
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an 710004, China.,Clinical Research Center of Shaanxi Province for Dental and Maxillofacial Diseases, College of Stomatology, Xi'an Jiaotong University, Xi'an 710004, China.,College of Stomatology, Xi'an Jiaotong University, Xi'an 710004, China
| | - Xueping Kang
- College of Stomatology, Xi'an Jiaotong University, Xi'an 710004, China
| | - Bei Gao
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an 710004, China.,Clinical Research Center of Shaanxi Province for Dental and Maxillofacial Diseases, College of Stomatology, Xi'an Jiaotong University, Xi'an 710004, China.,College of Stomatology, Xi'an Jiaotong University, Xi'an 710004, China
| | - Haoyu Shi
- Xi'an Jiaotong University Health Science Center, Xi'an 710061, China
| | - Shuyang Zhao
- Xi'an Jiaotong University Health Science Center, Xi'an 710061, China
| | - Lin Niu
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an 710004, China.,Clinical Research Center of Shaanxi Province for Dental and Maxillofacial Diseases, College of Stomatology, Xi'an Jiaotong University, Xi'an 710004, China.,College of Stomatology, Xi'an Jiaotong University, Xi'an 710004, China
| | - Rui Zou
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an 710004, China.,Clinical Research Center of Shaanxi Province for Dental and Maxillofacial Diseases, College of Stomatology, Xi'an Jiaotong University, Xi'an 710004, China.,College of Stomatology, Xi'an Jiaotong University, Xi'an 710004, China
| |
Collapse
|
6
|
Ihog proteins contribute to integrin-mediated focal adhesions. SCIENCE CHINA. LIFE SCIENCES 2023; 66:366-375. [PMID: 36103028 DOI: 10.1007/s11427-022-2154-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 06/27/2022] [Indexed: 10/14/2022]
Abstract
Integrin expression forms focal adhesions, but how this process is physiologically regulated is unclear. Ihog proteins are evolutionarily conserved, playing roles in Hedgehog signaling and serving as trans-homophilic adhesion molecules to mediate cell-cell interactions. Whether these proteins are also engaged in other cell adhesion processes remains unknown. Here, we report that Drosophila Ihog proteins function in the integrin-mediated adhesions. Removal of Ihog proteins causes blister and spheroidal muscle in wings and embryos, respectively. We demonstrate that Ihog proteins interact with integrin via the extracellular portion and that their removal perturbs integrin distribution. Finally, we show that Boc, a mammalian Ihog protein, rescues the embryonic defects caused by removing its Drosophila homologs. We thus propose that Ihog proteins contribute to integrin-mediated focal adhesions.
Collapse
|
7
|
Liao H, Wang F, Lu K, Ma X, Yan J, Luo L, Sun Y, Liang X. Requirement for PINCH in skeletal myoblast differentiation. Cell Tissue Res 2023; 391:205-215. [PMID: 36385586 PMCID: PMC9839796 DOI: 10.1007/s00441-022-03701-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 11/06/2022] [Indexed: 11/18/2022]
Abstract
PINCH, an adaptor of focal adhesion complex, plays essential roles in multiple cellular processes and organogenesis. Here, we ablated PINCH1 or both of PINCH1 and PINCH2 in skeletal muscle progenitors using MyoD-Cre. Double ablation of PINCH1 and PINCH2 resulted in early postnatal lethality with reduced size of skeletal muscles and detachment of diaphragm muscles from the body wall. PINCH mutant myofibers failed to undergo multinucleation and exhibited disrupted sarcomere structures. The mutant myoblasts in culture were able to adhere to newly formed myotubes but impeded in cell fusion and subsequent sarcomere genesis and cytoskeleton organization. Consistent with this, expression of integrin β1 and some cytoskeleton proteins and phosphorylation of ERK and AKT were significantly reduced in PINCH mutants. However, N-cadherin was correctly expressed at cell adhesion sites in PINCH mutant cells, suggesting that PINCH may play a direct role in myoblast fusion. Expression of MRF4, the most highly expressed myogenic factor at late stages of myogenesis, was abolished in PINCH mutants that could contribute to observed phenotypes. In addition, mice with PINCH1 being ablated in myogenic progenitors exhibited only mild centronuclear myopathic changes, suggesting a compensatory role of PINCH2 in myogenic differentiation. Our results revealed a critical role of PINCH proteins in myogenic differentiation.
Collapse
Affiliation(s)
- Huimin Liao
- Key Laboratory of Arrhythmia, Ministry of Education, East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China
| | - Fei Wang
- Key Laboratory of Arrhythmia, Ministry of Education, East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China
| | - Ke Lu
- Key Laboratory of Arrhythmia, Ministry of Education, East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China
| | - Xiaolei Ma
- Key Laboratory of Arrhythmia, Ministry of Education, East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China
| | - Jie Yan
- Key Laboratory of Arrhythmia, Ministry of Education, East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China
| | - Lina Luo
- Key Laboratory of Arrhythmia, Ministry of Education, East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China
| | - Yunfu Sun
- Key Laboratory of Arrhythmia, Ministry of Education, East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China.
| | - Xingqun Liang
- Key Laboratory of Arrhythmia, Ministry of Education, East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China.
| |
Collapse
|
8
|
Keramidioti A, Golegou E, Psarra E, Paschalidis N, Kalodimou K, Yamamoto S, Delidakis C, Vakaloglou KM, Zervas CG. Epithelial morphogenesis in the Drosophila egg chamber requires Parvin and ILK. Front Cell Dev Biol 2022; 10:951082. [PMID: 36531940 PMCID: PMC9752845 DOI: 10.3389/fcell.2022.951082] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 11/21/2022] [Indexed: 03/11/2024] Open
Abstract
Integrins are the major family of transmembrane proteins that mediate cell-matrix adhesion and have a critical role in epithelial morphogenesis. Integrin function largely depends on the indirect connection of the integrin cytoplasmic tail to the actin cytoskeleton through an intracellular protein network, the integrin adhesome. What is currently unknown is the role of individual integrin adhesome components in epithelia dynamic reorganization. Drosophila egg chamber consists of the oocyte encircled by a monolayer of somatic follicle epithelial cells that undergo specific cell shape changes. Egg chamber morphogenesis depends on a developmental array of cell-cell and cell-matrix signalling events. Recent elegant work on the role of integrins in the Drosophila egg chamber has indicated their essential role in the early stages of oogenesis when the pre-follicle cells assemble into the follicle epithelium. Here, we have focused on the functional requirement of two key integrin adhesome components, Parvin and Integrin-Linked Kinase (ILK). Both proteins are expressed in the developing ovary from pupae to the adult stage and display enriched expression in terminal filament and stalk cells, while their genetic removal from early germaria results in severe disruption of the subsequent oogenesis, leading to female sterility. Combining genetic mosaic analysis of available null alleles for both Parvin and Ilk with conditional rescue utilizing the UAS/Gal4 system, we found that Parvin and ILK are required in pre-follicle cells for germline cyst encapsulation and stalk cell morphogenesis. Collectively, we have uncovered novel developmental functions for both Parvin and ILK, which closely synergize with integrins in epithelia.
Collapse
Affiliation(s)
- Athina Keramidioti
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, Athens, Greece
- Department of Biochemistry and Biotechnology, University of Thessaly, Larissa, Greece
| | - Evgenia Golegou
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Eleni Psarra
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Nikolaos Paschalidis
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Konstantina Kalodimou
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Shinya Yamamoto
- Department of Molecular and Human Genetics, Department of Neuroscience (BCM), The Development Disease Models and Therapeutics Graduate Program, Baylor College of Medicine (BCM), Texas Children’s Hospital (TCH), Program in Developmental Biology (BCM), Jan and Dan Duncan Neurological Research Institute, Houston, TX, United States
| | - Christos Delidakis
- Department of Biology, University of Crete, Iraklio, Greece
- Foundation for Research and Technology-Hellas (FORTH), Institute of Molecular Biology and Biotechnology (IMBB), Iraklio, Greece
| | - Katerina M. Vakaloglou
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Christos G. Zervas
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| |
Collapse
|
9
|
Qu M, Yu K, Rehman Aziz AU, Zhang H, Zhang Z, Li N, Liu B. The role of Actopaxin in tumor metastasis. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2022; 175:90-102. [PMID: 36150525 DOI: 10.1016/j.pbiomolbio.2022.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 08/06/2022] [Accepted: 09/13/2022] [Indexed: 06/16/2023]
Abstract
Actopaxin is a newly discovered focal adhesions (FAs) protein, actin-binding protein and pseudopodia-enriched molecule. It can not only bind to a variety of FAs proteins (such as Paxillin, ILK and PINCH) and non-FAs proteins (such as TESK1, CdGAP, β2-adaptin, G3BP2, ADAR1 and CD29), but also participates in multiple signaling pathways. Thus, it plays a crucial role in regulating important processes of tumor metastasis, including matrix degradation, migration, and invasion, etc. This review covers the latest progress in the structure and function of Actopaxin, its interaction with other proteins as well as its involvement in regulating tumor development and metastasis. Additionally, the current limitations for Actopaxin related studies and the possible research directions on it in the future are also discussed. It is hoped that this review can assist relevant researchers to obtain a deep understanding of the role that Actopaxin plays in tumor progression, and also enlighten further research and development of therapeutic approaches for the treatment of tumor metastasis.
Collapse
Affiliation(s)
- Manrong Qu
- School of Biomedical Engineering, Dalian University of Technology, Key Laboratory for Integrated Circuit and Biomedical Electronic System of Liaoning Province, Dalian, 116024, China
| | - Kehui Yu
- School of Biomedical Engineering, Dalian University of Technology, Key Laboratory for Integrated Circuit and Biomedical Electronic System of Liaoning Province, Dalian, 116024, China
| | - Aziz Ur Rehman Aziz
- School of Biomedical Engineering, Dalian University of Technology, Key Laboratory for Integrated Circuit and Biomedical Electronic System of Liaoning Province, Dalian, 116024, China
| | - Hangyu Zhang
- School of Biomedical Engineering, Dalian University of Technology, Key Laboratory for Integrated Circuit and Biomedical Electronic System of Liaoning Province, Dalian, 116024, China
| | - Zhengyao Zhang
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, 124221, China
| | - Na Li
- School of Biomedical Engineering, Dalian University of Technology, Key Laboratory for Integrated Circuit and Biomedical Electronic System of Liaoning Province, Dalian, 116024, China.
| | - Bo Liu
- School of Biomedical Engineering, Dalian University of Technology, Key Laboratory for Integrated Circuit and Biomedical Electronic System of Liaoning Province, Dalian, 116024, China.
| |
Collapse
|
10
|
Draicchio F, Behrends V, Tillin NA, Hurren NM, Sylow L, Mackenzie R. Involvement of the extracellular matrix and integrin signalling proteins in skeletal muscle glucose uptake. J Physiol 2022; 600:4393-4408. [PMID: 36054466 PMCID: PMC9826115 DOI: 10.1113/jp283039] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 08/03/2022] [Indexed: 01/11/2023] Open
Abstract
Whole-body euglycaemia is partly maintained by two cellular processes that encourage glucose uptake in skeletal muscle, the insulin- and contraction-stimulated pathways, with research suggesting convergence between these two processes. The normal structural integrity of the skeletal muscle requires an intact actin cytoskeleton as well as integrin-associated proteins, and thus those structures are likely fundamental for effective glucose uptake in skeletal muscle. In contrast, excessive extracellular matrix (ECM) remodelling and integrin expression in skeletal muscle may contribute to insulin resistance owing to an increased physical barrier causing reduced nutrient and hormonal flux. This review explores the role of the ECM and the actin cytoskeleton in insulin- and contraction-mediated glucose uptake in skeletal muscle. This is a clinically important area of research given that defects in the structural integrity of the ECM and integrin-associated proteins may contribute to loss of muscle function and decreased glucose uptake in type 2 diabetes.
Collapse
Affiliation(s)
- Fulvia Draicchio
- School of Life and Health SciencesWhitelands CollegeUniversity of RoehamptonLondonUK
| | - Volker Behrends
- School of Life and Health SciencesWhitelands CollegeUniversity of RoehamptonLondonUK
| | - Neale A. Tillin
- School of Life and Health SciencesWhitelands CollegeUniversity of RoehamptonLondonUK
| | - Nicholas M. Hurren
- School of Life and Health SciencesWhitelands CollegeUniversity of RoehamptonLondonUK
| | - Lykke Sylow
- Molecular Metabolism in Cancer & Ageing Research GroupDepartment of Biomedical SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Richard Mackenzie
- School of Life and Health SciencesWhitelands CollegeUniversity of RoehamptonLondonUK
| |
Collapse
|
11
|
Kausar S, Abbas MN, Gul I, Liu Y, Tang BP, Maqsood I, Liu QN, Dai LS. Integrins in the Immunity of Insects: A Review. Front Immunol 2022; 13:906294. [PMID: 35757717 PMCID: PMC9218073 DOI: 10.3389/fimmu.2022.906294] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 05/02/2022] [Indexed: 12/30/2022] Open
Abstract
Integrins are a large group of cell-surface proteins that are classified as transmembrane proteins. Integrins are classified into different types based on sequence variations, leading to structural and functional diversity. They are broadly distributed in animals and have a wide range of biological functions such as cell-to-cell communication, intracellular cytoskeleton organization, cellular signaling, immune responses, etc. Integrins are among the most abundant cell surface proteins in insects, exhibiting their indispensability in insect physiology. Because of their critical biological involvement in physiological processes, they appear to be a novel target for designing effective pest control strategies. In the current literature review, we first discuss the discovery and expression responses of integrins against various types of pathogens. Secondly, we examine the specific biological roles of integrins in controlling microbial pathogens, such as phagocytosis, encapsulation, nodulation, immune signaling, and so on. Finally, we describe the possible uses of integrins to control agricultural insect pests.
Collapse
Affiliation(s)
- Saima Kausar
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, China
| | - Muhammad Nadeem Abbas
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, China
| | - Isma Gul
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, China
| | - Yu Liu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Bo-Ping Tang
- Jiangsu Key Laboratory for Bioresources of Saline Soils, Jiangsu Synthetic Innovation Center for Coastal Bio-agriculture, Jiangsu Provincial Key Laboratory of Coastal Wetland Bioresources and Environmental Protection, School of Wetlands, Yancheng Teachers University, Yancheng, China
| | - Iram Maqsood
- Department of Zoology, Shaheed Benazir Bhutto Woman University, Peshawar, Pakistan
| | - Qiu-Ning Liu
- Jiangsu Key Laboratory for Bioresources of Saline Soils, Jiangsu Synthetic Innovation Center for Coastal Bio-agriculture, Jiangsu Provincial Key Laboratory of Coastal Wetland Bioresources and Environmental Protection, School of Wetlands, Yancheng Teachers University, Yancheng, China.,Key Laboratory of Insect Developmental and Evolutionary Biology, Chinese Academy of Sciences (CAS) Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
| | - Li-Shang Dai
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
12
|
Górska A, Mazur AJ. Integrin-linked kinase (ILK): the known vs. the unknown and perspectives. Cell Mol Life Sci 2022; 79:100. [PMID: 35089438 PMCID: PMC8799556 DOI: 10.1007/s00018-021-04104-1] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 11/29/2021] [Accepted: 12/17/2021] [Indexed: 02/08/2023]
Abstract
Integrin-linked kinase (ILK) is a multifunctional molecular actor in cell-matrix interactions, cell adhesion, and anchorage-dependent cell growth. It combines functions of a signal transductor and a scaffold protein through its interaction with integrins, then facilitating further protein recruitment within the ILK-PINCH-Parvin complex. ILK is involved in crucial cellular processes including proliferation, survival, differentiation, migration, invasion, and angiogenesis, which reflects on systemic changes in the kidney, heart, muscle, skin, and vascular system, also during the embryonal development. Dysfunction of ILK underlies the pathogenesis of various diseases, including the pro-oncogenic activity in tumorigenesis. ILK localizes mostly to the cell membrane and remains an important component of focal adhesion. We do know much about ILK but a lot still remains either uncovered or unclear. Although it was initially classified as a serine/threonine-protein kinase, its catalytical activity is now questioned due to structural and functional issues, leaving the exact molecular mechanism of signal transduction by ILK unsolved. While it is known that the three isoforms of ILK vary in length, the presence of crucial domains, and modification sites, most of the research tends to focus on the main isoform of this protein while the issue of functional differences of ILK2 and ILK3 still awaits clarification. The activity of ILK is regulated on the transcriptional, protein, and post-transcriptional levels. The crucial role of phosphorylation and ubiquitylation has been investigated, but the functions of the vast majority of modifications are still unknown. In the light of all those open issues, here we present an extensive literature survey covering a wide spectrum of latest findings as well as a past-to-present view on controversies regarding ILK, finishing with pointing out some open questions to be resolved by further research.
Collapse
Affiliation(s)
- Agata Górska
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, ul. Joliot-Curie 14a, 50-383, Wrocław, Poland.
| | - Antonina Joanna Mazur
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, ul. Joliot-Curie 14a, 50-383, Wrocław, Poland.
| |
Collapse
|
13
|
Barrera-Velázquez M, Ríos-Barrera LD. Crosstalk between basal extracellular matrix adhesion and building of apical architecture during morphogenesis. Biol Open 2021; 10:bio058760. [PMID: 34842274 PMCID: PMC8649640 DOI: 10.1242/bio.058760] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Tissues build complex structures like lumens and microvilli to carry out their functions. Most of the mechanisms used to build these structures rely on cells remodelling their apical plasma membranes, which ultimately constitute the specialised compartments. In addition to apical remodelling, these shape changes also depend on the proper attachment of the basal plasma membrane to the extracellular matrix (ECM). The ECM provides cues to establish apicobasal polarity, and it also transduces forces that allow apical remodelling. However, physical crosstalk mechanisms between basal ECM attachment and the apical plasma membrane remain understudied, and the ones described so far are very diverse, which highlights the importance of identifying the general principles. Here, we review apicobasal crosstalk of two well-established models of membrane remodelling taking place during Drosophila melanogaster embryogenesis: amnioserosa cell shape oscillations during dorsal closure and subcellular tube formation in tracheal cells. We discuss how anchoring to the basal ECM affects apical architecture and the mechanisms that mediate these interactions. We analyse this knowledge under the scope of other morphogenetic processes and discuss what aspects of apicobasal crosstalk may represent widespread phenomena and which ones are used to build subsets of specialised compartments.
Collapse
Affiliation(s)
- Mariana Barrera-Velázquez
- Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, Mexico City 04510, Mexico
- Undergraduate Program on Genomic Sciences, Centro de Ciencias Genómicas, Universidad Nacional Autónoma de México, Cuernavaca, Morelos 62210, Mexico
| | - Luis Daniel Ríos-Barrera
- Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, Mexico City 04510, Mexico
| |
Collapse
|
14
|
Abstract
Integrin linked kinase (ILK) is a vital signaling protein ubiquitously expressed throughout the body. It binds to intracellular integrins to help promote signaling related to cell adhesion, apoptosis, proliferation, migration, and a plethora of other common cellular functions. In this review, ILKs role in the liver is detailed. Studies have shown ILK to be a major participant in hepatic ECM organization, liver regeneration, insulin resistance, and hepatocellular carcinoma.
Collapse
Affiliation(s)
- Nicole Martucci
- Department of Pathology, University of Pittsburgh School of MedicinePittsburgh, PAUSA
| | | | - Wendy M Mars
- Department of Pathology, University of Pittsburgh School of MedicinePittsburgh, PAUSA
| |
Collapse
|
15
|
Natarajaseenivasan K, Shanmughapriya S, Velusamy P, Sayre M, Garcia A, Gomez NM, Langford D. Inflammation-induced PINCH expression leads to actin depolymerization and mitochondrial mislocalization in neurons. Transl Neurodegener 2020; 9:32. [PMID: 32746944 PMCID: PMC7397656 DOI: 10.1186/s40035-020-00211-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 07/21/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Diseases and disorders with a chronic neuroinflammatory component are often linked with changes in brain metabolism. Among neurodegenerative disorders, people living with human immunodeficiency virus (HIV) and Alzheimer's disease (AD) are particularly vulnerable to metabolic disturbances, but the mechanistic connections of inflammation, neurodegeneration and bioenergetic deficits in the central nervous system (CNS) are poorly defined. The particularly interesting new cysteine histidine-rich-protein (PINCH) is nearly undetectable in healthy mature neurons, but is robustly expressed in tauopathy-associated neurodegenerative diseases including HIV infection and AD. Although robust PINCH expression has been reported in neurons in the brains of patients with HIV and AD, the molecular mechanisms and cellular consequences of increased PINCH expression in CNS disease remain largely unknown. METHODS We investigated the regulatory mechanisms responsible for PINCH protein-mediated changes in bioenergetics, mitochondrial subcellular localization and bioenergetic deficits in neurons exposed to physiological levels of TNFα or the HIV protein Tat. Changes in the PINCH-ILK-Parvin (PIP) complex association with cofilin and TESK1 were assessed to identify factors responsible for actin depolymerization and mitochondrial mislocalization. Lentiviral and pharmacological inhibition experiments were conducted to confirm PINCH specificity and to reinstate proper protein-protein complex communication. RESULTS We identified MEF2A as the PINCH transcription factor in neuroinflammation and determined the biological consequences of increased PINCH in neurons. TNFα-mediated activation of MEF2A via increased cellular calcium induced PINCH, leading to disruption of the PIP ternary complex, cofilin activation by TESK1 inactivation, and actin depolymerization. The disruption of actin led to perinuclear mislocalization of mitochondria by destabilizing the kinesin-dependent mitochondrial transport machinery, resulting in impaired neuronal metabolism. Blocking TNFα-induced PINCH expression preserved mitochondrial localization and maintained metabolic functioning. CONCLUSIONS This study reported for the first time the mechanistic and biological consequences of PINCH expression in CNS neurons in diseases with a chronic neuroinflammation component. Our findings point to the maintenance of PINCH at normal physiological levels as a potential new therapeutic target for neurodegenerative diseases with impaired metabolisms.
Collapse
Affiliation(s)
- Kalimuthusamy Natarajaseenivasan
- Department of Neurosciences and Center for Neurovirology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140 USA
- Department of Microbiology, Bharathidasan University, Tiruchirappalli, 620024 India
| | - Santhanam Shanmughapriya
- Heart and Vascular Institute, Department of Medicine, Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA 17033 USA
| | - Prema Velusamy
- Heart and Vascular Institute, Department of Medicine, Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA 17033 USA
| | - Matthew Sayre
- Department of Neurosciences and Center for Neurovirology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140 USA
| | - Alvaro Garcia
- Department of Neurosciences and Center for Neurovirology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140 USA
| | - Nestor Mas Gomez
- Department of Neurosciences and Center for Neurovirology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140 USA
| | - Dianne Langford
- Department of Neurosciences and Center for Neurovirology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140 USA
| |
Collapse
|
16
|
ILK silencing inhibits migration and invasion of more invasive glioblastoma cells by downregulating ROCK1 and Fascin-1. Mol Cell Biochem 2020; 471:143-153. [PMID: 32506247 DOI: 10.1007/s11010-020-03774-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 05/31/2020] [Indexed: 12/23/2022]
Abstract
Glioblastoma multiforme (GBM) is the most aggressive type of brain tumor and it is associated with poor survival. Integrin-linked kinase (ILK) is a serine/threonine protein pseudo-kinase that binds to the cytoplasmic domains of β1 and β3 integrins and has been previously shown to promote invasion and metastasis in many cancer types, including GBM. However, little is known regarding the exact molecular mechanism implicating ILK in GBM aggressiveness. In this study, we used two brain cell lines, the non-invasive neuroglioma H4 cells, and the highly invasive glioblastoma A172 cells, which express ILK in much higher levels than H4. We studied the effect of ILK silencing on the metastatic behavior of glioblastoma cells in vitro and elucidate the underlying molecular mechanism. We showed that siRNA-mediated silencing of ILK inhibits cell migration and invasion of the highly invasive A172 cells while it does not affect the migratory and invasive capacity of H4 cells. These data were also supported by respective changes in the expression of Rho-associated kinase 1 (ROCK1), fascin actin-bundling protein 1 (FSCN1), and matrix metalloproteinase 13 (MMP13), which are known to regulate cell migration and invasion. Our findings were further corroborated by analyzing the Cancer Genome Atlas Glioblastoma Multiforme (TCGA-GBM) dataset. We conclude that ILK promotes glioblastoma cell invasion through activation of ROCK1 and FSCN1 in vitro, providing a more exact molecular mechanism for its action.
Collapse
|
17
|
Gupta A, Gözen I, Taylor M. A cellular automaton for modeling non-trivial biomembrane ruptures. SOFT MATTER 2019; 15:4178-4186. [PMID: 31062781 DOI: 10.1039/c8sm02032a] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
A novel cellular automaton (CA) for simulating biological membrane rupture is proposed. Constructed via simple rules governing deformation, tension, and fracture, the CA incorporates ideas from standard percolation models and bond-based fracture methods. The model is demonstrated by comparing simulations with experimental results of a double bilayer lipid membrane expanding on a solid substrate. Results indicate that the CA can capture non-trivial rupture morphologies such as floral patterns and the saltatory dynamics of fractal avalanches observed in experiments. Moreover, the CA provides insight into the poorly understood role of inter-layer adhesion, supporting the hypothesis that the density of adhesion sites governs rupture morphology.
Collapse
Affiliation(s)
- Abhay Gupta
- Department of Mechanical Engineering, Santa Clara University, Santa Clara, California, USA.
| | | | | |
Collapse
|
18
|
Camp D, Haage A, Solianova V, Castle WM, Xu QA, Lostchuck E, Goult BT, Tanentzapf G. Direct binding of Talin to Rap1 is required for cell-ECM adhesion in Drosophila. J Cell Sci 2018; 131:jcs.225144. [PMID: 30446511 DOI: 10.1242/jcs.225144] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 11/08/2018] [Indexed: 12/27/2022] Open
Abstract
Attachment of cells to the extracellular matrix (ECM) via integrins is essential for animal development and tissue maintenance. The cytoplasmic protein Talin (encoded by rhea in flies) is necessary for linking integrins to the cytoskeleton, and its recruitment is a key step in the assembly of the adhesion complex. However, the mechanisms that regulate Talin recruitment to sites of adhesion in vivo are still not well understood. Here, we show that Talin recruitment to, and maintenance at, sites of integrin-mediated adhesion requires a direct interaction between Talin and the GTPase Rap1. A mutation that blocks the direct binding of Talin to Rap1 abolished Talin recruitment to sites of adhesion and the resulting phenotype phenocopies that seen with null alleles of Talin. Moreover, we show that Rap1 activity modulates Talin recruitment to sites of adhesion via its direct binding to Talin. These results identify the direct Talin-Rap1 interaction as a key in vivo mechanism for controlling integrin-mediated cell-ECM adhesion.
Collapse
Affiliation(s)
- Darius Camp
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada V6T 1Z3
| | - Amanda Haage
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada V6T 1Z3
| | - Veronika Solianova
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada V6T 1Z3
| | - William M Castle
- School of Biosciences, University of Kent, Canterbury CT2 7NJ, UK
| | - Qinyuan A Xu
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada V6T 1Z3
| | - Emily Lostchuck
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada V6T 1Z3
| | - Benjamin T Goult
- School of Biosciences, University of Kent, Canterbury CT2 7NJ, UK
| | - Guy Tanentzapf
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada V6T 1Z3
| |
Collapse
|
19
|
Urner S, Planas-Paz L, Hilger LS, Henning C, Branopolski A, Kelly-Goss M, Stanczuk L, Pitter B, Montanez E, Peirce SM, Mäkinen T, Lammert E. Identification of ILK as a critical regulator of VEGFR3 signalling and lymphatic vascular growth. EMBO J 2018; 38:embj.201899322. [PMID: 30518533 PMCID: PMC6331728 DOI: 10.15252/embj.201899322] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 11/05/2018] [Accepted: 11/07/2018] [Indexed: 12/20/2022] Open
Abstract
Vascular endothelial growth factor receptor‐3 (VEGFR3) signalling promotes lymphangiogenesis. While there are many reported mechanisms of VEGFR3 activation, there is little understanding of how VEGFR3 signalling is attenuated to prevent lymphatic vascular overgrowth and ensure proper lymph vessel development. Here, we show that endothelial cell‐specific depletion of integrin‐linked kinase (ILK) in mouse embryos hyper‐activates VEGFR3 signalling and leads to overgrowth of the jugular lymph sacs/primordial thoracic ducts, oedema and embryonic lethality. Lymphatic endothelial cell (LEC)‐specific deletion of Ilk in adult mice initiates lymphatic vascular expansion in different organs, including cornea, skin and myocardium. Knockdown of ILK in human LECs triggers VEGFR3 tyrosine phosphorylation and proliferation. ILK is further found to impede interactions between VEGFR3 and β1 integrin in vitro and in vivo, and endothelial cell‐specific deletion of an Itgb1 allele rescues the excessive lymphatic vascular growth observed upon ILK depletion. Finally, mechanical stimulation disrupts the assembly of ILK and β1 integrin, releasing the integrin to enable its interaction with VEGFR3. Our data suggest that ILK facilitates mechanically regulated VEGFR3 signalling via controlling its interaction with β1 integrin and thus ensures proper development of lymphatic vessels.
Collapse
Affiliation(s)
- Sofia Urner
- Institute of Metabolic Physiology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Lara Planas-Paz
- Institute of Metabolic Physiology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Laura Sophie Hilger
- Institute of Metabolic Physiology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Carina Henning
- Institute of Metabolic Physiology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Anna Branopolski
- Institute of Metabolic Physiology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany.,Division of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Molly Kelly-Goss
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Lukas Stanczuk
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Bettina Pitter
- Walter-Brendel-Center of Experimental Medicine, University Hospital, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Eloi Montanez
- Walter-Brendel-Center of Experimental Medicine, University Hospital, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Shayn M Peirce
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Taija Mäkinen
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Eckhard Lammert
- Institute of Metabolic Physiology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany .,Institute for Beta Cell Biology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany.,German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| |
Collapse
|
20
|
Non-catalytic signaling by pseudokinase ILK for regulating cell adhesion. Nat Commun 2018; 9:4465. [PMID: 30367047 PMCID: PMC6203859 DOI: 10.1038/s41467-018-06906-7] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 10/04/2018] [Indexed: 01/01/2023] Open
Abstract
Dynamic communication between integrin-containing complexes (focal adhesions, FAs) and actin filaments is critical for regulating cell adhesion. Pseudokinase ILK plays a key role in this process but the underlying mechanism remains highly elusive. Here we show that by recruiting FA adaptors PINCH and Parvin into a heterotrimeric complex (IPP), ILK triggers F-actin filament bundling - a process known to generate force/mechanical signal to promote cytoskeleton reassembly and dynamic cell adhesion. Structural, biochemical, and functional analyses revealed that the F-actin bundling is orchestrated by two previously unrecognized WASP-Homology-2 actin binding motifs within IPP, one from PINCH and the other from Parvin. Strikingly, this process is also sensitized to Mg-ATP bound to the pseudoactive site of ILK and its dysregulation severely impairs stress fibers formation, cell spreading, and migration. These data identify a crucial mechanism for ILK, highlighting its uniqueness as a pseudokinase to transduce non-catalytic signal and regulate cell adhesion.
Collapse
|
21
|
OSU-T315 as an Interesting Lead Molecule for Novel B Cell-Specific Therapeutics. J Immunol Res 2018; 2018:2505818. [PMID: 30276218 PMCID: PMC6157143 DOI: 10.1155/2018/2505818] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 08/09/2018] [Indexed: 11/17/2022] Open
Abstract
B cells are pathogenic in various disease processes and therefore represent an interesting target for the development of novel immunosuppressants. In the search for new therapeutic molecules, we utilized an in vitro B cell activation assay with ODN2006-stimulated Namalwa cells to screen a chemical library of small molecules for B cell modulating effects. OSU-T315, described as an inhibitor of integrin-linked kinase (ILK), was hereby identified as a hit. On human and murine primary B cells, OSU-T315 potently suppressed the proliferation and the production of antibodies and cytokines upon stimulation, suggesting that ILK could be a promising target in the modulation of B cell activity. Mice with B cell-specific knockout of ILK were generated. Surprisingly, knockout of ILK in murine B cells did not affect B cell function as assessed by several in vivo and ex vivo B cell assays and did not alter the B cell immunosuppressive activity of OSU-T315. In conclusion, OSU-T315 displays potency as B cell modulator, probably through a mechanism of action independent of ILK, and might serve as lead drug molecule for the development of novel B cell-selective drugs.
Collapse
|
22
|
Green HJ, Griffiths AGM, Ylänne J, Brown NH. Novel functions for integrin-associated proteins revealed by analysis of myofibril attachment in Drosophila. eLife 2018; 7:e35783. [PMID: 30028294 PMCID: PMC6092120 DOI: 10.7554/elife.35783] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 07/19/2018] [Indexed: 01/18/2023] Open
Abstract
We use the myotendinous junction of Drosophila flight muscles to explore why many integrin associated proteins (IAPs) are needed and how their function is coordinated. These muscles revealed new functions for IAPs not required for viability: Focal Adhesion Kinase (FAK), RSU1, tensin and vinculin. Genetic interactions demonstrated a balance between positive and negative activities, with vinculin and tensin positively regulating adhesion, while FAK inhibits elevation of integrin activity by tensin, and RSU1 keeps PINCH activity in check. The molecular composition of myofibril termini resolves into 4 distinct layers, one of which is built by a mechanotransduction cascade: vinculin facilitates mechanical opening of filamin, which works with the Arp2/3 activator WASH to build an actin-rich layer positioned between integrins and the first sarcomere. Thus, integration of IAP activity is needed to build the complex architecture of the myotendinous junction, linking the membrane anchor to the sarcomere.
Collapse
Affiliation(s)
- Hannah J Green
- Department of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUnited Kingdom
- Department of Biological and Environmental SciencesUniversity of JyväskyläJyväskyläFinland
- Nanoscience CenterUniversity of JyväskyläJyväskyläFinland
| | - Annabel GM Griffiths
- Department of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUnited Kingdom
| | - Jari Ylänne
- Department of Biological and Environmental SciencesUniversity of JyväskyläJyväskyläFinland
- Nanoscience CenterUniversity of JyväskyläJyväskyläFinland
| | - Nicholas H Brown
- Department of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUnited Kingdom
| |
Collapse
|
23
|
Xu F, Zhou X, Hao J, Dai H, Zhang J, He Y, Hao H. Lipoxin A 4 and its analog suppress hepatocarcinoma cell epithelial-mesenchymal transition, migration and metastasis via regulating integrin-linked kinase axis. Prostaglandins Other Lipid Mediat 2018; 137:9-19. [PMID: 29787808 DOI: 10.1016/j.prostaglandins.2018.05.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2018] [Revised: 04/05/2018] [Accepted: 05/09/2018] [Indexed: 12/22/2022]
Abstract
Epithelial-mesenchymal Transition (EMT) and migration play an important role in tumor progression, and lipoxin (LX), the 'stop signal' for inflammation, has been studied in basic research for its anti-inflammatory or inflammatory pro-resolving properties. Here, in the in vitro experiment, we showed that LXA4 could inhibit the EMT and migration in phorbol myristate acetate (PMA) or activated conditioned medium (ACM)-stimulated Hep3B cells by downregulation of integrin-linked kinase (ILK), a pseudokinase in cytoplasm and these effects were via inhibiting the phosphorylation of Akt and GSK3β. Morover, LXA4 could not affect the EMT and migration of PMA-stimulated Hep3B cells by knockdown of ILK. In the in vivo experiment, BML-111 (the analog of LXA4) could inhibit the EMT and metastasis of hepatocarcinoma cells. We also demonstrated that ILK siRNA inhibited phosphorylation of downstream signaling targets Akt and GSK3β, decreased expression of MMP-2 and MMP-9. These results showed that LXA4 could be a possible candidate for liver cancer therapy, and blocking ILK axis would be an effective drug target.
Collapse
Affiliation(s)
- Fen Xu
- Department of General Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, PR China
| | - Xiaoyan Zhou
- Department of Pathophysiology, Medical College of Nanchang University, Nanchang, Jiangxi, 330006, PR China
| | - Jian Hao
- Department of Critical Care Medicine, The Third People's Hospital of Bengbu City, Bengbu, Anhui, 233000, PR China
| | - Hua Dai
- Department of Pathology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, PR China
| | - Jian Zhang
- Department of Pathology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, PR China
| | - Yuanqiao He
- Department of Laboratory Animal Science, Medical College of Nanchang University, Nanchang, Jiangxi, 330006, PR China
| | - Hua Hao
- Department of Pathology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, PR China.
| |
Collapse
|
24
|
Bulgakova NA, Wellmann J, Brown NH. Diverse integrin adhesion stoichiometries caused by varied actomyosin activity. Open Biol 2018; 7:rsob.160250. [PMID: 28446705 PMCID: PMC5413901 DOI: 10.1098/rsob.160250] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 03/17/2017] [Indexed: 12/14/2022] Open
Abstract
Cells in an organism are subjected to numerous sources of external and internal forces, and are able to sense and respond to these forces. Integrin-mediated adhesion links the extracellular matrix outside cells to the cytoskeleton inside, and participates in sensing, transmitting and responding to forces. While integrin adhesion rapidly adapts to changes in forces in isolated migrating cells, it is not known whether similar or more complex responses occur within intact, developing tissues. Here, we studied changes in integrin adhesion composition upon different contractility conditions in Drosophila embryonic muscles. We discovered that all integrin adhesion components tested were still present at muscle attachment sites (MASs) when either cytoplasmic or muscle myosin II was genetically removed, suggesting a primary role of a developmental programme in the initial assembly of integrin adhesions. Contractility does, however, increase the levels of integrin adhesion components, suggesting a mechanism to balance the strength of muscle attachment to the force of muscle contraction. Perturbing contractility in distinct ways, by genetic removal of either cytoplasmic or muscle myosin II or eliminating muscle innervation, each caused unique alterations to the stoichiometry at MASs. This suggests that different integrin-associated proteins are added to counteract different kinds of force increase.
Collapse
Affiliation(s)
- Natalia A Bulgakova
- Department of Physiology, Development and Neuroscience and The Gurdon Institute, University of Cambridge, Downing Street, Cambridge CB2 3DY, UK
| | - Jutta Wellmann
- Department of Physiology, Development and Neuroscience and The Gurdon Institute, University of Cambridge, Downing Street, Cambridge CB2 3DY, UK
| | - Nicholas H Brown
- Department of Physiology, Development and Neuroscience and The Gurdon Institute, University of Cambridge, Downing Street, Cambridge CB2 3DY, UK
| |
Collapse
|
25
|
Integrin-linked kinase: A new actor in the ageing process? Exp Gerontol 2017; 100:87-90. [PMID: 29101014 DOI: 10.1016/j.exger.2017.10.026] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 10/27/2017] [Accepted: 10/29/2017] [Indexed: 02/05/2023]
Abstract
Integrin-linked kinase (ILK) is a protein located in focal adhesion complexes that is linked to the cytoplasmic domain of integrin receptors. Together with PINCH and parvin, ILK forms the IPP complex, which is associated with conserved intracellular signalling pathways and integrin regulation of the actin cytoskeleton. ILK plays an essential role in a wide variety of cellular functions, including cell migration, differentiation, survival, and division. The present review summarizes recent evidence, suggesting a new role for ILK in organismal ageing and cellular senescence, indicating that ILK is a key regulator of longevity and premature cellular senescence induced by extracellular stressors.
Collapse
|
26
|
Arvind V, Huang AH. Mechanobiology of limb musculoskeletal development. Ann N Y Acad Sci 2017; 1409:18-32. [PMID: 28833194 DOI: 10.1111/nyas.13427] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 05/30/2017] [Accepted: 06/07/2017] [Indexed: 12/26/2022]
Abstract
While there has been considerable progress in identifying molecular regulators of musculoskeletal development, the role of physical forces in regulating induction, differentiation, and patterning events is less well understood. Here, we highlight recent findings in this area, focusing primarily on model systems that test the mechanical regulation of skeletal and tendon development in the limb. We also discuss a few of the key signaling pathways and mechanisms that have been implicated in mechanotransduction and highlight current gaps in knowledge and opportunities for further research in the field.
Collapse
Affiliation(s)
- Varun Arvind
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Alice H Huang
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
27
|
Maartens AP, Wellmann J, Wictome E, Klapholz B, Green H, Brown NH. Drosophila vinculin is more harmful when hyperactive than absent, and can circumvent integrin to form adhesion complexes. J Cell Sci 2016; 129:4354-4365. [PMID: 27737911 PMCID: PMC5201009 DOI: 10.1242/jcs.189878] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 10/07/2016] [Indexed: 12/12/2022] Open
Abstract
Vinculin is a highly conserved protein involved in cell adhesion and mechanotransduction, and both gain and loss of its activity causes defective cell behaviour. Here, we examine how altering vinculin activity perturbs integrin function within the context of Drosophila development. Whereas loss of vinculin produced relatively minor phenotypes, gain of vinculin activity, through a loss of head–tail autoinhibition, caused lethality. The minimal domain capable of inducing lethality is the talin-binding D1 domain, and this appears to require talin-binding activity, as lethality was suppressed by competition with single vinculin-binding sites from talin. Activated Drosophila vinculin triggered the formation of cytoplasmic adhesion complexes through the rod of talin, but independently of integrin. These complexes contain a subset of adhesion proteins but no longer link the membrane to actin. The negative effects of hyperactive vinculin were segregated into morphogenetic defects caused by its whole head domain and lethality caused by its D1 domain. These findings demonstrate the crucial importance of the tight control of the activity of vinculin. Summary: Development is more sensitive to gain of vinculin activity than its loss, and vinculin can promote cytoplasmic adhesion complexes independently of the usual integrin cue.
Collapse
Affiliation(s)
- Aidan P Maartens
- Department of Physiology, Development and Neuroscience, and the Gurdon Institute, University of Cambridge, Downing St., Cambridge CB2 1DY, UK
| | - Jutta Wellmann
- Department of Physiology, Development and Neuroscience, and the Gurdon Institute, University of Cambridge, Downing St., Cambridge CB2 1DY, UK
| | - Emma Wictome
- Department of Physiology, Development and Neuroscience, and the Gurdon Institute, University of Cambridge, Downing St., Cambridge CB2 1DY, UK
| | - Benjamin Klapholz
- Department of Physiology, Development and Neuroscience, and the Gurdon Institute, University of Cambridge, Downing St., Cambridge CB2 1DY, UK
| | - Hannah Green
- Department of Physiology, Development and Neuroscience, and the Gurdon Institute, University of Cambridge, Downing St., Cambridge CB2 1DY, UK
| | - Nicholas H Brown
- Department of Physiology, Development and Neuroscience, and the Gurdon Institute, University of Cambridge, Downing St., Cambridge CB2 1DY, UK
| |
Collapse
|
28
|
Huang H, Kornberg TB. Cells must express components of the planar cell polarity system and extracellular matrix to support cytonemes. eLife 2016; 5. [PMID: 27591355 PMCID: PMC5030081 DOI: 10.7554/elife.18979] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 08/31/2016] [Indexed: 01/10/2023] Open
Abstract
Drosophila dorsal air sac development depends on Decapentaplegic (Dpp) and Fibroblast growth factor (FGF) proteins produced by the wing imaginal disc and transported by cytonemes to the air sac primordium (ASP). Dpp and FGF signaling in the ASP was dependent on components of the planar cell polarity (PCP) system in the disc, and neither Dpp- nor FGF-receiving cytonemes extended over mutant disc cells that lacked them. ASP cytonemes normally navigate through extracellular matrix (ECM) composed of collagen, laminin, Dally and Dally-like (Dlp) proteins that are stratified in layers over the disc cells. However, ECM over PCP mutant cells had reduced levels of laminin, Dally and Dlp, and whereas Dpp-receiving ASP cytonemes navigated in the Dally layer and required Dally (but not Dlp), FGF-receiving ASP cytonemes navigated in the Dlp layer, requiring Dlp (but not Dally). These findings suggest that cytonemes interact directly and specifically with proteins in the stratified ECM.
Collapse
Affiliation(s)
- Hai Huang
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, United States
| | - Thomas B Kornberg
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, United States
| |
Collapse
|
29
|
Kundu S. Stochastic modelling suggests that an elevated superoxide anion - hydrogen peroxide ratio can drive extravascular phagocyte transmigration by lamellipodium formation. J Theor Biol 2016; 407:143-154. [PMID: 27380944 DOI: 10.1016/j.jtbi.2016.07.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 07/01/2016] [Indexed: 11/24/2022]
Abstract
Chemotaxis, integrates diverse intra- and inter-cellular molecular processes into a purposeful patho-physiological response; the operatic rules of which, remain speculative. Here, I surmise, that superoxide anion induced directional motility, in a responding cell, results from a quasi pathway between the stimulus, surrounding interstitium, and its biochemical repertoire. The epochal event in the mounting of an inflammatory response, is the extravascular transmigration of a phagocyte competent cell towards the site of injury, secondary to the development of a lamellipodium. This stochastic-to-markovian process conversion, is initiated by the cytosolic-ROS of the damaged cell, but is maintained by the inverse association of a de novo generated pool of self-sustaining superoxide anions and sub-critical hydrogen peroxide levels. Whilst, the exponential rise of O2(.-) is secondary to the focal accumulation of higher order lipid raft-Rac1/2-actin oligomers; O2(.-) mediated inactivation and redistribution of ECSOD, accounts for the minimal concentration of H2O2 that the phagocyte experiences. The net result of this reciprocal association between ROS/ RNS members, is the prolonged perturbation and remodeling of the cytoskeleton and plasma membrane, a prelude to chemotactic migration. The manuscript also describes the significance of stochastic modeling, in the testing of plausible molecular hypotheses of observable phenomena in complex biological systems.
Collapse
Affiliation(s)
- Siddhartha Kundu
- Department of Biochemistry, Dr. Baba Saheb Ambedkar Medical College & Hospital, Government of NCT Delhi, Sector - 6, Rohini, Delhi 110085, India; Mathematical and Computational Biology, Information Technology Research Academy (ITRA), Media Lab Asia, 2nd Floor, Block 2, C-DOT Campus, Mehrauli, New Delhi 110030, India; School of Computational and Integrative Sciences, Jawaharlal Nehru University, New Mehrauli Road, New Delhi 110067, India.
| |
Collapse
|
30
|
Tesfay L, Schulz VV, Frank SB, Lamb LE, Miranti CK. Receptor tyrosine kinase Met promotes cell survival via kinase-independent maintenance of integrin α3β1. Mol Biol Cell 2016; 27:2493-504. [PMID: 27307589 PMCID: PMC4966988 DOI: 10.1091/mbc.e15-09-0649] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 06/08/2016] [Indexed: 01/13/2023] Open
Abstract
This study identifies a new mechanism by which the receptor tyrosine kinase Met promotes cell survival. The ectodomain and transmembrane domain of Met, independently of kinase activity, are required to maintain integrin α3β1 on the cell surface to prevent activation of intrinsic and extrinsic cell death pathways and maintain autophagic flux. Matrix adhesion via integrins is required for cell survival. Adhesion of epithelial cells to laminin via integrin α3β1 was previously shown to activate at least two independent survival pathways. First, integrin α3β1 is required for autophagy-induced cell survival after growth factor deprivation. Second, integrin α3β1 independently activates two receptor tyrosine kinases, EGFR and Met, in the absence of ligands. EGFR signaling to Erk promotes survival independently of autophagy. To determine how Met promotes cell survival, we inhibited Met kinase activity or blocked its expression with RNA interference. Loss of Met expression, but not inhibition of Met kinase activity, induced apoptosis by reducing integrin α3β1 levels, activating anoikis, and blocking autophagy. Met was specifically required for the assembly of autophagosomes downstream of LC3II processing. Reexpression of wild-type Met, kinase-dead Met, or integrin α3 was sufficient to rescue death upon removal of endogenous Met. Integrin α3β1 coprecipitated and colocalized with Met in cells. The extracellular and transmembrane domain of Met was required to fully rescue cell death and restore integrin α3 expression. Thus Met promotes survival of laminin-adherent cells by maintaining integrin α3β1 via a kinase-independent mechanism.
Collapse
Affiliation(s)
- Lia Tesfay
- Laboratory of Integrin Signaling and Tumorigenesis, Van Andel Research Institute, Grand Rapids, MI 49503
| | - Veronique V Schulz
- Laboratory of Integrin Signaling and Tumorigenesis, Van Andel Research Institute, Grand Rapids, MI 49503
| | - Sander B Frank
- Laboratory of Integrin Signaling and Tumorigenesis, Van Andel Research Institute, Grand Rapids, MI 49503
| | - Laura E Lamb
- Laboratory of Integrin Signaling and Tumorigenesis, Van Andel Research Institute, Grand Rapids, MI 49503
| | - Cindy K Miranti
- Laboratory of Integrin Signaling and Tumorigenesis, Van Andel Research Institute, Grand Rapids, MI 49503
| |
Collapse
|
31
|
IPP Complex Reinforces Adhesion by Relaying Tension-Dependent Signals to Inhibit Integrin Turnover. Cell Rep 2016; 14:2668-82. [DOI: 10.1016/j.celrep.2016.02.052] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Revised: 01/05/2016] [Accepted: 02/08/2016] [Indexed: 12/19/2022] Open
|
32
|
Hákonardóttir GK, López-Ceballos P, Herrera-Reyes AD, Das R, Coombs D, Tanentzapf G. In vivo quantitative analysis of Talin turnover in response to force. Mol Biol Cell 2015; 26:4149-62. [PMID: 26446844 PMCID: PMC4710244 DOI: 10.1091/mbc.e15-05-0304] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Accepted: 10/01/2015] [Indexed: 12/18/2022] Open
Abstract
Cell–ECM adhesion is regulated by mechanical force. Quantitative imaging and mathematical modeling are used to elucidate how the intracellular adhesion complex of integrin-based adhesions responds to force, revealing the molecular mechanisms that allow the adhesion complex to respond to force to stabilize cell–ECM adhesion over development. Cell adhesion to the extracellular matrix (ECM) allows cells to form and maintain three-dimensional tissue architecture. Cell–ECM adhesions are stabilized upon exposure to mechanical force. In this study, we used quantitative imaging and mathematical modeling to gain mechanistic insight into how integrin-based adhesions respond to increased and decreased mechanical forces. A critical means of regulating integrin-based adhesion is provided by modulating the turnover of integrin and its adhesion complex (integrin adhesion complex [IAC]). The turnover of the IAC component Talin, a known mechanosensor, was analyzed using fluorescence recovery after photobleaching. Experiments were carried out in live, intact flies in genetic backgrounds that increased or decreased the force applied on sites of adhesion. This analysis showed that when force is elevated, the rate of assembly of new adhesions increases such that cell–ECM adhesion is stabilized. Moreover, under conditions of decreased force, the overall rate of turnover, but not the proportion of adhesion complex components undergoing turnover, increases. Using point mutations, we identify the key functional domains of Talin that mediate its response to force. Finally, by fitting a mathematical model to the data, we uncover the mechanisms that mediate the stabilization of ECM-based adhesion during development.
Collapse
Affiliation(s)
- Guðlaug Katrín Hákonardóttir
- Department of Cellular and Physiological Sciences, Life Science Institute, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Pablo López-Ceballos
- Department of Cellular and Physiological Sciences, Life Science Institute, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Alejandra Donají Herrera-Reyes
- Department of Mathematics and Institute of Applied Mathematics, University of British Columbia, Vancouver, BC V6T 1Z2, Canada
| | - Raibatak Das
- Department of Integrative Biology, University of Colorado Denver, Denver, CO 80204
| | - Daniel Coombs
- Department of Mathematics and Institute of Applied Mathematics, University of British Columbia, Vancouver, BC V6T 1Z2, Canada
| | - Guy Tanentzapf
- Department of Cellular and Physiological Sciences, Life Science Institute, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| |
Collapse
|
33
|
Wang Y, Antunes M, Anderson AE, Kadrmas JL, Jacinto A, Galko MJ. Integrin Adhesions Suppress Syncytium Formation in the Drosophila Larval Epidermis. Curr Biol 2015; 25:2215-27. [PMID: 26255846 DOI: 10.1016/j.cub.2015.07.031] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Revised: 06/15/2015] [Accepted: 07/10/2015] [Indexed: 12/16/2022]
Abstract
Integrins are critical for barrier epithelial architecture. Integrin loss in vertebrate skin leads to blistering and wound healing defects. However, how integrins and associated proteins maintain the regular morphology of epithelia is not well understood. We found that targeted knockdown of the integrin focal adhesion (FA) complex components β-integrin, PINCH, and integrin-linked kinase (ILK) caused formation of multinucleate epidermal cells within the Drosophila larval epidermis. This phenotype was specific to the integrin FA complex and not due to secondary effects on polarity or junctional structures. The multinucleate cells resembled the syncytia caused by physical wounding. Live imaging of wound-induced syncytium formation in the pupal epidermis suggested direct membrane breakdown leading to cell-cell fusion and consequent mixing of cytoplasmic contents. Activation of Jun N-terminal kinase (JNK) signaling, which occurs upon wounding, also correlated with syncytium formation induced by PINCH knockdown. Further, ectopic JNK activation directly caused epidermal syncytium formation. No mode of syncytium formation, including that induced by wounding, genetic loss of FA proteins, or local JNK hyperactivation, involved misregulation of mitosis or apoptosis. Finally, the mechanism of epidermal syncytium formation following JNK hyperactivation and wounding appeared to be direct disassembly of FA complexes. In conclusion, the loss-of-function phenotype of integrin FA components in the larval epidermis resembles a wound. Integrin FA loss in mouse and human skin also causes a wound-like appearance. Our results reveal a novel and unexpected role for proper integrin-based adhesion in suppressing larval epidermal cell-cell fusion--a role that may be conserved in other epithelia.
Collapse
Affiliation(s)
- Yan Wang
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Marco Antunes
- CEDOC-Faculdade de Ciências Médicas, Universidade Nova de Lisboa Campo Mártires da Pátria, 130, 1169-056 Lisboa, Portugal; Instituto de Medicina Molecular Faculdade de Medicina da Universidade de Lisboa Edificio Egas Moniz, Av Prof Egas Moniz, 1649-028 Lisboa, Portugal
| | - Aimee E Anderson
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Julie L Kadrmas
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Antonio Jacinto
- CEDOC-Faculdade de Ciências Médicas, Universidade Nova de Lisboa Campo Mártires da Pátria, 130, 1169-056 Lisboa, Portugal; Instituto de Medicina Molecular Faculdade de Medicina da Universidade de Lisboa Edificio Egas Moniz, Av Prof Egas Moniz, 1649-028 Lisboa, Portugal; Instituto Gulbenkian de Ciência, Rua da Quinta Grande 6, 2780-156 Oeiras, Portugal
| | - Michael J Galko
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Genes and Development Graduate Program, The University of Texas Graduate School of Biomedical Sciences, Houston, TX 77030, USA.
| |
Collapse
|
34
|
Dyachuk VA, Maiorova MA, Odintsova NA. Identification of β integrin-like- and fibronectin-like proteins in the bivalve mollusk Mytilus trossulus. Dev Growth Differ 2015; 57:515-28. [PMID: 26183371 DOI: 10.1111/dgd.12234] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Revised: 06/04/2015] [Accepted: 06/14/2015] [Indexed: 01/28/2023]
Abstract
Integrins play a key role in the intermediation and coordination between cells and extracellular matrix components. In this study, we first determined the presence of the β integrin-like protein and its presumptive ligand, fibronectin-like protein, during development and in some adult tissues of the bivalve mollusc Mytilus trossulus. We found that β integrin-like protein expression correlated with the development and differentiation of the digestive system in larvae. Besides the presence of β integrin-like protein in the digestive epithelial larval cells, this protein was detected in the hemocytes and some adult tissues of M. trossulus. The fibronectin-like protein was detected firstly at the blastula stage and later, the FN-LP-immunoreactive cells were scattered in the trochophore larvae. The fibronectin-like protein was not expressed in the β integrin-positive cells of either the veliger stage larvae or the adult mussel tissues and the primary hemocyte cell culture. Despite the β integrin- and fibronectin-like proteins being expressed in different cell types of mussel larvae, we do not exclude the possibility of direct interaction between these two proteins during M. trossulus development or in adult tissues.
Collapse
Affiliation(s)
- Vyacheslav A Dyachuk
- A. V. Zhirmunsky Institute of Marine Biology, Far Eastern Branch of the Russian Academy of Sciences, Palchevsky Str. 17, 690041, Vladivostok, Russia.,Far Eastern Federal University, 690950, Vladivostok, Russia
| | - Maria A Maiorova
- A. V. Zhirmunsky Institute of Marine Biology, Far Eastern Branch of the Russian Academy of Sciences, Palchevsky Str. 17, 690041, Vladivostok, Russia.,Far Eastern Federal University, 690950, Vladivostok, Russia
| | - Nelly A Odintsova
- A. V. Zhirmunsky Institute of Marine Biology, Far Eastern Branch of the Russian Academy of Sciences, Palchevsky Str. 17, 690041, Vladivostok, Russia.,Far Eastern Federal University, 690950, Vladivostok, Russia
| |
Collapse
|
35
|
Maartens AP, Brown NH. The many faces of cell adhesion during Drosophila muscle development. Dev Biol 2015; 401:62-74. [DOI: 10.1016/j.ydbio.2014.12.038] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Revised: 12/17/2014] [Accepted: 12/19/2014] [Indexed: 10/24/2022]
|
36
|
Maartens AP, Brown NH. Anchors and signals: the diverse roles of integrins in development. Curr Top Dev Biol 2015; 112:233-72. [PMID: 25733142 DOI: 10.1016/bs.ctdb.2014.11.020] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Integrins mediate cell adhesion by providing a link between the actin cytoskeleton and the extracellular matrix. As well as acting to anchor cells, integrin adhesions provide sensory input via mechanotransduction and synergism with signaling pathways, and provide the cell with the conditions necessary for differentiation in a permissive manner. In this review, we explore how integrins contribute to development, and what this tells us about how they work. From a signaling perspective, the influence of integrins on cell viability and fate is muted in a developmental context as compared to cell culture. Integrin phenotypes tend to arise from a failure of normally specified cells to create tissues properly, due to defective adhesion. The diversity of integrin functions in development shows how cell adhesion is continuously adjusted, both within and between animals, to fit developmental purpose.
Collapse
Affiliation(s)
- Aidan P Maartens
- Department of Physiology, Development and Neuroscience, The Gurdon Institute, University of Cambridge, Cambridge, United Kingdom
| | - Nicholas H Brown
- Department of Physiology, Development and Neuroscience, The Gurdon Institute, University of Cambridge, Cambridge, United Kingdom.
| |
Collapse
|
37
|
Park Y, Ahn SJ, Vogel H, Kim Y. Integrin β subunit and its RNA interference in immune and developmental processes of the Oriental tobacco budworm, Helicoverpa assulta. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2014; 47:59-67. [PMID: 25008242 DOI: 10.1016/j.dci.2014.06.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Revised: 06/26/2014] [Accepted: 06/28/2014] [Indexed: 06/03/2023]
Abstract
Integrins are cell surface heterodimeric proteins interacting with the extracellular matrix and mediating environmental signals through cell membranes. A full-length cDNA sequence of the integrin β1 subunit gene (HaITGb1) was cloned from the Oriental tobacco budworm, Helicoverpa assulta, and analyzed for its physiological role in both immune response and development. HaITGb1 was expressed in all developmental stages from egg to adult and in all tested larval tissues of hemocytes, fat body, gut, and epidermis. Utilizing an RNA interference (RNAi) approach, injection of a specific double-stranded RNA (dsRNA) in larvae suppressed HaITGb1 transcript levels and significantly impaired hemocytes in their extracellular matrix adherence properties. Furthermore, the RNAi treatment significantly suppressed hemocyte nodule formation in response to bacterial challenge, which resulted in significantly enhanced susceptibility to both pathogenic and non-pathogenic bacteria. The RNAi treatment also interfered with H. assulta larval and pupal development. These results suggest that the extensive and constitutive expression of HaITGb1 is necessary for H. assulta to perform an efficient immune response against microbial pathogens and undergo normal immature development.
Collapse
Affiliation(s)
- Youngjin Park
- Department of Bioresource Sciences, Andong National University, Andong 760-749, Republic of Korea
| | - Seung-Joon Ahn
- Department of Entomology, Max Planck Institute for Chemical Ecology, Jena 07745, Germany
| | - Heiko Vogel
- Department of Entomology, Max Planck Institute for Chemical Ecology, Jena 07745, Germany
| | - Yonggyun Kim
- Department of Bioresource Sciences, Andong National University, Andong 760-749, Republic of Korea.
| |
Collapse
|
38
|
|
39
|
Abstract
Integrin-linked kinase (ILK), PINCH and Parvin proteins form the IPP-complex that has been established as a core component of the integrin-actin link. Our recent genetic studies on Drosophila parvin, reveal that loss of function mutant defects phenocopy those observed upon loss of ILK or PINCH in the muscle and the wing, strengthening the notion that these proteins function together in the organism. Our work identified that ILK is necessary and sufficient for parvin subcellular localization, corroborating previous data indicating a direct association between these two proteins. Further genetic epistasis analysis of the IPP-complex assembly at integrin adhesion sites reveals that depending on the cell context each component is required differently. At the muscle attachment sites of the embryo, ILK is placed upstream in the hierarchy of genetic interactions required for the IPP-complex assembly. By contrast, in the wing epithelium the three proteins are mutually interdependent. Finally, we uncovered a novel property for the CH1-domain of parvin: its recruitment at the integrin-containing junctions in an ILK-dependent manner. Apparently, this ability of the CH1-domain is controlled by the inter-CH linker region. Thus, an intramolecular interaction within parvin could serve as a putative regulatory mechanism controlling the ILK-Parvin interaction.
Collapse
Affiliation(s)
- Katerina Vakaloglou
- Biomedical Research Foundation; Academy of Athens (BRFAA); Division of Genetics; Athens, Greece
| | | |
Collapse
|
40
|
Gómez-Lamarca MJ, Cobreros-Reguera L, Ibáñez-Jiménez B, Palacios IM, Martín-Bermudo MD. Integrins regulate epithelial cell differentiation by modulating Notch activity. J Cell Sci 2014; 127:4667-78. [PMID: 25179603 DOI: 10.1242/jcs.153122] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Coordinating exit from the cell cycle with differentiation is crucial for proper development and tissue homeostasis. Failure to do so can lead to aberrant organogenesis and tumorigenesis. However, little is known about the developmental signals that regulate the switch from cell cycle exit to differentiation. Signals downstream of two key developmental pathways, Notch and Salvador-Warts-Hippo (SWH), and signals downstream of myosin activity regulate this switch during the development of the follicle cell epithelium of the Drosophila ovary. Here, we have identified a fourth player, the integrin signaling pathway. Elimination of integrin function blocks the mitosis-to-endocycle switch and differentiation in posterior follicle cells (PFCs), by regulation of the cyclin-dependent kinase inhibitor (CKI) dacapo. In addition, integrin-mutant PFCs show defective Notch signaling and endocytosis. Furthermore, integrins act in PFCs by modulating the activity of the Notch pathway, as reducing the amount of Hairless, the major antagonist of Notch, or misexpressing Notch intracellular domain rescues the cell cycle and differentiation defects. Taken together, our findings reveal a direct involvement of integrin signaling on the spatial and temporal regulation of epithelial cell differentiation during development.
Collapse
Affiliation(s)
- M Jesús Gómez-Lamarca
- Centro Andaluz de Biología del Desarrollo CSIC-University Pablo de Olavide, Sevilla 41013, Spain
| | - Laura Cobreros-Reguera
- Centro Andaluz de Biología del Desarrollo CSIC-University Pablo de Olavide, Sevilla 41013, Spain
| | - Beatriz Ibáñez-Jiménez
- Centro Andaluz de Biología del Desarrollo CSIC-University Pablo de Olavide, Sevilla 41013, Spain
| | - Isabel M Palacios
- The Department of Zoology, University of Cambridge, Downing Street, Cambridge CB2 3EJ, UK
| | - María D Martín-Bermudo
- Centro Andaluz de Biología del Desarrollo CSIC-University Pablo de Olavide, Sevilla 41013, Spain
| |
Collapse
|
41
|
Fukuda K, Bledzka K, Yang J, Perera HD, Plow EF, Qin J. Molecular basis of kindlin-2 binding to integrin-linked kinase pseudokinase for regulating cell adhesion. J Biol Chem 2014; 289:28363-75. [PMID: 25160619 DOI: 10.1074/jbc.m114.596692] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Integrin-linked kinase (ILK) is a distinct intracellular adaptor essential for integrin-mediated cell-extracellular matrix adhesion, cell spreading, and migration. Acting as a major docking platform in focal adhesions, ILK engages many proteins to dynamically link integrins with the cytoskeleton, but the underlying mechanism remains elusive. Here, we have characterized the interaction of ILK with kindlin-2, a key regulator for integrin bidirectional signaling. We show that human kindlin-2 binds to human ILK with high affinity. Using systematic mapping approaches, we have identified a major ILK binding site involving a 20-residue fragment (residues 339-358) in kindlin-2. NMR-based analysis reveals a helical conformation of this fragment that utilizes its leucine-rich surface to recognize the ILK pseudokinase domain in a mode that is distinct from another ILK pseudokinase domain binding protein, α-parvin. Structure-based mutational experiments further demonstrate that the kindlin-2 binding to ILK is crucial for the kindlin-2 localization to focal adhesions and cell spreading (integrin outside-in signaling) but dispensable for the kindlin-2-mediated integrin activation (integrin inside-out signaling). These data define a specific mode of the kindlin-2/ILK interaction with mechanistic implications as to how it spatiotemporally mediates integrin signaling and cell adhesion.
Collapse
Affiliation(s)
- Koichi Fukuda
- From the Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195
| | - Kamila Bledzka
- From the Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195
| | - Jun Yang
- From the Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195
| | - H Dhanuja Perera
- From the Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195
| | - Edward F Plow
- From the Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195
| | - Jun Qin
- From the Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195
| |
Collapse
|
42
|
Extensive nonmuscle expression and epithelial apicobasal localization of the Drosophila ALP/Enigma family protein, Zasp52. Gene Expr Patterns 2014; 15:67-79. [DOI: 10.1016/j.gep.2014.05.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Revised: 05/05/2014] [Accepted: 05/08/2014] [Indexed: 01/31/2023]
|
43
|
Bilousov O, Koval A, Keshelava A, Katanaev VL. Identification of novel elements of the Drosophila blisterome sheds light on potential pathological mechanisms of several human diseases. PLoS One 2014; 9:e101133. [PMID: 24968325 PMCID: PMC4072764 DOI: 10.1371/journal.pone.0101133] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Accepted: 06/03/2014] [Indexed: 12/16/2022] Open
Abstract
Main developmental programs are highly conserved among species of the animal kingdom. Improper execution of these programs often leads to progression of various diseases and disorders. Here we focused on Drosophila wing tissue morphogenesis, a fairly complex developmental program, one of the steps of which – apposition of the dorsal and ventral wing sheets during metamorphosis – is mediated by integrins. Disruption of this apposition leads to wing blistering which serves as an easily screenable phenotype for components regulating this process. By means of RNAi-silencing technique and the blister phenotype as readout, we identify numerous novel proteins potentially involved in wing sheet adhesion. Remarkably, our results reveal not only participants of the integrin-mediated machinery, but also components of other cellular processes, e.g. cell cycle, RNA splicing, and vesicular trafficking. With the use of bioinformatics tools, these data are assembled into a large blisterome network. Analysis of human orthologues of the Drosophila blisterome components shows that many disease-related genes may contribute to cell adhesion implementation, providing hints on possible mechanisms of these human pathologies.
Collapse
Affiliation(s)
- Oleksii Bilousov
- Department of Pharmacology and Toxicology, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Alexey Koval
- Department of Pharmacology and Toxicology, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Amiran Keshelava
- Department of Pharmacology and Toxicology, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Vladimir L. Katanaev
- Department of Pharmacology and Toxicology, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
- * E-mail:
| |
Collapse
|
44
|
Nishimura M, Kumsta C, Kaushik G, Diop SB, Ding Y, Bisharat-Kernizan J, Catan H, Cammarato A, Ross RS, Engler AJ, Bodmer R, Hansen M, Ocorr K. A dual role for integrin-linked kinase and β1-integrin in modulating cardiac aging. Aging Cell 2014; 13:431-40. [PMID: 24400780 PMCID: PMC4032615 DOI: 10.1111/acel.12193] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/25/2013] [Indexed: 12/19/2022] Open
Abstract
Cardiac performance decreases with age, which is a major risk factor for cardiovascular disease and mortality in the aging human population, but the molecular mechanisms underlying cardiac aging are still poorly understood. Investigating the role of integrin-linked kinase (ilk) and β1-integrin (myospheroid, mys) in Drosophila, which colocalize near cardiomyocyte contacts and Z-bands, we find that reduced ilk or mys function prevents the typical changes of cardiac aging seen in wildtype, such as arrhythmias. In particular, the characteristic increase in cardiac arrhythmias with age is prevented in ilk and mys heterozygous flies with nearly identical genetic background, and they live longer, in line with previous findings in Caenorhabditis elegans for ilk and in Drosophila for mys. Consistent with these findings, we observed elevated β1-integrin protein levels in old compared with young wild-type flies, and cardiac-specific overexpression of mys in young flies causes aging-like heart dysfunction. Moreover, moderate cardiac-specific knockdown of integrin-linked kinase (ILK)/integrin pathway-associated genes also prevented the decline in cardiac performance with age. In contrast, strong cardiac knockdown of ilk or ILK-associated genes can severely compromise cardiac integrity, including cardiomyocyte adhesion and overall heart function. These data suggest that ilk/mys function is necessary for establishing and maintaining normal heart structure and function, and appropriate fine-tuning of this pathway can retard the age-dependent decline in cardiac performance and extend lifespan. Thus, ILK/integrin-associated signaling emerges as an important and conserved genetic mechanism in longevity, and as a new means to improve age-dependent cardiac performance, in addition to its vital role in maintaining cardiac integrity.
Collapse
Affiliation(s)
- Mayuko Nishimura
- Development, Aging and Regeneration Program; Sanford-Burnham Medical Research Institute; 10901 North Torrey Pines Road La Jolla CA 92037 USA
| | - Caroline Kumsta
- Development, Aging and Regeneration Program; Sanford-Burnham Medical Research Institute; 10901 North Torrey Pines Road La Jolla CA 92037 USA
| | - Gaurav Kaushik
- Sanford Consortium for Regenerative Medicine; University of California at San Diego; 2880 Torrey Pines Scenic Drive La Jolla CA 92037 USA
| | - Soda B. Diop
- Development, Aging and Regeneration Program; Sanford-Burnham Medical Research Institute; 10901 North Torrey Pines Road La Jolla CA 92037 USA
| | - Yun Ding
- School of Medicine; VA San Diego Healthcare System; University of California at San Diego; 3350 La Jolla Village Drive, Cardiology Section 111A San Diego CA 92161 USA
| | - Jumana Bisharat-Kernizan
- Development, Aging and Regeneration Program; Sanford-Burnham Medical Research Institute; 10901 North Torrey Pines Road La Jolla CA 92037 USA
| | - Hannah Catan
- Development, Aging and Regeneration Program; Sanford-Burnham Medical Research Institute; 10901 North Torrey Pines Road La Jolla CA 92037 USA
| | - Anthony Cammarato
- Division of Cardiology; Department of Medicine; School of Medicine; Johns Hopkins University; Baltimore MD 21287 USA
| | - Robert S. Ross
- School of Medicine; VA San Diego Healthcare System; University of California at San Diego; 3350 La Jolla Village Drive, Cardiology Section 111A San Diego CA 92161 USA
| | - Adam J. Engler
- Sanford Consortium for Regenerative Medicine; University of California at San Diego; 2880 Torrey Pines Scenic Drive La Jolla CA 92037 USA
| | - Rolf Bodmer
- Development, Aging and Regeneration Program; Sanford-Burnham Medical Research Institute; 10901 North Torrey Pines Road La Jolla CA 92037 USA
| | - Malene Hansen
- Development, Aging and Regeneration Program; Sanford-Burnham Medical Research Institute; 10901 North Torrey Pines Road La Jolla CA 92037 USA
| | - Karen Ocorr
- Development, Aging and Regeneration Program; Sanford-Burnham Medical Research Institute; 10901 North Torrey Pines Road La Jolla CA 92037 USA
| |
Collapse
|
45
|
Kumsta C, Ching TT, Nishimura M, Davis AE, Gelino S, Catan HH, Yu X, Chu CC, Ong B, Panowski SH, Baird N, Bodmer R, Hsu AL, Hansen M. Integrin-linked kinase modulates longevity and thermotolerance in C. elegans through neuronal control of HSF-1. Aging Cell 2014; 13:419-30. [PMID: 24314125 PMCID: PMC4059541 DOI: 10.1111/acel.12189] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/24/2013] [Indexed: 12/18/2022] Open
Abstract
Integrin-signaling complexes play important roles in cytoskeletal organization and cell adhesion in many species. Components of the integrin-signaling complex have been linked to aging in both Caenorhabditis elegans and Drosophila melanogaster, but the mechanism underlying this function is unknown. Here, we investigated the role of integrin-linked kinase (ILK), a key component of the integrin-signaling complex, in lifespan determination. We report that genetic reduction of ILK in both C. elegans and Drosophila increased resistance to heat stress, and led to lifespan extension in C. elegans without majorly affecting cytoskeletal integrity. In C. elegans, longevity and thermotolerance induced by ILK depletion was mediated by heat-shock factor-1 (HSF-1), a major transcriptional regulator of the heat-shock response (HSR). Reduction in ILK levels increased hsf-1 transcription and activation, and led to enhanced expression of a subset of genes with roles in the HSR. Moreover, induction of HSR-related genes, longevity and thermotolerance caused by ILK reduction required the thermosensory neurons AFD and interneurons AIY, which are known to play a critical role in the canonical HSR. Notably, ILK was expressed in neighboring neurons, but not in AFD or AIY, implying that ILK reduction initiates cell nonautonomous signaling through thermosensory neurons to elicit a noncanonical HSR. Our results thus identify HSF-1 as a novel effector of the organismal response to reduced ILK levels and show that ILK inhibition regulates HSF-1 in a cell nonautonomous fashion to enhance stress resistance and lifespan in C. elegans.
Collapse
Affiliation(s)
- Caroline Kumsta
- Development, Aging and Regeneration Program, Sanford-Burnham Medical Research InstituteLa Jolla, CA, USA
| | - Tsui-Ting Ching
- Department of Internal Medicine, Division of Geriatric and Palliative Medicine, University of Michigan Medical SchoolAnn Arbor, MI, USA
- Institute of Biopharmaceutical Sciences, National Yang-Ming UniversityTaipei, Taiwan
| | - Mayuko Nishimura
- Development, Aging and Regeneration Program, Sanford-Burnham Medical Research InstituteLa Jolla, CA, USA
| | - Andrew E Davis
- Development, Aging and Regeneration Program, Sanford-Burnham Medical Research InstituteLa Jolla, CA, USA
| | - Sara Gelino
- Development, Aging and Regeneration Program, Sanford-Burnham Medical Research InstituteLa Jolla, CA, USA
| | - Hannah H Catan
- Development, Aging and Regeneration Program, Sanford-Burnham Medical Research InstituteLa Jolla, CA, USA
| | - Xiaokun Yu
- Department of Internal Medicine, Division of Geriatric and Palliative Medicine, University of Michigan Medical SchoolAnn Arbor, MI, USA
| | - Chu-Chiao Chu
- Development, Aging and Regeneration Program, Sanford-Burnham Medical Research InstituteLa Jolla, CA, USA
| | - Binnan Ong
- Development, Aging and Regeneration Program, Sanford-Burnham Medical Research InstituteLa Jolla, CA, USA
| | - Siler H Panowski
- The Glenn Center for Aging Research, The Salk Institute for Biological Studies, The Howard Hughes Medical InstituteLa Jolla, CA, USA
| | - Nathan Baird
- The Glenn Center for Aging Research, The Salk Institute for Biological Studies, The Howard Hughes Medical InstituteLa Jolla, CA, USA
| | - Rolf Bodmer
- Development, Aging and Regeneration Program, Sanford-Burnham Medical Research InstituteLa Jolla, CA, USA
| | - Ao-Lin Hsu
- Department of Internal Medicine, Division of Geriatric and Palliative Medicine, University of Michigan Medical SchoolAnn Arbor, MI, USA
- Department of Molecular and Integrative Physiology, University of Michigan Medical SchoolAnn Arbor, MI, USA
| | - Malene Hansen
- Development, Aging and Regeneration Program, Sanford-Burnham Medical Research InstituteLa Jolla, CA, USA
| |
Collapse
|
46
|
Lennon R, Randles MJ, Humphries MJ. The importance of podocyte adhesion for a healthy glomerulus. Front Endocrinol (Lausanne) 2014; 5:160. [PMID: 25352829 PMCID: PMC4196579 DOI: 10.3389/fendo.2014.00160] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Accepted: 09/21/2014] [Indexed: 12/23/2022] Open
Abstract
Podocytes are specialized epithelial cells that cover the outer surfaces of glomerular capillaries. Unique cell junctions, known as slit diaphragms, which feature nephrin and Neph family proteins in addition to components of adherens, tight, and gap junctions, connect adjacent podocyte foot processes. Single gene disorders affecting the slit diaphragm result in nephrotic syndrome in humans, characterized by massive loss of protein across the capillary wall. In addition to specialized cell junctions, interconnecting podocytes also adhere to the glomerular basement membrane (GBM) of the capillary wall. The GBM is a dense network of secreted, extracellular matrix (ECM) components and contains tissue-restricted isoforms of collagen IV and laminin in addition to other structural proteins and ECM regulators such as proteases and growth factors. The specialized niche of the GBM provides a scaffold for endothelial cells and podocytes to support their unique functions and human genetic mutations in GBM components lead to renal failure, thus highlighting the importance of cell-matrix interactions in the glomerulus. Cells adhere to ECM via adhesion receptors, including integrins, syndecans, and dystroglycan and in particular the integrin heterodimer α3β1 is required to maintain barrier integrity. Therefore, the sophisticated function of glomerular filtration relies on podocyte adhesion both at cell junctions and at the interface with the ECM. In health, the podocyte coordinates signals from cell junctions and cell-matrix interactions, in response to environmental cues in order to regulate filtration and as our understanding of mechanisms that control cell adhesion in the glomerulus develops, then insight into the effects of disease will improve. The ultimate goal will be to develop targeted therapies to prevent or repair defects in the filtration barrier and to restore glomerular function.
Collapse
Affiliation(s)
- Rachel Lennon
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences, The University of Manchester, Manchester, UK
- Institute of Human Development, Faculty of Medical and Human Sciences, The University of Manchester, Manchester, UK
- Department of Paediatric Nephrology, Manchester Academic Health Science Centre, Central Manchester University Hospitals NHS Foundation Trust, Manchester, UK
- *Correspondence: Rachel Lennon, Wellcome Trust Centre for Cell-Matrix Research, The University of Manchester, Michael Smith Building, Manchester M13 9PT, UK e-mail:
| | - Michael J. Randles
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences, The University of Manchester, Manchester, UK
- Institute of Human Development, Faculty of Medical and Human Sciences, The University of Manchester, Manchester, UK
| | - Martin J. Humphries
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences, The University of Manchester, Manchester, UK
| |
Collapse
|
47
|
ILK: a pseudokinase with a unique function in the integrin-actin linkage. Biochem Soc Trans 2013; 41:995-1001. [PMID: 23863169 DOI: 10.1042/bst20130062] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
ILK (integrin-linked kinase) is a central component of cell-matrix adhesions and an important regulator of integrin function. It forms a ternary complex with two other adaptor proteins, PINCH (particularly interesting cysteine- and histidine-rich protein) and parvin, forming the IPP (ILK-PINCH-parvin) complex that regulates the integrin-actin linkage as well as microtubule dynamics. These functions are essential for processes such as cell migration and matrix remodelling. The present review discusses the recent advances on the structural and functional characterization of ILK and the long-standing debate regarding its reported kinase activity.
Collapse
|
48
|
Dai X, Jiang W, Zhang Q, Xu L, Geng P, Zhuang S, Petrich BG, Jiang C, Peng L, Bhattacharya S, Evans SM, Sun Y, Chen J, Liang X. Requirement for integrin-linked kinase in neural crest migration and differentiation and outflow tract morphogenesis. BMC Biol 2013; 11:107. [PMID: 24131868 PMCID: PMC3906977 DOI: 10.1186/1741-7007-11-107] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Accepted: 10/07/2013] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Neural crest defects lead to congenital heart disease involving outflow tract malformation. Integrin-linked-kinase (ILK) plays important roles in multiple cellular processes and embryogenesis. ILK is expressed in the neural crest, but its role in neural crest and outflow tract morphogenesis remains unknown. RESULTS We ablated ILK specifically in the neural crest using the Wnt1-Cre transgene. ILK ablation resulted in abnormal migration and overpopulation of neural crest cells in the pharyngeal arches and outflow tract and a significant reduction in the expression of neural cell adhesion molecule (NCAM) and extracellular matrix components. ILK mutant embryos exhibited an enlarged common arterial trunk and ventricular septal defect. Reduced smooth muscle differentiation, but increased ossification and neurogenesis/innervation were observed in ILK mutant outflow tract that may partly be due to reduced transforming growth factor β2 (TGFβ2) but increased bone morphogenetic protein (BMP) signaling. Consistent with these observations, microarray analysis of fluorescence-activated cell sorting (FACS)-sorted neural crest cells revealed reduced expression of genes associated with muscle differentiation, but increased expression of genes of neurogenesis and osteogenesis. CONCLUSIONS Our results demonstrate that ILK plays essential roles in neural crest and outflow tract development by mediating complex crosstalk between cell matrix and multiple signaling pathways. Changes in these pathways may collectively result in the unique neural crest and outflow tract phenotypes observed in ILK mutants.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Yunfu Sun
- Key Laboratory of Arrhythmia, Ministry of Education, East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai 200120, China.
| | | | | |
Collapse
|
49
|
Liu ZC, Odell N, Geisbrecht ER. Drosophila importin-7 functions upstream of the Elmo signaling module to mediate the formation and stability of muscle attachments. J Cell Sci 2013; 126:5210-23. [PMID: 24046451 DOI: 10.1242/jcs.132241] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Establishment and maintenance of stable muscle attachments is essential for coordinated body movement. Studies in Drosophila have pioneered a molecular understanding of the morphological events in the conserved process of muscle attachment formation, including myofiber migration, muscle-tendon signaling, and stable junctional adhesion between muscle cells and their corresponding target insertion sites. In both Drosophila and vertebrate models, integrin complexes play a key role in the biogenesis and stability of muscle attachments through the interactions of integrins with extracellular matrix (ECM) ligands. We show that Drosophila importin-7 (Dim7) is an upstream regulator of the conserved Elmo-Mbc→Rac signaling pathway in the formation of embryonic muscle attachment sites (MASs). Dim7 is encoded by the moleskin (msk) locus and was identified as an Elmo-interacting protein. Both Dim7 and Elmo localize to the ends of myofibers coincident with the timing of muscle-tendon attachment in late myogenesis. Phenotypic analysis of elmo mutants reveal muscle attachment defects similar to those previously described for integrin mutants. Furthermore, Elmo and Dim7 interact both biochemically and genetically in the developing musculature. The muscle detachment phenotype resulting from mutations in the msk locus can be rescued by components in the Elmo signaling pathway, including the Elmo-Mbc complex, an activated Elmo variant, or a constitutively active form of Rac. In larval muscles, the localization of Dim7 and activated Elmo to the sites of muscle attachment is attenuated upon RNAi knockdown of integrin heterodimer complex components. Our results show that integrins function as upstream signals to mediate Dim7-Elmo enrichment to the MASs.
Collapse
Affiliation(s)
- Ze Cindy Liu
- Division of Cell Biology and Biophysics, School of Biological Sciences, University of Missouri, Kansas City, MO 64110, USA
| | | | | |
Collapse
|
50
|
αV-integrins are required for mechanotransduction in MDCK epithelial cells. PLoS One 2013; 8:e71485. [PMID: 23977051 PMCID: PMC3747215 DOI: 10.1371/journal.pone.0071485] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2013] [Accepted: 06/29/2013] [Indexed: 01/12/2023] Open
Abstract
The properties of epithelial cells within tissues are regulated by their immediate microenvironment, which consists of neighboring cells and the extracellular matrix (ECM). Integrin heterodimers orchestrate dynamic assembly and disassembly of cell-ECM connections and thereby convey biochemical and mechanical information from the ECM into cells. However, the specific contributions and functional hierarchy between different integrin heterodimers in the regulation of focal adhesion dynamics in epithelial cells are incompletely understood. Here, we have studied the functions of RGD-binding αV-integrins in a Madin Darby Canine Kidney (MDCK) cell model and found that αV-integrins regulate the maturation of focal adhesions (FAs) and cell spreading. αV-integrin-deficient MDCK cells bound collagen I (Col I) substrate via α2β1-integrins but failed to efficiently recruit FA components such as talin, focal adhesion kinase (FAK), vinculin and integrin-linked kinase (ILK). The apparent inability to mature α2β1-integrin-mediated FAs and link them to cellular actin cytoskeleton led to disrupted mechanotransduction in αV-integrin deficient cells seeded onto Col I substrate.
Collapse
|