1
|
Katoh K. Integrin and Its Associated Proteins as a Mediator for Mechano-Signal Transduction. Biomolecules 2025; 15:166. [PMID: 40001469 PMCID: PMC11853369 DOI: 10.3390/biom15020166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 01/11/2025] [Accepted: 01/20/2025] [Indexed: 02/27/2025] Open
Abstract
Mechano-signal transduction is a process in which cells perceive extracellular mechanical signals, convert them into intracellular biochemical signals, and produce a response. Integrins are cell surface receptors that sense the extracellular mechanical cues and bind to the extracellular matrix (ECM). This binding induces integrin clustering and activation. Cytoplasmic tails of activated integrins interact and induce cytoskeleton tensions via several adaptor proteins. Integrins monitor extracellular stiffness via cytoskeleton tensions and modulate ECM stiffness via downstream signaling pathways regulating the expression of genes of ECM components. Integrin-mediated mechano-transduction is very crucial for the cell as it regulates the cell physiology both in normal and diseased conditions according to extracellular mechanical cues. It regulates cell proliferation, survival, and migration. Abnormal mechanical cues such as extreme and prolonged mechanical stress result in pathological conditions including fibrosis, cancers, skin, and autoimmune disorders. This paper aims to explore the role of integrins and their associated proteins in mechano-signal transduction. It highlights the integrins and their associated proteins as targets for therapy development. Furthermore, it also presents the challenges to the targeted drug development, which can be drug resistance and cytotoxicity. It is concluded in this paper that research on integrin-mediated mechano-signal transduction and its relationship with cell physiology and pathologies will be an important step towards the development of effective therapies.
Collapse
Affiliation(s)
- Kazuo Katoh
- Laboratory of Human Anatomy and Cell Biology, Faculty of Health Sciences, Tsukuba University of Technology, Tsukuba 305-8521, Japan
| |
Collapse
|
2
|
Li J, Xu Y, Jiang Y, Li N, Li Z, Kong D, Guo X, Zhang J, Zuo R. Nongenomic effects and mechanistic study of butyl benzyl phthalate-induced thyroid disruption: Based on integrated in vitro, in silico assays and proteome analysis. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 836:155715. [PMID: 35525365 DOI: 10.1016/j.scitotenv.2022.155715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 04/19/2022] [Accepted: 05/01/2022] [Indexed: 06/14/2023]
Abstract
Based on in vitro and in silico assays as well as proteome analysis, this study explored the nongenomic mechanism for butyl benzyl phthalate (BBP)-induced thyroid disruption. Molecular docking simulations showed that BBP could dock into the Arg-Gly-Asp (RGD) domain of integrin αvβ3 and form hydrogen bonds with a docking energy of -35.80 kcal/mol. This chemical enhanced rat pituitary tumor cell (GH3) proliferation and exhibited thyroid hormone-disrupting effects at 5-10 μmol/L. Meanwhile, BBP upregulated β3 gene expression and activated the downstream mitogen-activated protein kinase (MAPK) pathway in GH3 cells. Interestingly, GH3 cell proliferation was attenuated by integrin αvβ3 inhibitor (RGD peptide) or ERK1/2 inhibitor (PD98059), suggesting that the disruptions might be partly attributed to its interaction with integrin αvβ3 and activation of MAPK. Furthermore, quantitative proteomic analysis of zebrafish embryos exposed to BBP at an environmentally relevant concentration of 0.3 μmol/L revealed that BBP perturbed proteins and pathways related to cell communication (e.g., integrin binding) and signal transduction (e.g., MAPK signaling pathway). Taken together, our results supported that the biological effects of BBP-activated integrin αvβ3 mediated by the nongenomic pathway play an important role in its thyroid disruption. CAPSULE: The nongenomic pathway plays a vital role in the thyroid disruption-inducing actions of BBP.
Collapse
Affiliation(s)
- Jian Li
- Engineering Research Center of Groundwater Pollution Control and Remediation, Ministry of Education, College of Water Sciences, Beijing Normal University, Beijing 100875, China
| | - Ying Xu
- Engineering Research Center of Groundwater Pollution Control and Remediation, Ministry of Education, College of Water Sciences, Beijing Normal University, Beijing 100875, China
| | - Yuhao Jiang
- Key Laboratory of Radiopharmaceuticals of Ministry of Education, College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Na Li
- Key Laboratory of Drinking Water Science and Technology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Zhanjie Li
- Engineering Research Center of Groundwater Pollution Control and Remediation, Ministry of Education, College of Water Sciences, Beijing Normal University, Beijing 100875, China
| | - Dongdong Kong
- Engineering Research Center of Groundwater Pollution Control and Remediation, Ministry of Education, College of Water Sciences, Beijing Normal University, Beijing 100875, China
| | - Xueru Guo
- Engineering Research Center of Groundwater Pollution Control and Remediation, Ministry of Education, College of Water Sciences, Beijing Normal University, Beijing 100875, China
| | - Junbo Zhang
- Key Laboratory of Radiopharmaceuticals of Ministry of Education, College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Rui Zuo
- Engineering Research Center of Groundwater Pollution Control and Remediation, Ministry of Education, College of Water Sciences, Beijing Normal University, Beijing 100875, China.
| |
Collapse
|
3
|
Tamai S, Ichinose T, Tsutsui T, Tanaka S, Garaeva F, Sabit H, Nakada M. Tumor Microenvironment in Glioma Invasion. Brain Sci 2022; 12:brainsci12040505. [PMID: 35448036 PMCID: PMC9031400 DOI: 10.3390/brainsci12040505] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 04/11/2022] [Accepted: 04/12/2022] [Indexed: 02/05/2023] Open
Abstract
A major malignant trait of gliomas is their remarkable infiltration capacity. When glioma develops, the tumor cells have already reached the distant part. Therefore, complete removal of the glioma is impossible. Recently, research on the involvement of the tumor microenvironment in glioma invasion has advanced. Local hypoxia triggers cell migration as an environmental factor. The transcription factor hypoxia-inducible factor (HIF) -1α, produced in tumor cells under hypoxia, promotes the transcription of various invasion related molecules. The extracellular matrix surrounding tumors is degraded by proteases secreted by tumor cells and simultaneously replaced by an extracellular matrix that promotes infiltration. Astrocytes and microglia become tumor-associated astrocytes and glioma-associated macrophages/microglia, respectively, in relation to tumor cells. These cells also promote glioma invasion. Interactions between glioma cells actively promote infiltration of each other. Surgery, chemotherapy, and radiation therapy transform the microenvironment, allowing glioma cells to invade. These findings indicate that the tumor microenvironment may be a target for glioma invasion. On the other hand, because the living body actively promotes tumor infiltration in response to the tumor, it is necessary to reconsider whether the invasion itself is friend or foe to the brain.
Collapse
|
4
|
Trimarchi H, Ortiz A, Sánchez-Niño MD. Lyso-Gb3 Increases αvβ3 Integrin Gene Expression in Cultured Human Podocytes in Fabry Nephropathy. J Clin Med 2020; 9:jcm9113659. [PMID: 33203029 PMCID: PMC7696179 DOI: 10.3390/jcm9113659] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 11/02/2020] [Accepted: 11/09/2020] [Indexed: 12/23/2022] Open
Abstract
Background: Podocyturia in Fabry nephropathy leads to glomerulosclerosis and kidney disease progression. Integrins are involved in podocyte attachment to the glomerular basement membrane. We hypothesized that in Fabry nephropathy, lyso-Gb3 could modulate αvβ3 expression in podocytes. Together with UPAR, the αvβ3 integrin is a key mechanism involved in podocyte detachment and podocyturia. Methods: In cultured human podocytes stimulated with lyso-Gb3, the mRNA expression of the ITGAV and ITGB3 genes encoding integrins αv and β3, respectively, was analyzed by RT-qPCR. Results: In cultured human podocytes, lyso-Gb3 at concentrations encountered in the serum of Fabry patients increased ITGAV and ITGB3 mRNA levels within 3 to 6 h. This pattern of gene expression is similar to that previously observed for PLAUR (UPAR) gene expression but is in contrast to the delayed (24 h) upregulation of other markers of podocyte stress and mediators of injury, such as CD80, TGFβ1, CD74, Notch1, and HES. Conclusions: Human podocyte stress in response to glycolipid overload in Fabry nephropathy, exemplified by lyso-Gb3, is characterized by an early increase in the expression of components of the αvβ3/UPAR system, which contrasts with the delayed rise in the expression of other mediators of podocyte injury. This suggests that the αvβ3/UPAR system may be a therapeutic target in Fabry nephropathy.
Collapse
Affiliation(s)
- Hernán Trimarchi
- Nephrology Service, Hospital Británico de Buenos Aires, 1280 Buenos Aires, Argentina
- Correspondence: ; Tel.: +1280-5411-4309-6400
| | - Alberto Ortiz
- IIS-Fundación Jimenez Díaz, School of Medicine, UAM, 28040 Madrid, Spain; (A.O.); (M.D.S.-N.)
- Spanish Renal Research Network (REDINREN), 28004 Madrid, Spain
| | - Maria Dolores Sánchez-Niño
- IIS-Fundación Jimenez Díaz, School of Medicine, UAM, 28040 Madrid, Spain; (A.O.); (M.D.S.-N.)
- Spanish Renal Research Network (REDINREN), 28004 Madrid, Spain
- Pharmacology and Therapeutics Department, Universidad Autonoma de Madrid, 28049 Madrid, Spain
| |
Collapse
|
5
|
Expression of uPAR in Urinary Podocytes of Patients with Fabry Disease. Int J Nephrol 2017; 2017:1287289. [PMID: 28523190 PMCID: PMC5420917 DOI: 10.1155/2017/1287289] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2017] [Revised: 03/11/2017] [Accepted: 03/19/2017] [Indexed: 12/17/2022] Open
Abstract
Background. Despite enzyme replacement therapy, Fabry nephropathy still progresses. Podocyturia is an irreversible event that antedates proteinuria and leads to chronic renal failure. We evaluated a potential mechanism of podocyte detachment via the expression of the urokinase-type Plasminogen Activator Receptor (uPAR) in urinary podocytes of Fabry patients. Methods. This is a cross-sectional study that included controls (n = 20) and Fabry patients (n = 44) either untreated (n = 23) or treated with agalsidase-β (n = 21). Variables. Variables are estimated glomerular filtration rate (eGFR), urinary protein : creatinine ratio, and urinary uPAR+ podocyte : creatinine ratio. uPAR mRNA expression in response to lyso-Gb3, a bioactive glycolipid accumulated in Fabry disease, was studied in cultured human podocytes. Results. Controls and Fabry patients had similar age, gender, and renal function. Urinary uPAR+ podocytes were higher in patients than in controls. Untreated patients were significantly younger; had more females, and presented lower urinary protein : creatinine ratios and significantly higher urinary uPAR+ podocytes than treated subjects. In treated patients, urinary uPAR+ podocytes correlated with urinary protein : creatinine ratio (ρ = 0.5; p = 0.02). Lyso-Gb3 at concentrations found in the circulation of Fabry patients increased uPAR expression in cultured podocytes. Conclusions. Urinary podocytes expressing uPAR are increased in Fabry patients, especially in untreated patients. The potential contribution of uPAR expression to podocyte detachment merits further studies.
Collapse
|
6
|
Dzobo K, Vogelsang M, Parker MI. Wnt/β-Catenin and MEK-ERK Signaling are Required for Fibroblast-Derived Extracellular Matrix-Mediated Endoderm Differentiation of Embryonic Stem Cells. Stem Cell Rev Rep 2016; 11:761-73. [PMID: 26022506 DOI: 10.1007/s12015-015-9598-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Human embryonic stem cells (hESCs) have the potential to differentiate into all cells of the three germ layers, thus making them an attractive source of cells for use in regenerative medicine. The greatest challenge lies in regulating the differentiation of hESCs into specific cell lineages by both intrinsic and extrinsic factors. In this study we determined the effect of a fibroblast-derived extracellular matrix (fd-ECM) on hESCs differentiation. We demonstrate that growth of hESCs on fd-ECM results in hESCs losing their stemness and proliferation potential. As the stem cells differentiate they attain gene expression profiles similar to the primitive streak of the in vivo embryo. The activation of both the MEK-ERK and Wnt/β-catenin signaling pathways is required for the fd-ECM-mediated differentiation of hESCs towards the endoderm and involves integrins α1, α2, α3 and β1. This study illustrates the importance of the cellular microenvironment in directing stem cell fate and that the nature and composition of the extracellular matrix is a crucial determining factor.
Collapse
Affiliation(s)
- Kevin Dzobo
- Cape Town Component, Wernher and Beit Building (South), UCT Campus, International Centre for Genetic Engineering and Biotechnology (ICGEB), Anzio Road, Observatory, 7925, Cape Town, South Africa
| | | | | |
Collapse
|
7
|
Litan A, Langhans SA. Cancer as a channelopathy: ion channels and pumps in tumor development and progression. Front Cell Neurosci 2015; 9:86. [PMID: 25852478 PMCID: PMC4362317 DOI: 10.3389/fncel.2015.00086] [Citation(s) in RCA: 221] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 02/23/2015] [Indexed: 12/23/2022] Open
Abstract
Increasing evidence suggests that ion channels and pumps not only regulate membrane potential, ion homeostasis, and electric signaling in excitable cells but also play important roles in cell proliferation, migration, apoptosis and differentiation. Consistent with a role in cell signaling, channel proteins and ion pumps can form macromolecular complexes with growth factors, and cell adhesion and other signaling molecules. And while cancer is still not being cataloged as a channelopathy, as the non-traditional roles of ion pumps and channels are being recognized, it is increasingly being suggested that ion channels and ion pumps contribute to cancer progression. Cancer cell migration requires the regulation of adhesion complexes between migrating cells and surrounding extracellular matrix (ECM) proteins. Cell movement along solid surfaces requires a sequence of cell protrusions and retractions that mainly depend on regulation of the actin cytoskeleton along with contribution of microtubules and molecular motor proteins such as mysoin. This process is triggered and modulated by a combination of environmental signals, which are sensed and integrated by membrane receptors, including integrins and cadherins. Membrane receptors transduce these signals into downstream signaling pathways, often involving the Rho GTPase protein family. These pathways regulate the cytoskeletal rearrangements necessary for proper timing of adhesion, contraction and detachment of cells in order to find their way through extracellular spaces. Migration and adhesion involve continuous modulation of cell motility, shape and volume, in which ion channels and pumps play major roles. Research on cancer cells suggests that certain ion channels may be involved in aberrant tumor growth and channel inhibitors often lead to growth arrest. This review will describe recent research into the role of ion pumps and ion channels in cell migration and adhesion, and how they may contribute to tumor development.
Collapse
Affiliation(s)
- Alisa Litan
- Nemours Center for Childhood Cancer Research, Alfred I. duPont Hospital for Children Wilmington, DE, USA
| | - Sigrid A Langhans
- Nemours Center for Childhood Cancer Research, Alfred I. duPont Hospital for Children Wilmington, DE, USA
| |
Collapse
|
8
|
Wang L, Zhang C, Su X, Lin D. Kcnj10 is a major type of K+ channel in mouse corneal epithelial cells and plays a role in initiating EGFR signaling. Am J Physiol Cell Physiol 2014; 307:C710-7. [PMID: 25099735 DOI: 10.1152/ajpcell.00040.2014] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We used primary mouse corneal epithelial cells (pMCE) to examine the role of Kcnj10 in determining membrane K(+) conductance and cell membrane potential and in regulating EGF/TGFA release. Western blot, immunostaining, and RT-PCR detected the expression of Kcnj10 in mouse cornea. The single channel recording identified the 20-pS inwardly rectifying K(+) channels in pMCE of WT mice, but these channels were absent in Kcnj10(-/-). Moreover, the whole cell recording demonstrates that deletion of Kcnj10 largely abolished the inward K(+) currents and depolarized the cell membrane K(+) reversal potential (an index of the cell membrane potential). This suggests that Kcnj10 is a main contributor to the cell K(+) conductance and it is pivotal in generating membrane potential in cornea. Furthermore, to test the hypothesis that Kcnj10 expression plays a key role in the stimulation of growth factors release, we employed an immortalized human corneal epithelial cell line (HCE) transfected with siRNA-Kcnj10 or siRNA-control. Levels of TGFA and EGF secreted in the medium were measured by ELISA. Coimmunoprecipitation, biotinylation, and pull-down assay were used to examine the expression of EGFR and the GTP bound form of Rac1 (active Rac1). Downregulation of Kcnj10 activated Rac1 and enhanced EGF/TGFA release, which further contributed to the upregulation of EGFR phosphorylation and surface expression. We conclude that Kcnj10 is a main K(+) channel expressed in corneal epithelial cells and the inhibition of Kcnj10 resulted in depolarization, which in turn induced an EGF-like effect.
Collapse
Affiliation(s)
- Lijun Wang
- Department of Pharmacology, New York Medical College, Valhalla, New York
| | - Chengbiao Zhang
- Department of Pharmacology, New York Medical College, Valhalla, New York
| | - Xiaotong Su
- Department of Pharmacology, New York Medical College, Valhalla, New York
| | - Daohong Lin
- Department of Pharmacology, New York Medical College, Valhalla, New York
| |
Collapse
|
9
|
Gardinier JD, Gangadharan V, Wang L, Duncan RL. Hydraulic Pressure during Fluid Flow Regulates Purinergic Signaling and Cytoskeleton Organization of Osteoblasts. Cell Mol Bioeng 2014; 7:266-277. [PMID: 24910719 DOI: 10.1007/s12195-014-0329-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
During physiological activities, osteoblasts experience a variety of mechanical forces that stimulate anabolic responses at the cellular level necessary for the formation of new bone. Previous studies have primarily investigated the osteoblastic response to individual forms of mechanical stimuli. However in this study, we evaluated the response of osteoblasts to two simultaneous, but independently controlled stimuli; fluid flow-induced shear stress (FSS) and static or cyclic hydrostatic pressure (SHP or CHP, respectively). MC3T3-E1 osteoblasts-like cells were subjected to 12dyn/cm2 FSS along with SHP or CHP of varying magnitudes to determine if pressure enhances the anabolic response of osteoblasts during FSS. For both SHP and CHP, the magnitude of hydraulic pressure that induced the greatest release of ATP during FSS was 15 mmHg. Increasing the hydraulic pressure to 50 mmHg or 100 mmHg during FSS attenuated the ATP release compared to 15 mmHg during FSS. Decreasing the magnitude of pressure during FSS to atmospheric pressure reduced ATP release to that of basal ATP release from static cells and inhibited actin reorganization into stress fibers that normally occurred during FSS with 15 mmHg of pressure. In contrast, translocation of nuclear factor kappa B (NFκB) to the nucleus was independent of the magnitude of hydraulic pressure and was found to be mediated through the activation of phospholipase-C (PLC), but not src kinase. In conclusion, hydraulic pressure during FSS was found to regulate purinergic signaling and actin cytoskeleton reorganization in the osteoblasts in a biphasic manner, while FSS alone appeared to stimulate NFκB translocation. Understanding the effects of hydraulic pressure on the anabolic responses of osteoblasts during FSS may provide much needed insights into the physiologic effects of coupled mechanical stimuli on osteogenesis.
Collapse
Affiliation(s)
- Joseph D Gardinier
- Biomechanics and Movement Science, University of Delaware, Newark, DE, 19716 ; Department of Biological and Materials Science, University of Michigan, Ann Arbor, MI 48109
| | | | - Liyun Wang
- Biomechanics and Movement Science, University of Delaware, Newark, DE, 19716 ; Department of Mechanical Engineering, University of Delaware, Newark, DE, 19716
| | - Randall L Duncan
- Biomechanics and Movement Science, University of Delaware, Newark, DE, 19716 ; Department of Mechanical Engineering, University of Delaware, Newark, DE, 19716 ; Biological Sciences, University of Delaware, Newark, DE, 19716
| |
Collapse
|
10
|
Crociani O, Zanieri F, Pillozzi S, Lastraioli E, Stefanini M, Fiore A, Fortunato A, D'Amico M, Masselli M, De Lorenzo E, Gasparoli L, Chiu M, Bussolati O, Becchetti A, Arcangeli A. hERG1 channels modulate integrin signaling to trigger angiogenesis and tumor progression in colorectal cancer. Sci Rep 2013; 3:3308. [PMID: 24270902 PMCID: PMC3839040 DOI: 10.1038/srep03308] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2013] [Accepted: 11/05/2013] [Indexed: 12/12/2022] Open
Abstract
Angiogenesis is a potential target for cancer therapy. We identified a novel signaling pathway that sustains angiogenesis and progression in colorectal cancer (CRC). This pathway is triggered by β1 integrin-mediated adhesion and leads to VEGF-A secretion. The effect is modulated by the human ether-à-go-go related gene 1 (hERG1) K(+) channel. hERG1 recruits and activates PI3K and Akt. This in turn increases the Hypoxia Inducible Factor (HIF)-dependent transcription of VEGF-A and other tumour progression genes. This signaling pathway has novel features in that the integrin- and hERG1-dependent activation of HIF (i) is triggered in normoxia, especially after CRC cells have experienced a hypoxic stage, (ii) involves NF-kB and (iii) is counteracted by an active p53. Blocking hERG1 switches this pathway off also in vivo, by inhibiting cell growth, angiogenesis and metastatic spread. This suggests that non-cardiotoxic anti-hERG1 drugs might be a fruitful therapeutic strategy to prevent the failure of anti-VEGF therapy.
Collapse
Affiliation(s)
- Olivia Crociani
- Department of Experimental and Clinical Medicine, Section of Internal Medicine, University of Florence and Istituto Toscano Tumori (ITT), Florence, Italy
| | - Francesca Zanieri
- Department of Experimental and Clinical Medicine, Section of Internal Medicine, University of Florence and Istituto Toscano Tumori (ITT), Florence, Italy
| | - Serena Pillozzi
- Department of Experimental and Clinical Medicine, Section of Internal Medicine, University of Florence and Istituto Toscano Tumori (ITT), Florence, Italy
| | - Elena Lastraioli
- Department of Experimental and Clinical Medicine, Section of Internal Medicine, University of Florence and Istituto Toscano Tumori (ITT), Florence, Italy
| | - Matteo Stefanini
- Department of Experimental and Clinical Medicine, Section of Internal Medicine, University of Florence and Istituto Toscano Tumori (ITT), Florence, Italy
| | - Antonella Fiore
- Department of Experimental and Clinical Medicine, Section of Internal Medicine, University of Florence and Istituto Toscano Tumori (ITT), Florence, Italy
| | - Angelo Fortunato
- Department of Experimental and Clinical Medicine, Section of Internal Medicine, University of Florence and Istituto Toscano Tumori (ITT), Florence, Italy
| | - Massimo D'Amico
- Department of Experimental and Clinical Medicine, Section of Internal Medicine, University of Florence and Istituto Toscano Tumori (ITT), Florence, Italy
| | - Marika Masselli
- Department of Experimental and Clinical Medicine, Section of Internal Medicine, University of Florence and Istituto Toscano Tumori (ITT), Florence, Italy
| | - Emanuele De Lorenzo
- Department of Experimental and Clinical Medicine, Section of Internal Medicine, University of Florence and Istituto Toscano Tumori (ITT), Florence, Italy
| | - Luca Gasparoli
- Department of Experimental and Clinical Medicine, Section of Internal Medicine, University of Florence and Istituto Toscano Tumori (ITT), Florence, Italy
| | - Martina Chiu
- Unit of General Pathology Department of Biomedical, Biotechnological and Translational Sciences (SBiBiT) University of Parma
| | - Ovidio Bussolati
- Unit of General Pathology Department of Biomedical, Biotechnological and Translational Sciences (SBiBiT) University of Parma
| | - Andrea Becchetti
- Department of Biotechnology and Biosciences, University of Milano Bicocca, Milano, Italy
| | - Annarosa Arcangeli
- Department of Experimental and Clinical Medicine, Section of Internal Medicine, University of Florence and Istituto Toscano Tumori (ITT), Florence, Italy
| |
Collapse
|
11
|
Mainali D, Smith EA. Select cytoplasmic and membrane proteins increase the percentage of immobile integrins but do not affect the average diffusion coefficient of mobile integrins. Anal Bioanal Chem 2013; 405:8561-8. [DOI: 10.1007/s00216-013-7279-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2013] [Revised: 07/23/2013] [Accepted: 07/26/2013] [Indexed: 12/25/2022]
|
12
|
Denadai MV, Viana LS, Affonso RJ, Silva SR, Oliveira ID, Toledo SR, Matos D. Expression of integrin genes and proteins in progression and dissemination of colorectal adenocarcinoma. BMC Clin Pathol 2013; 13:16. [PMID: 23705994 PMCID: PMC3671190 DOI: 10.1186/1472-6890-13-16] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2012] [Accepted: 05/13/2013] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND This study aimed to evaluate the relationship between the expression levels of selected integrin genes and proteins and cell differentiation, TNM stage, histological type and other variables potentially associated with the progression and dissemination of colorectal carcinoma (CRC). METHODS A total of 114 patients (63 men and 51 women) were treated for CRC between 2006 and 2009, including 25 (21.9%) TNM I, 39 (34.2%) TNM II, 34 (29.8%) TNM III, and 16 (14.1%) TNM IV. Regarding grade, 91 (79.8%) were grade II, 14 (12.2%) were grade III and nine (7.8%) were grade I. Reverse-transcription polymerase chain reaction (RT-PCR) and tissue microarray (TMA) methods were used to examine the expression levels of the genes ITGAV, ITGA3, ITGA5, ITGB5, and ITGA6, and their proteins, respectively. RESULTS In relation to TNM staging, ITGB5 and ITGA3 were over-expressed in stages III versus I. These results were confirmed by TMA analysis. In terms of age, ITGA5 was under-expressed according to RT-PCR, but over-expressed by TMA in patients over 60 years, while ITGA5 gene and protein levels were increased in mucinous carcinomas. In addition ITGAV gene and protein levels were elevated in tumors with neural invasion, and ITGA6 gene and protein were over-expressed in cases with venous invasion. All these results were significant at P < 0.05. CONCLUSION The results of this study suggest that over-expression of some integrins is associated with TNM III stage, increased risk of vascular and neural invasion, and mucinous histology in patients with CRC.
Collapse
Affiliation(s)
- Marcos Va Denadai
- Interdisciplinary Surgery, Federal University of São Paulo, São Paulo, SP, Brazil ; Barretos Cancer Hospital, Fundação Pio XII, Barretos, SP, Brazil
| | - Luciano S Viana
- Interdisciplinary Surgery, Federal University of São Paulo, São Paulo, SP, Brazil ; Barretos Cancer Hospital, Fundação Pio XII, Barretos, SP, Brazil
| | - Renato J Affonso
- Interdisciplinary Surgery, Federal University of São Paulo, São Paulo, SP, Brazil ; Barretos Cancer Hospital, Fundação Pio XII, Barretos, SP, Brazil
| | - Sandra R Silva
- Interdisciplinary Surgery, Federal University of São Paulo, São Paulo, SP, Brazil ; Barretos Cancer Hospital, Fundação Pio XII, Barretos, SP, Brazil
| | - Indhira D Oliveira
- Genetics Laboratory (GRAACC), Federal University of São Paulo, São Paulo, SP, Brazil
| | - Silvia R Toledo
- Genetics Laboratory (GRAACC), Federal University of São Paulo, São Paulo, SP, Brazil
| | - Delcio Matos
- Interdisciplinary Surgery, Federal University of São Paulo, São Paulo, SP, Brazil
| |
Collapse
|
13
|
Arora N, Mainali D, Smith EA. Unraveling the role of membrane proteins Notch, Pvr, and EGFR in altering integrin diffusion and clustering. Anal Bioanal Chem 2012; 404:2339-48. [DOI: 10.1007/s00216-012-6362-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2012] [Revised: 08/07/2012] [Accepted: 08/14/2012] [Indexed: 02/07/2023]
|
14
|
|
15
|
Uematsu T, Konishi C, Hoshino D, Han X, Tomari T, Egawa N, Takada Y, Isobe T, Seiki M, Koshikawa N. Identification of proteins that associate with integrin α2 by proteomic analysis in human fibrosarcoma HT-1080 cells. J Cell Physiol 2012; 227:3072-9. [DOI: 10.1002/jcp.23054] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
16
|
Becchetti A. Ion channels and transporters in cancer. 1. Ion channels and cell proliferation in cancer. Am J Physiol Cell Physiol 2011; 301:C255-65. [DOI: 10.1152/ajpcell.00047.2011] [Citation(s) in RCA: 134] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Progress through the cell mitotic cycle requires precise timing of the intrinsic molecular steps and tight coordination with the environmental signals that maintain a cell into the proper physiological context. Because of their great functional flexibility, ion channels coordinate the upstream and downstream signals that converge on the cell cycle machinery. Both voltage- and ligand-gated channels have been implicated in the control of different cell cycle checkpoints in normal as well as neoplastic cells. Ion channels mediate the calcium signals that punctuate the mitotic process, the cell volume oscillations typical of cycling cells, and the exocytosis of autocrine or angiogenetic factors. Other functions of ion channels in proliferation are still matter of debate. These may or may not depend on ion transport, as the channel proteins can form macromolecular complexes with growth factor and cell adhesion receptors. Direct conformational coupling with the cytoplasmic regulatory proteins is also possible. Derangement or relaxed control of the above processes can promote neoplasia. Specific types of ion channels have turned out to participate in the different stages of the tumor progression, in which cell heterogeneity is increased by the selection of malignant cell clones expressing the ion channel types that better support unrestrained growth. However, a comprehensive mechanistic picture of the functional relations between ion channels and cell proliferation is yet not available, partly because of the considerable experimental challenges offered by studying these processes in living mammalian cells. No doubt, such studies will constitute one of the most fruitful research fields for the next generation of cell physiologists.
Collapse
Affiliation(s)
- Andrea Becchetti
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milano, Italy
| |
Collapse
|
17
|
Beauvais DM, Rapraeger AC. Syndecan-1 couples the insulin-like growth factor-1 receptor to inside-out integrin activation. J Cell Sci 2011; 123:3796-807. [PMID: 20971705 DOI: 10.1242/jcs.067645] [Citation(s) in RCA: 94] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Syndecan-1 (Sdc1) engages and activates the αvβ3 (and/or αvβ5) integrin when clustered in human carcinoma and endothelial cells. Although the engagement is extracellular, the activation mechanism is cytoplasmic. This talin-dependent, inside-out signaling pathway is activated downstream of the insulin-like growth factor-1 receptor (IGF1R), whose kinase activity is triggered by Sdc1 clustering. In vitro binding assays using purified receptors suggest that association of the Sdc1 ectodomain with the integrin provides a 'docking face' for IGF1R. IGF1R docking and activation of the associated integrin is blocked by synstatin (SSTN(92-119)), a peptide derived from the integrin engagement site in Sdc1. IGF1R colocalizes with αvβ3 integrin and Sdc1 in focal contacts, but fails to associate with or activate the integrin in cells either lacking Sdc1 or expressing Sdc1(Δ67-121), a mutant that is unable to form the Sdc1-integrin-IGF1R ternary complex. Integrin activation is also blocked by IGF1R inhibitors or by silencing IGF1R or talin expression with small-interfering RNAs (siRNAs). In both cases, expression of the constitutively active talin F23 head domain rescues integrin activation. We recently reported that SSTN(92-119) blocks angiogenesis and impairs tumor growth in mice, therefore this Sdc1-mediated integrin regulatory mechanism might be a crucial regulator of disease processes known to rely on these integrins, including tumor cell metastasis and tumor-induced angiogenesis.
Collapse
Affiliation(s)
- DeannaLee M Beauvais
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, 1111 Highland Avenue, Madison, WI 53705, USA
| | | |
Collapse
|
18
|
Yamashita R, Kotani N, Ishiura Y, Higashiyama S, Honke K. Spatiotemporally-regulated interaction between β1 integrin and ErbB4 that is involved in fibronectin-dependent cell migration. ACTA ACUST UNITED AC 2011; 149:347-55. [DOI: 10.1093/jb/mvq148] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
|
19
|
Costa P, Parsons M. New insights into the dynamics of cell adhesions. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2010; 283:57-91. [PMID: 20801418 DOI: 10.1016/s1937-6448(10)83002-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Adhesion to the extracellular matrix (ECM) and to adjacent cells is a fundamental requirement for survival, differentiation, and migration of numerous cell types during both embryonic development and adult homeostasis. Different types of adhesion structures have been classified within different cell types or tissue environments. Much is now known regarding the complexity of protein composition of these critical points of cell contact with the extracellular environment. It has become clear that adhesions are highly ordered, dynamic structures under tight spatial control at the subcellular level to enable localized responses to extracellular cues. However, it is only in the last decade that the relative dynamics of these adhesion proteins have been closely studied. Here, we provide an overview of the recent data arising from such studies of cell-matrix and cell-cell contact and an overview of the imaging strategies that have been developed and implemented to study the intricacies and hierarchy of protein turnover within adhesions.
Collapse
Affiliation(s)
- Patricia Costa
- Randall Division of Cell and Molecular Biophysics, King's College London, New Hunts House, Guys Campus, London, United Kingdom
| | | |
Collapse
|
20
|
Abstract
Major advances have been made in the understanding and treatment of pulmonary hypertension in the last few years. Without treatment (medication) for idiopathic pulmonary arterial hypertension, which is a rare and potentially fatal condition, the survival time is only about 3 years after diagnosis. However, if pulmonary hypertension is secondary to other causes such as congenital heart disease, it is possible to survive for 30 years or more without treatment. The condition can affect children at any age, from fetal life to adulthood. Patients with pulmonary hypertension can present to the respiratory pediatrician with unresponsive asthma, to the neurologist with faints, or to the general pediatrician with failure to thrive. Over the last few years there have been significant developments in the available therapy for managing this complicated disease. There is now a generally recognized ladder of long-term therapy for chronic pulmonary hypertension. Treatment can start with oxygen at home at night or even during the day. Next is the use of oral phosphodiesterase inhibitors, mostly type V, such as sildenafil, which enhance endogenous nitric oxide. More potent are the endothelin receptor antagonists and the most potent are the prostanoids, especially epoprostenol, which is given by constant intravenous infusion. In addition to interventional catheterization with atrial septostomy, these agents have improved the prognostic outlook. This article reviews the current knowledge about the etiology, investigation, and treatment of children with pulmonary hypertension in the clinical setting.
Collapse
Affiliation(s)
- Robert Tulloh
- Department of Congenital Heart Disease, Bristol Royal Hospital for Children and Bristol Royal Infirmary, Bristol, England.
| |
Collapse
|
21
|
The role of hERG1 K+ channels and a functional link between hERG1 K+ channels and SDF-1 in acute leukemic cell migration. Exp Cell Res 2009; 315:2256-64. [DOI: 10.1016/j.yexcr.2009.04.017] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2009] [Revised: 04/13/2009] [Accepted: 04/21/2009] [Indexed: 11/22/2022]
|
22
|
Bass R, Wagstaff L, Ravenhill L, Ellis V. Binding of extracellular maspin to beta1 integrins inhibits vascular smooth muscle cell migration. J Biol Chem 2009; 284:27712-20. [PMID: 19638634 DOI: 10.1074/jbc.m109.038919] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Maspin is a serpin that has multiple effects on cell behavior, including inhibition of migration. How maspin mediates these diverse effects remains unclear, as it is devoid of protease inhibitory activity. We have previously shown that maspin rapidly inhibits the migration of vascular smooth muscle cells (VSMC), suggesting the involvement of direct interactions with cell surface proteins. Here, using immunofluorescence microscopy, we demonstrate that maspin binds specifically to the surface of VSMC in the dedifferentiated, but not the differentiated, phenotype. Ligand blotting of VSMC lysates revealed the presence of several maspin-binding proteins, with a protein of 150 kDa differentially expressed between the two VSMC phenotypes. Western blotting suggested that this protein was the beta1 integrin subunit, and subsequently both alpha3beta1 and alpha5beta1, but not alphavbeta3, were shown to associate with maspin by coimmunoprecipitation. Specific binding of these integrins was also observed using maspin-affinity chromatography, using HT1080 cell lysates. Direct binding of maspin to alpha5beta1 was confirmed using a recombinant alpha5beta1-Fc fusion protein. Using conformation-dependent anti-beta1 antibodies, maspin binding to VSMC was found to lead to a decrease in the activation status of the integrin. The functional involvement of alpha5beta1 in mediating the effect of maspin was established by the inhibition of migration of CHO cells overexpressing human alpha5 integrin, but not those lacking alpha5 expression. Our observations suggest that maspin engages in specific interactions with a limited number of integrins on VSMC, leading to their inactivation, and that these interactions are responsible for the effects of maspin in the pericellular environment.
Collapse
Affiliation(s)
- Rosemary Bass
- School of Biological Sciences, Biomedical Research Centre, University of East Anglia, Norwich NR4 7TJ, United Kingdom
| | | | | | | |
Collapse
|
23
|
Beauvais DM, Ell BJ, McWhorter AR, Rapraeger AC. Syndecan-1 regulates alphavbeta3 and alphavbeta5 integrin activation during angiogenesis and is blocked by synstatin, a novel peptide inhibitor. ACTA ACUST UNITED AC 2009; 206:691-705. [PMID: 19255147 PMCID: PMC2699122 DOI: 10.1084/jem.20081278] [Citation(s) in RCA: 222] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Syndecan-1 (Sdc1) is a matrix receptor shown to associate via its extracellular domain with the αvβ3 and αvβ5 integrins, potentially regulating cell adhesion, spreading, and invasion of cells expressing these integrins. Using Sdc1 deletion mutants expressed in human mammary carcinoma cells, we identified the active site within the Sdc1 core protein and derived a peptide inhibitor called synstatin (SSTN) that disrupts Sdc1's interaction with these integrins. Because the αvβ3 and αvβ5 integrins are critical in angiogenesis, a process in which a role for Sdc1 has been uncertain, we used human vascular endothelial cells in vitro to show that the Sdc1 regulatory mechanism is also required for integrin activation on these cells. We found Sdc1 expressed in the vascular endothelium during microvessel outgrowth from aortic explants in vitro and in mouse mammary tumors in vivo. Moreover, we show that SSTN blocks angiogenesis in vitro or when delivered systemically in a mouse model of angiogenesis in vivo, and impairs mammary tumor growth in an orthotopic mouse tumor model. Thus, Sdc1 is a critical regulator of these two important integrins during angiogenesis and tumorigenesis, and is inhibited by the novel SSTN peptide.
Collapse
Affiliation(s)
- DeannaLee M Beauvais
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI 53706, USA
| | | | | | | |
Collapse
|
24
|
de Souza Andrade ES, Miguel MCDC, de Almeida Freitas R, Pereira Pinto L, Batista de Souza L. Immunoexpression of integrins in ameloblastoma, adenomatoid odontogenic tumor, and human tooth germs. Int J Surg Pathol 2008; 16:277-85. [PMID: 18573784 DOI: 10.1177/1066896907306084] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The expression of integrins alpha2beta1, alpha3beta1, and alpha5beta1 in 30 ameloblastomas (20 solid and 10 unicystic tumors), 12 adenomatoid odontogenic tumors (AOTs), and 5 human tooth germs in different stages of odontogenesis was analyzed. The distribution, location, pattern, and intensity of immunohistochemical expression were evaluated. Intensity was analyzed using scores (0 = absence, 1 = weak staining, and 2 = strong staining). No difference in the immunoexpression of the integrins was observed between solid and unicystic ameloblastomas. When these two ameloblastoma types were pooled into a single group, the following significant differences were found: immunoexpression of integrin alpha2beta1 was stronger in ameloblastomas than in AOTs and tooth germs, and the expression of integrin alpha5beta1 was stronger in ameloblastomas than in AOTs. The lack of detection of integrin alpha3beta1 in tooth germs and its detection in the odontogenic tumors studied suggest that this integrin might be used as a marker of neoplastic transformation in odontogenic tissues.
Collapse
|
25
|
Abstract
Formation of protrusions and protein segregation on the membrane is of a great importance for the functioning of the living cell. This is most evident in recent experiments that show the effects of the mechanical properties of the surrounding substrate on cell morphology. We propose a mechanism for the formation of membrane protrusions and protein phase separation, which may lay behind this effect. In our model, the fluid cell membrane has a mobile but constant population of proteins with a convex spontaneous curvature. Our basic assumption is that these membrane proteins represent small adhesion complexes, and also include proteins that activate actin polymerization. Such a continuum model couples the membrane and protein dynamics, including cell-substrate adhesion and protrusive actin force. Linear stability analysis shows that sufficiently strong adhesion energy and actin polymerization force can bring about phase separation of the membrane protein and the appearance of protrusions. Specifically, this occurs when the spontaneous curvature and aggregation potential alone (passive system) do not cause phase separation. Finite-size patterns may appear in the regime where the spontaneous curvature energy is a strong factor. Different instability characteristics are calculated for the various regimes, and are compared to various types of observed protrusions and phase separations, both in living cells and in artificial model systems. A number of testable predictions are proposed.
Collapse
Affiliation(s)
- Alex Veksler
- Department of Chemical Physics, The Weizmann Institute of Science, Rehovot, Israel.
| | | |
Collapse
|
26
|
Meyer S, Orsó E, Schmitz G, Landthaler M, Vogt T. Lubrol-RAFTs in Melanoma Cells: A Molecular Platform for Tumor-Promoting Ephrin-B2–Integrin-β1 Interaction. J Invest Dermatol 2007; 127:1615-21. [PMID: 17380111 DOI: 10.1038/sj.jid.5700778] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Ephrins control cell motility and matrix adhesion. These functions play a pivotal role in cancer progression, for example, in malignant melanomas. We have previously shown that the ephrin-B2-tumor-promoting action is partly mediated by integrin-beta1 interaction. However, the subcellular prerequisites for molecular interaction like molecular proximity and co-compartmentalization have not been elucidated yet. Specific cholesterol-rich microdomains, termed lipid rafts (RAFTs), are known to be essential for functional ephrin-B2 signalling and integrin-mediated effects. Therefore, we addressed the question whether RAFT co-compartmentalization of both molecules could provide the molecular platform for their tumor-promoting interaction. In this study, we show that overexpressed ephrin-B2 is not only compartmentalized to classical Triton X-100 RAFTs in B16 melanoma cells, but also to the recently defined Lubrol-RAFTs. Interestingly, in the melanoma cells investigated, integrin-beta1 is also preferentially detected in such Lubrol-RAFTs. Accordingly, the presence of ephrin-B2 and integrin-beta1 in RAFTs and their function in cell migration and matrix attachment are highly sensitive to RAFT disruption by cholesterol depletion. Confocal fluorescence microscopy analyses also support the concept of a close molecular proximity and functional interplay of ephrin-B2 and integrin-beta1 in the plasma membrane. We conclude that Lubrol-RAFTs probably represent the platform for tumor-promoting ephrin-B2-integrin-beta1 interaction, which could become an interesting target for future antitumoral therapies.
Collapse
Affiliation(s)
- Stefanie Meyer
- Department of Dermatology, University Hospital of Regensburg, Regensburg, Germany.
| | | | | | | | | |
Collapse
|
27
|
Pillozzi S, Brizzi MF, Bernabei PA, Bartolozzi B, Caporale R, Basile V, Boddi V, Pegoraro L, Becchetti A, Arcangeli A. VEGFR-1 (FLT-1), beta1 integrin, and hERG K+ channel for a macromolecular signaling complex in acute myeloid leukemia: role in cell migration and clinical outcome. Blood 2007; 110:1238-50. [PMID: 17420287 DOI: 10.1182/blood-2006-02-003772] [Citation(s) in RCA: 143] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Leukemia cell motility and transendothelial migration into extramedullary sites are regulated by angiogenic factors and are considered unfavorable prognostic factors in acute leukemias. We have studied cross talk among (1) the vascular endothelial growth factor receptor-1, FLT-1; (2) the human eag-related gene 1 (hERG1) K(+) channels; and (3) integrin receptors in acute myeloid leukemia (AML) cells. FLT-1, hERG1, and the beta(1) integrin were found to form a macromolecular signaling complex. The latter mostly recruited the hERG1B isoform of hERG1 channels, and its assembly was necessary for FLT-1 signaling activation and AML cell migration. Both effects were inhibited when hERG1 channels were specifically blocked. A FLT-1/hERG1/beta(1) complex was also observed in primary AML blasts, obtained from a population of human patients. The co-expression of FLT-1 and hERG1 conferred a pro-migratory phenotype to AML blasts. Such a phenotype was also observed in vivo. The hERG1-positive blasts were more efficient in invading the peripheral circulation and the extramedullary sites after engraftment into immunodeficient mice. Moreover, hERG1 expression in leukemia patients correlated with a higher probability of relapse and shorter survival periods. We conclude that in AML, hERG1 channels mediate the FLT-1-dependent cell migration and invasion, and hence confer a greater malignancy.
Collapse
Affiliation(s)
- Serena Pillozzi
- Department of Experimental Pathology and Oncology, University of Firenze, Firenze, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Seker A, Yildirim O, Kurtkaya O, Sav A, Günel M, Pamir MN, Kiliç T. Expression of integrins in cerebral arteriovenous and cavernous malformations. Neurosurgery 2006; 58:159-68; discussion 159-68. [PMID: 16385340 DOI: 10.1227/01.neu.0000192174.55131.09] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE To assess and compare levels and patterns of expression for integrins alphavbeta1, alphavbeta3, and alphavbeta5 in arteriovenous malformations (AVMs) and cavernous malformations (CCMs) of the brain. MATERIALS AND METHODS Specimens from 10 AVM and 10 CCM lesions were selected from 112 patients with AVMs and 97 patients with CCMs who were treated microsurgically in the Department of Neurosurgery, Marmara University, Istanbul, Turkey. Sections were immunohistochemically stained with antibodies for integrins alphavbeta1, alphavbeta3, and alphavbeta5. Separate histological layers of the vascular wall were evaluated, and levels of expression were graded using a four-tier system. RESULTS Integrin alphavbeta1 was more strongly expressed in AVMs than in CCMs. This difference was most pronounced in the endothelium and subendothelium/media. Integrin alphavbeta3 was more strongly expressed in CCM endothelium than in AVM endothelium (average grades, 0.9 and 0.4, respectively). All 10 of the CCM lesions expressed integrin alphavbeta5 in the endothelium, whereas only five of the AVMs showed minimal expression of this molecule in the endothelium. CONCLUSION Current scientific understanding of the roles integrins play in angiogenesis is far from complete. The levels and patterns of expression for these molecules in the histological layers of the vascular walls of AVMs and CCMs provide some clues about the complex biological activities of integrins in these lesions. If one accepts the premise that immunohistochemistry has its inherent methodological problems, integrins alphavbeta1, alphavbeta3, and alphavbeta5 are expressed in AVMs and CCMs in different ways that may be linked to stages of angiogenic maturation. Integrin alphavbeta1 is expressed more strongly in endothelium and subendothelium/media of AVMs than in the corresponding layers of CCMs. Integrins alphavbeta3 and alphavbeta5 are expressed more strongly in CCM endothelium than in AVM endothelium. In addition, integrin alphavbeta5 staining was stronger in CCM subendothelium than AVM subendothelium/media.
Collapse
Affiliation(s)
- Aşkin Seker
- Department of Neurosurgery, Institute of Neurological Sciences, Marmara University, Istanbul, Turkey
| | | | | | | | | | | | | |
Collapse
|
29
|
Staunton DE, Lupher ML, Liddington R, Gallatin WM. Targeting integrin structure and function in disease. Adv Immunol 2006; 91:111-57. [PMID: 16938539 DOI: 10.1016/s0065-2776(06)91003-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Initially linked to the pathogenesis of inflammatory and hematologic diseases, integrins have become validated drug targets with the approval of five drugs. Moreover, there are several promising drug candidates in preclinical and clinical stages of development for multiple clinical indications. Integrins are attractive drug targets as their antagonism can block several steps in disease progression or maintenance. Integrin inhibitors can block the proliferation, migration, or tissue localization of inflammatory, angiogenic, and tumor cells, as well as signaling and gene expression contributing to disease. There has been a rapid increase in the elucidation of integrin structure, their allosteric mechanisms of bidirectional signaling, and the structure of complexes with drugs. This information brings greater focus to how integrins support various cellular functions and how they have been and may be targeted to develop novel drugs. Here we review conformational switches, including an internal ligand, which allosterically regulate the transition from low- to high-affinity ligand binding. We address some of the successes, disappointments, and challenges in targeting competitive or allosteric sites to develop therapeutics. We also discuss new opportunities, including a structure-based approach to discover novel drugs to treat inflammatory and other diseases. This approach targets structural relatives of the von Willebrand factor A-domain present in integrins and many functionally diverse proteins.
Collapse
|
30
|
Phillips JA, Bonassar LJ. Matrix metalloproteinase activity synergizes with alpha2beta1 integrins to enhance collagen remodeling. Exp Cell Res 2005; 310:79-87. [PMID: 16098964 DOI: 10.1016/j.yexcr.2005.03.039] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2004] [Revised: 03/22/2005] [Accepted: 03/23/2005] [Indexed: 11/30/2022]
Abstract
Cell-matrix interactions transmit a wealth of information about the extracellular environment. In return, a variety of responses from the cell are initiated by changes in the matrix. One such response involves the positive regulation of matrix metalloproteinases (MMPs) by alpha2beta1 integrin attaching to a specific extracellular matrix component, collagen. This study explores the relationship between mechanical and biochemical functions of alpha2beta1 integrins as it pertains to regulating matrix remodeling. To understand this relationship, the individual influences of MMP activity and alpha2beta1 integrin function on collagen gel contraction were studied. We have observed little evidence of mutual participation in matrix remodeling by the alpha2beta1 integrin and MMP activity in cell models where alpha2 is minimally expressed. In cells expressing high levels of alpha2, we see an increase in gel contraction that is enhanced by MMP activity. Measuring tension as it builds within the gel reveals that alpha2beta1 integrin presence correlates with force output but is insensitive to MMP activity. These data strongly suggest that alpha2beta1 regulates collagen gel remodeling through multiple simultaneous mechanisms including force generation and modulation of MMP activity.
Collapse
Affiliation(s)
- Jonathan A Phillips
- University of Massachusetts, Graduate School of Biomedical Sciences, Worcester, MA 02125, USA
| | | |
Collapse
|
31
|
Affiliation(s)
- C Brakebusch
- Max Planck Institute of Biochemistry, Junior Group Regulation of Cytoskeletal Organization, Martinsried, Germany
| | | |
Collapse
|
32
|
Cherubini A, Hofmann G, Pillozzi S, Guasti L, Crociani O, Cilia E, Di Stefano P, Degani S, Balzi M, Olivotto M, Wanke E, Becchetti A, Defilippi P, Wymore R, Arcangeli A. Human ether-a-go-go-related gene 1 channels are physically linked to beta1 integrins and modulate adhesion-dependent signaling. Mol Biol Cell 2005; 16:2972-83. [PMID: 15800067 PMCID: PMC1142440 DOI: 10.1091/mbc.e04-10-0940] [Citation(s) in RCA: 124] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Adhesive receptors of the integrin family are primarily involved in cell-extracellular matrix adhesion. Additionally, integrins trigger multiple signaling pathways that are involved in cell migration, proliferation, survival, and differentiation. We previously demonstrated that the activation of integrins containing the beta(1) subunit leads to a selective increase in potassium currents carried by the human ether-a-go-go-related gene (hERG) channels in neuroblastoma and leukemia cells; this current activation modulates adhesion-dependent differentiation in these cells. We hypothesized that the cross-talk between integrins and hERG channels could be traced back to the assembly of a macromolecular signaling complex comprising the two proteins. We tested this hypothesis in both SH-SY5Y neuroblastoma cells and in human embryonic kidney 293 cells stably transfected with hERG1 and, therefore, expressing only the full-length hERG1 protein on the plasma membrane. The beta(1) integrin and hERG1 coprecipitate in these cells and colocalize in both intracellular and surface membrane compartments. The two proteins also coprecipitate with caveolin-1, suggesting the localization of the complex in lipid rafts/caveolae. hERG1-transfected cells undergo an activation of hERG currents after beta(1) integrin-mediated adhesion to fibronectin; concomitant with this activation, the focal adhesion kinase associates with the hERG1 protein and becomes tyrosine phosphorylated. Using hERG1-specific inhibitors, we show that the tyrosine phosphorylation of focal adhesion kinase is strictly dependent on hERG channel activity. Similarly, the activity of the small GTPase Rac1 turned out to be dependent on hERG currents. On the whole, these data indicate that the hERG1 protein associates with beta(1) integrins and modulates adhesion receptor signaling.
Collapse
Affiliation(s)
- Alessia Cherubini
- Department of Experimental Pathology and Oncology, University of Firenze, 50134 Firenze, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Li SS, Forslöw A, Sundqvist KG. Autocrine Regulation of T Cell Motility by Calreticulin-Thrombospondin-1 Interaction. THE JOURNAL OF IMMUNOLOGY 2005; 174:654-61. [PMID: 15634883 DOI: 10.4049/jimmunol.174.2.654] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The mechanisms regulating T lymphocyte migration within the extracellular matrix are not understood. We show in this study that the thrombospondin-1 binding site of calreticulin, spanning aa 19-32, is a major triggering factor for T cell motility and migration within a three-dimensional collagen type 1 matrix, and that exogenous motogenic factors such as chemokines can stimulate migration via a calreticulin-thrombospondin-1 pathway. Endogenous calreticulin binding to the N-terminal domain of endogenous thrombospondin-1 elicited a motogenic signal to the T cells through the C-terminal domain of thrombospondin-1 and its cell surface receptor integrin-associated protein (CD47). Our data further revealed that thrombospondin-1 was expressed on the cell surface with a high turnover, and that PI3K and the Janus family of tyrosine kinases were required for T cell motility mediated through calreticulin, thrombospondin-1, and CD47. These results unveil an autocrine mechanism of calreticulin-thrombospondin-1-CD47 interaction for the control of T cell motility and migration within three-dimensional extracellular matrix substrata.
Collapse
Affiliation(s)
- Shu Shun Li
- Division of Clinical Immunology, Department of Clinical Microbiology, Umeå University, Umeå, Sweden
| | | | | |
Collapse
|
34
|
Bialkowska K, Saido TC, Fox JEB. SH3 domain of spectrin participates in the activation of Rac in specialized calpain-induced integrin signaling complexes. J Cell Sci 2005; 118:381-95. [PMID: 15632109 DOI: 10.1242/jcs.01625] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
In this study, we used cultured cells spreading on beta3 integrin substrates to examine the possibility that spectrin is involved in signal transduction. Spectrin clustered with specialized calpain-induced beta3 integrin signaling complexes that mediate the initial attachment of cells and initiate Rac activation and lamellipodia extension. It was absent from focal complexes and focal adhesions, the integrin complexes that mediate adhesion in lamellipodia and fully spread cells. Spectrin contains a Src homology (SH3) domain of unknown function. Cells overexpressing this domain adhered and calpain-induced integrin signaling complexes formed. However, Rac activation, lamellipodia extension and cell spreading were inhibited. Spreading was restored by overexpression of constitutively active Rac. These studies point to a previously unrecognized role for spectrin and its SH3 domain in initiating Rac activation in the specialized integrin clusters that initiate cell adhesion and spreading. Thus, spectrin may have a pivotal role in initiating integrin-induced physiological and pathological events such as development, proliferation, cell survival, wound healing, metastasis and atherosclerosis.
Collapse
Affiliation(s)
- Katarzyna Bialkowska
- Joseph J. Jacobs Center for Thrombosis and Vascular Biology, Department of Molecular Cardiology, The Lerner Research Institute, The Cleveland Clinic Foundation, Cleveland, OH 44195, USA
| | | | | |
Collapse
|
35
|
Arcangeli A, Becchetti A, Cherubini A, Crociani O, Defilippi P, Guasti L, Hofmann G, Pillozzi S, Olivotto M, Wanke E. Physical and functional interaction between integrins and hERG potassium channels. Biochem Soc Trans 2004; 32:826-7. [PMID: 15494025 DOI: 10.1042/bst0320826] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Integrins are adhesion receptors capable of transmitting intracellular signals that regulate many different cellular functions. Among integrin-mediated signals, the activation of ion channels can be included. We demonstrated that a long-lasting activation of hERG (human ether-a-go-go-related gene) potassium channels occurs in both human neuroblastoma and leukaemia cells after the activation of the β1 integrin subunit. This activation is apparently a determining factor inducing neurite extension and osteoclastic differentiation in both the cell types. More recently, we provided evidences that β1 integrins and hERG channels co-precipitate in both the cell types. Preliminary results suggest that a macromolecular signalling complex indeed occurs between integrins and the hERG1 protein and that hERG channel activity can modulate integrin downstream signalling.
Collapse
Affiliation(s)
- A Arcangeli
- Department of Experimental Pathology and Oncology, University of Firenze, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Funaro A, Ortolan E, Ferranti B, Gargiulo L, Notaro R, Luzzatto L, Malavasi F. CD157 is an important mediator of neutrophil adhesion and migration. Blood 2004; 104:4269-78. [PMID: 15328157 DOI: 10.1182/blood-2004-06-2129] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
CD157, a glycosylphosphatidylinositol (GPI)-anchored protein encoded by a member of the CD38 NADase/ADP-ribosyl cyclase gene family, is expressed on the surface of most human circulating neutrophils. This work demonstrates that CD157 is a receptor that induces reorganization of the cytoskeleton and significant changes in cell shape, and that signals mediated by CD157 act through modulation of cytosolic Ca(2+) concentration. These signals are independent of the products of CD157's enzymatic activities (ie, cyclic adenosine diphosphate [ADP]-ribose and ADP-ribose). Indeed, the enzymatic activities of CD157 in circulating neutrophils as well as in dimethyl sulfoxide (DMSO)-differentiated (CD157(+)/CD38(-)) HL-60 cells, are hardly detectable. This work also shows that the receptorial activity relies on cross-talk between CD157 and beta(2) integrin. CD157 localizes in GM1-enriched lipid rafts and, upon activation, it migrates to the uropod, a structure specialized in motility and adhesive functions. Indeed, CD157 is involved in adhesion to extracellular matrix proteins and in chemotaxis induced in vitro by formyl-methionyl-leucyl-phenylalanine (fMLP). These findings were consistent with the results obtained in neutrophils from patients with paroxysmal nocturnal hemoglobinuria (PNH), in which CD157 is deficient. These neutrophils showed constant defects in adhesion and migration. Our data attribute specific and crucial roles to CD157 in the regulation of innate immunity during inflammation.
Collapse
Affiliation(s)
- Ada Funaro
- Laboratory of Immunogenetics, Department of Genetics, Biology and Biochemistry, University of Torino, Via Santena 19, 10126 Torino, Italy.
| | | | | | | | | | | | | |
Collapse
|
37
|
Numakawa T, Ishimoto T, Suzuki S, Numakawa Y, Adachi N, Matsumoto T, Yokomaku D, Koshimizu H, Fujimori KE, Hashimoto R, Taguchi T, Kunugi H. Neuronal roles of the integrin-associated protein (IAP/CD47) in developing cortical neurons. J Biol Chem 2004; 279:43245-53. [PMID: 15297459 DOI: 10.1074/jbc.m406733200] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Little is known about the role of the integrin-associated protein (IAP, or CD47) in neuronal development and its function in the central nervous system. We investigated neuronal responses in IAP-overexpressing cortical neurons using a virus-gene transfer system. We found that dendritic outgrowth was significantly enhanced in IAP (form 4)-transfected neurons. Furthermore, synaptic proteins including synaptotagmin, syntaxin, synapsin I, and SNAP25 (25-kDa synaptosomal associated protein) were up-regulated. In accordance with this finding, the release of the excitatory transmitter glutamate and the frequencies of Ca2+ oscillations (glutamate-mediated synaptic transmission) were increased. Interestingly, the overexpression of IAP activated mitogen-activated protein kinase (MAPK), and this activation was required for the IAP-dependent biological effects. After down-regulation of the endogenous IAP by small interfering RNA, MAPK activity, synaptic protein levels, and glutamate release decreased. These observations suggest that the IAP plays important roles in dendritic outgrowth and synaptic transmission in developing cortical neurons through the activation of MAPK.
Collapse
Affiliation(s)
- Tadahiro Numakawa
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry of Japan, 4-1-1 Ogawahigashi, Kodaira, Tokyo 187-8502, Japan.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Bellis SL. Variant glycosylation: an underappreciated regulatory mechanism for β1 integrins. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2004; 1663:52-60. [PMID: 15157607 DOI: 10.1016/j.bbamem.2004.03.012] [Citation(s) in RCA: 125] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2004] [Revised: 03/24/2004] [Accepted: 03/31/2004] [Indexed: 01/07/2023]
Abstract
Although it has been known for many years that beta1 integrins undergo differential glycosylation in accordance with changes in cell phenotype, the potential role of N-glycosylation as a modulator of integrin function has received little attention. One reason for the relatively limited interest in this topic likely relates to the fact that much of the prior research was correlative in nature. However, new results now bolster the hypothesis that there is a causal relationship between variant glycosylation and altered integrin activity. In this review, the evidence for variant glycosylation as a regulatory mechanism for beta1 integrins are summarized, with particular emphasis on: (1). outlining the instances in which cell phenotypic variation is associated with differential beta1 glycosylation, (2). describing the specific alterations in glycan structure that accompany phenotypic changes and (3). presenting potential mechanisms by which variant glycosylation might regulate integrin function.
Collapse
Affiliation(s)
- Susan L Bellis
- Department of Physiology and Biophysics, University of Alabama at Birmingham, MCLM 982A, 1918 University Boulevard, Birmingham, AL 35294, USA.
| |
Collapse
|
39
|
Hintermann E, Quaranta V. Epithelial cell motility on laminin-5: regulation by matrix assembly, proteolysis, integrins and erbB receptors. Matrix Biol 2004; 23:75-85. [PMID: 15246107 DOI: 10.1016/j.matbio.2004.03.001] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2004] [Accepted: 03/03/2004] [Indexed: 01/10/2023]
Abstract
Cell migration plays a central role in a wide variety of biological events, including embryogenesis, inflammatory immune response, wound healing, or cancer invasion. Tight regulation of cell motility is a prerequisite for normal development and maintenance of an organism, and to avoid metastatic spread of tumor cells. An important determinant of migratory efficiency is the substrate over which a cell migrates. Laminin-5 (Ln-5) is an extracellular matrix component prominent in basement membranes and as such it is a substrate in direct contact with epithelial cells. Interestingly, Ln-5 has been shown to both stimulate and downregulate epithelial cell migration. In this article, we plan to give an overview on the different mechanisms cells employ to regulate their migratory behavior on Ln-5. We will discuss how proteolytic processing of Ln-5 acts as posttranslational modification that plays a major role in the regulation of cell migration. The different proteolytic Ln-5 species may bind to distinct cell surface receptors called integrins, which translate substrate binding into a specific cellular response that triggers cell motility. Furthermore, interaction between Ln-5-binding integrins and other transmembrane and cytoplasmic proteins increases complexity and may allow fine-tuning of cell migration in response to the cellular environment.
Collapse
Affiliation(s)
- Edith Hintermann
- Department of Cell Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | | |
Collapse
|
40
|
McDonald JF, Zheleznyak A, Frazier WA. Cholesterol-independent Interactions with CD47 Enhance αvβ3 Avidity. J Biol Chem 2004; 279:17301-11. [PMID: 14966135 DOI: 10.1074/jbc.m312782200] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Expression in OV10 cells of either wild-type CD47 or its extracellular IgV domain linked to a glycosylphosphatidylinositol anchor-(IgV-GPI) enhanced ligand-induced alpha(v)beta(3) activation as detected by the binding of LIBS1 and LIBS6 mAbs. The amplitude of LIBS binding was greater with both CD47 and IgV-GPI expression, indicating an increase in the population of "activable" integrin molecules. Expression of either CD47 species also increased alpha(v)beta(3)-mediated adhesion to vitronectin, and to surfaces coated with the anti-beta(3) antibody AP3, because of enhanced clustering of alpha(v)beta(3) as confirmed by chemical cross-linking. Cholesterol depletion with methyl-beta-cyclodextrin did not prevent the increase in anti-LIBS binding, but reduced cell adhesion to vitronectin and AP3. However, cells expressing CD47 were partially insulated against this disruption, and IgV-GPI was even more effective. Both CD47 and IgV-GPI were found in cholesterol-rich rafts prepared in the absence of detergent, but only CD47 could recruit alpha(v)beta(3) and its associated signaling molecules to these domains. Thus CD47-alpha(v)beta(3) complexes in cholesterol-rich raft domains appear to engage in G(i)-dependent signaling whereas CD47-alpha(v)beta(3) interactions that lead to integrin clustering are also detergent resistant, but are insensitive to cholesterol depletion and do not require the transmembrane region of CD47.
Collapse
Affiliation(s)
- John F McDonald
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | | | | |
Collapse
|
41
|
N'Diaye EN, Brown EJ. The ubiquitin-related protein PLIC-1 regulates heterotrimeric G protein function through association with Gbetagamma. ACTA ACUST UNITED AC 2004; 163:1157-65. [PMID: 14662753 PMCID: PMC2173627 DOI: 10.1083/jcb.200307155] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
PLIC-1, a newly described ubiquitin-related protein, inhibited both Jurkat migration toward SDF-1alpha and A431 wound healing, but the closely related PLIC-2 did not. PLIC-1 prevented the SDF-1alpha-induced activation of phospholipase C, decreased ligand-induced internalization of SDF-1alpha receptor CXCR4 and inhibited chemotaxis signaled by a transfected Gi-coupled receptor. However, PLIC-1 had no effect on Gs-mediated adenylyl cyclase activation, and inhibited only the Gbetagamma-dependent component of Gq-initiated increase in [Ca2+]i, which is consistent with selective inhibition of Gbetagamma function. PLIC-1 colocalized with G proteins in lamellae and pseudopods, and precipitated Gbetagamma in pull downs. Interaction with Gbetagamma did not require PLIC-1's ubiquitin-like or ubiquitin-associated domains, and proteasome inhibition had no effect on SDF-1alpha activation of phospholipase C, indicating that PLIC-1's inhibition of Gbetagamma did not result from effects on proteasome function. Thus, PLIC-1 inhibits Gi signaling by direct association with Gbetagamma; because it also interacts with CD47, a modulator of integrin function, it likely has a role integrating adhesion and signaling components of cell migration.
Collapse
Affiliation(s)
- Elsa-Noah N'Diaye
- Program in Host-Pathogen Interactions, University of California, San Francisco, Campus Box 2140, 600 16th St., San Francisco, CA 94143-2140, USA
| | | |
Collapse
|
42
|
Abstract
Integrins play crucial roles in cell adhesion, migration, and signaling by providing transmembrane links between the extracellular matrix and the cytoskeleton. Integrins cluster in macromolecular complexes to generate cell-matrix adhesions such as focal adhesions. In this mini-review, we compare certain integrin-based biological responses and signaling during cell interactions with standard 2D cell culture versus 3D matrices. Besides responding to the composition of the matrix, cells sense and react to physical properties that include three-dimensionality and rigidity. In routine cell culture, fibroblasts and mesenchymal cells appear to use focal adhesions as anchors. They then use intracellular actomyosin contractility and dynamic, directional integrin movements to stretch cell-surface fibronectin and to generate characteristic long fibrils of fibronectin in "fibrillar adhesions". Some cells in culture proceed to produce dense, three-dimensional matrices similar to in vivo matrix, as opposed to the flat, rigid, two-dimensional surfaces habitually used for cell culture. Cells within such more natural 3D matrices form a distinctive class of adhesion termed "3D-matrix adhesions". These 3D adhesions show distinctive morphology and molecular composition. Their formation is heavily dependent on interactions between integrin alpha 5 beta 1 and fibronectin. Cells adhere much more rapidly to 3D matrices. They also show more rapid morphological changes, migration, and proliferation compared to most 2D matrices or 3D collagen gels. Particularly notable are low levels of tyrosine phosphorylation of focal adhesion kinase and moderate increases in activated mitogen-activated protein kinase. These findings underscore the importance of the dimensionality and dynamics of matrix substrates in cellular responses to the extracellular matrix.
Collapse
Affiliation(s)
- K M Yamada
- Craniofacial Developmental Biology and Regeneration Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892-4370, USA.
| | | | | |
Collapse
|
43
|
Fujimoto TT, Katsutani S, Shimomura T, Fujimura K. Thrombospondin-bound integrin-associated protein (CD47) physically and functionally modifies integrin alphaIIbbeta3 by its extracellular domain. J Biol Chem 2003; 278:26655-65. [PMID: 12736272 DOI: 10.1074/jbc.m302194200] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Integrin-associated protein (IAP/CD47) is a receptor for the C-terminal cell binding domain of thrombospondin (TS). A peptide from the C-terminal cell binding domain, KRFYVVMWKK (4N1K) binds to IAP and stimulates the integrin-dependent cell functions, including platelet aggregation. We investigated the mechanism by which TS-bound IAP modulates the affinity of platelet integrin, alphaIIbbeta3. Platelet aggregation induced by 4N1K was not completely inhibited by energy depletion with sodium azide and 2-deoxy-d-glucose, although ADP or collagen-induced platelet response was completely inhibited. The binding of ligand-mimetic antibody PAC1 to alphaIIbbeta3 was also induced in the energy-depleted platelets. In the transfected Namalwa cells, 4N1K induced activation of the alphaIIbbeta3 with mutated beta3 (Ser-752 to Pro), which is a non-responsive form to inside-out signaling, as well as wild type alphaIIbbeta3. The truncated form of IAP with only the extracellular immunoglobulin-like (Ig) domain was sufficient for the activation of alphaIIbbeta3 in Chinese hamster ovary cells, although the IAP-mediated intracellular signaling was abolished, which was monitored by the absence of down-regulation of mitogen-activated protein kinase phosphorylation. Furthermore, the soluble recombinant Ig domain of IAP induced PAC1 binding to alphaIIbbeta3 on Chinese hamster ovary cells when added with 4N1K. Physical association between the soluble recombinant Ig domain of IAP and purified alphaIIbbeta3 was detected in the presence of 4N1K. These data indicate that the extracellular Ig domain of IAP, when bound to TS, interacts with alphaIIbbeta3 and can change alphaIIbbeta3 in a high affinity state without the requirement of intracellular signaling. This extracellular event would be a novel mechanism of affinity modulation of integrin.
Collapse
Affiliation(s)
- Tetsuro-Takahiro Fujimoto
- Department of Hematology and Oncology, Division of Clinical Pharmacotherapeutics, Graduate School of Biomedical Sciences, Hiroshima University, Hiroshima 734-8551, Japan.
| | | | | | | |
Collapse
|
44
|
Affiliation(s)
- Alexander Dityatev
- Zentrum für Molekulare Neurobiologie, University of Hamburg, Martinistr. 52, 20246 Hamburg, Germany.
| | | |
Collapse
|