1
|
Liu YL, Mei YM, Xun JQ, Lv ZY, He Q, Liu ZBR, Li L, Xie F, Dai RC. The biological function of integrin-linked kinase on bone formation. Bone Rep 2025; 25:101834. [PMID: 40171447 PMCID: PMC11957501 DOI: 10.1016/j.bonr.2025.101834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 01/30/2025] [Accepted: 03/08/2025] [Indexed: 04/03/2025] Open
Abstract
Bone remodeling process is the basis for maintaining normal bone microstructure and promoting fracture repair. Recent studies have proven that integrins can promote bone formation and fracture repair. Integrin-linked kinase (ILK), as the proximal effector of the integrin receptor, is a key protein factor linking integrin and cytoskeleton. It is involved in crucial cellular processes including proliferation, survival, differentiation, migration, invasion, and angiogenesis reflects on systemic changes in the kidney, heart, muscle, skin, and vascular system. At present, the regulation effect of ILK in bone formation attracts the attention of researchers. This review emphasizes that ILK as a key molecule affects the functions of bone marrow stromal cells (BMSCs) and osteoblasts, and regulates bone formation. Additionally, ILK plays a key role in the process of"angiogenic-osteogenic coupling ". The present role of ILK in the pathogenesis of osteoporosis is also described. Strategies that target ILK may as a new prospective treatment for osteoporosis (OP).
Collapse
Affiliation(s)
- Yu-ling Liu
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Yue-ming Mei
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Jing-qiong Xun
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Zhuo-yue Lv
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Qian He
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Zhou-bo-ran Liu
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Lin Li
- Department of Endocrinology and Metabolism, The Affiliated Changsha Hospital of Xiangya School of Medicine, Central South University, Changsha 410005, Hunan, China
| | - Fen Xie
- Medicine School, Changsha Social Work College, Changsha 410004, Hunan, China
| | - Ru-chun Dai
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| |
Collapse
|
2
|
Shi R, Farnsworth DA, Febres-Aldana CA, Chow JLM, Sheena R, Atwal T, Gomez Marti JL, Li S, Thomas KN, Lee CM, Awrey SJ, McDonald PC, Somwar R, Dedhar S, Ladanyi M, Bennewith KL, Lockwood WW. Drug tolerance and persistence to EGFR inhibitor treatment are mediated by an ILK-SFK-YAP signaling axis in lung adenocarcinoma. Oncogene 2025:10.1038/s41388-025-03461-6. [PMID: 40450112 DOI: 10.1038/s41388-025-03461-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 05/09/2025] [Accepted: 05/20/2025] [Indexed: 06/03/2025]
Abstract
Combating resistance to targeted therapy remains a major challenge to improving lung cancer care. Epithelial-mesenchymal transition (EMT) in tumour cells is an established non-genetic resistance mechanism to EGFR tyrosine kinase inhibitors (TKI) that is also associated with worse outcome in patients. Here we demonstrate that integrin-linked kinase (ILK) is an important driver of EMT-mediated TKI resistance in lung adenocarcinoma (LUAD) by promoting a drug-tolerant persister (DTP) cell phenotype. Our results indicate that high ILK expression is associated with EMT in LUAD patients and that genetic suppression of ILK can limit EMT progression and reduce the viability of DTP cells by impairing YAP activation, ultimately improving osimertinib (Osi) sensitivity in LUAD cells. Importantly, LUAD cells with high ILK expression are able to persist during EGFR-TKI treatment, acquiring additional genetic and phenotypic alterations to develop EGFR-TKI resistance. To improve clinical translatability of our findings, we showed that pharmacological inhibition of ILK can suppress EMT and improve Osi response in LUAD cells. Lastly, we found that strong immunohistochemistry staining of ILK in patient biopsies was significantly associated with and may be used to predict receptor tyrosine kinase-independent mechanisms of EGFR-TKI resistance. Overall, our results suggest that ILK is an important regulator of EGFR-TKI response and may be exploited as a predictor for acquired resistance, providing evidence for co-targeting ILK with EGFR to better control minimal residual disease and EGFR-TKI resistance in lung cancer.
Collapse
Affiliation(s)
- Rocky Shi
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Dylan A Farnsworth
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Christopher A Febres-Aldana
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Justine L M Chow
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Ravinder Sheena
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Tejveer Atwal
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Juan Luis Gomez Marti
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Samantha Li
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Kiersten N Thomas
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Che-Min Lee
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Shannon J Awrey
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Paul C McDonald
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Romel Somwar
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Shoukat Dedhar
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| | - Marc Ladanyi
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kevin L Bennewith
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
- Department of Pathology & Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - William W Lockwood
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada.
- Department of Pathology & Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
3
|
Sankhe CS, Sacco JL, Lawton J, Fair RA, Soares DVR, Aldahdooh MKR, Gomez ED, Gomez EW. Breast Cancer Cells Exhibit Mesenchymal-Epithelial Plasticity Following Dynamic Modulation of Matrix Stiffness. Adv Biol (Weinh) 2024; 8:e2400087. [PMID: 38977422 DOI: 10.1002/adbi.202400087] [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: 02/17/2024] [Revised: 05/31/2024] [Indexed: 07/10/2024]
Abstract
Mesenchymal-epithelial transition (MET) is essential for tissue and organ development and is thought to contribute to cancer by enabling the establishment of metastatic lesions. Despite its importance in both health and disease, there is a lack of in vitro platforms to study MET and little is known about the regulation of MET by mechanical cues. Here, hyaluronic acid-based hydrogels with dynamic and tunable stiffnesses mimicking that of normal and tumorigenic mammary tissue are synthesized. The platform is then utilized to examine the response of mammary epithelial cells and breast cancer cells to dynamic modulation of matrix stiffness. Gradual softening of the hydrogels reduces proliferation and increases apoptosis of breast cancer cells. Moreover, breast cancer cells exhibit temporal changes in cell morphology, cytoskeletal organization, and gene expression that are consistent with mesenchymal-epithelial plasticity as the stiffness of the matrix is reduced. A reduction in matrix stiffness attenuates the expression of integrin-linked kinase, and inhibition of integrin-linked kinase impacts proliferation, apoptosis, and gene expression in cells cultured on stiff and dynamic hydrogels. Overall, these findings reveal intermediate epithelial/mesenchymal states as cells move along a matrix stiffness-mediated MET trajectory and suggest an important role for matrix mechanics in regulating mesenchymal-epithelial plasticity.
Collapse
Affiliation(s)
- Chinmay S Sankhe
- Department of Chemical Engineering, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Jessica L Sacco
- Department of Chemical Engineering, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Jacob Lawton
- Department of Chemical Engineering, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Ryan A Fair
- Department of Materials Science and Engineering, The Pennsylvania State University, University Park, PA, 16802, USA
| | | | - Mohammed K R Aldahdooh
- Department of Chemistry, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Enrique D Gomez
- Department of Chemical Engineering, The Pennsylvania State University, University Park, PA, 16802, USA
- Department of Materials Science and Engineering, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Esther W Gomez
- Department of Chemical Engineering, The Pennsylvania State University, University Park, PA, 16802, USA
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, 16802, USA
| |
Collapse
|
4
|
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
|
5
|
Wu X, Lin S, Liao R, Yao Q, Lin L, Zou X, Xiao G. Brief research report: Effects of Pinch deficiency on cartilage homeostasis in adult mice. Front Cell Dev Biol 2023; 11:1116128. [PMID: 36743414 PMCID: PMC9892552 DOI: 10.3389/fcell.2023.1116128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 01/11/2023] [Indexed: 01/20/2023] Open
Abstract
Pinch1 and Pinch2 are LIM domain-containing proteins with crucial functions in mediating focal adhesion formation. Our previous studies have demonstrated that Pinch1/2 expression is essential for cartilage and bone formation during skeletal development in mice. Loss of Pinch expression (Prx1Cre; Pinch1flox/flox; Pinch2-/-) inhibits chondrocyte proliferation and promotes chondrocyte apoptosis, resulting in severe chondrodysplasia and limb shortening. Based on these observations, we wonder if Pinch proteins have a role in adult cartilage and whether Pinch deficiency will compromise cartilage homeostasis and promote osteoarthritis (OA)-related defects in adult mice. To this end, we generated the AggrecanCreERT2; Pinch1flox/flox; Pinch2-/- mice, in which the Pinch1 gene can be inducibly deleted in aggrecan-expressing chondrocytes by tamoxifen and the Pinch2 gene is globally inactivated. Immunofluorescent staining confirmed that the expression of Pinch proteins was significantly decreased in articular cartilage in tamoxifen-treated adult AggrecanCreERT2; Pinch1flox/flox; Pinch2-/- mice. Unexpectedly, our results showed that Pinch loss did not induce marked abnormalities in articular cartilage and other joint tissues in the knee joints of either adult (10-month-old) mice or aged (17-month-old) mice. In a destabilization of the medial meniscus (DMM)-induced OA model, the surgically-induced OA lesions were comparable between Pinch-deficient mice and control mice. Given the fact that Pinch proteins are essential for chondrogenesis and cartilage formation during skeletal development, these findings suggest that Pinch expression is seemingly not indispensable for adult cartilage homeostasis in mice.
Collapse
Affiliation(s)
- Xiaohao Wu
- Department of Biochemistry, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Sixiong Lin
- Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Department of Spine Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Rongdong Liao
- Department of Orthopaedics, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Qing Yao
- Department of Biochemistry, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Lijun Lin
- Department of Orthopaedics, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xuenong Zou
- Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Department of Spine Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Guozhi Xiao
- Department of Biochemistry, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
6
|
Paes de Faria J, Vale-Silva RS, Fässler R, Werner HB, Relvas JB. Pinch2 regulates myelination in the mouse central nervous system. Development 2022; 149:275524. [DOI: 10.1242/dev.200597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 05/16/2022] [Indexed: 11/20/2022]
Abstract
ABSTRACT
The extensive morphological changes of oligodendrocytes during axon ensheathment and myelination involve assembly of the Ilk-Parvin-Pinch (IPP) heterotrimeric complex of proteins to relay essential mechanical and biochemical signals between integrins and the actin cytoskeleton. Binding of Pinch1 and Pinch2 isoforms to Ilk is mutually exclusive and allows the formation of distinct IPP complexes with specific signaling properties. Using tissue-specific conditional gene ablation in mice, we reveal an essential role for Pinch2 during central nervous system myelination. Unlike Pinch1 gene ablation, loss of Pinch2 in oligodendrocytes results in hypermyelination and in the formation of pathological myelin outfoldings in white matter regions. These structural changes concur with inhibition of Rho GTPase RhoA and Cdc42 activities and phenocopy aspects of myelin pathology observed in corresponding mouse mutants. We propose a dual role for Pinch2 in preventing an excess of myelin wraps through RhoA-dependent control of membrane growth and in fostering myelin stability via Cdc42-dependent organization of cytoskeletal septins. Together, these findings indicate that IPP complexes containing Pinch2 act as a crucial cell-autonomous molecular hub ensuring synchronous control of key signaling networks during developmental myelination.
Collapse
Affiliation(s)
- Joana Paes de Faria
- Department of Neurobiology and Neurological Disease, Glial Cell Biology Laboratory, Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto 1 , 4200-135 Porto , Portugal
- Department of Neurobiology and Neurological Disease, Glial Cell Biology Laboratory, Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto 2 , 4200-135 Porto , Portugal
| | - Raquel S. Vale-Silva
- Department of Neurobiology and Neurological Disease, Glial Cell Biology Laboratory, Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto 1 , 4200-135 Porto , Portugal
- Department of Neurobiology and Neurological Disease, Glial Cell Biology Laboratory, Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto 2 , 4200-135 Porto , Portugal
- Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto 3 , 4050-313 Porto , Portugal
| | - Reinhard Fässler
- Department of Molecular Medicine, Max Planck Institute of Biochemistry 4 , 82152 Martinsried , Germany
| | - Hauke B. Werner
- Max Planck Institute of Experimental Medicine 5 Department of Neurogenetics , , D-37075 Gottingen , Germany
| | - João B. Relvas
- Department of Neurobiology and Neurological Disease, Glial Cell Biology Laboratory, Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto 1 , 4200-135 Porto , Portugal
- Department of Neurobiology and Neurological Disease, Glial Cell Biology Laboratory, Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto 2 , 4200-135 Porto , Portugal
- Faculty of Medicine, Universidade do Porto 6 Department of Biomedicine , , 4200-319 Porto , Portugal
| |
Collapse
|
7
|
Mierke CT, Hayn A, Fischer T. PINCH1 Promotes Fibroblast Migration in Extracellular Matrices and Influences Their Mechanophenotype. Front Cell Dev Biol 2022; 10:869563. [PMID: 35652097 PMCID: PMC9149598 DOI: 10.3389/fcell.2022.869563] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 04/15/2022] [Indexed: 11/13/2022] Open
Abstract
Cell migration performs a critical function in numerous physiological processes, including tissue homeostasis or wound healing after tissue injury, as well as pathological processes that include malignant progression of cancer. The efficiency of cell migration and invasion appears to be based on the mechano-phenotype of the cytoskeleton. The properties of the cytoskeleton depend on internal cytoskeletal and external environmental factors. A reason for this are connections between the cell and its local matrix microenvironment, which are established by cell-matrix adhesion receptors. Upon activation, focal adhesion proteins such as PINCH1 are recruited to sites where focal adhesions form. PINCH1 specifically couples through interactions with ILK, which binds to cell matrix receptors and the actomyosin cytoskeleton. However, the role of PINCH1 in cell mechanics regulating cellular motility in 3D collagen matrices is still unclear. PINCH1 is thought to facilitate 3D motility by regulating cellular mechanical properties, such as stiffness. In this study, PINCH1 wild-type and knock-out cells were examined for their ability to migrate in dense extracellular 3D matrices. Indeed, PINCH1 wild-type cells migrated more numerously and deeper in 3D matrices, compared to knock-out cells. Moreover, cellular deformability was determined, e.g., elastic modulus (stiffness). PINCH1 knock-out cells are more deformable (compliable) than PINCH1 wild-type cells. Migration of both PINCH1−/− cells and PINCH1fl/fl cells was decreased by Latrunculin A inhibition of actin polymerization, suggesting that actin cytoskeletal differences are not responsible for the discrepancy in invasiveness of the two cell types. However, the mechanical phenotype of PINCH1−/− cells may be reflected by Latrunculin A treatment of PINCH1fl/fl cells, as they exhibit resembling deformability to untreated PINCH1−/− cells. Moreover, an apparent mismatch exists between the elongation of the long axis and the contraction of the short axis between PINCH1fl/fl cells and PINCH1−/− cells following Latrunculin A treatment. There is evidence of this indicating a shift in the proxy values for Poisson’s ratio in PINCH1−/− cells compared with PINCH1fl/fl cells. This is probably attributable to modifications in cytoskeletal architecture. The non-muscle myosin II inhibitor Blebbistatin also reduced the cell invasiveness in 3D extracellular matrices but instead caused a stiffening of the cells. Finally, PINCH1 is apparently essential for providing cellular mechanical stiffness through the actin cytoskeleton, which regulates 3D motility.
Collapse
|
8
|
Zhao R, Trainor PA. Epithelial to mesenchymal transition during mammalian neural crest cell delamination. Semin Cell Dev Biol 2022; 138:54-67. [PMID: 35277330 DOI: 10.1016/j.semcdb.2022.02.018] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 02/08/2022] [Accepted: 02/21/2022] [Indexed: 11/18/2022]
Abstract
Epithelial to mesenchymal transition (EMT) is a well-defined cellular process that was discovered in chicken embryos and described as "epithelial to mesenchymal transformation" [1]. During EMT, epithelial cells lose their epithelial features and acquire mesenchymal character with migratory potential. EMT has subsequently been shown to be essential for both developmental and pathological processes including embryo morphogenesis, wound healing, tissue fibrosis and cancer [2]. During the past 5 years, interest and study of EMT especially in cancer biology have increased exponentially due to the implied role of EMT in multiple aspects of malignancy such as cell invasion, survival, stemness, metastasis, therapeutic resistance and tumor heterogeneity [3]. Since the process of EMT in embryogenesis and cancer progression shares similar phenotypic changes, core transcription factors and molecular mechanisms, it has been proposed that the initiation and development of carcinoma could be attributed to abnormal activation of EMT factors usually required for normal embryo development. Therefore, developmental EMT mechanisms, whose timing, location, and tissue origin are strictly regulated, could prove useful for uncovering new insights into the phenotypic changes and corresponding gene regulatory control of EMT under pathological conditions. In this review, we initially provide an overview of the phenotypic and molecular mechanisms involved in EMT and discuss the newly emerging concept of epithelial to mesenchymal plasticity (EMP). Then we focus on our current knowledge of a classic developmental EMT event, neural crest cell (NCC) delamination, highlighting key differences in our understanding of NCC EMT between mammalian and non-mammalian species. Lastly, we highlight available tools and future directions to advance our understanding of mammalian NCC EMT.
Collapse
Affiliation(s)
- Ruonan Zhao
- Stowers Institute for Medical Research, Kansas City, MO, USA; Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Paul A Trainor
- Stowers Institute for Medical Research, Kansas City, MO, USA; Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, USA.
| |
Collapse
|
9
|
Ortega MA, Chaowen C, Fraile-Martinez O, García-Montero C, Saez MA, Cruza I, Pereda-Cerquella C, Alvarez-Mon MA, Guijarro LG, Fatych Y, Menor-Salván C, Alvarez-Mon M, De Leon-Luis J, Buján J, Garcia-Honduvilla N, Bravo C, Asúnsolo-del-Barco A. Chronic Venous Disease in Pregnant Women Causes an Increase in ILK in the Placental Villi Associated with a Decrease in E-Cadherin. J Pers Med 2022; 12:jpm12020277. [PMID: 35207765 PMCID: PMC8875350 DOI: 10.3390/jpm12020277] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 02/08/2022] [Accepted: 02/11/2022] [Indexed: 02/06/2023] Open
Abstract
Chronic venous disease (CVD) is a multifactorial vascular disorder frequently manifested in lower limbs in the form of varicose veins (VVs). Women are a vulnerable population for suffering from CVD, especially during pregnancy, when a plethora of changes occur in their cardiovascular system. Previous studies have indicated a worrisome association between CVD in pregnancy with the placental structure and function. Findings include an altered cellular behavior and extracellular matrix (ECM) composition. Integrin-linked kinase (ILK) is a critical molecule involved in multiple physiological and pathological conditions, and together with cadherins, is essential to mediate cell to ECM and cell to cell interplay, respectively. Thus, the aim of this study was to evaluate the implication of ILK and a set of cadherins (e-cadherin, cadherin-6 and cadherin-17) in placentas of women with CVD in order to unravel the possible pathophysiological role of these components. Gene expression (RT-qPCR) and protein expression (immunohistochemistry) studies were performed. Our results show a significant increase in the gene and protein expression of ILK, cadherin-6 and cadherin-17 and a decrease of e-cadherin in the placenta of women with CVD. Overall, this work shows that an abnormal expression of ILK, e-cadherin, cadherin-6 and cadherin-17 may be implicated in the pathological changes occurring in the placental tissue. Further studies should be conducted to determine the possible associations of these changes with maternal and fetal well-being.
Collapse
Affiliation(s)
- Miguel A. Ortega
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (O.F.-M.); (C.G.-M.); (M.A.S.); (I.C.); (C.P.-C.); (M.A.A.-M.); (M.A.-M.); (J.B.); (N.G.-H.)
- Ramón y Cajal Institute of Healthcare Research (IRYCIS), 28001 Madrid, Spain; (L.G.G.); (A.A.-d.-B.)
- Cancer Registry and Pathology Department, Hospital Universitario Principe de Asturias, 28801 Alcalá de Henares, Spain
- Correspondence: (M.A.O.); (C.B.); Tel.: +34-91-885-45-40 (M.A.O.); Fax: +34-91-885-48-85 (M.A.O.)
| | - Chen Chaowen
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain;
| | - Oscar Fraile-Martinez
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (O.F.-M.); (C.G.-M.); (M.A.S.); (I.C.); (C.P.-C.); (M.A.A.-M.); (M.A.-M.); (J.B.); (N.G.-H.)
- Ramón y Cajal Institute of Healthcare Research (IRYCIS), 28001 Madrid, Spain; (L.G.G.); (A.A.-d.-B.)
| | - Cielo García-Montero
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (O.F.-M.); (C.G.-M.); (M.A.S.); (I.C.); (C.P.-C.); (M.A.A.-M.); (M.A.-M.); (J.B.); (N.G.-H.)
- Ramón y Cajal Institute of Healthcare Research (IRYCIS), 28001 Madrid, Spain; (L.G.G.); (A.A.-d.-B.)
| | - Miguel A. Saez
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (O.F.-M.); (C.G.-M.); (M.A.S.); (I.C.); (C.P.-C.); (M.A.A.-M.); (M.A.-M.); (J.B.); (N.G.-H.)
- Ramón y Cajal Institute of Healthcare Research (IRYCIS), 28001 Madrid, Spain; (L.G.G.); (A.A.-d.-B.)
- Pathological Anatomy Service, Central University Hospital of Defence-UAH, 28001 Madrid, Spain
| | - Iris Cruza
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (O.F.-M.); (C.G.-M.); (M.A.S.); (I.C.); (C.P.-C.); (M.A.A.-M.); (M.A.-M.); (J.B.); (N.G.-H.)
| | - Claude Pereda-Cerquella
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (O.F.-M.); (C.G.-M.); (M.A.S.); (I.C.); (C.P.-C.); (M.A.A.-M.); (M.A.-M.); (J.B.); (N.G.-H.)
| | - Miguel Angel Alvarez-Mon
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (O.F.-M.); (C.G.-M.); (M.A.S.); (I.C.); (C.P.-C.); (M.A.A.-M.); (M.A.-M.); (J.B.); (N.G.-H.)
- Ramón y Cajal Institute of Healthcare Research (IRYCIS), 28001 Madrid, Spain; (L.G.G.); (A.A.-d.-B.)
| | - Luis G. Guijarro
- Ramón y Cajal Institute of Healthcare Research (IRYCIS), 28001 Madrid, Spain; (L.G.G.); (A.A.-d.-B.)
- Unit of Biochemistry and Molecular Biology (CIBEREHD), Department of System Biology, University of Alcalá, 28801 Alcalá de Henares, Spain; (Y.F.); (C.M.-S.)
| | - Yuliia Fatych
- Unit of Biochemistry and Molecular Biology (CIBEREHD), Department of System Biology, University of Alcalá, 28801 Alcalá de Henares, Spain; (Y.F.); (C.M.-S.)
| | - César Menor-Salván
- Unit of Biochemistry and Molecular Biology (CIBEREHD), Department of System Biology, University of Alcalá, 28801 Alcalá de Henares, Spain; (Y.F.); (C.M.-S.)
| | - Melchor Alvarez-Mon
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (O.F.-M.); (C.G.-M.); (M.A.S.); (I.C.); (C.P.-C.); (M.A.A.-M.); (M.A.-M.); (J.B.); (N.G.-H.)
- Ramón y Cajal Institute of Healthcare Research (IRYCIS), 28001 Madrid, Spain; (L.G.G.); (A.A.-d.-B.)
- Immune System Diseases-Rheumatology and Oncology Service, University Hospital Príncipe de Asturias, CIBEREHD, 28801 Alcalá de Henares, Spain
| | - Juan De Leon-Luis
- Department of Public and Maternal and Child Health, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain;
- Department of Obstetrics and Gynecology, University Hospital Gregorio Marañón, 28009 Madrid, Spain
- Health Research Institute Gregorio Marañón, 28009 Madrid, Spain
| | - Julia Buján
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (O.F.-M.); (C.G.-M.); (M.A.S.); (I.C.); (C.P.-C.); (M.A.A.-M.); (M.A.-M.); (J.B.); (N.G.-H.)
- Ramón y Cajal Institute of Healthcare Research (IRYCIS), 28001 Madrid, Spain; (L.G.G.); (A.A.-d.-B.)
| | - Natalio Garcia-Honduvilla
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (O.F.-M.); (C.G.-M.); (M.A.S.); (I.C.); (C.P.-C.); (M.A.A.-M.); (M.A.-M.); (J.B.); (N.G.-H.)
- Ramón y Cajal Institute of Healthcare Research (IRYCIS), 28001 Madrid, Spain; (L.G.G.); (A.A.-d.-B.)
| | - Coral Bravo
- Department of Public and Maternal and Child Health, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain;
- Department of Obstetrics and Gynecology, University Hospital Gregorio Marañón, 28009 Madrid, Spain
- Health Research Institute Gregorio Marañón, 28009 Madrid, Spain
- Correspondence: (M.A.O.); (C.B.); Tel.: +34-91-885-45-40 (M.A.O.); Fax: +34-91-885-48-85 (M.A.O.)
| | - Angel Asúnsolo-del-Barco
- Ramón y Cajal Institute of Healthcare Research (IRYCIS), 28001 Madrid, Spain; (L.G.G.); (A.A.-d.-B.)
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain;
| |
Collapse
|
10
|
Bang ML, Bogomolovas J, Chen J. Understanding the molecular basis of cardiomyopathy. Am J Physiol Heart Circ Physiol 2022; 322:H181-H233. [PMID: 34797172 PMCID: PMC8759964 DOI: 10.1152/ajpheart.00562.2021] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/16/2021] [Accepted: 11/16/2021] [Indexed: 02/03/2023]
Abstract
Inherited cardiomyopathies are a major cause of mortality and morbidity worldwide and can be caused by mutations in a wide range of proteins located in different cellular compartments. The present review is based on Dr. Ju Chen's 2021 Robert M. Berne Distinguished Lectureship of the American Physiological Society Cardiovascular Section, in which he provided an overview of the current knowledge on the cardiomyopathy-associated proteins that have been studied in his laboratory. The review provides a general summary of the proteins in different compartments of cardiomyocytes associated with cardiomyopathies, with specific focus on the proteins that have been studied in Dr. Chen's laboratory.
Collapse
Affiliation(s)
- Marie-Louise Bang
- Institute of Genetic and Biomedical Research (IRGB), National Research Council (CNR), Milan Unit, Milan, Italy
- IRCCS Humanitas Research Hospital, Rozzano (Milan), Italy
| | - Julius Bogomolovas
- Division of Cardiovascular Medicine, Department of Medicine Cardiology, University of California, San Diego, La Jolla, California
| | - Ju Chen
- Division of Cardiovascular Medicine, Department of Medicine Cardiology, University of California, San Diego, La Jolla, California
| |
Collapse
|
11
|
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
|
12
|
Li G, Qian H. Hydroxymethylation of protein-encoding genes in the testes involved in precocious puberty of Eriocheir sinensis. Gene 2019; 683:18-27. [PMID: 30315924 DOI: 10.1016/j.gene.2018.10.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Revised: 09/04/2018] [Accepted: 10/09/2018] [Indexed: 10/28/2022]
Abstract
To investigate the possible effects of epigenetic modification of testis protein-encoding genes on precocious puberty of Eriocheir sinensis, we used MeDIP-seq and hMeDIP-seq techniques to compare the methylation and hydroxymethylation of 263 E. sinensis protein-encoding genes known in the NCBI database in precocious testes with those in normally developing testes. The results showed that total methylation level of those genes was lower than their total hydroxymethylation level. Moreover, their total hydroxymethylation level in precocious testes was significantly lower than that in normal testes. In addition, no methylated genes had significant difference, but there were 37 different hydroxymethylated genes (DhMGs) in the precocious testes compared to the normal ones. Among the DhMGs, 21 were hypo-hydroxymethylated and 16 were hyper-hydroxymethylated. The hypo-hydroxymethylated DhMGs were associated with development, cell structural and cytoskeletal proteins, and response to stress. However, the hyper-hydroxymethylated DhMGs included immune-related genes, free radicals removement-related genes, protein folding-related genes, and so on. In addition, some DhMGs were hyper-hydroxymethylated while their homologous DhMGs were hypo-hydroxymethylated. The results of a qRT-PCR assay showed that the expression levels of 5 DhMGs randomly chosen presented a positive correlation with their hydroxymethylation levels. It can be seen that hydroxymethylation might regulate the expression of genes and be involved in precocious puberty to cause high mortality of crabs. Therefore, the hydroxymethylation level of DhMGs may be used as an evaluation index with economically meaningful growth and breeding traits.
Collapse
Affiliation(s)
- Genliang Li
- Youjiang Medical University for Nationalities, Baise 533000, Guangxi, People's Republic of China.
| | - Hui Qian
- Youjiang Medical University for Nationalities, Baise 533000, Guangxi, People's Republic of China
| |
Collapse
|
13
|
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
|
14
|
Kadry YA, Huet-Calderwood C, Simon B, Calderwood DA. Kindlin-2 interacts with a highly conserved surface of ILK to regulate focal adhesion localization and cell spreading. J Cell Sci 2018; 131:jcs.221184. [PMID: 30254023 DOI: 10.1242/jcs.221184] [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: 06/06/2018] [Accepted: 09/17/2018] [Indexed: 12/27/2022] Open
Abstract
The integrin-associated adaptor proteins integrin-linked kinase (ILK) and kindlin-2 play central roles in integrin signaling and control of cell morphology. A direct ILK-kindlin-2 interaction is conserved across species and involves the F2PH subdomain of kindlin-2 and the pseudokinase domain (pKD) of ILK. However, complete understanding of the ILK-kindlin-2 interaction and its role in integrin-mediated signaling has been impeded by difficulties identifying the binding site for kindlin-2 on ILK. We used conservation-guided mapping to dissect the interaction between ILK and kindlin-2 and identified a previously unknown binding site for kindlin-2 on the C-lobe of the pKD of ILK. Mutations at this site inhibit binding to kindlin-2 while maintaining structural integrity of the pKD. Importantly, kindlin-binding-defective ILK mutants exhibit impaired focal adhesion localization and fail to fully rescue the spreading defects seen in ILK knockdown cells. Furthermore, kindlin-2 mutants with impaired ILK binding are also unable to fully support cell spreading. Thus, the interaction between ILK and kindlin-2 is critical for cell spreading and focal adhesion localization, representing a key signaling axis downstream of integrins.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Yasmin A Kadry
- From the Department of Pharmacology, Yale University, New Haven CT 06510, USA
| | | | - Bertrand Simon
- From the Department of Pharmacology, Yale University, New Haven CT 06510, USA
| | - David A Calderwood
- From the Department of Pharmacology, Yale University, New Haven CT 06510, USA .,Department of Cell Biology, Yale University, New Haven CT 06510, USA
| |
Collapse
|
15
|
|
16
|
Liu C, Russin J, Heck C, Kawata K, Adiga R, Yen W, Lambert J, Stear B, Law M, Marquez Y, Crino P, Millett D, Langford D. Dysregulation of PINCH signaling in mesial temporal epilepsy. J Clin Neurosci 2017; 36:43-52. [PMID: 27838154 PMCID: PMC6492941 DOI: 10.1016/j.jocn.2016.10.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 09/26/2016] [Accepted: 10/12/2016] [Indexed: 01/15/2023]
Abstract
Mounting evidence suggests that inflammation is important in epileptogenesis. Particularly Interesting New Cysteine Histidine-rich (PINCH) protein is a highly conserved, LIM-domain protein known to interact with hyperphosphorylated Tau. We assessed PINCH expression in resected epileptogenic human hippocampi and further explored the relationships among PINCH, hpTau and associated kinases. Resected hippocampal tissue from 7 patients with mesial temporal lobe epilepsy (MTLE) was assessed by Western analyses to measure levels of PINCH and hyperphosphorylated Tau, as well as changes in phosphorylation levels of associated kinases AKT and GSK3β in comparison to normal control tissue. Immunolabeling was also conducted to evaluate PINCH and hpTau patterns of expression, co-localization and cell-type specific expression. Hippocampal PINCH was increased by 2.6 fold in the epilepsy cases over controls and hpTau was increased 10 fold over control. Decreased phospho-AKT and phospho-GSK3β in epilepsy tissue suggested involvement of this pathway in MTLE. PINCH and hpTau co-localized in some neurons in MTLE tissue. While PINCH was expressed by both neurons and astrocytes in MTLE tissue, hpTau was extracellular or associated with neurons. PINCH was absent from the serum of control subjects but readily detectable from the serum of patients with chronic epilepsy. Our study describes the expression of PINCH and points to AKT/GSK3β signaling dysregulation as a possible pathway in hpTau formation in MTLE. In view of the interactions between hpTau and PINCH, understanding the role of PINCH in MTLE may provide increased understanding of mechanisms leading to inflammation and MTLE epileptogenesis and a potential biomarker for drug-resistant epilepsy.
Collapse
Affiliation(s)
- Charles Liu
- Department of Neurological Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Jon Russin
- Department of Neurological Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Christianne Heck
- Department of Neurological Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Keisuke Kawata
- Department of Neuroscience, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA; Department of Kinesiology, College of Public Health, Philadelphia, PA, USA; Department of Kinesiology, University of Indiana, Philadelphia, PA, USA
| | - Radhika Adiga
- Department of Neuroscience, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - William Yen
- Department of Neuroscience, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Jonathan Lambert
- Department of Pharmacology, Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA, USA
| | - Benjamin Stear
- Department of Neuroscience, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Meng Law
- Department of Neurological Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Yvette Marquez
- Department of Neurological Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Peter Crino
- Department of Neurology, Temple University School of Medicine, and Shriners Hospitals Pediatric Research Center, Philadelphia, PA, USA
| | - David Millett
- Department of Neurological Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Dianne Langford
- Department of Neuroscience, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA.
| |
Collapse
|
17
|
Xu H, Cao H, Xiao G. Signaling via PINCH: Functions, binding partners and implications in human diseases. Gene 2016; 594:10-15. [PMID: 27590440 DOI: 10.1016/j.gene.2016.08.039] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 08/24/2016] [Indexed: 12/16/2022]
Abstract
Particularly interesting new cysteine-histidine-rich protein (PINCH) is a LIM-domain-only adaptor that plays important roles in cytoskeletal organization and extracellular matrix adhesion, migration, proliferation and survival. Mammalian cells have two functional PINCH proteins, PINCH1 and PINCH2. PINCH not only binds to Nck2 and engages in the signaling of growth factor receptors, but also forms a ternary complex with ILK and parvin (IPP complex). Normally, the IPP complex locates to focal adhesions participating in the signaling of integrins and mediating the interaction of cytoskeleton and extracellular matrix (ECM). Accumulative evidence indicates that abnormalities in PINCH signaling are involved in the pathogenesis of important diseases, such as cancers, renal diseases, cardiomyopathy, and HIV. Therefore, clarifying the functions of PINCH and its interactions with key factors is important for better understanding of signaling events both in health and disease.
Collapse
Affiliation(s)
- Huamin Xu
- Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing 100191, China; Department of Biology and Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen 518055, China
| | - Huiling Cao
- Department of Biology and Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen 518055, China
| | - Guozhi Xiao
- Department of Biology and Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen 518055, China; Department of Biochemistry, Rush University Medical Center, Chicago, IL 60612, United States.
| |
Collapse
|
18
|
Xu D, Jalal SI, Sledge GW, Meroueh SO. Small-molecule binding sites to explore protein-protein interactions in the cancer proteome. MOLECULAR BIOSYSTEMS 2016; 12:3067-87. [PMID: 27452673 DOI: 10.1039/c6mb00231e] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The Cancer Genome Atlas (TCGA) offers an unprecedented opportunity to identify small-molecule binding sites on proteins with overexpressed mRNA levels that correlate with poor survival. Here, we analyze RNA-seq and clinical data for 10 tumor types to identify genes that are both overexpressed and correlate with patient survival. Protein products of these genes were scanned for binding sites that possess shape and physicochemical properties that can accommodate small-molecule probes or therapeutic agents (druggable). These binding sites were classified as enzyme active sites (ENZ), protein-protein interaction sites (PPI), or other sites whose function is unknown (OTH). Interestingly, the overwhelming majority of binding sites were classified as OTH. We find that ENZ, PPI, and OTH binding sites often occurred on the same structure suggesting that many of these OTH cavities can be used for allosteric modulation of enzyme activity or protein-protein interactions with small molecules. We discovered several ENZ (PYCR1, QPRT, and HSPA6) and PPI (CASC5, ZBTB32, and CSAD) binding sites on proteins that have been seldom explored in cancer. We also found proteins that have been extensively studied in cancer that have not been previously explored with small molecules that harbor ENZ (PKMYT1, STEAP3, and NNMT) and PPI (HNF4A, MEF2B, and CBX2) binding sites. All binding sites were classified by the signaling pathways to which the protein that harbors them belongs using KEGG. In addition, binding sites were mapped onto structural protein-protein interaction networks to identify promising sites for drug discovery. Finally, we identify pockets that harbor missense mutations previously identified from analysis of TCGA data. The occurrence of mutations in these binding sites provides new opportunities to develop small-molecule probes to explore their function in cancer.
Collapse
Affiliation(s)
- David Xu
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA
| | | | | | | |
Collapse
|
19
|
Rooney N, Wang P, Brennan K, Gilmore AP, Streuli CH. The Integrin-Mediated ILK-Parvin-αPix Signaling Axis Controls Differentiation in Mammary Epithelial Cells. J Cell Physiol 2016; 231:2408-17. [PMID: 27019299 PMCID: PMC5053222 DOI: 10.1002/jcp.25390] [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: 03/23/2016] [Accepted: 03/24/2016] [Indexed: 02/03/2023]
Abstract
Epithelial cell adhesion to the surrounding extracellular matrix is necessary for their proper behavior and function. During pregnancy and lactation, mammary epithelial cells (MECs) receive signals from their interaction with laminin via β1‐integrin (β1‐itg) to establish apico‐basal polarity and to differentiate in response to prolactin. Downstream of β1‐itg, the scaffold protein Integrin Linked Kinase (ILK) has been identified as the key signal transducer that is required for both lactational differentiation and the establishment of apico‐basal polarity. ILK is an adaptor protein that forms the IPP complex with PINCH and Parvins, which are central to its adaptor functions. However, it is not known how ILK and its interacting partners control tissue‐specific gene expression. Expression of ILK mutants, which weaken the interaction between ILK and Parvin, revealed that Parvins have a role in mammary epithelial differentiation. This conclusion was supported by shRNA‐mediated knockdown of the Parvins. In addition, shRNA knockdown of the Parvin‐binding guanine nucleotide exchange factor αPix prevented prolactin‐induced differentiation. αPix depletion did not disrupt focal adhesions, MEC proliferation, or polarity. This suggests that αPix represents a differentiation‐specific bifurcation point in β1‐itg‐ILK adhesive signaling. In summary, this study has identified a new role for Parvin and αPix downstream of the integrin‐ILK signaling axis for MEC differentiation. J. Cell. Physiol. 231: 2408–2417, 2016. © 2016 The Authors. Journal of Cellular Physiology Published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Nicholas Rooney
- The Wellcome Trust Centre for Cell-Matrix Research and Manchester Breast Centre, Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom
| | - Pengbo Wang
- The Wellcome Trust Centre for Cell-Matrix Research and Manchester Breast Centre, Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom
| | - Keith Brennan
- The Wellcome Trust Centre for Cell-Matrix Research and Manchester Breast Centre, Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom
| | - Andrew P Gilmore
- The Wellcome Trust Centre for Cell-Matrix Research and Manchester Breast Centre, Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom
| | - Charles H Streuli
- The Wellcome Trust Centre for Cell-Matrix Research and Manchester Breast Centre, Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
20
|
Abstract
Biodegradable zinc (Zn) metals, a new generation of biomaterials, have attracted much attention due to their excellent biodegradability, bioabsorbability, and adaptability to tissue regeneration. Compared with magnesium (Mg) and iron (Fe), Zn exhibits better corrosion and mechanical behaviors in orthopedic and stent applications. After implantation, Zn containing material will slowly degrade, and Zn ions (Zn2+) will be released to the surrounding tissue. For stent applications, the local Zn2+concentration near endothelial tissue/cells could be high. However, it is unclear how endothelia will respond to such high concentrations of Zn2+, which is pivotal to vascular remodeling and regeneration. Here, we evaluated the short-term cellular behaviors of primary human coronary artery endothelial cells (HCECs) exposed to a concentration gradient (0-140 μM) of extracellular Zn2+. Zn2+ had an interesting biphasic effect on cell viability, proliferation, spreading, and migration. Generally, low concentrations of Zn2+ promoted viability, proliferation, adhesion, and migration, while high concentrations of Zn2+ had opposite effects. For gene expression profiles, the most affected functional genes were related to cell adhesion, cell injury, cell growth, angiogenesis, inflammation, vessel tone, and coagulation. These results provide helpful information and guidance for Zn-based alloy design as well as the controlled release of Zn2+in stent and other related medical applications.
Collapse
Affiliation(s)
- Jun Ma
- Department of Chemical, Biological and Bio-Engineering, North Carolina Agricultural and Technical State University, 1601 East Market Street, Greensboro, North Carolina 27411, United States
- NSF-ERC for Revolutionizing Metallic Biomaterial, North Carolina Agricultural and Technical State University, 1601 East Market Street, Greensboro, North Carolina 27411, United States
| | - Nan Zhao
- Department of Chemical, Biological and Bio-Engineering, North Carolina Agricultural and Technical State University, 1601 East Market Street, Greensboro, North Carolina 27411, United States
- NSF-ERC for Revolutionizing Metallic Biomaterial, North Carolina Agricultural and Technical State University, 1601 East Market Street, Greensboro, North Carolina 27411, United States
| | - Donghui Zhu
- Department of Chemical, Biological and Bio-Engineering, North Carolina Agricultural and Technical State University, 1601 East Market Street, Greensboro, North Carolina 27411, United States
- NSF-ERC for Revolutionizing Metallic Biomaterial, North Carolina Agricultural and Technical State University, 1601 East Market Street, Greensboro, North Carolina 27411, United States
| |
Collapse
|
21
|
Kim YC, Gonzalez-Nieves R, Cutler ML. Rsu1 contributes to cell adhesion and spreading in MCF10A cells via effects on P38 map kinase signaling. Cell Adh Migr 2014; 9:227-32. [PMID: 25482629 PMCID: PMC4594256 DOI: 10.4161/19336918.2014.972775] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The ILK, PINCH, Parvin (IPP) complex regulates adhesion and migration via binding of ILK to β1 integrin and α−parvin thus linking focal adhesions to actin cytoskeleton. ILK also binds the adaptor protein PINCH which connects signaling proteins including Rsu1 to the complex. A recent study of Rsu1 and PINCH1 in non-transformed MCF10A human mammary epithelial cells revealed that the siRNA-mediated depletion of either Rsu1 or PINCH1 decreased the number of focal adhesions (FAs) and altered the distribution and localization of FA proteins. This correlated with reduced adhesion, failure to spread or migrate in response to EGF and a loss of actin stress fibers and caveolae. The depletion of Rsu1 caused significant reduction in PINCH1 implying that Rsu1 may function in part by regulating levels of PINCH1. However, Rsu1, but not PINCH1, was required for EGF-induced activation of p38 Map kinase and ATF2 phosphorylation, suggesting a Rsu1 function independent from the IPP complex. Reconstitution of Rsu1-depleted cells with a Rsu1 mutant (N92D) that does not bind to PINCH1 failed to restore FAs or migration but did promote IPP-independent spreading and constitutive as well as EGF-induced p38 activation. In this commentary we discuss p38 activity in adhesion and how Rsu1 expression may be linked to Map kinase kinase (MKK) activation and detachment-induced stress kinase signaling.
Collapse
Affiliation(s)
- Yong-Chul Kim
- a Department of Pathology; F. Edward Hebert School of Medicine ; Uniformed Services University of the Health Sciences ; Bethesda , MD USA
| | | | | |
Collapse
|
22
|
Jones CI, Tucker KL, Sasikumar P, Sage T, Kaiser WJ, Moore C, Emerson M, Gibbins JM. Integrin-linked kinase regulates the rate of platelet activation and is essential for the formation of stable thrombi. J Thromb Haemost 2014; 12:1342-52. [PMID: 24888521 DOI: 10.1111/jth.12620] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Accepted: 05/21/2014] [Indexed: 12/18/2022]
Abstract
BACKGROUND Integrin-linked kinase (ILK) and its associated complex of proteins are involved in many cellular activation processes, including cell adhesion and integrin signaling. We have previously demonstrated that mice with induced platelet ILK deficiency show reduced platelet activation and aggregation, but only a minor bleeding defect. Here, we explore this apparent disparity between the cellular and hemostatic phenotypes. METHODS The impact of ILK inhibition on integrin αII b β3 activation and degranulation was assessed with the ILK-specific inhibitor QLT0267, and a conditional ILK-deficient mouse model was used to assess the impact of ILK deficiency on in vivo platelet aggregation and thrombus formation. RESULTS Inhibition of ILK reduced the rate of both fibrinogen binding and α-granule secretion, but was accompanied by only a moderate reduction in the maximum extent of platelet activation or aggregation in vitro. The reduction in the rate of fibrinogen binding occurred prior to degranulation or translocation of αII b β3 to the platelet surface. The change in the rate of platelet activation in the absence of functional ILK led to a reduction in platelet aggregation in vivo, but did not change the size of thrombi formed following laser injury of the cremaster arteriole wall in ILK-deficient mice. It did, however, result in a marked decrease in the stability of thrombi formed in ILK-deficient mice. CONCLUSION Taken together, the findings of this study indicate that, although ILK is not essential for platelet activation, it plays a critical role in facilitating rapid platelet activation, which is essential for stable thrombus formation.
Collapse
Affiliation(s)
- C I Jones
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, Reading, UK
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Honda S, Shirotani-Ikejima H, Tadokoro S, Tomiyama Y, Miyata T. The integrin-linked kinase-PINCH-parvin complex supports integrin αIIbβ3 activation. PLoS One 2013; 8:e85498. [PMID: 24376884 PMCID: PMC3871693 DOI: 10.1371/journal.pone.0085498] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2013] [Accepted: 12/05/2013] [Indexed: 12/22/2022] Open
Abstract
Integrin-linked kinase (ILK) is an important signaling regulator that assembles into the heteroternary complex with adaptor proteins PINCH and parvin (termed the IPP complex). We recently reported that ILK is important for integrin activation in a Chinese hamster ovary (CHO) cell system. We previously established parental CHO cells expressing a constitutively active chimeric integrin (αIIbα6Bβ3) and mutant CHO cells expressing inactive αIIbα6Bβ3 due to ILK deficiency. In this study, we further investigated the underlying mechanisms for ILK-dependent integrin activation. ILK-deficient mutant cells had trace levels of PINCH and α-parvin, and transfection of ILK cDNA into the mutant cells increased not only ILK but also PINCH and α-parvin, resulting in the restoration of αIIbα6Bβ3 activation. In the parental cells expressing active αIIbα6Bβ3, ILK, PINCH, and α-parvin were co-immunoprecipitated, indicating the formation of the IPP complex. Moreover, short interfering RNA (siRNA) experiments targeting PINCH-1 or both α- and β-parvin mRNA in the parent cells impaired the αIIbα6Bβ3 activation as well as the expression of the other components of the IPP complex. In addition, ILK mutants possessing defects in either PINCH or parvin binding failed to restore αIIbα6Bβ3 activation in the mutant cells. Kindlin-2 siRNA in the parental cells impaired αIIbα6Bβ3 activation without disturbing the expression of ILK. For CHO cells stably expressing wild-type αIIbβ3 that is an inactive form, overexpression of a talin head domain (THD) induced αIIbβ3 activation and the THD-induced αIIbβ3 activation was impaired by ILK siRNA through a significant reduction in the expression of the IPP complex. In contrast, overexpression of all IPP components in the αIIbβ3-expressing CHO cells further augmented THD-induced αIIbβ3 activation, whereas they did not induce αIIbβ3 activation without THD. These data suggest that the IPP complex rather than ILK plays an important role and supports integrin activation probably through stabilization of the active conformation.
Collapse
Affiliation(s)
- Shigenori Honda
- Department of Molecular Pathogenesis, National Cerebral and Cardiovascular Center, Suita, Japan
- * E-mail:
| | | | - Seiji Tadokoro
- Department of Hematology and Oncology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Yoshiaki Tomiyama
- Department of Hematology and Oncology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
- Department of Blood Transfusion, Osaka University Hospital, Suita, Osaka, Japan
| | - Toshiyuki Miyata
- Department of Molecular Pathogenesis, National Cerebral and Cardiovascular Center, Suita, Japan
| |
Collapse
|
24
|
Akhshi TK, Wernike D, Piekny A. Microtubules and actin crosstalk in cell migration and division. Cytoskeleton (Hoboken) 2013; 71:1-23. [DOI: 10.1002/cm.21150] [Citation(s) in RCA: 110] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Revised: 10/02/2013] [Accepted: 10/06/2013] [Indexed: 12/22/2022]
Affiliation(s)
| | - Denise Wernike
- Department of Biology; Concordia University; Montreal Quebec Canada
| | - Alisa Piekny
- Department of Biology; Concordia University; Montreal Quebec Canada
| |
Collapse
|
25
|
Gonzalez-Nieves R, Desantis AI, Cutler ML. Rsu1 contributes to regulation of cell adhesion and spreading by PINCH1-dependent and - independent mechanisms. J Cell Commun Signal 2013; 7:279-93. [PMID: 23765260 PMCID: PMC3889256 DOI: 10.1007/s12079-013-0207-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2013] [Accepted: 05/28/2013] [Indexed: 01/29/2023] Open
Abstract
Cell adhesion and migration are complex processes that require integrin activation, the formation and dissolution of focal adhesion (FAs), and linkage of actin cytoskeleton to the FAs. The IPP (ILK, PINCH, Parvin) complex regulates FA formation via binding of the adaptor protein ILK to β1 integrin, PINCH and parvin. The signaling protein Rsu1 is linked to the complex via binding PINCH1. The role of Rsu1 and PINCH1 in adhesion and migration was examined in non-transformed mammary epithelial cells. Confocal microscopy revealed that the depletion of either Rsu1 or PINCH1 by siRNA in MCF10A cells decreased the number of focal adhesions and altered the distribution and localization of β1 integrin, vinculin, talin and paxillin without affecting the levels of FA protein expression. This correlated with reduced adhesion, failure to spread or migrate in response to EGF and a loss of actin stress fibers and caveolae. In addition, constitutive phosphorylation of actin regulatory proteins occurred in the absence of PINCH1. The depletion of Rsu1 caused significant reduction in PINCH1 implying that Rsu1 may function by regulating levels of PINCH1. However, while both Rsu1- or PINCH1-depleted cells retained the ability to activate adhesion signaling in response to EGF stimulation, only Rsu1 was required for EGF-induced p38 Map Kinase phosphorylation and ATF2 activation, suggesting an Rsu1 function independent from the IPP complex. Reconstitution of Rsu1-depleted cells with an Rsu1 mutant that does not bind to PINCH1 failed to restore FAs or migration but did promote spreading and constitutive p38 activation. These data show that Rsu1-PINCH1 association with ILK and the IPP complex is required for regulation of adhesion and migration but that Rsu1 has a critical role in linking integrin-induced adhesion to activation of p38 Map kinase signaling and cell spreading. Moreover, it suggests that Rsu1 may regulate p38 signaling from the IPP complex affecting other functions including survival.
Collapse
Affiliation(s)
- Reyda Gonzalez-Nieves
- Department of Pathology, F. Edward Hebert School of Medicine, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA
| | | | | |
Collapse
|
26
|
Yoshigi M, Pronovost SM, Kadrmas JL. Interactions by 2D Gel Electrophoresis Overlap (iGEO): a novel high fidelity approach to identify constituents of protein complexes. Proteome Sci 2013; 11:21. [PMID: 23663728 PMCID: PMC3688448 DOI: 10.1186/1477-5956-11-21] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2013] [Accepted: 05/08/2013] [Indexed: 01/29/2023] Open
Abstract
Background Here we describe a novel approach used to identify the constituents of protein complexes with high fidelity, using the integrin-associated scaffolding protein PINCH as a test case. PINCH is comprised of five LIM domains, zinc-finger protein interaction modules. In Drosophila melanogaster, PINCH has two known high-affinity binding partners—Integrin-linked kinase (ILK) that binds to LIM1 and Ras Suppressor 1 (RSU1) that binds to LIM5—but has been postulated to bind additional proteins as well. Results To purify PINCH complexes, in parallel we fused different affinity tags (Protein A and Flag) to different locations within the PINCH sequence (N- and C-terminus). We expressed these tagged versions of PINCH both in cell culture (overexpressed in Drosophila S2 cell culture in the presence of endogenous PINCH) and in vivo (at native levels in Drosophila lacking endogenous PINCH). After affinity purification, we analyzed PINCH complexes by a novel 2D-gel electrophoresis analysis, iGEO (interactions by 2D Gel Electrophoresis Overlap), with mass spectrometric identification of individual spots of interest. iGEO allowed the identification of protein partners that associate with PINCH under two independent purification strategies, providing confidence in the significance of the interaction. Proteins identified by iGEO were validated against a highly inclusive list of candidate PINCH interacting proteins identified in previous analyses by MuDPIT mass spectrometry. Conclusions The iGEO strategy confirmed a core complex comprised of PINCH, RSU1, ILK, and ILK binding partner Parvin. Our iGEO method also identified five novel protein partners that specifically interacted with PINCH in Drosophila S2 cell culture. Because of the improved reproducibility of 2D-GE methodology and the increasing affordability of the required labeling reagents, iGEO is a method that is accessible to most moderately well-equipped biological laboratories. The biochemical co-purifications inherent in iGEO allow for rapid and unambiguous identification of the constituents of protein complexes, without the need for extensive follow-up experiments.
Collapse
Affiliation(s)
- Masaaki Yoshigi
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, 84112, USA.
| | | | | |
Collapse
|
27
|
Pronovost SM, Beckerle MC, Kadrmas JL. Elevated expression of the integrin-associated protein PINCH suppresses the defects of Drosophila melanogaster muscle hypercontraction mutants. PLoS Genet 2013; 9:e1003406. [PMID: 23555310 PMCID: PMC3610608 DOI: 10.1371/journal.pgen.1003406] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2012] [Accepted: 02/07/2013] [Indexed: 01/05/2023] Open
Abstract
A variety of human diseases arise from mutations that alter muscle contraction. Evolutionary conservation allows genetic studies in Drosophila melanogaster to be used to better understand these myopathies and suggest novel therapeutic strategies. Integrin-mediated adhesion is required to support muscle structure and function, and expression of Integrin adhesive complex (IAC) proteins is modulated to adapt to varying levels of mechanical stress within muscle. Mutations in flapwing (flw), a catalytic subunit of myosin phosphatase, result in non-muscle myosin hyperphosphorylation, as well as muscle hypercontraction, defects in size, motility, muscle attachment, and subsequent larval and pupal lethality. We find that moderately elevated expression of the IAC protein PINCH significantly rescues flw phenotypes. Rescue requires PINCH be bound to its partners, Integrin-linked kinase and Ras suppressor 1. Rescue is not achieved through dephosphorylation of non-muscle myosin, suggesting a mechanism in which elevated PINCH expression strengthens integrin adhesion. In support of this, elevated expression of PINCH rescues an independent muscle hypercontraction mutant in muscle myosin heavy chain, MhcSamba1. By testing a panel of IAC proteins, we show specificity for PINCH expression in the rescue of hypercontraction mutants. These data are consistent with a model in which PINCH is present in limiting quantities within IACs, with increasing PINCH expression reinforcing existing adhesions or allowing for the de novo assembly of new adhesion complexes. Moreover, in myopathies that exhibit hypercontraction, strategic PINCH expression may have therapeutic potential in preserving muscle structure and function. A wide variety of diseases of the muscle are caused by mutations that alter either the actin and myosin contractile machinery or its regulation. One class of mutations of interest results in hypercontraction of the muscle—actin and myosin fibers contract, but cannot efficiently relax. We have used the fruit fly as a model to study these mutations because of the striking similarity of fly and human muscle and because of the many genetic techniques that are available in the fly. Using a genetic approach we identified a protein, PINCH, whose increased expression can rescue the defects observed in hypercontraction mutants. PINCH is a component of integrin adhesion complexes, responsible for anchoring cells in their environment. This suggests that strengthening the anchorage of muscles via PINCH may be an effective strategy to prevent or reduce the muscle damage that occurs in diseases of muscle hypercontraction.
Collapse
Affiliation(s)
- Stephen M. Pronovost
- Huntsman Cancer Institute, Departments of Biology and Oncological Sciences, University of Utah, Salt Lake City, Utah, United States of America
| | - Mary C. Beckerle
- Huntsman Cancer Institute, Departments of Biology and Oncological Sciences, University of Utah, Salt Lake City, Utah, United States of America
| | - Julie L. Kadrmas
- Huntsman Cancer Institute, Departments of Biology and Oncological Sciences, University of Utah, Salt Lake City, Utah, United States of America
- * E-mail:
| |
Collapse
|
28
|
Integrin-linked kinase regulates interphase and mitotic microtubule dynamics. PLoS One 2013; 8:e53702. [PMID: 23349730 PMCID: PMC3549953 DOI: 10.1371/journal.pone.0053702] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2012] [Accepted: 12/03/2012] [Indexed: 01/25/2023] Open
Abstract
Integrin-linked kinase (ILK) localizes to both focal adhesions and centrosomes in distinct multiprotein complexes. Its dual function as a kinase and scaffolding protein has been well characterized at focal adhesions, where it regulates integrin-mediated cell adhesion, spreading, migration and signaling. At the centrosomes, ILK regulates mitotic spindle organization and centrosome clustering. Our previous study showed various spindle defects after ILK knockdown or inhibition that suggested alteration in microtubule dynamics. Since ILK expression is frequently elevated in many cancer types, we investigated the effects of ILK overexpression on microtubule dynamics. We show here that overexpressing ILK in HeLa cells was associated with a shorter duration of mitosis and decreased sensitivity to paclitaxel, a chemotherapeutic agent that suppresses microtubule dynamics. Measurement of interphase microtubule dynamics revealed that ILK overexpression favored microtubule depolymerization, suggesting that microtubule destabilization could be the mechanism behind the decreased sensitivity to paclitaxel, which is known to stabilize microtubules. Conversely, the use of a small molecule inhibitor selective against ILK, QLT-0267, resulted in suppressed microtubule dynamics, demonstrating a new mechanism of action for this compound. We further show that treatment of HeLa cells with QLT-0267 resulted in higher inter-centromere tension in aligned chromosomes during mitosis, slower microtubule regrowth after cold depolymerization and the presence of a more stable population of spindle microtubules. These results demonstrate that ILK regulates microtubule dynamics in both interphase and mitotic cells.
Collapse
|
29
|
Gkretsi V, Papanikolaou V, Dubos S, Papathanasiou I, Giotopoulou N, Valiakou V, Wu C, Malizos KN, Tsezou A. Migfilin's elimination from osteoarthritic chondrocytes further promotes the osteoarthritic phenotype via β-catenin upregulation. Biochem Biophys Res Commun 2012; 430:494-9. [PMID: 23237804 DOI: 10.1016/j.bbrc.2012.12.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2012] [Accepted: 12/04/2012] [Indexed: 01/18/2023]
Abstract
Osteoarthritis (OA) is a debilitating disease of the joints characterized by cartilage degradation but to date there is no available pharmacological treatment to inhibit disease progression neither is there any available biomarker to predict its development. In the present study, we examined the expression level and possible involvement of novel cell-ECM adhesion-related molecules such as Iintegrin Linked Kinase (ILK), PINCH, parvin, Mig-2 and Migfilin in OA pathogenesis using primary human articular chondrocytes from healthy individuals and OA patients. Our findings show that only ILK and Migfilin were upregulated in OA compared to the normal chondrocytes. Interestingly, Migfilin silencing in OA chondrocytes rather exacerbated than ameliorated the osteoarthritic phenotype, as it resulted in even higher levels of catabolic and hypertrophic markers while at the same time induced reduction in ECM molecules such as aggrecan. Furthermore, we also provide a link between Migfilin and β-catenin activation in OA chondrocytes, showing Migfilin to be inversely correlated with β-catenin. Thus, the present study emphasizes for the first time to our knowledge the role of Migfilin in OA and highlights the importance of cell-ECM adhesion proteins in OA pathogenesis.
Collapse
Affiliation(s)
- Vasiliki Gkretsi
- Center for Research and Technology-Thessaly (CE.RE.TE.TH), Department of Biomedical Research and Technology, Larissa 41222, Greece.
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Davidson B, Holth A, Nguyen MTP, Tropé CG, Wu C. Migfilin, α-parvin and β-parvin are differentially expressed in ovarian serous carcinoma effusions, primary tumors and solid metastases. Gynecol Oncol 2012; 128:364-70. [PMID: 23099104 DOI: 10.1016/j.ygyno.2012.10.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2012] [Revised: 10/10/2012] [Accepted: 10/14/2012] [Indexed: 12/17/2022]
Abstract
OBJECTIVE The objective of this study is to analyze the expression and clinical role of integrin-linked kinase (ILK), α-parvin, β-parvin and migfilin in advanced-stage serous ovarian carcinoma. METHODS Expression of these 4 proteins was investigated in 205 effusions and in 94 patient-matched solid lesions (33 primary tumors and 61 solid metastases) using immunohistochemistry. Protein expression was analyzed for association with clinicopathologic parameters and survival. RESULTS ILK, α-parvin, β-parvin and migfilin were expressed in tumor cells in 53%, 2%, 28% and 53% of effusions and 57%, 20%, 83% and 25% of solid lesions, respectively. Statistical analysis showed significantly higher α-parvin and β-parvin expression in primary carcinomas (p=0.02 and p=0.001, respectively) and solid metastases (p=0.001 and p<0.001, respectively), compared to effusions, with opposite findings for migfilin (p=0.006 and p=0.008 for primary carcinomas and solid metastases, respectively). ILK expression was comparable at all anatomic sites. β-Parvin expression in effusions was associated with better response to chemotherapy at diagnosis (p=0.014), with no other significant association with clinicopathologic parameters or survival. Expression in primary tumors and solid metastases was similarly unrelated to clinicopathologic parameters or survival. CONCLUSIONS This study provides further evidence to our previous observations that the adhesion profile of ovarian serous carcinoma cells in effusions differs from their counterparts in primary carcinomas and solid metastases. β-Parvin may be a novel marker of chemoresponse in metastatic ovarian carcinoma.
Collapse
Affiliation(s)
- Ben Davidson
- Division of Pathology, Oslo University Hospital, Norwegian Radium Hospital, N-0424 Oslo, Norway.
| | | | | | | | | |
Collapse
|
31
|
Elias MC, Pronovost SM, Cahill KJ, Beckerle MC, Kadrmas JL. A crucial role for Ras suppressor-1 (RSU-1) revealed when PINCH and ILK binding is disrupted. J Cell Sci 2012; 125:3185-94. [PMID: 22467865 DOI: 10.1242/jcs.101386] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
PINCH, integrin-linked kinase (ILK) and Ras suppressor-1 (RSU-1) are molecular scaffolding proteins that form a physical complex downstream of integrins, and have overlapping roles in cellular adhesion. In Drosophila, PINCH and ILK colocalize in cells and have indistinguishable functions in maintaining wing adhesion and integrin to actin linkage in the muscle. We sought to determine whether the direct physical interaction between PINCH and ILK was essential for their functions using transgenic flies expressing a version of PINCH with a point mutation that disrupts ILK binding (PINCH(Q38A)). We demonstrate that the PINCH-ILK interaction is not required for viability, for integrin-mediated adhesion of the wing or muscle, or for maintaining appropriate localization or levels of either PINCH or ILK. These results suggest alternative modes for PINCH localization, stabilization and linkage to the actin cytoskeleton that are independent of a direct interaction with ILK. Furthermore, we identified a synthetic lethality in flies carrying both the PINCH(Q38A) mutation and a null mutation in the gene encoding RSU-1. This lethality does not result from PINCH mislocalization or destabilization, and illustrates a novel compensatory role for RSU-1 in maintaining viability in flies with compromised PINCH-ILK binding. Taken together, this work highlights the existence of redundant mechanisms in adhesion complex assembly that support integrin function in vivo.
Collapse
Affiliation(s)
- Maria C Elias
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT 84112, USA
| | | | | | | | | |
Collapse
|
32
|
Zervas CG, Psarra E, Williams V, Solomon E, Vakaloglou KM, Brown NH. A central multifunctional role of integrin-linked kinase at muscle attachment sites. J Cell Sci 2011; 124:1316-27. [PMID: 21444757 DOI: 10.1242/jcs.081422] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Integrin-linked kinase (ILK) is an essential component of a multiprotein complex that links actin to the plasma membrane. Here, we have used a genetic approach to examine the molecular interactions that are essential for the assembly of this ILK-containing complex at Drosophila muscle attachment sites (MASs). We show that, downstream of integrins, talin plays a decisive role in the recruitment of three proteins: ILK, PINCH and paxillin. The accumulation of ILK at MASs appears to follow an amplification mechanism, suggesting that numerous binding sites are generated by minimal levels of the upstream integrin and talin effectors. This property suggests that ILK functions as an essential hub in the assembly of its partner proteins at sites of integrin adhesion. We found that PINCH stability, and its subcellular localization at MASs, depends upon ILK function, but that ILK stability and localization is not dependent upon PINCH. An in vivo structure-function analysis of ILK demonstrated that each ILK domain has sufficient information for its independent recruitment at embryonic MASs, whereas at later developmental stages only the kinase domain was effectively recruited. Our data strengthen the view that the ILK complex is assembled sequentially at sites of integrin adhesion by employing multiple molecular interactions, which collectively stabilize the integrin-actin link.
Collapse
Affiliation(s)
- Christos G Zervas
- Biomedical Research Foundation, Academy of Athens, Division of Genetics, Soranou Efessiou 4, 11527 Athens, Greece.
| | | | | | | | | | | |
Collapse
|
33
|
Jatiani A, Pannizzo P, Gualco E, Del-Valle L, Langford D. Neuronal PINCH is regulated by TNF-α and is required for neurite extension. J Neuroimmune Pharmacol 2011; 6:330-40. [PMID: 20689998 PMCID: PMC3107369 DOI: 10.1007/s11481-010-9236-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2010] [Accepted: 07/18/2010] [Indexed: 12/18/2022]
Abstract
During HIV infection of the CNS, neurons are damaged by viral proteins, such as Tat and gp120, or by inflammatory factors, such as TNF-α, that are released from infected and/or activated glial cells. Host responses to this damage may include the induction of survival or repair mechanisms. In this context, previous studies report robust expression of a protein called particularly interesting new cysteine histidine-rich protein (PINCH), in the neurons of HIV patients' brains, compared with nearly undetectable levels in HIV-negative individuals (Rearden et al., J Neurosci Res 86:2535-2542, 2008), suggesting PINCH's involvement in neuronal signaling during HIV infection of the brain. To address potential triggers for PINCH induction in HIV patients' brains, an in vitro system mimicking some aspects of HIV infection of the CNS was utilized. We investigated neuronal PINCH expression, subcellular distribution, and biological consequences of PINCH sequestration upon challenge with Tat, gp120, and TNF-α. Our results indicate that in neurons, TNF-α stimulation increases PINCH expression and changes its subcellular localization. Furthermore, PINCH mobility is required to maintain neurite extension upon challenge with TNF-α. PINCH may function as a neuron-specific host-mediated response to challenge by HIV-related factors in the CNS.
Collapse
Affiliation(s)
- Asavari Jatiani
- Department of Neuroscience, Temple University School of Medicine, 3500 N. Broad St., MERB 750, Philadelphia, PA, USA
| | - Paola Pannizzo
- Department of Neuroscience, Temple University School of Medicine, 3500 N. Broad St., MERB 750, Philadelphia, PA, USA
| | - Elisa Gualco
- Department of Oncology, Biology and Genetics, University of Genoa, Genoa, Italy
| | - Luis Del-Valle
- Department of Pathology, Louisiana State University, New Orleans, LA, USA
| | - Dianne Langford
- Department of Neuroscience, Temple University School of Medicine, 3500 N. Broad St., MERB 750, Philadelphia, PA, USA,
| |
Collapse
|
34
|
Hwangbo C, Park J, Lee JH. mda-9/Syntenin protein positively regulates the activation of Akt protein by facilitating integrin-linked kinase adaptor function during adhesion to type I collagen. J Biol Chem 2011; 286:33601-12. [PMID: 21828040 DOI: 10.1074/jbc.m110.206789] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
The integrin-linked kinase (ILK)-PINCH1-α-parvin (IPP) complex functions as a signaling platform for integrins that modulates various cellular processes. ILK functions as a central adaptor for the assembly of IPP complex. We report here that mda-9/syntenin, a positive regulator of cancer metastasis, regulates the activation of Akt (also known as protein kinase B) by facilitating ILK adaptor function during adhesion to type I collagen (COL-I) in human breast cancer cells. COL-I stimulation induced the phosphorylation and plasma membrane translocation of Akt. Inhibition of mda-9/syntenin or expression of mutant ILK (E359K) significantly blocked the translocation of both ILK and Akt to the plasma membrane. mda-9/syntenin associated with ILK, and this association was increased at the plasma membrane by COL-I stimulation. Knockdown of mda-9/syntenin impaired COL-I-induced association of ILK with Akt and plasma membrane targeting of ILK-Akt complex. These results demonstrated that mda-9/syntenin regulates the activation of Akt by controlling the plasma membrane targeting of Akt via a mechanism that facilitates the association of Akt with ILK at the plasma membrane during adhesion to COL-I. On a striking note, inhibition of mda-9/syntenin impaired COL-I-induced plasma membrane translocation of the IPP complex and assembly of integrin β1-IPP signaling complexes. Thus, our study defines the role of mda-9/syntenin in ILK adaptor function and describes a new mechanism of mda-9/syntenin for regulation of cell migration.
Collapse
Affiliation(s)
- Cheol Hwangbo
- Medical and Biomaterials Research Center and Department of Biochemistry, College of Natural Sciences, Kangwon National University Chuncheon 200-701, Korea
| | | | | |
Collapse
|
35
|
Fukuda K, Knight JDR, Piszczek G, Kothary R, Qin J. Biochemical, proteomic, structural, and thermodynamic characterizations of integrin-linked kinase (ILK): cross-validation of the pseudokinase. J Biol Chem 2011; 286:21886-95. [PMID: 21524996 PMCID: PMC3122243 DOI: 10.1074/jbc.m111.240093] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2011] [Revised: 04/05/2011] [Indexed: 12/13/2022] Open
Abstract
Integrin-linked kinase (ILK) is one of the few evolutionarily conserved focal adhesion proteins involved in diverse cell adhesion-dependent physiological and pathological responses. Despite more than a decade of studies and extensive literature, the kinase function of ILK is controversial. ILK contains a highly degraded kinase active site but it has been argued that ILK may be an unusual manganese (Mn)-dependent serine-threonine kinase that targets specific substrates such as glycogen synthase kinase-3β (GSK-3β). In this study, we have tackled this issue by a systematic bottom-up biochemical, proteomic, structural, and thermodynamic analysis of ILK. We show that recombinant ILK from either bacteria or mammalian cells exhibits no kinase activity on GSK-3β in the presence of either Mn(2+) or the conventional kinase co-factor Mg(2+). A comprehensive and unbiased whole cell-based kinase assay using entire mammalian CG-4 and C2C12 cell lysate did not identify any specific ILK substrates. High resolution crystallographic structure analysis further confirmed that the Mn-bound ILK adopts the same pseudo active site conformation as that of the Mg-bound ILK. More importantly, thermodynamic analysis revealed that the K220M mutation, previously thought to inactivate ILK by disrupting ATP binding, significantly impairs the structural integrity and stability of ILK, which provides a new basis for understanding how this mutation caused renal agenesis, a failure of fetal kidney development. Collectively, our data provide strong evidence that ILK lacks intrinsic kinase function. It is a bona fide pseudokinase that likely evolved from an ancestral catalytic counterpart to act as a distinct scaffold to mediate protein-protein interactions during focal adhesion assembly and many other cellular events.
Collapse
Affiliation(s)
- Koichi Fukuda
- From the Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195
| | - James D. R. Knight
- the Ottawa Hospital Research Institute, Ottawa, Ontario K1H 8L6, Canada
- the Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada, and
| | - Grzegorz Piszczek
- the Biophysics Core Facility, NHLBI, National Institutes of Health, Bethesda, Maryland 20892
| | - Rashmi Kothary
- the Ottawa Hospital Research Institute, Ottawa, Ontario K1H 8L6, Canada
- the Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada, and
| | - Jun Qin
- From the Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195
| |
Collapse
|
36
|
Rooney N, Streuli CH. How integrins control mammary epithelial differentiation: a possible role for the ILK-PINCH-Parvin complex. FEBS Lett 2011; 585:1663-72. [PMID: 21570968 DOI: 10.1016/j.febslet.2011.05.014] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2011] [Revised: 05/05/2011] [Accepted: 05/05/2011] [Indexed: 01/15/2023]
Abstract
Differentiation into tissue-specific cell types occurs in response to numerous external signals. Integrins impart signals from the extracellular matrix microenvironment that are required for cell differentiation. However, the precise cytoplasmic transducers of these signals are yet to be understood properly. In lactating mammary epithelial cells, integrin-linked kinase has been identified as an indispensable integrin-signalling adaptor that enables the activation of Rac1, which is necessary for prolactin-induced milk protein expression. Here we use examples from various tissues to summarise possible mechanisms by which ILK and its binding partners PINCH and Parvin (ILK-PINCH-Parvin complex) could be required for Rac activation and mammary epithelial differentiation.
Collapse
Affiliation(s)
- Nicholas Rooney
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences and Manchester Breast Centre, University of Manchester, Manchester, UK
| | | |
Collapse
|
37
|
Kovalevich J, Tracy B, Langford D. PINCH: More than just an adaptor protein in cellular response. J Cell Physiol 2011; 226:940-7. [PMID: 20945343 DOI: 10.1002/jcp.22437] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Particularly interesting new cysteine-histidine-rich protein (PINCH) is a LIM-domain-only adaptor protein involved in protein recruitment, subsequent assembly of multi-protein complexes, and subcellular localization of these complexes. PINCH is developmentally regulated and its expression is critical for proper cytoskeletal organization and extracellular matrix adhesion. Although PINCH has no catalytic abilities, the PIP (PINCH-ILK-parvin) complex serves as a link between integrins and components of growth factor receptor kinase and GTPase signaling pathways. Accordingly, PINCH-mediated signaling induces cell migration, spreading, and survival. Further research on the signaling cascades affected by PINCH is key to appreciating its biological significance in cell fate and systems maintenance, as the developmental functions of PINCH may extend to disease states and the cellular response to damage. PINCH is implicated in a diverse array of diseases including renal failure, cardiomyopathy, nervous system degeneration and demyelination, and tumorigenesis. This review presents evidence for PINCH's structural and functional importance in normal cellular processes and in pathogenesis. The current data for PINCH expression in nervous system disease is substantial, but due to the complex and ubiquitous nature of this protein, our understanding of its function in pathology remains unclear. In this review, an overview of studies identifying PINCH binding partners, their molecular interactions, and the potentially overlapping role(s) of PINCH in cancer and in nervous system diseases will be discussed. Many questions remain regarding PINCH's role in cells. What induces cell-specific PINCH expression? How does PINCH expression contribute to cell fate in the central nervous system? More broadly, is PINCH expression in disease a good thing? Clarifying the ambiguous functions of PINCH expression in the central nervous system and other systems is important to understand more clearly signaling events both in health and disease.
Collapse
Affiliation(s)
- Jane Kovalevich
- Temple University School of Medicine, Department of Neuroscience, Philadelphia, Pennsylvania 19140, USA
| | | | | |
Collapse
|
38
|
Yuan L, Sanders MA, Basson MD. ILK mediates the effects of strain on intestinal epithelial wound closure. Am J Physiol Cell Physiol 2011; 300:C356-C367. [PMID: 21084641 PMCID: PMC3043633 DOI: 10.1152/ajpcell.00273.2010] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2010] [Accepted: 11/15/2010] [Indexed: 01/02/2023]
Abstract
The intestinal epithelium is subjected to repetitive deformation during normal gut function by peristalsis and villous motility. Such repetitive strain promotes intestinal epithelial migration across fibronectin in vitro, but signaling mediators for this are poorly understood. We hypothesized that integrin-linked kinase (ILK) mediates strain-stimulated migration in intestinal epithelial cells cultured on fibronectin. ILK kinase activity increased rapidly 5 min after strain induction in both Caco-2 and intestinal epithelial cell-6 (IEC-6) cells. Wound closure in response to strain was reduced in ILK small interfering RNA (siRNA)-transfected Caco-2 cell monolayers when compared with control siRNA-transfected Caco-2 cells. Pharmacological blockade of phosphatidylinositol-3 kinase (PI3K) or Src or reducing Src by siRNA prevented strain activation of ILK. ILK coimmunoprecipitated with focal adhesion kinase (FAK), and this association was decreased by mutation of FAK Tyr925 but not FAK Tyr397. Strain induction of FAK Tyr925 phosphorylation but not FAK Tyr397 or FAK Tyr576 phosphorylation was blocked in ILK siRNA-transfected cells. ILK-Src association was stimulated by strain and was blocked by the Src inhibitor PP2. Finally, ILK reduction by siRNA inhibited strain-induced phosphorylation of myosin light chain and Akt. These results suggest a strain-dependent signaling pathway in which ILK association with FAK and Src mediates the subsequent downstream strain-induced motogenic response and suggest that ILK induction by repetitive deformation may contribute to recovery from mucosal injury and restoration of the mucosal barrier in patients with prolonged ileus. ILK may therefore be an important target for intervention to maintain the mucosa in such patients.
Collapse
Affiliation(s)
- Lisi Yuan
- Dept. of Surgery, Michigan State University, East Lansing, MI 48912, USA
| | | | | |
Collapse
|
39
|
Liu Y, Liu J, Chen J, Cheng L, Cao Q, Zhu L, Sun Y, Liu Q, Li J. Molecular cloning and characterization of a novel splice variant of the LIM domain family gene, PINCH 2, in human testis. Mol Biotechnol 2010; 35:109-18. [PMID: 17435276 DOI: 10.1007/bf02686105] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/1999] [Revised: 11/30/1999] [Accepted: 11/30/1999] [Indexed: 01/29/2023]
Abstract
By hybridizing human adult testis cDNA microarrays with human adult and embryo testis cDNA probes, we identified a novel human testis gene, PINCH 2. PINCH 2 expression was 3.4-fold higher in adult than in fetal testis. The full length of its cDNA was 963 bp, with a 354-bp open reading frame (ORF), encoding a 117-amino acid protein. PINCH 2 was a splicing isoform of PINCH. It shared one exon, which encoded the LIM domain, with PINCH gene in human genome. Multitissue reverse transcriptase-polymerase chain reaction (RTPCR) analysis revealed that this gene was expressed variably in a wide range of tissues, with high expression levels in human adult testis. These results suggest that PINCH 2, a novel LIM domain-containing gene, may play an important role in testicular development/spermatogenesis.
Collapse
Affiliation(s)
- Yun Liu
- Institute of Stomatology, Nanjing Medical University, 140 Han Zhong Road, Nanjing 210029, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Matthews BD, Thodeti CK, Tytell JD, Mammoto A, Overby DR, Ingber DE. Ultra-rapid activation of TRPV4 ion channels by mechanical forces applied to cell surface beta1 integrins. Integr Biol (Camb) 2010; 2:435-42. [PMID: 20725677 PMCID: PMC3147167 DOI: 10.1039/c0ib00034e] [Citation(s) in RCA: 211] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Integrins are ubiquitous transmembrane mechanoreceptors that elicit changes in intracellular biochemistry in response to mechanical force application, but these alterations generally proceed over seconds to minutes. Stress-sensitive ion channels represent another class of mechanoreceptors that are activated much more rapidly (within msec), and recent findings suggest that calcium influx through Transient Receptor Potential Vanilloid-4 (TRPV4) channels expressed in the plasma membrane of bovine capillary endothelial cells is required for mechanical strain-induced changes in focal adhesion assembly, cell orientation and directional migration. However, whether mechanically stretching a cell's extracellular matrix (ECM) adhesions might directly activate cell surface ion channels remains unknown. Here we show that forces applied to beta1 integrins result in ultra-rapid (within 4 msec) activation of calcium influx through TRPV4 channels. The TRPV4 channels were specifically activated by mechanical strain in the cytoskeletal backbone of the focal adhesion, and not by deformation of the lipid bilayer or submembranous cortical cytoskeleton alone. This early-immediate calcium signaling response required the distal region of the beta1 integrin cytoplasmic tail that contains a binding site for the integrin-associated transmembrane CD98 protein, and external force application to CD98 within focal adhesions activated the same ultra-rapid calcium signaling response. Local direct strain-dependent activation of TRPV4 channels mediated by force transfer from integrins and CD98 may therefore enable compartmentalization of calcium signaling within focal adhesions that is critical for mechanical control of many cell behaviors that underlie cell and tissue development.
Collapse
Affiliation(s)
- Benjamin D Matthews
- Department of Medicine, Harvard Medical School and Children's Hospital, Boston, MA 02115, USA
| | | | | | | | | | | |
Collapse
|
41
|
Eke I, Koch U, Hehlgans S, Sandfort V, Stanchi F, Zips D, Baumann M, Shevchenko A, Pilarsky C, Haase M, Baretton GB, Calleja V, Larijani B, Fässler R, Cordes N. PINCH1 regulates Akt1 activation and enhances radioresistance by inhibiting PP1alpha. J Clin Invest 2010; 120:2516-27. [PMID: 20530873 DOI: 10.1172/jci41078] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2009] [Accepted: 04/28/2010] [Indexed: 01/03/2023] Open
Abstract
Tumor cell resistance to ionizing radiation and chemotherapy is a major obstacle in cancer therapy. One factor contributing to this is integrin-mediated adhesion to ECM. The adapter protein particularly interesting new cysteine-histidine-rich 1 (PINCH1) is recruited to integrin adhesion sites and promotes cell survival, but the mechanisms underlying this effect are not well understood. Here we have shown that PINCH1 is expressed at elevated levels in human tumors of diverse origins relative to normal tissue. Furthermore, PINCH1 promoted cell survival upon treatment with ionizing radiation in vitro and in vivo by perpetuating Akt1 phosphorylation and activity. Mechanistically, PINCH1 was found to directly bind to protein phosphatase 1alpha (PP1alpha) - an Akt1-regulating protein - and inhibit PP1alpha activity, resulting in increased Akt1 phosphorylation and enhanced radioresistance. Thus, our data suggest that targeting signaling molecules such as PINCH1 that function downstream of focal adhesions (the complexes that mediate tumor cell adhesion to ECM) may overcome radio- and chemoresistance, providing new therapeutic approaches for cancer.
Collapse
Affiliation(s)
- Iris Eke
- OncoRay - Center for Radiation Research in Oncology, Medical Faculty Carl Gustav Carus, Dresden University of Technology, 01307 Dresden, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Schistosoma mansoni: signal transduction processes during the development of the reproductive organs. Parasitology 2010; 137:497-520. [PMID: 20163751 DOI: 10.1017/s0031182010000053] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Among the topics of considerable interest concerning our understanding of the unusual biology of schistosomes is the sexual maturation of the female. The identification of genes coding for signal transduction proteins controlling essential steps of the pairing-dependent differentiation of the reproductive organs, vitellarium and ovary will help to substantiate our knowledge about this unique parasite. Furthermore, such signalling proteins could be potential targets to interfere with the development of this parasite to combat schistosomiasis since its pathology is caused by the eggs. This review summarises first post-genomic steps to elucidate the function of gonad-specific signalling molecules which were identified by homology-based cloning strategies, by in silico identification or by yeast two-hybrid interaction analyses, using a combination of novel techniques. These include the in vitro culture of adult schistosomes, their treatment with chemical inhibitors to block enzyme activity, the use of RNAi to silence gene function post-transcriptionally, and confocal laser scanning microscopy to study the morphological consequences of these experimental approaches. Finally, we propose a first model of protein networks that are active in the ovary regulating mitogenic activity and differentiation. Some of these molecules are also active in the testes of males, probably fulfilling similar roles as in the ovary.
Collapse
|
43
|
Chiswell BP, Stiegler AL, Razinia Z, Nalibotski E, Boggon TJ, Calderwood DA. Structural basis of competition between PINCH1 and PINCH2 for binding to the ankyrin repeat domain of integrin-linked kinase. J Struct Biol 2009; 170:157-63. [PMID: 19963065 DOI: 10.1016/j.jsb.2009.12.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2009] [Revised: 11/30/2009] [Accepted: 12/02/2009] [Indexed: 11/16/2022]
Abstract
Formation of a heterotrimeric IPP complex composed of integrin-linked kinase (ILK), the LIM domain protein PINCH, and parvin is important for signaling through integrin adhesion receptors. Mammals possess two PINCH genes that are expressed simultaneously in many tissues. PINCH1 and PINCH2 have overlapping functions and can compensate for one another in many settings; however, isoform-specific functions have been reported and it is proposed that association with a PINCH1- or PINCH2-containing IPP complex may provide a bifurcation point in integrin signaling promoting different cellular responses. Here we report that the LIM1 domains of PINCH1 and PINCH2 directly compete for the same binding site on the ankyrin repeat domain (ARD) of ILK. We determined the 1.9A crystal structure of the PINCH2 LIM1 domain complexed with the ARD of ILK, and show that disruption of this interface by point mutagenesis reduces binding in vitro and alters localization of PINCH2 in cells. These studies provide further evidence for the role of the PINCH LIM1 domain in association with ILK and highlight direct competition as one mechanism for regulating which PINCH isoform predominates in IPP complexes. Differential regulation of PINCH1 and PINCH2 expression may therefore provide a means for altering cellular integrin signaling pathways.
Collapse
Affiliation(s)
- Brian P Chiswell
- Department of Pharmacology, 333 Cedar Street, Yale University School of Medicine, New Haven, CT 06520, USA
| | | | | | | | | | | |
Collapse
|
44
|
Conti FJ, Monkley SJ, Wood MR, Critchley DR, Müller U. Talin 1 and 2 are required for myoblast fusion, sarcomere assembly and the maintenance of myotendinous junctions. Development 2009; 136:3597-606. [PMID: 19793892 PMCID: PMC2761109 DOI: 10.1242/dev.035857] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/01/2009] [Indexed: 01/08/2023]
Abstract
Talin 1 and 2 connect integrins to the actin cytoskeleton and regulate the affinity of integrins for ligands. In skeletal muscle, talin 1 regulates the stability of myotendinous junctions (MTJs), but the function of talin 2 in skeletal muscle is not known. Here we show that MTJ integrity is affected in talin 2-deficient mice. Concomitant ablation of talin 1 and 2 leads to defects in myoblast fusion and sarcomere assembly, resembling defects in muscle lacking beta1 integrins. Talin 1/2-deficient myoblasts express functionally active beta1 integrins, suggesting that defects in muscle development are not primarily caused by defects in ligand binding, but rather by disruptions of the interaction of integrins with the cytoskeleton. Consistent with this finding, assembly of integrin adhesion complexes is perturbed in the remaining muscle fibers of talin 1/2-deficient mice. We conclude that talin 1 and 2 are crucial for skeletal muscle development, where they regulate myoblast fusion, sarcomere assembly and the maintenance of MTJs.
Collapse
Affiliation(s)
- Francesco J Conti
- The Scripps Research Institute, Department of Cell Biology and Institute of Childhood and Neglected Diseases, La Jolla, CA 92037, USA
| | | | | | | | | |
Collapse
|
45
|
Hugo HJ, Wafai R, Blick T, Thompson EW, Newgreen DF. Staurosporine augments EGF-mediated EMT in PMC42-LA cells through actin depolymerisation, focal contact size reduction and Snail1 induction - a model for cross-modulation. BMC Cancer 2009; 9:235. [PMID: 19604397 PMCID: PMC2717979 DOI: 10.1186/1471-2407-9-235] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2008] [Accepted: 07/15/2009] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND A feature of epithelial to mesenchymal transition (EMT) relevant to tumour dissemination is the reorganization of actin cytoskeleton/focal contacts, influencing cellular ECM adherence and motility. This is coupled with the transcriptional repression of E-cadherin, often mediated by Snail1, Snail2 and Zeb1/deltaEF1. These genes, overexpressed in breast carcinomas, are known targets of growth factor-initiated pathways, however it is less clear how alterations in ECM attachment cross-modulate to regulate these pathways. EGF induces EMT in the breast cancer cell line PMC42-LA and the kinase inhibitor staurosporine (ST) induces EMT in embryonic neural epithelial cells, with F-actin de-bundling and disruption of cell-cell adhesion, via inhibition of aPKC. METHODS PMC42-LA cells were treated for 72 h with 10 ng/ml EGF, 40 nM ST, or both, and assessed for expression of E-cadherin repressor genes (Snail1, Snail2, Zeb1/deltaEF1) and EMT-related genes by QRT-PCR, multiplex tandem PCR (MT-PCR) and immunofluorescence +/- cycloheximide. Actin and focal contacts (paxillin) were visualized by confocal microscopy. A public database of human breast cancers was assessed for expression of Snail1 and Snail2 in relation to outcome. RESULTS When PMC42-LA were treated with EGF, Snail2 was the principal E-cadherin repressor induced. With ST or ST+EGF this shifted to Snail1, with more extreme EMT and Zeb1/deltaEF1 induction seen with ST+EGF. ST reduced stress fibres and focal contact size rapidly and independently of gene transcription. Gene expression analysis by MT-PCR indicated that ST repressed many genes which were induced by EGF (EGFR, CAV1, CTGF, CYR61, CD44, S100A4) and induced genes which alter the actin cytoskeleton (NLF1, NLF2, EPHB4). Examination of the public database of breast cancers revealed tumours exhibiting higher Snail1 expression have an increased risk of disease-recurrence. This was not seen for Snail2, and Zeb1/deltaEF1 showed a reverse correlation with lower expression values being predictive of increased risk. CONCLUSION ST in combination with EGF directed a greater EMT via actin depolymerisation and focal contact size reduction, resulting in a loosening of cell-ECM attachment along with Snail1-Zeb1/deltaEF1 induction. This appeared fundamentally different to the EGF-induced EMT, highlighting the multiple pathways which can regulate EMT. Our findings add support for a functional role for Snail1 in invasive breast cancer.
Collapse
Affiliation(s)
- Honor J Hugo
- Embryology Laboratory, Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Australia
- VBCRC Invasion and Metastasis Unit, St. Vincent's Institute of Medical Research, Melbourne, Australia
| | - Razan Wafai
- University of Melbourne Department of Surgery, St. Vincent's Hospital, Melbourne, Australia
| | - Tony Blick
- VBCRC Invasion and Metastasis Unit, St. Vincent's Institute of Medical Research, Melbourne, Australia
| | - Erik W Thompson
- VBCRC Invasion and Metastasis Unit, St. Vincent's Institute of Medical Research, Melbourne, Australia
- University of Melbourne Department of Surgery, St. Vincent's Hospital, Melbourne, Australia
| | - Donald F Newgreen
- Embryology Laboratory, Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Australia
| |
Collapse
|
46
|
Stanchi F, Grashoff C, Nguemeni Yonga CF, Grall D, Fässler R, Van Obberghen-Schilling E. Molecular dissection of the ILK-PINCH-parvin triad reveals a fundamental role for the ILK kinase domain in the late stages of focal-adhesion maturation. J Cell Sci 2009; 122:1800-11. [PMID: 19435803 DOI: 10.1242/jcs.044602] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Integrin-linked kinase (ILK) and cytoplasmic adaptors of the PINCH and parvin families form a ternary complex, termed IPP, that localizes to integrin adhesions. We show here that deletion of the genes encoding ILK or PINCH1 similarly blocks maturation of focal adhesions to tensin-rich and phosphotyrosine-poor fibrillar adhesions (FBs) by downregulating expression or recruitment of tensin and destabilizing alpha5beta1-integrin-cytoskeleton linkages. As IPP components are interdependent for integrin targeting and protein stability, functional dissection of the complex was achieved by fusing ILK, PINCH, parvin or their individual motifs to the cytoplasmic tail of beta3 integrin, normally excluded from FBs. Using this novel gain-of-function approach, we demonstrated that expression of the C-terminal kinase domain of ILK can restore tensin recruitment and prompt focal-adhesion maturation in IPP-null cells. Debilitating mutations in the paxillin- or ATP-binding sites of ILK, together with alpha-parvin silencing, revealed a determinant role for ILK-parvin association, but not for direct paxillin binding, in this function. We propose a model in which the C-terminal domain of ILK promotes integrin sorting by reinforcing alpha5beta1-integrin-actin linkage and controls force transmission by targeting tensin to maturing adhesions.
Collapse
Affiliation(s)
- Fabio Stanchi
- Institute of Developmental Biology and Cancer Research, University of Nice-Sophia Antiopolis, CNRS-UMR6543, Centre Antoine Lacassagne, 33 Avenue de Valombrose, 06189 Nice, France
| | | | | | | | | | | |
Collapse
|
47
|
Zong H, Bastie CC, Xu J, Fassler R, Campbell KP, Kurland IJ, Pessin JE. Insulin resistance in striated muscle-specific integrin receptor beta1-deficient mice. J Biol Chem 2009; 284:4679-88. [PMID: 19064993 PMCID: PMC2640962 DOI: 10.1074/jbc.m807408200] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2008] [Revised: 12/01/2008] [Indexed: 01/19/2023] Open
Abstract
Integrin receptor plays key roles in mediating both inside-out and outside-in signaling between cells and the extracellular matrix. We have observed that the tissue-specific loss of the integrin beta1 subunit in striated muscle results in a near complete loss of integrin beta1 subunit protein expression concomitant with a loss of talin and to a lesser extent, a reduction in F-actin content. Muscle-specific integrin beta1-deficient mice had no significant difference in food intake, weight gain, fasting glucose, and insulin levels with their littermate controls. However, dynamic analysis of glucose homeostasis using euglycemichyperinsulinemic clamps demonstrated a 44 and 48% reduction of insulin-stimulated glucose infusion rate and glucose clearance, respectively. The whole body insulin resistance resulted from a specific inhibition of skeletal muscle glucose uptake and glycogen synthesis without any significant effect on the insulin suppression of hepatic glucose output or insulin-stimulated glucose uptake in adipose tissue. The reduction in skeletal muscle insulin responsiveness occurred without any change in GLUT4 protein expression levels but was associated with an impairment of the insulin-stimulated protein kinase B/Akt serine 473 phosphorylation but not threonine 308. The inhibition of insulin-stimulated serine 473 phosphorylation occurred concomitantly with a decrease in integrin-linked kinase expression but with no change in the mTOR.Rictor.LST8 complex (mTORC2). These data demonstrate an in vivo crucial role of integrin beta1 signaling events in mediating cross-talk to that of insulin action.
Collapse
Affiliation(s)
- Haihong Zong
- Departments of Medicine and Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | | | | | | | | | | | | |
Collapse
|
48
|
|
49
|
Yang Y, Wang X, Hawkins CA, Chen K, Vaynberg J, Mao X, Tu Y, Zuo X, Wang J, Wang YX, Wu C, Tjandra N, Qin J. Structural basis of focal adhesion localization of LIM-only adaptor PINCH by integrin-linked kinase. J Biol Chem 2008; 284:5836-44. [PMID: 19117955 DOI: 10.1074/jbc.m805319200] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
The LIM-only adaptor PINCH (the particularly interesting cysteine- and histidine-rich protein) plays a pivotal role in the assembly of focal adhesions (FAs), supramolecular complexes that transmit mechanical and biochemical information between extracellular matrix and actin cytoskeleton, regulating diverse cell adhesive processes such as cell migration, cell spreading, and survival. A key step for the PINCH function is its localization to FAs, which depends critically on the tight binding of PINCH to integrin-linked kinase (ILK). Here we report the solution NMR structure of the core ILK.PINCH complex (28 kDa, K(D) approximately 68 nm) involving the N-terminal ankyrin repeat domain (ARD) of ILK and the first LIM domain (LIM1) of PINCH. We show that the ILK ARD exhibits five sequentially stacked ankyrin repeat units, which provide a large concave surface to grip the two contiguous zinc fingers of the PINCH LIM1. The highly electrostatic interface is evolutionally conserved but differs drastically from those of known ARD and LIM bound to other types of protein domains. Consistently mutation of a hot spot in LIM1, which is not conserved in other LIM domains, disrupted the PINCH binding to ILK and abolished the PINCH targeting to FAs. These data provide atomic insight into a novel modular recognition and demonstrate how PINCH is specifically recruited by ILK to mediate the FA assembly and cell-extracellular matrix communication.
Collapse
Affiliation(s)
- Yanwu Yang
- Department of Molecular Cardiology, Lerner Research Institute, The Cleveland Clinic, Cleveland, Ohio 44195, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
The structural basis of integrin-linked kinase-PINCH interactions. Proc Natl Acad Sci U S A 2008; 105:20677-82. [PMID: 19074270 DOI: 10.1073/pnas.0811415106] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
The heterotrimeric complex between integrin-linked kinase (ILK), PINCH, and parvin is an essential signaling platform, serving as a convergence point for integrin and growth-factor signaling and regulating cell adhesion, spreading, and migration. We report a 1.6-A crystal structure of the ILK ankyrin repeat domain bound to the PINCH1 LIM1 domain, revealing the molecular basis of ILK-PINCH interactions and providing a structural description of this region of ILK. This structure identifies 5 ankyrin repeats in ILK, explains previous deletion mutagenesis data, permits identification of ILK and PINCH1 point mutations that disrupt the interaction, shows how zincs are coordinated by PINCH1 LIM1, and suggests that conformational flexibility and twisting between the 2 zinc fingers within the LIM1 domain may be important for ILK binding. These data provide an atomic-resolution description of a key interaction in the ILK-PINCH-parvin scaffolding complex.
Collapse
|