1
|
Morishita H, Kawai K, Egami Y, Honda K, Araki N. Live-cell imaging and CLEM reveal the existence of ACTN4-dependent ruffle-edge lamellipodia acting as a novel mode of cell migration. Exp Cell Res 2024; 442:114232. [PMID: 39222868 DOI: 10.1016/j.yexcr.2024.114232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 08/26/2024] [Accepted: 08/29/2024] [Indexed: 09/04/2024]
Abstract
α-Actinin-4 (ACTN4) expression levels are correlated with the invasive and metastatic potential of cancer cells; however, the underlying mechanism remains unclear. Here, we identified ACTN4-localized ruffle-edge lamellipodia using live-cell imaging and correlative light and electron microscopy (CLEM). BSC-1 cells expressing EGFP-ACTN4 showed that ACTN4 was most abundant in the leading edges of lamellipodia, although it was also present in stress fibers and focal adhesions. ACTN4 localization in lamellipodia was markedly diminished by phosphoinositide 3-kinase inhibition, whereas its localization in stress fibers and focal adhesions remained. Furthermore, overexpression of ACTN4, but not ACTN1, promoted lamellipodial formation. Live-cell analysis demonstrated that ACTN4-enriched lamellipodia are highly dynamic and associated with cell migration. CLEM revealed that ACTN4-enriched lamellipodia exhibit a characteristic morphology of multilayered ruffle-edges that differs from canonical flat lamellipodia. Similar ruffle-edge lamellipodia were observed in A549 and MDA-MB-231 invasive cancer cells. ACTN4 knockdown suppressed the formation of ruffle-edge lamellipodia and cell migration during wound healing in A549 monolayer cultures. Additionally, membrane-type 1 matrix metalloproteinase was observed in the membrane ruffles, suggesting that ruffle-edge lamellipodia have the ability to degrade the extracellular matrix and may contribute to active cell migration/invasion in certain cancer cell types.
Collapse
Affiliation(s)
- Haruka Morishita
- Department of Histology and Cell Biology, School of Medicine, Kagawa University, Kagawa, 761-0793, Miki, Japan
| | - Katsuhisa Kawai
- Department of Histology and Cell Biology, School of Medicine, Kagawa University, Kagawa, 761-0793, Miki, Japan
| | - Youhei Egami
- Department of Histology and Cell Biology, School of Medicine, Kagawa University, Kagawa, 761-0793, Miki, Japan
| | - Kazufumi Honda
- Department of Bioregulation, Graduate of Medicine, Nippon Medical School, Sendagi, Bunkyo-ku, 113-8602, Tokyo, Japan
| | - Nobukazu Araki
- Department of Histology and Cell Biology, School of Medicine, Kagawa University, Kagawa, 761-0793, Miki, Japan.
| |
Collapse
|
2
|
Katsuta H, Sokabe M, Hirata H. From stress fiber to focal adhesion: a role of actin crosslinkers in force transmission. Front Cell Dev Biol 2024; 12:1444827. [PMID: 39193363 PMCID: PMC11347286 DOI: 10.3389/fcell.2024.1444827] [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: 06/06/2024] [Accepted: 08/01/2024] [Indexed: 08/29/2024] Open
Abstract
The contractile apparatus, stress fiber (SF), is connected to the cell adhesion machinery, focal adhesion (FA), at the termini of SF. The SF-FA complex is essential for various mechanical activities of cells, including cell adhesion to the extracellular matrix (ECM), ECM rigidity sensing, and cell migration. This mini-review highlights the importance of SF mechanics in these cellular activities. Actin-crosslinking proteins solidify SFs by attenuating myosin-driven flows of actin and myosin filaments within the SF. In the solidified SFs, viscous slippage between actin filaments in SFs and between the filaments and the surrounding cytosol is reduced, leading to efficient transmission of myosin-generated contractile force along the SFs. Hence, SF solidification via actin crosslinking ensures exertion of a large force to FAs, enabling FA maturation, ECM rigidity sensing and cell migration. We further discuss intracellular mechanisms for tuning crosslinker-modulated SF mechanics and the potential relationship between the aberrance of SF mechanics and pathology including cancer.
Collapse
Affiliation(s)
- Hiroki Katsuta
- Department of Cardiovascular Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Masahiro Sokabe
- Human Information Systems Laboratories, Kanazawa Institute of Technology, Hakusan, Japan
| | - Hiroaki Hirata
- Department of Applied Bioscience, Kanazawa Institute of Technology, Hakusan, Japan
| |
Collapse
|
3
|
Gharaba S, Paz O, Feld L, Abashidze A, Weinrab M, Muchtar N, Baransi A, Shalem A, Sprecher U, Wolf L, Wolfenson H, Weil M. Perturbed actin cap as a new personalized biomarker in primary fibroblasts of Huntington's disease patients. Front Cell Dev Biol 2023; 11:1013721. [PMID: 36743412 PMCID: PMC9889876 DOI: 10.3389/fcell.2023.1013721] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Accepted: 01/03/2023] [Indexed: 01/19/2023] Open
Abstract
Primary fibroblasts from patient's skin biopsies are directly isolated without any alteration in the genome, retaining in culture conditions their endogenous cellular characteristics and biochemical properties. The aim of this study was to identify a distinctive cell phenotype for potential drug evaluation in fibroblasts from Huntington's Disease (HD) patients, using image-based high content analysis. We show that HD fibroblasts have a distinctive nuclear morphology associated with a nuclear actin cap deficiency. This in turn affects cell motility in a similar manner to fibroblasts from Hutchinson-Gilford progeria syndrome (HGPS) patients used as known actin cap deficient cells. Moreover, treatment of the HD cells with either Latrunculin B, used to disrupt actin cap formation, or the antioxidant agent Mitoquinone, used to improve mitochondrial activity, show expected opposite effects on actin cap associated morphological features and cell motility. Deep data analysis allows strong cluster classification within HD cells according to patients' disease severity score which is distinct from HGPS and matching controls supporting that actin cap is a biomarker in HD patients' cells correlated with HD severity status that could be modulated by pharmacological agents as tool for personalized drug evaluation.
Collapse
Affiliation(s)
- Saja Gharaba
- Laboratory for Personalized Medicine and Neurodegenerative Diseases, The Shmunis School of Biomedicine and Cancer Research, The George S. Wise Faculty for Life Sciences, Sagol School of Neurosciences, Tel Aviv University, Tel Aviv, Israel
| | - Omri Paz
- Laboratory for Personalized Medicine and Neurodegenerative Diseases, The Shmunis School of Biomedicine and Cancer Research, The George S. Wise Faculty for Life Sciences, Sagol School of Neurosciences, Tel Aviv University, Tel Aviv, Israel
| | - Lea Feld
- Department of Genetics and Developmental Biology, The Rappaport Faculty of Medicine and Research Institute, Technion—Israel Institute of Technology, Haifa, Israel
| | - Anastasia Abashidze
- The Blavatnik Center for Drug Discovery, Tel Aviv University, Tel Aviv, Israel
| | - Maydan Weinrab
- Laboratory for Personalized Medicine and Neurodegenerative Diseases, The Shmunis School of Biomedicine and Cancer Research, The George S. Wise Faculty for Life Sciences, Sagol School of Neurosciences, Tel Aviv University, Tel Aviv, Israel
| | - Noam Muchtar
- Laboratory for Personalized Medicine and Neurodegenerative Diseases, The Shmunis School of Biomedicine and Cancer Research, The George S. Wise Faculty for Life Sciences, Sagol School of Neurosciences, Tel Aviv University, Tel Aviv, Israel
| | - Adam Baransi
- Laboratory for Personalized Medicine and Neurodegenerative Diseases, The Shmunis School of Biomedicine and Cancer Research, The George S. Wise Faculty for Life Sciences, Sagol School of Neurosciences, Tel Aviv University, Tel Aviv, Israel
| | - Aviv Shalem
- Laboratory for Personalized Medicine and Neurodegenerative Diseases, The Shmunis School of Biomedicine and Cancer Research, The George S. Wise Faculty for Life Sciences, Sagol School of Neurosciences, Tel Aviv University, Tel Aviv, Israel
- The Blavatnik School of Computer Sciences, Tel Aviv University, Tel Aviv, Israel
- School of Electrical Engineering, Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel
| | - Uri Sprecher
- Laboratory for Personalized Medicine and Neurodegenerative Diseases, The Shmunis School of Biomedicine and Cancer Research, The George S. Wise Faculty for Life Sciences, Sagol School of Neurosciences, Tel Aviv University, Tel Aviv, Israel
| | - Lior Wolf
- The Blavatnik School of Computer Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Haguy Wolfenson
- Department of Genetics and Developmental Biology, The Rappaport Faculty of Medicine and Research Institute, Technion—Israel Institute of Technology, Haifa, Israel
| | - Miguel Weil
- Laboratory for Personalized Medicine and Neurodegenerative Diseases, The Shmunis School of Biomedicine and Cancer Research, The George S. Wise Faculty for Life Sciences, Sagol School of Neurosciences, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
4
|
Al-Fahad D, Alyaseen F, Al-Amery A, Singh G, Srinath M, Rehman HM, Abbas Y. Kinetic Changes of Ptdins (3,4,5) P3 within Fast and Slow Turnover Rates of Focal Adhesion. Rep Biochem Mol Biol 2022; 11:262-269. [PMID: 36164635 PMCID: PMC9455192 DOI: 10.52547/rbmb.11.2.262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Accepted: 01/02/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND The assembly and disassembly of the focal adhesions (FA) components occurs throughout life cycle of adhesion, with conservation of balance between removal and recruitment rate during temporal stages. Previous studies have demonstrated that phosphotidyilinositols play a role in regulating FA turnover. However, a little attention has been given to quantify the dynamics changes of Phosphatidylinositol 3,4,5-trisphosphate (PtdIns (3,4,5) P3) within and during fast and slow turnover rates of FA. METHODS In this study, we developed a protein purification MDA-MB-231 breast cancer cell line was used as a model in this study due to high metastatic and motile. These cells were co-transfected with GFP- paxillin/vinculin, as FA marker, and the GFP/mCherry-Btk-PH, as a biosensor to visualize PtdIns (3,4,5) P3. Confocal time-lapse images were used to monitor changes or differences in the local generation of PtdIns (3,4,5) P3 within and during assembly and disassembly of FA. Following transfection, immunostaining was used to examine the spatial co-localization between FA and PtdIns (3,4,5) P3. RESULTS Our data demonstrated that PtdIns (3,4,5) P3 co-localized with FAs and increase during assembly and decline during disassembly of FA which exhibits slow turnover rates and was in a constant level during assembly and disassembly of FA that displays fast turnover rates. DISCUSSION Our result suggested that the dynamic changes of PtdIns (3,4,5) P3, it may depend on components undergo turnover, such that early, nascent FA displays fast turnover rates and mature FA exhibits slow turnover rates. Thus, the local enrichment of PtdIns (3,4,5) P3 enhances FA assembly and disassembly activation.
Collapse
Affiliation(s)
- Dhurgham Al-Fahad
- Department of Pharmaceutical Sciences, College of Pharmacy University of Thi-Qar, Thi-Qar 64001, Iraq.
| | - Firas Alyaseen
- Department of Pharmaceutical Sciences, College of Pharmacy University of Thi-Qar, Thi-Qar 64001, Iraq.
| | - Ahmed Al-Amery
- Department of Physiology, College of Medicine, University of Thi-Qar, Iraq.
| | - Gagandeep Singh
- Viral Research and Diagnostic Laboratory, Department of Microbiology, Osmanian Medical College, Hyderabad, Telangana, India.
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, New Delhi, India.
| | - Mote Srinath
- Viral Research and Diagnostic Laboratory, Department of Microbiology, Osmanian Medical College, Hyderabad, Telangana, India.
| | | | - Yahya Abbas
- Department of Biology, College of Science, University of Thi-Qar, Thi-Qar, 64001 Iraq.
| |
Collapse
|
5
|
Murphy-Ullrich JE. Thrombospondin-1 Signaling Through the Calreticulin/LDL Receptor Related Protein 1 Axis: Functions and Possible Roles in Glaucoma. Front Cell Dev Biol 2022; 10:898772. [PMID: 35693935 PMCID: PMC9185677 DOI: 10.3389/fcell.2022.898772] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 05/10/2022] [Indexed: 11/13/2022] Open
Abstract
Thrombospondin-1 (TSP-1) is a matricellular extracellular matrix protein. Matricellular proteins are components of the extracellular matrix (ECM) that regulate key cellular functions and impact ECM organization, but which lack direct primary structural roles in the ECM. TSP-1 expression is upregulated in response to injury, hypoxia, growth factor stimulation, inflammation, glucose, and by reactive oxygen species. Relevant to glaucoma, TSP-1 is also a mechanosensitive molecule upregulated by mechanical stretch. TSP-1 expression is increased in ocular remodeling in glaucoma in both the trabecular meshwork and in the optic nerve head. The exact roles of TSP-1 in glaucoma remain to be defined, however. It plays important roles in cell behavior and in ECM remodeling during wound healing, fibrosis, angiogenesis, and in tumorigenesis and metastasis. At the cellular level, TSP-1 can modulate cell adhesion and migration, protease activity, growth factor activity, anoikis resistance, apoptosis, and collagen secretion and matrix assembly and cross-linking. These multiple functions and macromolecular and receptor interactions have been ascribed to specific domains of the TSP-1 molecule. In this review, we will focus on the cell regulatory activities of the TSP-1 N-terminal domain (NTD) sequence that binds to cell surface calreticulin (Calr) and which regulates cell functions via signaling through Calr complexed with LDL receptor related protein 1 (LRP1). We will describe TSP-1 actions mediated through the Calr/LRP1 complex in regulating focal adhesion disassembly and cytoskeletal reorganization, cell motility, anoikis resistance, and induction of collagen secretion and matrix deposition. Finally, we will consider the relevance of these TSP-1 functions to the pathologic remodeling of the ECM in glaucoma.
Collapse
Affiliation(s)
- Joanne E. Murphy-Ullrich
- Departments of Pathology, University of Alabama at Birmingham, Birmingham, AL, United States
- Department of Cell Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, United States
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, Birmingham, AL, United States
- *Correspondence: Joanne E. Murphy-Ullrich,
| |
Collapse
|
6
|
Kilbas PO, Can ND, Kizilboga T, Ezberci F, Doganay HL, Arisan ED, Dinler Doganay G. CRISPR/Cas9-mediated Bag-1 knockout increased mesenchymal characteristics of MCF-7 cells via Akt hyperactivation-mediated actin cytoskeleton remodeling. PLoS One 2022; 17:e0261062. [PMID: 34995286 PMCID: PMC8741009 DOI: 10.1371/journal.pone.0261062] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 11/23/2021] [Indexed: 11/18/2022] Open
Abstract
Bag-1 protein is a crucial target in cancer to increase the survival and proliferation of cells. The Bag-1 expression is significantly upregulated in primary and metastatic cancer patients compared to normal breast tissue. Overexpression of Bag-1 decreases the efficiency of conventional chemotherapeutic drugs, whereas Bag-1 silencing enhances the apoptotic efficiency of therapeutics, mostly in hormone-positive breast cancer subtypes. In this study, we generated stable Bag-1 knockout (KO) MCF-7 breast cancer cells to monitor stress-mediated cellular alterations in comparison to wild type (wt) and Bag-1 overexpressing (Bag-1 OE) MCF-7 cells. Validation and characterization studies of Bag-1 KO cells showed different cellular morphology with hyperactive Akt signaling, which caused stress-mediated actin reorganization, focal adhesion decrease and led to mesenchymal characteristics in MCF-7 cells. A potent Akt inhibitor, MK-2206, suppressed mesenchymal transition in Bag-1 KO cells. Similar results were obtained following the recovery of Bag-1 isoforms (Bag-1S, M, or L) in Bag-1 KO cells. The findings of this study emphasized that Bag-1 is a mediator of actin-mediated cytoskeleton organization through regulating Akt activation.
Collapse
Affiliation(s)
- Pelin Ozfiliz Kilbas
- Department of Molecular Biology Genetics and Biotechnology, Istanbul Technical University, Istanbul, Turkey
- Department of Molecular Biology and Genetics, Istanbul Kultur University, Istanbul, Turkey
| | - Nisan Denizce Can
- Department of Molecular Biology Genetics and Biotechnology, Istanbul Technical University, Istanbul, Turkey
| | - Tugba Kizilboga
- Department of Molecular Biology Genetics and Biotechnology, Istanbul Technical University, Istanbul, Turkey
| | - Fikret Ezberci
- Department of General Surgery, Umraniye Teaching and Research Hospital, Istanbul, Turkey
| | - Hamdi Levent Doganay
- Genomic Laboratory (GLAB), Umraniye Teaching And Research Hospital, University of Health Sciences, Istanbul, Turkey
| | - Elif Damla Arisan
- Institute of Biotechnology, Gebze Technical University, Kocaeli, Turkey
| | - Gizem Dinler Doganay
- Department of Molecular Biology Genetics and Biotechnology, Istanbul Technical University, Istanbul, Turkey
| |
Collapse
|
7
|
Transcriptome profiling analysis of muscle tissue reveals potential candidate genes affecting water holding capacity in Chinese Simmental beef cattle. Sci Rep 2021; 11:11897. [PMID: 34099805 PMCID: PMC8184995 DOI: 10.1038/s41598-021-91373-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Accepted: 05/26/2021] [Indexed: 11/12/2022] Open
Abstract
Water holding capacity (WHC) is an important sensory attribute that greatly influences meat quality. However, the molecular mechanism that regulates the beef WHC remains to be elucidated. In this study, the longissimus dorsi (LD) muscles of 49 Chinese Simmental beef cattle were measured for meat quality traits and subjected to RNA sequencing. WHC had significant correlation with 35 kg water loss (r = − 0.99, p < 0.01) and IMF content (r = 0.31, p < 0.05), but not with SF (r = − 0.20, p = 0.18) and pH (r = 0.11, p = 0.44). Eight individuals with the highest WHC (H-WHC) and the lowest WHC (L-WHC) were selected for transcriptome analysis. A total of 865 genes were identified as differentially expressed genes (DEGs) between two groups, of which 633 genes were up-regulated and 232 genes were down-regulated. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment revealed that DEGs were significantly enriched in 15 GO terms and 96 pathways. Additionally, based on protein–protein interaction (PPI) network, animal QTL database (QTLdb), and relevant literature, the study not only confirmed seven genes (HSPA12A, HSPA13, PPARγ, MYL2, MYPN, TPI, and ATP2A1) influenced WHC in accordance with previous studies, but also identified ATP2B4, ACTN1, ITGAV, TGFBR1, THBS1, and TEK as the most promising novel candidate genes affecting the WHC. These findings could offer important insight for exploring the molecular mechanism underlying the WHC trait and facilitate the improvement of beef quality.
Collapse
|
8
|
Tousley A, Iuliano M, Weisman E, Sapp E, Zhang N, Vodicka P, Alexander J, Aviolat H, Gatune L, Reeves P, Li X, Khvorova A, Ellerby LM, Aronin N, DiFiglia M, Kegel-Gleason KB. Rac1 Activity Is Modulated by Huntingtin and Dysregulated in Models of Huntington's Disease. J Huntingtons Dis 2020; 8:53-69. [PMID: 30594931 PMCID: PMC6398565 DOI: 10.3233/jhd-180311] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Background: Previous studies suggest that Huntingtin, the protein mutated in Huntington’s disease (HD), is required for actin based changes in cell morphology, and undergoes stimulus induced targeting to plasma membranes where it interacts with phospholipids involved in cell signaling. The small GTPase Rac1 is a downstream target of growth factor stimulation and PI 3-kinase activity and is critical for actin dependent membrane remodeling. Objective: To determine if Rac1 activity is impaired in HD or regulated by normal Huntingtin. Methods: Analyses were performed in differentiated control and HD human stem cells and HD Q140/Q140 knock-in mice. Biochemical methods included SDS-PAGE, western blot, immunoprecipitation, affinity chromatography, and ELISA based Rac activity assays. Results: Basal Rac1 activity increased following depletion of Huntingtin with Huntingtin specific siRNA in human primary fibroblasts and in human control neuron cultures. Human cells (fibroblasts, neural stem cells, and neurons) with the HD mutation failed to increase Rac1 activity in response to growth factors. Rac1 activity levels were elevated in striatum of 1.5-month-old HD Q140/Q140 mice and in primary embryonic cortical neurons from HD mice. Affinity chromatography analysis of striatal lysates showed that Huntingtin is in a complex with Rac1, p85α subunit of PI 3-kinase, and the actin bundling protein α-actinin and interacts preferentially with the GTP bound form of Rac1. The HD mutation reduced Huntingtin interaction with p85α. Conclusions: These findings suggest that Huntingtin regulates Rac1 activity as part of a coordinated response to growth factor signaling and this function is impaired early in HD.
Collapse
Affiliation(s)
- Adelaide Tousley
- Department of Neurology, Laboratory of Cellular Neurobiology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Maria Iuliano
- Department of Neurology, Laboratory of Cellular Neurobiology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Elizabeth Weisman
- Department of Neurology, Laboratory of Cellular Neurobiology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Ellen Sapp
- Department of Neurology, Laboratory of Cellular Neurobiology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Ningzhe Zhang
- Buck Institute for Research on Aging, Novato, CA, USA
| | - Petr Vodicka
- Department of Neurology, Laboratory of Cellular Neurobiology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Jonathan Alexander
- Department of Neurology, Laboratory of Cellular Neurobiology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Hubert Aviolat
- Department of Neurology, Laboratory of Cellular Neurobiology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Leah Gatune
- Department of Neurology, Laboratory of Cellular Neurobiology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Patrick Reeves
- Department of Neurology, Laboratory of Cellular Neurobiology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Xueyi Li
- Department of Neurology, Laboratory of Cellular Neurobiology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Anastasia Khvorova
- RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, MA, USA
| | | | - Neil Aronin
- RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, MA, USA.,Department of Medicine and Cell Biology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Marian DiFiglia
- Department of Neurology, Laboratory of Cellular Neurobiology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Kimberly B Kegel-Gleason
- Department of Neurology, Laboratory of Cellular Neurobiology, Massachusetts General Hospital, Charlestown, MA, USA
| |
Collapse
|
9
|
Tousley A, Iuliano M, Weisman E, Sapp E, Richardson H, Vodicka P, Alexander J, Aronin N, DiFiglia M, Kegel-Gleason KB. Huntingtin associates with the actin cytoskeleton and α-actinin isoforms to influence stimulus dependent morphology changes. PLoS One 2019; 14:e0212337. [PMID: 30768638 PMCID: PMC6377189 DOI: 10.1371/journal.pone.0212337] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 01/31/2019] [Indexed: 01/09/2023] Open
Abstract
One response of cells to growth factor stimulus involves changes in morphology driven by the actin cytoskeleton and actin associated proteins which regulate functions such as cell adhesion, motility and in neurons, synaptic plasticity. Previous studies suggest that Huntingtin may be involved in regulating morphology however, there has been limited evidence linking endogenous Huntingtin localization or function with cytoplasmic actin in cells. We found that depletion of Huntingtin in human fibroblasts reduced adhesion and altered morphology and these phenotypes were made worse with growth factor stimulation, whereas the presence of the Huntington's Disease mutation inhibited growth factor induced changes in morphology and increased numbers of vinculin-positive focal adhesions. Huntingtin immunoreactivity localized to actin stress fibers, vinculin-positive adhesion contacts and membrane ruffles in fibroblasts. Interactome data from others has shown that Huntingtin can associate with α-actinin isoforms which bind actin filaments. Mapping studies using a cDNA encoding α-actinin-2 showed that it interacts within Huntingtin aa 399-969. Double-label immunofluorescence showed Huntingtin and α-actinin-1 co-localized to stress fibers, membrane ruffles and lamellar protrusions in fibroblasts. Proximity ligation assays confirmed a close molecular interaction between Huntingtin and α-actinin-1 in human fibroblasts and neurons. Huntingtin silencing with siRNA in fibroblasts blocked the recruitment of α-actinin-1 to membrane foci. These studies support the idea that Huntingtin is involved in regulating adhesion and actin dependent functions including those involving α-actinin.
Collapse
Affiliation(s)
- Adelaide Tousley
- Laboratory of Cellular Neurobiology, Department of Neurology, Massachusetts General Hospital, Charlestown, Massachusetts, United States of America
| | - Maria Iuliano
- Laboratory of Cellular Neurobiology, Department of Neurology, Massachusetts General Hospital, Charlestown, Massachusetts, United States of America
| | - Elizabeth Weisman
- Laboratory of Cellular Neurobiology, Department of Neurology, Massachusetts General Hospital, Charlestown, Massachusetts, United States of America
| | - Ellen Sapp
- Laboratory of Cellular Neurobiology, Department of Neurology, Massachusetts General Hospital, Charlestown, Massachusetts, United States of America
| | - Heather Richardson
- Laboratory of Cellular Neurobiology, Department of Neurology, Massachusetts General Hospital, Charlestown, Massachusetts, United States of America
| | - Petr Vodicka
- Laboratory of Cellular Neurobiology, Department of Neurology, Massachusetts General Hospital, Charlestown, Massachusetts, United States of America
| | - Jonathan Alexander
- Laboratory of Cellular Neurobiology, Department of Neurology, Massachusetts General Hospital, Charlestown, Massachusetts, United States of America
| | - Neil Aronin
- Department of Medicine and Cell Biology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Marian DiFiglia
- Laboratory of Cellular Neurobiology, Department of Neurology, Massachusetts General Hospital, Charlestown, Massachusetts, United States of America
| | - Kimberly B. Kegel-Gleason
- Laboratory of Cellular Neurobiology, Department of Neurology, Massachusetts General Hospital, Charlestown, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
10
|
Thomas DG, Robinson DN. The fifth sense: Mechanosensory regulation of alpha-actinin-4 and its relevance for cancer metastasis. Semin Cell Dev Biol 2017; 71:68-74. [PMID: 28579451 DOI: 10.1016/j.semcdb.2017.05.024] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 05/26/2017] [Accepted: 05/30/2017] [Indexed: 12/18/2022]
Abstract
Metastatic cancer cells invading through dense tumor stroma experience internal and external forces that are sensed through a variety of mechanosensory proteins that drive adaptations for specific environments. Alpha-actinin-4 (ACTN4) is a member of the α-actinin family of actin crosslinking proteins that is upregulated in several types of cancers. It shares 86% protein similarity with α-actinin-1, another non-muscle ACTN isoform, which appears to have a more modest role, if any, in cancer progression. While they share regulatory mechanisms, such as phosphorylation, calcium binding, phosphatidyl inositol binding, and calpain cleavage, α-actinin-4 exhibits a unique mechanosensory regulation that α-actinin-1 does not. This behavior is mediated, at least in part, by each protein's actin-binding affinity as well as the catch-slip-bond behavior of the actin binding domains. We will discuss currently known modes of ACTN4 regulation, their interactions, and how mechanosensation may provide major therapeutic targeting potential for cancer metastasis.
Collapse
Affiliation(s)
- Dustin G Thomas
- Department of Cell Biology, Johns Hopkins University, Baltimore, MD, 21205, USA.
| | - Douglas N Robinson
- Department of Cell Biology, Johns Hopkins University, Baltimore, MD, 21205, USA; Department of Pharmacology and Molecular Science, Johns Hopkins University,Baltimore, MD, 21205, USA; Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, 21205, USA; Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA.
| |
Collapse
|
11
|
Ballek O, Valečka J, Dobešová M, Broučková A, Manning J, Řehulka P, Stulík J, Filipp D. TCR Triggering Induces the Formation of Lck-RACK1-Actinin-1 Multiprotein Network Affecting Lck Redistribution. Front Immunol 2016; 7:449. [PMID: 27833610 PMCID: PMC5081367 DOI: 10.3389/fimmu.2016.00449] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 10/10/2016] [Indexed: 02/02/2023] Open
Abstract
The initiation of T-cell signaling is critically dependent on the function of the member of Src family tyrosine kinases, Lck. Upon T-cell antigen receptor (TCR) triggering, Lck kinase activity induces the nucleation of signal-transducing hubs that regulate the formation of complex signaling network and cytoskeletal rearrangement. In addition, the delivery of Lck function requires rapid and targeted membrane redistribution, but the mechanism underpinning this process is largely unknown. To gain insight into this process, we considered previously described proteins that could assist in this process via their capacity to interact with kinases and regulate their intracellular translocations. An adaptor protein, receptor for activated C kinase 1 (RACK1), was chosen as a viable option, and its capacity to bind Lck and aid the process of activation-induced redistribution of Lck was assessed. Our microscopic observation showed that T-cell activation induces a rapid, concomitant, and transient co-redistribution of Lck and RACK1 into the forming immunological synapse. Consistent with this observation, the formation of transient RACK1-Lck complexes were detectable in primary CD4+ T-cells with their maximum levels peaking 10 s after TCR-CD4 co-aggregation. Moreover, RACK1 preferentially binds to a pool of kinase active pY394Lck, which co-purifies with high molecular weight cellular fractions. The formation of RACK1-Lck complexes depends on functional SH2 and SH3 domains of Lck and includes several other signaling and cytoskeletal elements that transiently bind the complex. Notably, the F-actin-crosslinking protein, α-actinin-1, binds to RACK1 only in the presence of kinase active Lck suggesting that the formation of RACK1-pY394Lck-α-actinin-1 complex serves as a signal module coupling actin cytoskeleton bundling with productive TCR/CD4 triggering. In addition, the treatment of CD4+ T-cells with nocodazole, which disrupts the microtubular network, also blocked the formation of RACK1-Lck complexes. Importantly, activation-induced Lck redistribution was diminished in primary CD4+ T-cells by an adenoviral-mediated knockdown of RACK1. These results demonstrate that in T cells, RACK1, as an essential component of the multiprotein complex which upon TCR engagement, links the binding of kinase active Lck to elements of the cytoskeletal network and affects the subcellular redistribution of Lck.
Collapse
Affiliation(s)
- Ondřej Ballek
- Laboratory of Immunobiology, Institute of Molecular Genetics AS CR , Prague , Czech Republic
| | - Jan Valečka
- Laboratory of Immunobiology, Institute of Molecular Genetics AS CR , Prague , Czech Republic
| | - Martina Dobešová
- Laboratory of Immunobiology, Institute of Molecular Genetics AS CR , Prague , Czech Republic
| | - Adéla Broučková
- Laboratory of Immunobiology, Institute of Molecular Genetics AS CR , Prague , Czech Republic
| | - Jasper Manning
- Laboratory of Immunobiology, Institute of Molecular Genetics AS CR , Prague , Czech Republic
| | - Pavel Řehulka
- Faculty of Military Health Sciences, Institute of Molecular Pathology , Hradec Králové , Czech Republic
| | - Jiří Stulík
- Faculty of Military Health Sciences, Institute of Molecular Pathology , Hradec Králové , Czech Republic
| | - Dominik Filipp
- Laboratory of Immunobiology, Institute of Molecular Genetics AS CR , Prague , Czech Republic
| |
Collapse
|
12
|
Lei W, He Y, Shui X, Li G, Yan G, Zhang Y, Huang S, Chen C, Ding Y. Expression and analyses of the HIF-1 pathway in the lungs of humans with pulmonary arterial hypertension. Mol Med Rep 2016; 14:4383-4390. [PMID: 27667582 DOI: 10.3892/mmr.2016.5752] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2015] [Accepted: 07/22/2016] [Indexed: 11/06/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is characterized by endothelial dysfunction and structural remodeling of the pulmonary vasculature, mediated initially by reduced oxygen availability in the lungs. Hypoxia inducible factor (HIF), consisting of the functional subunit, HIF‑1α, and the constitutively expressed HIF‑1β, is involved in the pathological processes associated with hypoxia. In the current study, the sequences of cDNAs and amino acids of HIF were characterized and analyzed using online bioinformatics tools. To further evaluate whether HIF accounts for the occurrence of PAH, the present study determine the expression and phosphorylation levels of HIF and its associated pathways, including extracellular signal‑regulated kinase (Erk)1/2 and phosphoinositide 3‑kinase (PI3K)/Akt, in the lungs of patients with PAH by reverse transcription‑quantitative polymerase chain reaction and western blotting. The mRNA expression levels of PI3K, Erk2, and HIF‑1α in the patients with PAH were significantly higher, compared with those in the control group, by 3.6‑fold (P<0.01), 4.06‑fold and 2.64‑fold (P<0.05), respectively. No significant differences were found in the mRNA and protein levels of Akt between the two groups (P>0.05). The protein levels of phosphorylated (p‑)Akt, Erk1/2, p‑Erk1/2, HIF‑1α and HIF‑1β were significantly increased by 5.89‑, 0.5‑, 0.59‑, 1.46‑ and 0.92‑fold, respectively, in the patients with PAH, compared with those in the controls group (P<0.01 for p‑Akt, Erk1/2; P<0.05 for p‑Erk1/2, HIF‑1α and HIF‑1β). These findings suggested that the mitogen‑activated protein kinase and PI3K/Akt signaling pathways, and HIF‑1 may perform a specific function in the pathogenesis of PAH.
Collapse
Affiliation(s)
- Wei Lei
- Laboratory of Cardiovascular Diseases, Guangdong Medical University, Zhanjiang, Guangdong 524000, P.R. China
| | - Yuan He
- Laboratory of Cardiovascular Diseases, Guangdong Medical University, Zhanjiang, Guangdong 524000, P.R. China
| | - Xiaorong Shui
- Laboratory of Vascular Surgery, Guangdong Medical University, Zhanjiang, Guangdong 524000, P.R. China
| | - Guoming Li
- Cardiovascular Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524000, P.R. China
| | - Guosen Yan
- Laboratory of Cardiovascular Diseases, Guangdong Medical University, Zhanjiang, Guangdong 524000, P.R. China
| | - Yu Zhang
- Cardiovascular Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524000, P.R. China
| | - Shian Huang
- Cardiovascular Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524000, P.R. China
| | - Can Chen
- Laboratory of Cardiovascular Diseases, Guangdong Medical University, Zhanjiang, Guangdong 524000, P.R. China
| | - Yuanlin Ding
- Institute of Medical Systems Biology, Guangdong Medical University, Dongguan, Guangdong 523808, P.R. China
| |
Collapse
|
13
|
Modulation of the extracellular matrix patterning of thrombospondins by actin dynamics and thrombospondin oligomer state. Biosci Rep 2015; 35:BSR20140168. [PMID: 26182380 PMCID: PMC4613707 DOI: 10.1042/bsr20140168] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Accepted: 04/21/2015] [Indexed: 01/01/2023] Open
Abstract
Thrombospondins (TSPs) are evolutionarily-conserved, secreted glycoproteins that interact with cell surfaces and extracellular matrix (ECM) and have complex roles in cell interactions. Unlike the structural components of the ECM that form networks or fibrils, TSPs are deposited into ECM as arrays of nanoscale puncta. The cellular and molecular mechanisms for the patterning of TSPs in ECM are poorly understood. In the present study, we investigated whether the mechanisms of TSP patterning in cell-derived ECM involves actin cytoskeletal pathways or TSP oligomer state. From tests of a suite of pharmacological inhibitors of small GTPases, actomyosin-based contractility, or actin microfilament integrity and dynamics, cytochalasin D and jasplakinolide treatment of cells were identified to result in altered ECM patterning of a model TSP1 trimer. The strong effect of cytochalasin D indicated that mechanisms controlling puncta patterning depend on global F-actin dynamics. Similar spatial changes were obtained with endogenous TSPs after cytochalasin D treatment, implicating physiological relevance. Under matched experimental conditions with ectopically-expressed TSPs, the magnitude of the effect was markedly lower for pentameric TSP5 and Drosophila TSP, than for trimeric TSP1 or dimeric Ciona TSPA. To distinguish between the variables of protein sequence or oligomer state, we generated novel, chimeric pentamers of TSP1. These proteins accumulated within ECM at higher levels than TSP1 trimers, yet the effect of cytochalasin D on the spatial distribution of puncta was reduced. These findings introduce a novel concept that F-actin dynamics modulate the patterning of TSPs in ECM and that TSP oligomer state is a key determinant of this process.
Collapse
|
14
|
Abstract
α-Actinins are a major class of actin filament cross-linking proteins expressed in virtually all cells. In muscle, actinins cross-link thin filaments from adjacent sarcomeres. In non-muscle cells, different actinin isoforms play analogous roles in cross-linking actin filaments and anchoring them to structures such as cell-cell and cell-matrix junctions. Although actinins have long been known to play roles in cytokinesis, cell adhesion and cell migration, recent studies have provided further mechanistic insights into these functions. Roles for actinins in synaptic plasticity and membrane trafficking events have emerged more recently, as has a 'non-canonical' function for actinins in transcriptional regulation in the nucleus. In the present paper we review recent advances in our understanding of these diverse cell biological functions of actinins in non-muscle cells, as well as their roles in cancer and in genetic disorders affecting platelet and kidney physiology. We also make two proposals with regard to the actinin nomenclature. First, we argue that naming actinin isoforms according to their expression patterns is problematic and we suggest a more precise nomenclature system. Secondly, we suggest that the α in α-actinin is superfluous and can be omitted.
Collapse
|
15
|
Kojima S, Koitabashi K, Iizuka N, Okamoto K, Arito M, Sato T, Kurokawa MS, Suematsu N, Shibagaki Y, Yasuda T, Kimura K, Kato T. Proteomic analysis of whole glomeruli in patients with IgA nephropathy using microsieving. Am J Nephrol 2014; 39:36-45. [PMID: 24434790 DOI: 10.1159/000357788] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Accepted: 12/04/2013] [Indexed: 12/20/2022]
Abstract
BACKGROUND To promote understanding of immunoglobulin A nephropathy (IgAN) pathophysiology, we tried to elucidate glomerular protein profiles in IgAN, using microsieving that we established recently to isolate glomeruli from renal biopsy samples and proteomic approaches. METHODS Glomeruli were isolated from renal biopsy samples of patients with IgAN (n = 5) and with minimal change nephrotic syndrome (MCNS; n = 5) using microsieving. Proteins extracted from the isolated glomeruli were separated by 2-dimensional differential gel electrophoresis (2D-DIGE). Proteins with different amounts between the two groups were identified by mass spectrometry. One of the identified proteins, α-actinin-4 (ACTN4), was further analyzed by Western blotting, RT-polymerase chain reaction (PCR), and immunohistochemistry. RESULTS By 2D-DIGE, 72 out of the detected 1,170 protein spots showed significantly different intensity between the two groups (p < 0.05). Thirty-four out of the 72 protein spots showed more than 1.5-fold or less than 1/1.5-fold intensity, out of which 16 protein spots were successfully identified. No microbial protein was identified. ACTN4 molecules with a low molecular weight of approximately 77 kDa were found to increase in the IgAN group. Lack of an N-terminal part of ACTN4 was demonstrated by Western blotting. No defect of mRNA for ACTN4 was evidenced by RT-PCR. Predominant existence of ACTN4 in capillary walls of glomeruli of IgAN patients was demonstrated by immunohistochemistry in glomerular sections of patients with IgAN. CONCLUSION Use of microsieving enabled us to biochemically analyze glomerular proteins in renal biopsy samples from patients with glomerular diseases. With this method, we demonstrated skewed glomerular protein profiles in IgAN.
Collapse
Affiliation(s)
- Shigeki Kojima
- Clinical Proteomics and Molecular Medicine, St. Marianna University Graduate School of Medicine, Kawasaki, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Roca-Cusachs P, del Rio A, Puklin-Faucher E, Gauthier NC, Biais N, Sheetz MP. Integrin-dependent force transmission to the extracellular matrix by α-actinin triggers adhesion maturation. Proc Natl Acad Sci U S A 2013; 110:E1361-70. [PMID: 23515331 PMCID: PMC3625291 DOI: 10.1073/pnas.1220723110] [Citation(s) in RCA: 211] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Focal adhesions are mechanosensitive elements that enable mechanical communication between cells and the extracellular matrix. Here, we demonstrate a major mechanosensitive pathway in which α-actinin triggers adhesion maturation by linking integrins to actin in nascent adhesions. We show that depletion of the focal adhesion protein α-actinin enhances force generation in initial adhesions on fibronectin, but impairs mechanotransduction in a subsequent step, preventing adhesion maturation. Expression of an α-actinin fragment containing the integrin binding domain, however, dramatically reduces force generation in depleted cells. This behavior can be explained by a competition between talin (which mediates initial adhesion and force generation) and α-actinin for integrin binding. Indeed, we show in an in vitro assay that talin and α-actinin compete for binding to β3 integrins, but cooperate in binding to β1 integrins. Consistently, we find opposite effects of α-actinin depletion and expression of mutants on substrates that bind β3 integrins (fibronectin and vitronectin) versus substrates that only bind β1 integrins (collagen). We thus suggest that nascent adhesions composed of β3 integrins are initially linked to the actin cytoskeleton by talin, and then α-actinin competes with talin to bind β3 integrins. Force transmitted through α-actinin then triggers adhesion maturation. Once adhesions have matured, α-actinin recruitment correlates with force generation, suggesting that α-actinin is the main link transmitting force between integrins and the cytoskeleton in mature adhesions. Such a multistep process enables cells to adjust forces on matrices, unveiling a role of α-actinin that is different from its well-studied function as an actin cross-linker.
Collapse
Affiliation(s)
- Pere Roca-Cusachs
- Institute for Bioengineering of Catalonia, 08028 Barcelona, Spain
- Department of Physiological Sciences I, University of Barcelona, 08036 Barcelona, Spain
| | - Armando del Rio
- Department of Biological Sciences, Columbia University, New York, NY 10027
- Center for Biophysical Studies, Autonomous University of Barcelona, 08193 Bellaterra, Spain
| | | | - Nils C. Gauthier
- Department of Biological Sciences, Columbia University, New York, NY 10027
- Mechanobiology Institute of Singapore, National University of Singapore, Singapore 117411; and
| | - Nicolas Biais
- Department of Biological Sciences, Columbia University, New York, NY 10027
- Department of Biology, Brooklyn College, City University of New York, Brooklyn, NY 11210
| | - Michael P. Sheetz
- Department of Biological Sciences, Columbia University, New York, NY 10027
- Mechanobiology Institute of Singapore, National University of Singapore, Singapore 117411; and
| |
Collapse
|
17
|
Abstract
Alpha-actinins (ACTNs) were originally identified as cytoskeletal proteins which cross-link filamentous actin to establish cytoskeletal architect that protects cells from mechanical stress and controls cell movement. Notably, unlike other ACTNs, alpha-actinin 4 (ACTN4) displays unique characteristics in signaling transduction, nuclear translocation, and gene expression regulation. Initial reports indicated that ACTN4 is part of the breast cancer cell motile apparatus and is highly expressed in the nucleus. These results imply that ACTN4 plays a role in breast cancer tumorigenesis. While several observations in breast cancer and other cancers support this hypothesis, little direct evidence links the tumorigenic phenotype with ACTN4-mediated pathological mechanisms. Recently, several studies have demonstrated that in addition to its role in coordinating cytoskeleton, ACTN4 interacts with signaling mediators, chromatin remodeling factors, and transcription factors including nuclear receptors. Thus, ACTN4 functions as a versatile promoter for breast cancer tumorigenesis and appears to be an ideal drug target for future therapeutic development.
Collapse
Affiliation(s)
- Kuo-Sheng Hsu
- Department of Biochemistry, School of Medicine, Case Western Reserve University-CWRU, The Comprehensive Cancer Center of CWRU, Cleveland, Ohio, USA
| | | |
Collapse
|
18
|
Sweetwyne MT, Murphy-Ullrich JE. Thrombospondin1 in tissue repair and fibrosis: TGF-β-dependent and independent mechanisms. Matrix Biol 2012; 31:178-86. [PMID: 22266026 DOI: 10.1016/j.matbio.2012.01.006] [Citation(s) in RCA: 172] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2011] [Revised: 12/20/2011] [Accepted: 12/22/2011] [Indexed: 12/16/2022]
Abstract
Thrombospondin 1 (TSP1) plays major roles in both physiologic and pathologic tissue repair. TSP1 through its type 1 repeats is a known regulator of latent TGF-β activation and plays a role in wound healing and fibrosis. Binding of the TSP N-terminal domain to cell surface calreticulin in complex with LDL-receptor related protein 1 stimulates intermediate cell adhesion, cell migration, anoikis resistance, collagen expression and matrix deposition in an in vivo model of the foreign body response. There is also emerging evidence that TSP EGF-like repeats alter endothelial cell-cell interactions and stimulate epithelial migration through transactivation of EGF receptors. The mechanisms underlying these functions of TSP1 and the implications for physiologic and pathologic wound repair and fibrosis will be discussed.
Collapse
Affiliation(s)
- Mariya T Sweetwyne
- Department of Animal Biology, School of Veterinary Medicine, University of Pennsylvania, United States
| | | |
Collapse
|
19
|
Slabbaert JR, Khuong TM, Verstreken P. Phosphoinositides at the Neuromuscular Junction of Drosophila melanogaster: A Genetic Approach. Methods Cell Biol 2012; 108:227-47. [DOI: 10.1016/b978-0-12-386487-1.00012-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
20
|
Gu W, Katz Z, Wu B, Park HY, Li D, Lin S, Wells AL, Singer RH. Regulation of local expression of cell adhesion and motility-related mRNAs in breast cancer cells by IMP1/ZBP1. J Cell Sci 2012; 125:81-91. [PMID: 22266909 PMCID: PMC3269024 DOI: 10.1242/jcs.086132] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/21/2011] [Indexed: 02/05/2023] Open
Abstract
Metastasis involves tumor cell detachment from the primary tumor, and acquisition of migratory and invasive capabilities. These capabilities are mediated by multiple events, including loss of cell-cell contact, an increase in focal adhesion turnover and failure to maintain a normal cell polarity. We have previously reported that silencing of the expression of the zipcode-binding protein IMP1/ZBP1 in breast tumor patients is associated with metastasis. IMP1/ZBP1 selectively binds to a group of mRNAs that encode important mediators for cell adhesion and motility. Here, we show that in both T47D and MDA231 human breast carcinoma cells IMP1/ZBP1 functions to suppress cell invasion. Binding of ZBP1 to the mRNAs encoding E-cadherin, β-actin, α-actinin and the Arp2/3 complex facilitates localization of the mRNAs, which stabilizes cell-cell connections and focal adhesions. Our studies suggest a novel mechanism through which IMP1/ZBP1 simultaneously regulates the local expression of many cell-motility-related mRNAs to maintain cell adherence and polarity, decrease focal adhesion turnover and maintain a persistent and directional motility.
Collapse
Affiliation(s)
- Wei Gu
- Department of Pathophysiology, The Key Immunopathology Laboratory of Guangdong Province, Shantou University Medical College, Shantou, Guangdong Province, 515031, China
- Authors for correspondence (; )
| | - Zachary Katz
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx NY 10461, USA
| | - Bin Wu
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx NY 10461, USA
| | - Hye Yoon Park
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx NY 10461, USA
| | - Deling Li
- Department of Pathophysiology, The Key Immunopathology Laboratory of Guangdong Province, Shantou University Medical College, Shantou, Guangdong Province, 515031, China
| | - Stanley Lin
- Department of Pathophysiology, The Key Immunopathology Laboratory of Guangdong Province, Shantou University Medical College, Shantou, Guangdong Province, 515031, China
| | - Amber L. Wells
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx NY 10461, USA
- Department of Medicine, Albert Einstein College of Medicine, Bronx NY 10461, USA
| | - Robert H. Singer
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx NY 10461, USA
- Authors for correspondence (; )
| |
Collapse
|
21
|
Scales TME, Parsons M. Spatial and temporal regulation of integrin signalling during cell migration. Curr Opin Cell Biol 2011; 23:562-8. [DOI: 10.1016/j.ceb.2011.05.008] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2011] [Revised: 05/29/2011] [Accepted: 05/30/2011] [Indexed: 11/26/2022]
|
22
|
Abstract
Thrombospondins are evolutionarily conserved, calcium-binding glycoproteins that undergo transient or longer-term interactions with other extracellular matrix components. They share properties with other matrix molecules, cytokines, adaptor proteins, and chaperones, modulate the organization of collagen fibrils, and bind and localize an array of growth factors or proteases. At cell surfaces, interactions with an array of receptors activate cell-dependent signaling and phenotypic outcomes. Through these dynamic, pleiotropic, and context-dependent pathways, mammalian thrombospondins contribute to wound healing and angiogenesis, vessel wall biology, connective tissue organization, and synaptogenesis. We overview the domain organization and structure of thrombospondins, key features of their evolution, and their cell biology. We discuss their roles in vivo, associations with human disease, and ongoing translational applications. In many respects, we are only beginning to appreciate the important roles of these proteins in physiology and pathology.
Collapse
Affiliation(s)
- Josephine C Adams
- School of Biochemistry, University of Bristol, Bristol BS8 1TD, United Kingdom.
| | | |
Collapse
|
23
|
Qu H, Tu Y, Shi X, Larjava H, Saleem MA, Shattil SJ, Fukuda K, Qin J, Kretzler M, Wu C. Kindlin-2 regulates podocyte adhesion and fibronectin matrix deposition through interactions with phosphoinositides and integrins. J Cell Sci 2011; 124:879-91. [PMID: 21325030 DOI: 10.1242/jcs.076976] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Kindlin-2 is a FERM and PH domain-containing integrin-binding protein that is emerging as an important regulator of integrin activation. How kindlin-2 functions in integrin activation, however, is not known. We report here that kindlin-2 interacts with multiple phosphoinositides, preferentially with phosphatidylinositol 3,4,5-trisphosphate. Although integrin-binding is essential for focal adhesion localization of kindlin-2, phosphoinositide-binding is not required for this process. Using biologically and clinically relevant glomerular podocytes as a model system, we show that integrin activation and dependent processes are tightly regulated by kindlin-2: depletion of kindlin-2 reduced integrin activation, matrix adhesion and fibronectin matrix deposition, whereas overexpression of kindlin-2 promoted these processes. Furthermore, we provide evidence showing that kindlin-2 is involved in phosphoinositide-3-kinase-mediated regulation of podocyte-matrix adhesion and fibronectin matrix deposition. Mechanistically, kindlin-2 promotes integrin activation and integrin-dependent processes through interacting with both integrins and phosphoinositides. TGF-β1, a mediator of progressive glomerular failure, markedly increased the level of kindlin-2 and fibronectin matrix deposition, and the latter process was reversed by depletion of kindlin-2. Our results reveal important functions of kindlin-2 in the regulation of podocyte-matrix adhesion and matrix deposition and shed new light on the mechanism whereby kindlin-2 functions in these processes.
Collapse
Affiliation(s)
- Hong Qu
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Mitsios JV, Prévost N, Kasirer-Friede A, Gutierrez E, Groisman A, Abrams CS, Wang Y, Litvinov RI, Zemljic-Harpf A, Ross RS, Shattil SJ. What is vinculin needed for in platelets? J Thromb Haemost 2010; 8:2294-304. [PMID: 20670372 PMCID: PMC2965783 DOI: 10.1111/j.1538-7836.2010.03998.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
UNLABELLED Summary. BACKGROUND Vinculin links integrins to the cell cytoskeleton by virtue of its binding to proteins such as talin and F-actin. It has been implicated in the transmission of mechanical forces from the extracellular matrix to the cytoskeleton of migrating cells. Vinculin's function in platelets is unknown. OBJECTIVE To determine whether vinculin is required for the functions of platelets and their major integrin, α(IIb) β(3) . METHODS The murine vinculin gene (Vcl) was deleted in the megakaryocyte/platelet lineage by breeding Vcl fl/fl mice with Pf4-Cre mice. Platelet and integrin functions were studied in vivo and ex vivo. RESULTS Vinculin was undetectable in platelets from Vcl fl/fl Cre(+) mice, as determined by immunoblotting and fluorescence microscopy. Vinculin-deficient megakaryocytes exhibited increased membrane tethers in response to mechanical pulling on α(IIb) β(3) with laser tweezers, suggesting that vinculin helps to maintain membrane cytoskeleton integrity. Surprisingly, vinculin-deficient platelets displayed normal agonist-induced fibrinogen binding to α(IIb) β(3) , aggregation, spreading, actin polymerization/organization, clot retraction and the ability to form a procoagulant surface. Furthermore, vinculin-deficient platelets adhered to immobilized fibrinogen or collagen normally, under both static and flow conditions. Tail bleeding times were prolonged in 59% of vinculin-deficient mice. However, these mice exhibited no spontaneous bleeding and they formed occlusive platelet thrombi comparable to those in wild-type littermates in response to carotid artery injury with FeCl(3) . CONCLUSION Despite promoting membrane cytoskeleton integrity when mechanical force is applied to α(IIb) β(3) , vinculin is not required for the traditional functions of α(IIb) β(3) or the platelet actin cytoskeleton.
Collapse
Affiliation(s)
- John V. Mitsios
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Nicolas Prévost
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Ana Kasirer-Friede
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Edgar Gutierrez
- Department of Physics, University of California, San Diego, La Jolla, CA 92093
| | - Alex Groisman
- Department of Physics, University of California, San Diego, La Jolla, CA 92093
| | - Charles S. Abrams
- Department of Medicine and Cell, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
| | - Yanfeng Wang
- Department of Medicine and Cell, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
| | - Rustem I. Litvinov
- Department of Developmental Biology, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
| | - Alice Zemljic-Harpf
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093
- VA Healthcare San Diego, San Diego, CA 92161
| | - Robert S. Ross
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093
- VA Healthcare San Diego, San Diego, CA 92161
| | - Sanford J. Shattil
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093
| |
Collapse
|
25
|
Skalski M, Sharma N, Williams K, Kruspe A, Coppolino MG. SNARE-mediated membrane traffic is required for focal adhesion kinase signaling and Src-regulated focal adhesion turnover. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2010; 1813:148-58. [PMID: 20888376 DOI: 10.1016/j.bbamcr.2010.09.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2010] [Revised: 09/10/2010] [Accepted: 09/15/2010] [Indexed: 01/27/2023]
Abstract
Integrin signaling is central to cell growth and differentiation, and critical for the processes of apoptosis, cell migration and wound repair. Previous research has demonstrated a requirement for SNARE-dependent membrane traffic in integrin trafficking, as well as cell adhesion and migration. The goal of the present research was to ascertain whether SNARE-dependent membrane trafficking is required specifically for integrin-mediated signaling. Membrane traffic was inhibited in Chinese hamster ovary cells by expression of dominant-negative (E329Q) N-ethylmaleimide-sensitive fusion protein (NSF) or a truncated form of the SNARE SNAP23. Integrin signaling was monitored as cells were plated on fibronectin under serum-free conditions. E329Q-NSF expression inhibited phosphorylation of focal adhesion kinase (FAK) on Tyr397 at early time points of adhesion. Phosphorylation of FAK on Tyr576, Tyr861 and Tyr925 was also impaired by expression of E329Q-NSF or truncated SNAP23, as was trafficking, localization and activation of Src and its interaction with FAK. Decreased FAK-Src interaction coincided with reduced Rac activation, decreased focal adhesion turnover, reduced Akt phosphorylation and lower phosphatidylinositol 3,4,5-trisphosphate levels in the cell periphery. Over-expression of plasma membrane-targeted Src or phosphatidylinositol 3-kinase (PI3K) rescued cell spreading and focal adhesion turnover. The results suggest that SNARE-dependent trafficking is required for integrin signaling through a FAK/Src/PI3K-dependent pathway.
Collapse
Affiliation(s)
- Michael Skalski
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| | | | | | | | | |
Collapse
|
26
|
Lee CS, Choi CK, Shin EY, Schwartz MA, Kim EG. Myosin II directly binds and inhibits Dbl family guanine nucleotide exchange factors: a possible link to Rho family GTPases. ACTA ACUST UNITED AC 2010; 190:663-74. [PMID: 20713598 PMCID: PMC2928003 DOI: 10.1083/jcb.201003057] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The activity of Rho GTPases in migrating cells is regulated by binding of myosin II to GEFs. Cell migration requires the coordinated spatiotemporal regulation of actomyosin contraction and cell protrusion/adhesion. Nonmuscle myosin II (MII) controls Rac1 and Cdc42 activation, and cell protrusion and focal complex formation in migrating cells. However, these mechanisms are poorly understood. Here, we show that MII interacts specifically with multiple Dbl family guanine nucleotide exchange factors (GEFs). Binding is mediated by the conserved tandem Dbl homology–pleckstrin homology module, the catalytic site of these GEFs, with dissociation constants of ∼0.3 µM. Binding to the GEFs required assembly of the MII into filaments and actin-stimulated ATPase activity. Binding of MII suppressed GEF activity. Accordingly, inhibition of MII ATPase activity caused release of GEFs and activation of Rho GTPases. Depletion of βPIX GEF in migrating NIH3T3 fibroblasts suppressed lamellipodial protrusions and focal complex formation induced by MII inhibition. The results elucidate a functional link between MII and Rac1/Cdc42 GTPases, which may regulate protrusion/adhesion dynamics in migrating cells.
Collapse
Affiliation(s)
- Chan-Soo Lee
- Department of Biochemistry and Medical Research Center, Chungbuk National University College of Medicine, Cheongju, South Korea
| | | | | | | | | |
Collapse
|
27
|
Cadherins and Pak1 control contact inhibition of proliferation by Pak1-betaPIX-GIT complex-dependent regulation of cell-matrix signaling. Mol Cell Biol 2010; 30:1971-83. [PMID: 20154149 DOI: 10.1128/mcb.01247-09] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
It is crucial for organ homeostasis that epithelia have effective mechanisms to restrict motility and cell proliferation in order to maintain tissue architecture. On the other hand, epithelial cells need to rapidly and transiently acquire a more mesenchymal phenotype, with high levels of cell motility and proliferation, in order to repair epithelia upon injury. Cross talk between cell-cell and cell-matrix signaling is crucial for regulating these transitions. The Pak1-betaPIX-GIT complex is an effector complex downstream of the small GTPase Rac1. We previously showed that translocation of this complex from cell-matrix to cell-cell adhesion sites was required for the establishment of contact inhibition of proliferation. In this study, we provide evidence that this translocation depends on cadherin function. Cadherins do not recruit the complex by direct interaction. Rather, we found that inhibition of the normal function of cadherin or Pak1 leads to defects in focal adhesion turnover and to increased signaling by phosphatidylinositol 3-kinase. We propose that cadherins are involved in regulation of contact inhibition by controlling the function of the Pak1-betaPIX-GIT complex at focal contacts.
Collapse
|
28
|
Abstract
Integrins are cell surface transmembrane receptors that recognize and bind to extracellular matrix proteins and counter receptors. Binding of activated integrins to their ligands induces a vast number of structural and signaling changes within the cell. Large, multimolecular complexes assemble onto the cytoplasmic tails of activated integrins to engage and organize the cytoskeleton, and activate signaling pathways that ultimately lead to changes in gene expression. Additionally, integrin-mediated signaling intersects with growth factor-mediated signaling through various levels of cross-talk. This review discusses recent work that has tremendously broadened our understanding of the complexity of integrin-mediated signaling.
Collapse
|
29
|
Glycine-rich region regulates cysteine-rich protein 1 binding to actin cytoskeleton. Biochem Biophys Res Commun 2009; 380:484-8. [PMID: 19284992 DOI: 10.1016/j.bbrc.2009.01.125] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2009] [Accepted: 01/16/2009] [Indexed: 11/22/2022]
Abstract
Cysteine-rich protein 1 (CRP1) has a unique structure with two well separated LIM domains, each followed by a glycine-rich region. Although CRP1 has been shown to interact with actin-binding proteins and actin filaments, the mechanism regulating localization to the actin cytoskeleton in cells is not clear. Experiments using truncated forms showed that the first LIM domain and glycine-rich region are necessary for CRP1 bundling of actin filaments and localization to the actin cytoskeleton. Furthermore, domain swapping experiments replacing the first glycine-rich region with the second resulted in the loss of CRP1 bundling activity and localization to the actin cytoskeleton, identifying seven critical amino acid residues. These results highlight the importance of the first glycine-rich region for CRP1 bundling activity and localization to the actin cytoskeleton. In addition, this work identifies the first LIM domain and glycine-rich region as a distinct actin filament bundling module.
Collapse
|
30
|
Sprague CR, Fraley TS, Jang HS, Lal S, Greenwood JA. Phosphoinositide binding to the substrate regulates susceptibility to proteolysis by calpain. J Biol Chem 2008; 283:9217-23. [PMID: 18258589 DOI: 10.1074/jbc.m707436200] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Calpain-mediated proteolysis regulates cytoskeletal dynamics and is altered during aging and the progression of numerous diseases or pathological conditions. Although several cytoskeletal proteins have been identified as substrates, how localized calpain activity is regulated and the mechanisms controlling substrate recognition are not clear. In this study, we report that phosphoinositide binding regulates the susceptibility of the cytoskeletal adhesion protein alpha-actinin to proteolysis by calpains 1 and 2. At first, alpha-actinin did not appear to be a substrate for calpain 2; however, phosphatidylinositol 3,4,5-trisphosphate (PtdIns(3,4,5)P(3)) binding to alpha-actinin resulted in nearly complete proteolysis of the full-length protein, producing stable breakdown products. Calpain 1 was able to cleave alpha-actinin in the absence of phosphoinositide binding; however, PtdIns(3,4,5)P(3) binding increased the rate of proteolysis, and phosphatidylinositol 4,5-diphosphate (PtdIns(4,5)P(2)) binding significantly inhibited cleavage. Phosphoinositide binding appeared to regulate calpain proteolysis of alpha-actinin by modulating the exposure of a highly sensitive cleavage site within the calponin homology 2 domain. In U87MG glioblastoma cells, which contain elevated levels of PtdIns(3,4,5)P(3), alpha-actinin colocalized with calpain within dynamic actin cytoskeletal structures. Furthermore, proteolysis of alpha-actinin producing stable breakdown products was observed in U87MG cells treated with calcium ionophore to activate the calcium-dependent calpains. Additional evidence of PtdIns(3,4,5)P(3)-mediated calpain proteolysis of alpha-actinin was observed in rat embryonic fibroblasts. These results suggest that PtdIns(3,4,5)P(3) binding is a critical determinant for alpha-actinin proteolysis by calpain. In conclusion, phosphoinositide binding to the substrate is a potential mechanism for regulating susceptibility to proteolysis by calpain.
Collapse
Affiliation(s)
- Chelsea R Sprague
- Department of Biochemistry and Biophysics, Oregon State University, Corvallis, OR 97331, USA
| | | | | | | | | |
Collapse
|
31
|
RelA/NF-kappaB transcription factor associates with alpha-actinin-4. Exp Cell Res 2007; 314:1030-8. [PMID: 18215660 DOI: 10.1016/j.yexcr.2007.12.001] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2007] [Revised: 11/13/2007] [Accepted: 12/03/2007] [Indexed: 11/23/2022]
Abstract
The NF-kappaB/RelA family of transcription factors regulates inducible transcription of a large number of genes in response to diverse stimuli. Little is known, however, about the location of NF-kappaB in the cytoplasm and the transport mechanism to the nucleus. We found that NF-kappaB is associated with the actin-binding protein alpha-actinin-4. NF-kappaB and alpha-actinin-4 co-localized along actin stress fibers and in membrane lamellae in A431 cells. After a 30-min stimulation with EGF or TNF-alpha, alpha-actinin-4 and p65 were found in the nucleus. Disruption of cytoskeleton by cytochalasin D prior to treatment with TNF-alpha led to increase of p65 nuclear translocation. Antibodies to p65 subunit of NF-kappaB co-immunoprecipitated alpha-actinin-4 from A431 cell lysates and nuclear extracts, but alpha-actinin-1 and beta-actin were not found in the precipitates. Affinity chromatography experiments displayed that p65 and p50 subunits of NF-kappaB can bind to matrix-bound chicken gizzard alpha-actinin. We suggest that the alpha-actinin-4 is important for the NF-kappaB nuclear translocation and its functions inside the nucleus.
Collapse
|
32
|
Full SJ, Deinzer ML, Ho PS, Greenwood JA. Phosphoinositide binding regulates alpha-actinin CH2 domain structure: analysis by hydrogen/deuterium exchange mass spectrometry. Protein Sci 2007; 16:2597-604. [PMID: 17965186 DOI: 10.1110/ps.073146807] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
alpha-Actinin is an actin bundling protein that regulates cell adhesion by directly linking actin filaments to integrin adhesion receptors. Phosphatidylinositol (4,5)-diphosphate (PtdIns (4,5)-P(2)) and phosphatidylinositol (3,4,5)-triphosphate (PtdIns (3,4,5)-P(3)) bind to the calponin homology 2 domain of alpha-actinin, regulating its interactions with actin filaments and integrin receptors. In this study, we examine the mechanism by which phosphoinositide binding regulates alpha-actinin function using mass spectrometry to monitor hydrogen-deuterium (H/D) exchange within the calponin homology 2 domain. The overall level of H/D exchange for the entire protein showed that PtdIns (3,4,5)-P(3) binding alters the structure of the calponin homology 2 domain increasing deuterium incorporation, whereas PtdIns (4,5)-P(2) induces changes in the structure decreasing deuterium incorporation. Analysis of peptic fragments from the calponin homology 2 domain showed decreased local H/D exchange within the loop region preceding helix F with both phosphoinositides. However, the binding of PtdIns (3,4,5)-P(3) also induced increased exchange within helix E. This suggests that the phosphate groups on the fourth and fifth position of the inositol head group of the phosphoinositides constrict the calponin homology 2 domain, thereby altering the orientation of actin binding sequence 3 and decreasing the affinity of alpha-actinin for filamentous actin. In contrast, the phosphate group on the third position of the inositol head group of PtdIns (3,4,5)-P(3) perturbs the calponin homology 2 domain, altering the interaction between the N and C terminus of the full-length alpha-actinin antiparallel homodimer, thereby disrupting bundling activity and interaction with integrin receptors.
Collapse
Affiliation(s)
- Stephen J Full
- Department of Biochemistry and Biophysics, Oregon State University, Corvallis, Oregon 97331, USA
| | | | | | | |
Collapse
|
33
|
Harper L, Kashiwagi Y, Pusey CD, Hendry BM, Domin J. Platelet-derived growth factor reorganizes the actin cytoskeleton through 3-phosphoinositide-dependent and 3-phosphoinositide-independent mechanisms in human mesangial cells. Nephron Clin Pract 2007; 107:p45-56. [PMID: 17804914 DOI: 10.1159/000107805] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2006] [Accepted: 05/08/2007] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Platelet-derived growth factor (PDGF) is a potent activator of mesangial cell proliferation and migration. Although phosphoinositide 3-kinase (PI3K) enzymes are important downstream targets of the PDGF receptor, the contribution made by their 3-phosphoinositide products in the reorganization of actin cytoskeleton and focal adhesions has been questioned. METHODS AND RESULTS Pharmacological inhibition of the PI3K activity blocks PDGF-induced migration of human primary mesangial cells using an in vitro scrape wound healing assay. Acute (<10 min) inhibition of the PI3K activity did not alter the effect of PDGF on either stress fibre dissolution or reorganization of focal adhesions. However, at later times (>30 min), PDGF-stimulated responses were inhibited. In contrast, PDGF-stimulated membrane ruffling remained insensitive to PI3K inhibitors throughout. Inhibition of protein kinase C and Erk also attenuated PDGF-stimulated mesangial cell migration; however, neither signaling pathway was responsible for the initial effects on filamentous actin and focal adhesions. CONCLUSIONS We propose that following PDGF stimulation of mesangial cells, reorganization of the actin cytoskeleton occurs in a biphasic manner. The mechanism responsible for mesangial cell migration that occurs immediately following PDGF stimulation may serve to 'prime' for the subsequent 3-phosphoinositide-, protein-kinase-C-, and Erk-dependent migration.
Collapse
|
34
|
Cantiello HF, Montalbetti N, Li Q, Chen XZ. The Cytoskeletal Connection to Ion Channels as a Potential Mechanosensory Mechanism: Lessons from Polycystin-2 (TRPP2). CURRENT TOPICS IN MEMBRANES 2007; 59:233-96. [PMID: 25168140 DOI: 10.1016/s1063-5823(06)59010-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Mechanosensitivity of ion channels, or the ability to transfer mechanical forces into a gating mechanism of channel regulation, is split into two main working (not mutually exclusive) hypotheses. One is that elastic and/or structural changes in membrane properties act as a transducing mechanism of channel regulation. The other hypothesis involves tertiary elements, such as the cytoskeleton which, itself by dynamic interactions with the ion channel, may convey conformational changes, including those ascribed to mechanical forces. This hypothesis is supported by numerous instances of regulatory changes in channel behavior by alterations in cytoskeletal structures/interactions. However, only recently, the molecular nature of these interactions has slowly emerged. Recently, a surge of evidence has emerged to indicate that transient receptor potential (TRP) channels are key elements in the transduction of a variety of environmental signals. This chapter describes the molecular linkage and regulatory elements of polycystin-2 (PC2), a TRP-type (TRPP2) nonselective cation channel whose mutations cause autosomal dominant polycystic kidney disease (ADPKD). The chapter focuses on the involvement of cytoskeletal structures in the regulation of PC2 and discusses how these connections are the transducing mechanism of environmental signals to its channel function.
Collapse
Affiliation(s)
- Horacio F Cantiello
- Renal Unit, Massachusetts General Hospital East, Charlestown, Massachusetts 02129; Department of Medicine, Harvard Medical School, Boston, Massachusetts 02115; Laboratorio de Canales Iónicos, Departamento de Fisicoquímica y Química Analítica, Facultad de Farmacia y Bioquímica, Buenos Aires 1113, Argentina
| | - Nicolás Montalbetti
- Laboratorio de Canales Iónicos, Departamento de Fisicoquímica y Química Analítica, Facultad de Farmacia y Bioquímica, Buenos Aires 1113, Argentina
| | - Qiang Li
- Department of Physiology, University of Alberta, Edmonton T6G 2H7, Canada
| | - Xing-Zhen Chen
- Department of Physiology, University of Alberta, Edmonton T6G 2H7, Canada
| |
Collapse
|
35
|
Araki N, Egami Y, Watanabe Y, Hatae T. Phosphoinositide metabolism during membrane ruffling and macropinosome formation in EGF-stimulated A431 cells. Exp Cell Res 2007; 313:1496-507. [PMID: 17368443 DOI: 10.1016/j.yexcr.2007.02.012] [Citation(s) in RCA: 98] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2006] [Revised: 01/30/2007] [Accepted: 02/13/2007] [Indexed: 11/30/2022]
Abstract
Inhibitors of phosphoinositide 3-kinase (PI3K) were found to perturb macropinosome formation without affecting the membrane ruffling and actin polymerization in epidermal growth factor-stimulated A431 cells. Live-cell imaging and quantitative image analysis of the fluorescence intensity ratio of the YFP-tagged phospholipase Cdelta1-pleckstrin homology domain (YFP-PLC-PH) relative to membrane-targeted CFP (CFP-Mem) demonstrated that the concentration of PI(4,5)P(2) in the membrane ruffles forming macropinocytic cups increased to more than double that in planar plasma membranes. The PI(4,5)P(2) level in the membrane reached its maximum just before macropinosome closure and rapidly fell as the macropinocytic cups closed. In contrast, the PI(3,4,5)P(3) concentrations visualized based on the YFP-Akt-PH or YFP-Bruton's tyrosine kinase (Btk)-PH/CFP-Mem ratio increased locally at the site of macropinosome formation and peaked at the time of macropinosome closure. The kinetics of PI(4,5)P(2) and PI(3,4,5)P(3) appeared to be mechanistically linked to actin remodeling during macropinocytosis. From the pharmacological data using inhibitors and synthetic phosphoinositides and other data, it could be concluded that both PI(4,5)P(2) elimination and PI(3,4,5)P(3) production by PI3K might be crucial for macropinosome formation from membrane ruffles. This study emphasizes that locally controlled levels of phosphoinositides are important for regulating the function of actin-binding proteins which effect changes in the membrane architecture.
Collapse
Affiliation(s)
- Nobukazu Araki
- Department of Histology and Cell Biology, School of Medicine, Kagawa University, Miki, Kagawa 761-0793, Japan.
| | | | | | | |
Collapse
|
36
|
Guvakova MA. Insulin-like growth factors control cell migration in health and disease. Int J Biochem Cell Biol 2007; 39:890-909. [PMID: 17113337 DOI: 10.1016/j.biocel.2006.10.013] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2006] [Revised: 10/13/2006] [Accepted: 10/19/2006] [Indexed: 12/30/2022]
Abstract
Insulin-like growth factors I and II (IGF-I and IGF-II) have an ancient origin and play essential roles in fundamental biological processes. Although IGFs are principally known for their roles in regulating cell growth and survival, their ability to influence cell motility is just as significant. In the past 20 years, research has provided indisputable evidence for the regulatory role of IGFs in the migration of various cell types. Cell migration is crucial for reproduction, development, and tissue regeneration; IGFs play an important role in coordinating these processes. Moreover, studies continue to uncover the IGFs' role in stimulating cancer cell migration, invasion and metastasis. This review surveys current knowledge on the cell migration-modulating properties of IGFs and the biochemical pathways by which these peptides regulate cell movement in both physiological and pathological conditions.
Collapse
Affiliation(s)
- Marina A Guvakova
- Division of Endocrine and Oncologic Surgery, Department of Surgery, School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
37
|
Wahlström G, Norokorpi HL, Heino TI. Drosophila alpha-actinin in ovarian follicle cells is regulated by EGFR and Dpp signalling and required for cytoskeletal remodelling. Mech Dev 2006; 123:801-18. [PMID: 17008069 DOI: 10.1016/j.mod.2006.08.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2005] [Revised: 08/04/2006] [Accepted: 08/16/2006] [Indexed: 01/09/2023]
Abstract
alpha-Actinin is an evolutionarily conserved actin filament crosslinking protein with functions in both muscle and non-muscle cells. In non-muscle cells, interactions between alpha-actinin and its many binding partners regulate cell adhesion and motility. In Drosophila, one non-muscle and two muscle-specific alpha-actinin isoforms are produced by alternative splicing of a single gene. In wild-type ovaries, alpha-actinin is ubiquitously expressed. The non-muscle alpha-actinin mutant Actn(Delta233), which is viable and fertile, lacks alpha-actinin expression in ovarian germline cells, while somatic follicle cells express alpha-actinin at late oogenesis. Here we show that this latter population of alpha-actinin, termed FC-alpha-actinin, is absent from the dorsoanterior follicle cells, and we present evidence that this is the result of a negative regulation by combined Epidermal growth factor receptor (EGFR) and Decapentaplegic signalling. Furthermore, EGFR signalling increased the F-actin bundling activity of ectopically expressed muscle-specific alpha-actinin. We also describe a novel morphogenetic event in the follicle cells that occurs during egg elongation. This event involves a transient repolarisation of the basal actin fibres and the assembly of a posterior beta-integrin-dependent adhesion site accumulating alpha-actinin and Enabled. Clonal analysis using Actn null alleles demonstrated that although alpha-actinin was not necessary for actin fibre formation or maintenance, the cytoskeletal remodelling was perturbed, and Enabled did not localise in the posterior adhesion site. Nevertheless, epithelial morphogenesis proceeded normally. This work provides the first evidence that alpha-actinin is involved in the organisation of the cytoskeleton in a non-muscle tissue in Drosophila.
Collapse
Affiliation(s)
- Gudrun Wahlström
- Developmental Biology Programme/Institute of Biotechnology, Viikki Biocenter, P.O. Box 56 (Viikinkaari 9), FIN-00014, University of Helsinki, Finland.
| | | | | |
Collapse
|
38
|
Zhang W, Gunst SJ. Dynamic association between alpha-actinin and beta-integrin regulates contraction of canine tracheal smooth muscle. J Physiol 2006; 572:659-76. [PMID: 16513669 PMCID: PMC1780001 DOI: 10.1113/jphysiol.2006.106518] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
The adhesion junctions of smooth muscle cells may be dynamically regulated during smooth muscle contraction, and this dynamic regulation may be important for the development of active tension. In the present study, the role of alpha-actinin during smooth muscle contraction was evaluated in tracheal smooth muscle tissues and freshly dissociated cells. Stimulation with acetylcholine (ACh) increased the localization of alpha-actinin at the membrane of freshly dissociated smooth muscle cells, and increased the amount of beta1 integrin that coprecipitated with alpha-actinin from muscle tissue homogenates. GFP-alpha-actinin fusion proteins were expressed in muscle tissues and visualized in live freshly dissociated cells. GFP-alpha-actinin translocated to the membrane within seconds of stimulation of the cells with ACh. Expression of the integrin-binding rod domain of alpha-actinin in smooth muscle tissues depressed active contraction in response to ACh. Expression of the alpha-actinin rod domain also inhibited the translocation of endogenous alpha-actinin to the membrane, and inhibited the association of endogenous alpha-actinin with beta1-integrin in alpha-actinin immunoprecipitates from tissue extracts. However, the expression of alpha-actinin rod domain peptides did not inhibit increases in myosin light chain phosphorylation or actin polymerization in response to stimulation with ACh. Results suggest that contractile stimulation of smooth muscle causes the rapid recruitment of alpha-actinin to beta-integrin complexes at the membrane, and that the recruitment of alpha-actinin to integrin complexes is necessary for active tension development in smooth muscle.
Collapse
Affiliation(s)
- Wenwu Zhang
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, 635 Barnhill Drive, Indianapolis, IN 46202, USA
| | | |
Collapse
|
39
|
Ziegler WH, Liddington RC, Critchley DR. The structure and regulation of vinculin. Trends Cell Biol 2006; 16:453-60. [PMID: 16893648 DOI: 10.1016/j.tcb.2006.07.004] [Citation(s) in RCA: 353] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2006] [Revised: 06/25/2006] [Accepted: 07/24/2006] [Indexed: 01/09/2023]
Abstract
Vinculin is a ubiquitously expressed actin-binding protein frequently used as a marker for both cell-cell and cell-extracellular matrix (focal adhesion) adherens-type junctions, but its function has remained elusive. Vinculin is made up of a globular head linked to a tail domain by a short proline-rich sequence, and an intramolecular interaction between the head and tail masks the numerous ligand-binding sites in the protein. Determination of the crystal structure of vinculin has shed new light on the way that these ligand-binding sites are regulated. The picture that emerges is one in which vinculin stabilizes focal adhesions and thereby suppresses cell migration, an effect that is relieved by transient changes in the local concentrations of inositol phospholipids. However, the finding that vinculin modulates the signalling pathways involved in apoptosis suggests that additional roles for vinculin remain to be discovered.
Collapse
Affiliation(s)
- Wolfgang H Ziegler
- IZKF Leipzig, Faculty of Medicine, University of Leipzig, 04103 Leipzig, Germany
| | | | | |
Collapse
|
40
|
Friedrich C, Endlich N, Kriz W, Endlich K. Podocytes are sensitive to fluid shear stress in vitro. Am J Physiol Renal Physiol 2006; 291:F856-65. [PMID: 16684926 DOI: 10.1152/ajprenal.00196.2005] [Citation(s) in RCA: 109] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Podocytes are exposed to mechanical forces arising from glomerular capillary pressure and filtration. It has been shown that stretch affects podocyte biology in vitro and plays a significant role in the development of glomerulosclerosis in vivo. However, whether podocytes are sensitive to fluid shear stress is completely unknown. In the present study, we therefore exposed cells of a recently generated conditionally immortalized mouse podocyte cell line to defined fluid shear stress in a flow chamber, mimicking flow of the glomerular ultrafiltrate over the surface of podocytes in Bowman's space. Shear stress above 0.25 dyne/cm(2) resulted in dramatic loss of podocytes but not of proximal tubular epithelial cells (LLC-PK(1) cells) after 20 h. At 0.015-0.25 dyne/cm(2), lamellipodia formation in podocytes was enhanced and the actin nucleation protein cortactin was redistributed to the cell margins. Shear stress further diminished stress fibers and the presence of vinculin in focal adhesions. Linear zonula occludens-1 distribution at cell-cell contacts remained unaffected at low shear stress. At 0.25 dyne/cm(2), the monolayer was broken up and remaining cell-cell contacts were reinforced by F-actin and alpha-actinin. Because the cytoskeletal changes induced by shear stress suggested the involvement of tyrosine kinases (TKs), we tested several TK inhibitors that were all without effect on podocyte number under static conditions. At 0.25 dyne/cm(2), however, the TK inhibitors genistein and AG 82 were associated with marked podocyte loss. Our data demonstrate that podocytes are highly sensitive to fluid shear stress. Shear stress induces a reorganization of the actin cytoskeleton and activates specific tyrosine kinases that are required to withstand fluid shear stress.
Collapse
Affiliation(s)
- Colin Friedrich
- Department of Anatomy and Cell Biology I, University of Heidelberg, Heidelberg, Germany
| | | | | | | |
Collapse
|
41
|
Saunders RM, Holt MR, Jennings L, Sutton DH, Barsukov IL, Bobkov A, Liddington RC, Adamson EA, Dunn GA, Critchley DR. Role of vinculin in regulating focal adhesion turnover. Eur J Cell Biol 2006; 85:487-500. [PMID: 16584805 DOI: 10.1016/j.ejcb.2006.01.014] [Citation(s) in RCA: 146] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2005] [Revised: 12/30/2005] [Accepted: 01/02/2006] [Indexed: 11/15/2022] Open
Abstract
Although vinculin (-/-) mouse embryo fibroblasts assemble focal adhesions (FAs), they spread more slowly, less extensively, and close a wound more rapidly than vinculin (+/+) cells. To investigate the structure and dynamics of FAs in these cells, we used real-time interference reflection microscopy (IRM) thus avoiding the need to express exogenous GFP-tagged FA proteins which may be misregulated. This showed that the FAs were smaller, less abundant and turned over more rapidly in vinculin null compared to wild-type cells. Expression of vinculin rescued the spreading defect and resulted in larger and more stable FAs. Phosphatidylinositol 4,5-bisphosphate (PIP2) is thought to play a role in vinculin activation by relieving an intramolecular association between the vinculin head (Vh) and tail (Vt) that masks the ligand binding sites in Vh and Vt. To investigate the role of the vinculin/PIP2 interaction in FA dynamics, we used a vinculin mutant lacking the C-terminal arm (residues 1053-1066) and referred to as the deltaC mutation. This mutation reduced PIP2 binding to a Vt deltaC polypeptide by >90% compared to wild type without affecting binding to Vh or F-actin. Interestingly, cells expressing the vinculin deltaC mutant assembled remarkably stable FAs. The results suggest that vinculin inhibits cell migration by stabilising FAs, and that binding of inositol phospholipids to Vt plays an important role in FA turnover.
Collapse
Affiliation(s)
- Ruth M Saunders
- Department of Biochemistry, University of Leicester, University Road, Leicester LE1 7RH, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Foster LJ, Rudich A, Talior I, Patel N, Huang X, Furtado LM, Bilan PJ, Mann M, Klip A. Insulin-dependent interactions of proteins with GLUT4 revealed through stable isotope labeling by amino acids in cell culture (SILAC). J Proteome Res 2006; 5:64-75. [PMID: 16396496 DOI: 10.1021/pr0502626] [Citation(s) in RCA: 92] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The insulin-regulated glucose transporter (GLUT4) translocates to the plasma membrane in response to insulin in order to facilitate the postprandial uptake of glucose into fat and muscle cells. While early insulin receptor signaling steps leading to this translocation are well defined, the integration of signaling and regulation of GLUT4 traffic remains elusive. Several lines of evidence suggest an important role for the actin cytoskeleton and for protein-protein interactions in regulating GLUT4 localization by insulin. Here, we applied stable isotope labeling by amino acids in cell culture (SILAC) to identify proteins that interact with GLUT4 in an insulin-regulated manner. Myc-tagged GLUT4 (GLUT4myc) stably expressed in L6 myotubes was immunoprecipitated via the myc epitope from total membranes isolated from basal and insulin-stimulated cells grown in medium containing normal isotopic abundance leucine or deuterated leucine, respectively. Proteins coprecipitating with GLUT4myc were analyzed by liquid chromatography/ tandem mass spectrometry. Of 603 proteins quantified, 36 displayed an insulin-dependent change of their interaction with GLUT4myc of more than 1.5-fold in either direction. Several cytoskeleton-related proteins were elevated in immunoprecipates from insulin-treated cells, whereas components of the ubiquitin-proteasome degradation system were generally reduced. Proteins participating in vesicle traffic also displayed insulin-regulated association. Of cytoskeleton-related proteins, alpha-actinin-4 recovery in GLUT4 immunoprecipitates rose in response to insulin 2.1 +/- 0.5-fold by SILAC and 2.9 +/- 0.8-fold by immunoblotting. Insulin caused GLUT4 and alpha-actinin-4 co-localization as revealed by confocal immunofluorescence microscopy. We conclude that insulin elicits changes in interactions between diverse proteins and GLUT4, and that cytoskeletal proteins, notably alpha-actinin-4, associate with the transporter, potentially to facilitate its routing to the plasma membrane.
Collapse
Affiliation(s)
- Leonard J Foster
- Center for Experimental BioInformatics (CEBI), Department of Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, DK-5230 Odense M, Denmark
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Abstract
Phosphoinositide phosphates (PIPs) correspond to phosphorylated derivatives of phosphatidylinositol (PI). Despite their relatively low abundance in the plasma membrane, PIPs play a crucial role as precursors of second messengers and are themselves important signaling and targeting molecules. Indeed, modulation of levels of PIPs affects, for example, cortical actin organization, membrane dynamics, and cell migration. The focus of this review is on selected interesting targets of PIPs. Those proteins that bind PIPs and are involved in regulation of actin assembly, actin membrane linkage, and actin contractility are discussed, as well as those that are involved in signaling, such as small GTPases, protein kinases, and phosphatases, or in regulation of membrane dynamics.
Collapse
Affiliation(s)
- Verena Niggli
- Department of Pathology, University of Bern, CH-3010 Bern, Switzerland.
| |
Collapse
|
44
|
Tran TC, Singleton C, Fraley TS, Greenwood JA. Cysteine-rich protein 1 (CRP1) regulates actin filament bundling. BMC Cell Biol 2005; 6:45. [PMID: 16336664 PMCID: PMC1318456 DOI: 10.1186/1471-2121-6-45] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2005] [Accepted: 12/08/2005] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Cysteine-rich protein 1 (CRP1) is a LIM domain containing protein localized to the nucleus and the actin cytoskeleton. CRP1 has been demonstrated to bind the actin-bundling protein alpha-actinin and proposed to modulate the actin cytoskeleton; however, specific regulatory mechanisms have not been identified. RESULTS CRP1 expression increased actin bundling in rat embryonic fibroblasts. Although CRP1 did not affect the bundling activity of alpha-actinin, CRP1 was found to stabilize the interaction of alpha-actinin with actin bundles and to directly bundle actin microfilaments. Using confocal and photobleaching fluorescence resonance energy transfer (FRET) microscopy, we demonstrate that there are two populations of CRP1 localized along actin stress fibers, one associated through interaction with alpha-actinin and one that appears to bind the actin filaments directly. Consistent with a role in regulating actin filament cross-linking, CRP1 also localized to the membrane ruffles of spreading and PDGF treated fibroblasts. CONCLUSION CRP1 regulates actin filament bundling by directly cross-linking actin filaments and stabilizing the interaction of alpha-actinin with actin filament bundles.
Collapse
Affiliation(s)
- Thuan C Tran
- Department of Biochemistry and Biophysics, Oregon State University, Corvallis, Oregon 97331, USA
| | - CoreyAyne Singleton
- Department of Biochemistry and Biophysics, Oregon State University, Corvallis, Oregon 97331, USA
| | - Tamara S Fraley
- Department of Biochemistry and Biophysics, Oregon State University, Corvallis, Oregon 97331, USA
| | - Jeffrey A Greenwood
- Department of Biochemistry and Biophysics, Oregon State University, Corvallis, Oregon 97331, USA
| |
Collapse
|
45
|
Abstract
Alpha-actinin and vinculin orchestrate reorganization of the actin cytoskeleton following the formation of adhesion junctions. alpha-Actinin interacts with vinculin through the binding of an alpha-helix (alphaVBS) present within the R4 spectrin repeat of its central rod domain to vinculin's N-terminal seven-helical bundle domain (Vh1). The Vh1:alphaVBS structure suggests that alphaVBS first unravels from its buried location in the triple-helical R4 repeat to allow it to bind to vinculin. alphaVBS binding then induces novel conformational changes in the N-terminal helical bundle of Vh1, which disrupt its intramolecular association with vinculin's tail domain and which differ from the alterations in Vh1 provoked by the binding of talin. Surprisingly, alphaVBS binds to Vh1 in an inverted orientation compared to the binding of talin's VBSs to vinculin. Importantly, the binding of alphaVBS and talin's VBSs to vinculin's Vh1 domain appear to also trigger distinct conformational changes in full-length vinculin, opening up distant regions that are buried in the inactive molecule. The data suggest a model where vinculin's Vh1 domain acts as a molecular switch that undergoes distinct structural changes provoked by talin and alpha-actinin binding in focal adhesions versus adherens junctions, respectively.
Collapse
Affiliation(s)
- Philippe R J Bois
- Department of Hematology-Oncology, St. Jude Children's Research Hospital, 332 North Lauderdale Street, Memphis, Tennessee 38105, USA
| | | | | | | |
Collapse
|
46
|
Zhang J, Mruk DD, Cheng CY. Myotubularin phosphoinositide phosphatases, protein phosphatases, and kinases: their roles in junction dynamics and spermatogenesis. J Cell Physiol 2005; 204:470-83. [PMID: 15690393 DOI: 10.1002/jcp.20303] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Spermatogenesis in the seminiferous epithelium of the mammalian testis is a dynamic cellular event. It involves extensive restructuring at the Sertoli-germ cell interface, permitting germ cells to traverse the epithelium from basal to adluminal compartment. As such, Sertoli-germ cell actin-based adherens junctions (AJ), such as ectoplasmic specializations (ES), must disassemble and reassemble to facilitate this event. Recent studies have shown that AJ dynamics are regulated by intricate interactions between AJ integral membrane proteins (e.g., cadherins, alpha6beta1 integrins and nectins), phosphatases, kinases, adaptors, and the underlying cytoskeleton network. For instance, the myotubularin (MTM) phosphoinositide (PI) phosphatases, such as MTM related protein 2 (MTMR2), can form a functional complex with c-Src (a non-receptor protein tyrosine kinase). In turn, this phosphatase/kinase complex associates with beta-catenin, a constituent of the N-cadherin/beta-catenin functional unit at the AJ site. This MTMR2-c-Src-beta-catenin complex apparently regulates the phosphorylation status of beta-catenin, which determines cell adhesive function conferred by the cadherin-catenin protein complex in the seminiferous epithelium. In this review, we discuss the current status of research on selected phosphatases and kinases, and how these proteins potentially interact with adaptors at AJ in the seminiferous epithelium to regulate cell adhesion in the testis. Specific research areas that are open for further investigation are also highlighted.
Collapse
Affiliation(s)
- Jiayi Zhang
- Population Council, Center for Biomedical Research, New York, New York, USA
| | | | | |
Collapse
|
47
|
Abstract
Trypanosoma cruzi, the protozoan parasite that causes Chagas' disease in humans, is capable of invading and replicating within a wide variety of nucleated mammalian cell types. Host cell invasion by infective T. cruzi trypomastigotes is governed by parasite-triggered activation of host cell signaling pathways. Recent studies highlighting a role for host cell phosphatidylinositol 3-kinases (PI3Ks) in the T. cruzi invasion process have revealed surprising new insights into the mechanism of host cell invasion by this pathogen. In this Perspective, we discuss these findings and propose alternative models of T. cruzi invasion that incorporate this new information.
Collapse
Affiliation(s)
- Barbara A Burleigh
- Department of Immunology and Infectious Diseases, Harvard School of Public Health, 665 Huntington Avenue, Building I, Room 817, Boston, MA 02115, USA.
| |
Collapse
|
48
|
Gallant ND, Michael KE, García AJ. Cell adhesion strengthening: contributions of adhesive area, integrin binding, and focal adhesion assembly. Mol Biol Cell 2005; 16:4329-40. [PMID: 16000373 PMCID: PMC1196341 DOI: 10.1091/mbc.e05-02-0170] [Citation(s) in RCA: 295] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Mechanical interactions between a cell and its environment regulate migration, contractility, gene expression, and cell fate. We integrated micropatterned substrates to engineer adhesive area and a hydrodynamic assay to analyze fibroblast adhesion strengthening on fibronectin. Independently of cell spreading, integrin binding and focal adhesion assembly resulted in rapid sevenfold increases in adhesion strength to steady-state levels. Adhesive area strongly modulated adhesion strength, integrin binding, and vinculin and talin recruitment, exhibiting linear increases for small areas. However, above a threshold area, adhesion strength and focal adhesion assembly reached a saturation limit, whereas integrin binding transitioned from a uniform distribution to discrete complexes. Adhesion strength exhibited exponential increases with bound integrin numbers as well as vinculin and talin recruitment, and the relationship between adhesion strength and these biochemical events was accurately described by a simple mechanical model. Furthermore, adhesion strength was regulated by the position of an adhesive patch, comprised of bound integrins and cytoskeletal elements, which generated a constant 200-nN adhesive force. Unexpectedly, focal adhesion assembly, in particular vinculin recruitment, contributed only 30% of the adhesion strength. This work elucidates the roles of adhesive complex size and position in the generation of cell-extracellular matrix forces.
Collapse
Affiliation(s)
- Nathan D Gallant
- Woodruff School of Mechanical Engineering, Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332-0363, USA
| | | | | |
Collapse
|
49
|
Burnham CAD, Shokoples SE, Tyrrell GJ. Phosphoglycerate kinase inhibits epithelial cell invasion by group B streptococci. Microb Pathog 2005; 38:189-200. [PMID: 15925270 DOI: 10.1016/j.micpath.2005.02.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2004] [Revised: 01/20/2005] [Accepted: 02/02/2005] [Indexed: 10/25/2022]
Abstract
Group B streptococci (GBS) are opportunistic human pathogens that cause infection and invasive disease in newborns, pregnant women and non-pregnant adults. The internalization of GBS into eukaryotic cells occurs in an actin-microfilament dependent process. The objective of our study was to understand what host cell and/or bacterial factors may be involved in this process. We focused on alpha-actinin, an actin binding protein closely associated with cytoplasmic F-actin in the eukaryotic cell, to determine if it is involved in actin recruitment upon GBS internalization. Initial work revealed that GBS does not recruit alpha-actinin. However, it was found that alpha-actinin antibodies bound to the surface of the GBS, suggesting GBS possess surface-exposed actin binding protein(s). Slide agglutination experiments revealed that when the bacteria were emulsified with F-actin, visible agglutination occurred, further suggesting the presence of an actin binding protein on the GBS cell. Western blot analysis found that anti-alpha-actinin antibodies bound to a 42 kDa protein; mass spectra analysis identified this protein as GBS phosphoglycerate kinase (PGK). Competitive binding assays suggest that the PGK-actin interaction is not a factor in the initial binding of GBS to epithelial cells, however, treating epithelial cells with PGK prior to performing an invasion assay inhibited GBS internalization. This occurred in a dose dependent manner with 10 microg/mL of PGK inhibiting invasion by over 70%, and 50 microg/mL PGK inhibits GBS invasion completely.
Collapse
Affiliation(s)
- Carey-Ann D Burnham
- The Department of Laboratory Medicine and Pathology, The University of Alberta, Edmonton, Alberta, Canada
| | | | | |
Collapse
|
50
|
Li Q, Montalbetti N, Shen PY, Dai XQ, Cheeseman CI, Karpinski E, Wu G, Cantiello HF, Chen XZ. Alpha-actinin associates with polycystin-2 and regulates its channel activity. Hum Mol Genet 2005; 14:1587-603. [PMID: 15843396 DOI: 10.1093/hmg/ddi167] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Polycystin-2 (PC2) is the product of the PKD2 gene, which is mutated in 10-15% patients of autosomal dominant polycystic kidney disease (ADPKD). PC2 is an integral transmembrane protein and acts as a calcium-permeable cation channel. The functional modulation of this channel by other protein partners remains largely unknown. In the present study, using a yeast two-hybrid approach, we discovered that both intracellular N- and C-termini of PC2 associate with alpha-actinins, actin-binding and actin-bundling proteins important in cytoskeleton organization, cell adhesion, proliferation and migration. The PC2-alpha-actinin association was confirmed by in vitro glutathione S-transferase pull-down and dot blot overlay assays. In addition, the in vivo interaction between endogenous PC2 and alpha-actinins was demonstrated by co-immunoprecipitation in human embryonic kidney 293 and Madin-Darby canine kidney (MDCK) cells, rat kidney and heart tissues and human syncytiotrophoblast (hST) apical membrane vesicles. Immunofluorescence experiments showed that PC2 and alpha-actinin were partially co-localized in epithelial MDCK and inner medullary collecting duct cells, NIH 3T3 fibroblasts and hST vesicles. We studied the functional modulation of PC2 by alpha-actinin in a lipid bilayer electrophysiology system using in vitro translated PC2 and found that alpha-actinin substantially stimulated the channel activity of reconstituted PC2. A similar stimulatory effect of alpha-actinin on PC2 was also observed when hST vesicles were reconstituted in lipid bilayer. Thus, physical and functional interactions between PC2 and alpha-actinin may play an important role in abnormal cell adhesion, proliferation and migration observed in ADPKD.
Collapse
Affiliation(s)
- Qiang Li
- Membrane Protein Research Group, Department of Physiology, University of Alberta, Edmonton, Alberta, T6G 2H7 Canada
| | | | | | | | | | | | | | | | | |
Collapse
|