1
|
Lambert MW. Critical role of alpha spectrin in DNA repair: the importance of μ-calpain and Fanconi anemia proteins. Exp Biol Med (Maywood) 2025; 250:10537. [PMID: 40375875 PMCID: PMC12078185 DOI: 10.3389/ebm.2025.10537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Accepted: 04/17/2025] [Indexed: 05/18/2025] Open
Abstract
Nonerythroid spectrins are proteins important in maintaining the structural integrity and flexibility of the cell and nuclear membranes and are essential for a number of functionally important cellular processes. One of these proteins, nonerythroid α spectrin (αSpII), plays a critical role in DNA repair, specifically repair of DNA interstrand crosslinks (ICLs), where it acts as a scaffold, recruiting repair proteins to sites of damage. Loss or breakdown of αSpII is an important factor in a number of disorders. One of these is Fanconi anemia (FA), a genetic disorder characterized by bone marrow failure, chromosome instability, cancer predisposition, congenital abnormalities and a defect in DNA ICL repair. Significantly, breakdown of αSpII occurs in cells from a number of FA complementation groups, due to excessive cleavage by the protease, μ-calpain, leading to defective repair of DNA ICLs in telomeric and non-telomeric DNA. Knockdown of μ-calpain in FA cells by μ-calpain siRNA results in restoration of αSpII levels to normal and repair of DNA ICLs in telomeric and non-telomeric DNA, demonstrating the importance of αSpII stability in the repair process. It is hypothesized that there is a mechanistic link between excessive cleavage of αSpII by μ-calpain and defective DNA ICL repair in FA and that FA proteins, which are deficient in FA, play a key role in maintaining the stability of αSpII and preventing its cleavage by μ-calpain. All of these events are proposed to be important key factors involved in the pathophysiology of FA and suggest new avenues for potential therapeutic intervention.
Collapse
|
2
|
Schmidt N, Kumar A, Korf L, Dinh-Fricke AV, Abendroth F, Koide A, Linne U, Rakwalska-Bange M, Koide S, Essen LO, Vázquez O, Hantschel O. Development of mirror-image monobodies targeting the oncogenic BCR::ABL1 kinase. Nat Commun 2024; 15:10724. [PMID: 39715735 PMCID: PMC11666773 DOI: 10.1038/s41467-024-54901-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 10/28/2024] [Indexed: 12/25/2024] Open
Abstract
Mirror-image proteins, composed of D-amino acids, are an attractive therapeutic modality, as they exhibit high metabolic stability and lack immunogenicity. Development of mirror-image binding proteins is achieved through chemical synthesis of D-target proteins, phage display library selection of L-binders and chemical synthesis of (mirror-image) D-binders that consequently bind the physiological L-targets. Monobodies are well-established synthetic (L-)binding proteins and their small size (~90 residues) and lack of endogenous cysteine residues make them particularly accessible to chemical synthesis. Here, we develop monobodies with nanomolar binding affinities against the D-SH2 domain of the leukemic tyrosine kinase BCR::ABL1. Two crystal structures of heterochiral monobody-SH2 complexes reveal targeting of the pY binding pocket by an unconventional binding mode. We then prepare potent D-monobodies by either ligating two chemically synthesized D-peptides or by self-assembly without ligation. Their proper folding and stability are determined and high-affinity binding to the L-target is shown. D-monobodies are protease-resistant, show long-term plasma stability, inhibit BCR::ABL1 kinase activity and bind BCR::ABL1 in cell lysates and permeabilized cells. Hence, we demonstrate that functional D-monobodies can be developed readily. Our work represents an important step towards possible future therapeutic use of D-monobodies when combined with emerging methods to enable cytoplasmic delivery of monobodies.
Collapse
Affiliation(s)
- Nina Schmidt
- Institute of Physiological Chemistry, Faculty of Medicine, Philipps University of Marburg, Marburg, Germany
| | - Amit Kumar
- Institute of Physiological Chemistry, Faculty of Medicine, Philipps University of Marburg, Marburg, Germany
| | - Lukas Korf
- Faculty of Chemistry and Unit for Structural Biology, Philipps University of Marburg, Marburg, Germany
| | | | - Frank Abendroth
- Faculty of Chemistry and Unit for Chemical Biology, Philipps University of Marburg, Marburg, Germany
| | - Akiko Koide
- Department of Medicine, New York University School of Medicine, New York, NY, USA
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY, USA
| | - Uwe Linne
- Faculty of Chemistry and Unit for Mass Spectrometry, Philipps University of Marburg, Marburg, Germany
| | - Magdalena Rakwalska-Bange
- Institute of Physiological Chemistry, Faculty of Medicine, Philipps University of Marburg, Marburg, Germany
| | - Shohei Koide
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY, USA
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY, USA
| | - Lars-Oliver Essen
- Faculty of Chemistry and Unit for Structural Biology, Philipps University of Marburg, Marburg, Germany
| | - Olalla Vázquez
- Faculty of Chemistry and Unit for Chemical Biology, Philipps University of Marburg, Marburg, Germany.
- Center for Synthetic Microbiology (SYNMIKRO), Philipps University of Marburg, Marburg, Germany.
| | - Oliver Hantschel
- Institute of Physiological Chemistry, Faculty of Medicine, Philipps University of Marburg, Marburg, Germany.
| |
Collapse
|
3
|
Petersen M, Dubielecka P. Adaptor protein Abelson interactor 1 in homeostasis and disease. Cell Commun Signal 2024; 22:468. [PMID: 39354505 PMCID: PMC11446139 DOI: 10.1186/s12964-024-01738-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 07/04/2024] [Indexed: 10/03/2024] Open
Abstract
Dysregulation of Abelson interactor 1 (ABI1) is associated with various states of disease including developmental defects, pathogen infections, and cancer. ABI1 is an adaptor protein predominantly known to regulate actin cytoskeleton organization processes such as those involved in cell adhesion, migration, and shape determination. Linked to cytoskeleton via vasodilator-stimulated phosphoprotein (VASP), Wiskott-Aldrich syndrome protein family (WAVE), and neural-Wiskott-Aldrich syndrome protein (N-WASP)-associated protein complexes, ABI1 coordinates regulation of various cytoplasmic protein signaling complexes dysregulated in disease states. The roles of ABI1 beyond actin cytoskeleton regulation are much less understood. This comprehensive, protein-centric review describes molecular roles of ABI1 as an adaptor molecule in the context of its dysregulation and associated disease outcomes to better understand disease state-specific protein signaling and affected interconnected biological processes.
Collapse
Affiliation(s)
- Max Petersen
- Division of Hematology/Oncology, Department of Medicine, Warren Alpert Medical School of Brown University and Rhode Island Hospital, Providence, RI, USA
- Center for the Biology of Aging, Brown University, Providence, RI, USA
- Legoretta Cancer Center, Brown University, Providence, RI, USA
| | - Pat Dubielecka
- Division of Hematology/Oncology, Department of Medicine, Warren Alpert Medical School of Brown University and Rhode Island Hospital, Providence, RI, USA.
- Center for the Biology of Aging, Brown University, Providence, RI, USA.
- Legoretta Cancer Center, Brown University, Providence, RI, USA.
| |
Collapse
|
4
|
Khalili D, Kunc M, Herbrich S, Müller AM, Theopold U. Chitinase-like proteins promoting tumorigenesis through disruption of cell polarity via enlarged endosomal vesicles. Front Oncol 2023; 13:1170122. [PMID: 37188187 PMCID: PMC10175591 DOI: 10.3389/fonc.2023.1170122] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 04/05/2023] [Indexed: 05/17/2023] Open
Abstract
Introduction Chitinase-like proteins (CLPs) are associated with tissue-remodeling and inflammation but also with several disorders, including fibrosis, atherosclerosis, allergies, and cancer. However, CLP's role in tumors is far from clear. Methods Here, we utilize Drosophila melanogaster and molecular genetics to investigate the function of CLPs (imaginal disc growth factors; Idgf's) in RasV12 dysplastic salivary glands. Results and discussion We find one of the Idgf's members, Idgf3, is transcriptionally induced in a JNK-dependent manner via a positive feedback loop mediated by reactive oxygen species (ROS). Moreover, Idgf3 accumulates in enlarged endosomal vesicles (EnVs) that promote tumor progression by disrupting cytoskeletal organization. The process is mediated via the downstream component, aSpectrin, which localizes to the EnVs. Our data provide new insight into CLP function in tumors and identifies specific targets for tumor control.
Collapse
|
5
|
Faulkner J, Jiang P, Farris D, Walker R, Dai Z. CRISPR/CAS9-mediated knockout of Abi1 inhibits p185 Bcr-Abl-induced leukemogenesis and signal transduction to ERK and PI3K/Akt pathways. J Hematol Oncol 2020; 13:34. [PMID: 32276588 PMCID: PMC7147029 DOI: 10.1186/s13045-020-00867-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 03/27/2020] [Indexed: 02/07/2023] Open
Abstract
Background Abl interactor 1 (Abi1) is a downstream target of Abl tyrosine kinases and a component of the WAVE regulatory complex (WRC) that plays an important role in regulating actin cytoskeleton remodeling and membrane receptor signaling. While studies using short hairpin RNA (shRNA) have suggested that Abi1 plays a critical role in Bcr-Abl-induced leukemogenesis, the mechanism involved is not clear. Methods In this study, we knocked out Abi1 expression in p185Bcr-Abl-transformed hematopoietic cells using CRISPR/Cas9-mediated gene editing technology. The effects of Abi1 deficiency on actin cytoskeleton remodeling, the Bcr-Abl signaling, IL-3 independent growth, and SDF-induced chemotaxis in these cells were examined by various in vitro assays. The leukemogenic activity of these cells was evaluated by a syngeneic mouse transplantation model. Results We show here that Abi1 deficiency reduced the IL3-independent growth and SDF-1α-mediated chemotaxis in p185Bcr-Abl-transformed hematopoietic cells and inhibited Bcr-Abl-induced abnormal actin remodeling. Depletion of Abi1 also impaired the Bcr-Abl signaling to the ERK and PI3 kinase/Akt pathways. Remarkably, the p185Bcr-Abl-transformed cells with Abi1 deficiency lost their ability to develop leukemia in syngeneic mice. Even though these cells developed drug tolerance in vitro after prolonged selection with imatinib as their parental cells, the imatinib-tolerant cells remain incapable of leukemogenesis in vivo. Conclusions Together, this study highlights an essential role of Abi1 in Bcr-Abl-induced leukemogenesis and provides a model system for dissecting the Abi1 signaling in Bcr-Abl-positive leukemia.
Collapse
Affiliation(s)
- James Faulkner
- Department of Internal Medicine, Texas Tech University Health Sciences Center School of Medicine, 1406 Coulter St, Amarillo, TX, 79106, USA
| | - Peixin Jiang
- Department of Internal Medicine, Texas Tech University Health Sciences Center School of Medicine, 1406 Coulter St, Amarillo, TX, 79106, USA
| | - Delaney Farris
- Department of Internal Medicine, Texas Tech University Health Sciences Center School of Medicine, 1406 Coulter St, Amarillo, TX, 79106, USA
| | - Ryan Walker
- Department of Internal Medicine, Texas Tech University Health Sciences Center School of Medicine, 1406 Coulter St, Amarillo, TX, 79106, USA
| | - Zonghan Dai
- Department of Internal Medicine, Texas Tech University Health Sciences Center School of Medicine, 1406 Coulter St, Amarillo, TX, 79106, USA.
| |
Collapse
|
6
|
Machnicka B, Ponceau A, Picot J, Colin Y, Lecomte MC. Deficiency of αII-spectrin affects endothelial cell-matrix contact and migration leading to impairment of angiogenesis in vitro. Cell Mol Biol Lett 2020; 25:3. [PMID: 32042281 PMCID: PMC6998227 DOI: 10.1186/s11658-020-0200-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Accepted: 01/20/2020] [Indexed: 12/15/2022] Open
Abstract
Background Precise coordination of cytoskeletal components and dynamic control of cell adhesion and migration are required for crucial cell processes such as differentiation and morphogenesis. We investigated the potential involvement of αII-spectrin, a ubiquitous scaffolding element of the membrane skeleton, in the adhesion and angiogenesis mechanism. Methods The cell models were primary human umbilical vein endothelial cells (HUVECs) and a human dermal microvascular endothelial cell line (HMEC-1). After siRNA- and shRNA-mediated knockdown of αII-spectrin, we assessed its expression and that of its partners and adhesion proteins using western blotting. The phenotypes of the control and spectrin-depleted cells were examined using immunofluorescence and video microscopy. Capillary tube formation was assessed using the thick gel Matrigel matrix-based method and a microscope equipped with a thermostatic chamber and a Nikon Biostation System camera. Results Knockdown of αII-spectrin leads to: modified cell shape; actin cytoskeleton organization with the presence of peripheral actin patches; and decreased formation of stress fibers. Spectrin deficiency affects cell adhesion on laminin and fibronectin and cell motility. This included modification of the localization of adhesion molecules, such as αVβ3- and α5-integrins, and organization of adhesion structures, such as focal points. Deficiency of αII-spectrin can also affect the complex mechanism of in vitro capillary tube formation, as demonstrated in a model of angiogenesis. Live imaging revealed that impairment of capillary tube assembly was mainly associated with a significant decrease in cell projection length and stability. αII-spectrin depletion is also associated with significantly decreased expression of three proteins involved in capillary tube formation and assembly: VE-cadherin, MCAM and β3-integrin. Conclusion Our data confirm the role of αII-spectrin in the control of cell adhesion and spreading. Moreover, our findings further support the participation of αII-spectrin in capillary tube formation in vitro through control of adhesion molecules, such as integrins. This indicates a new function of αII-spectrin in angiogenesis.
Collapse
Affiliation(s)
- Beata Machnicka
- 1University of Zielona Góra, Institute of Biological Sciences, Zielona Góra, Poland
| | - Aurélie Ponceau
- 2Biologie Intégrée du Globule Rouge, UMR_S1134, BIGR, INSERM, Université de Paris, F-75015 Paris, France.,3Institut National de la Transfusion Sanguine, F-75015 Paris, France.,Laboratoire d'Excellence GR-Ex, Paris, France
| | - Julien Picot
- 2Biologie Intégrée du Globule Rouge, UMR_S1134, BIGR, INSERM, Université de Paris, F-75015 Paris, France.,3Institut National de la Transfusion Sanguine, F-75015 Paris, France.,Laboratoire d'Excellence GR-Ex, Paris, France
| | - Yves Colin
- 2Biologie Intégrée du Globule Rouge, UMR_S1134, BIGR, INSERM, Université de Paris, F-75015 Paris, France.,3Institut National de la Transfusion Sanguine, F-75015 Paris, France.,Laboratoire d'Excellence GR-Ex, Paris, France
| | - Marie-Christine Lecomte
- 2Biologie Intégrée du Globule Rouge, UMR_S1134, BIGR, INSERM, Université de Paris, F-75015 Paris, France.,3Institut National de la Transfusion Sanguine, F-75015 Paris, France.,Laboratoire d'Excellence GR-Ex, Paris, France
| |
Collapse
|
7
|
Lambert MW. The functional importance of lamins, actin, myosin, spectrin and the LINC complex in DNA repair. Exp Biol Med (Maywood) 2019; 244:1382-1406. [PMID: 31581813 PMCID: PMC6880146 DOI: 10.1177/1535370219876651] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Three major proteins in the nucleoskeleton, lamins, actin, and spectrin, play essential roles in maintenance of nuclear architecture and the integrity of the nuclear envelope, in mechanotransduction and mechanical coupling between the nucleoskeleton and cytoskeleton, and in nuclear functions such as regulation of gene expression, transcription and DNA replication. Less well known, but critically important, are the role these proteins play in DNA repair. The A-type and B-type lamins, nuclear actin and myosin, spectrin and the LINC (linker of nucleoskeleton and cytoskeleton) complex each function in repair of DNA damage utilizing various repair pathways. The lamins play a role in repair of DNA double-strand breaks (DSBs) by nonhomologous end joining (NHEJ) or homologous recombination (HR). Actin is involved in repair of DNA DSBs and interacts with myosin in facilitating relocalization of these DSBs in heterochromatin for HR repair. Nonerythroid alpha spectrin (αSpII) plays a critical role in repair of DNA interstrand cross-links (ICLs) where it acts as a scaffold in recruitment of repair proteins to sites of damage and is important in the initial damage recognition and incision steps of the repair process. The LINC complex contributes to the repair of DNA DSBs and ICLs. This review will address the important functions of these proteins in the DNA repair process, their mechanism of action, and the profound impact a defect or deficiency in these proteins has on cellular function. The critical roles of these proteins in DNA repair will be further emphasized by discussing the human disorders and the pathophysiological changes that result from or are related to deficiencies in these proteins. The demonstrated function for each of these proteins in the DNA repair process clearly indicates that there is another level of complexity that must be considered when mechanistically examining factors crucial for DNA repair.
Collapse
Affiliation(s)
- Muriel W Lambert
- Department of Pathology, Immunology and Laboratory
Medicine, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| |
Collapse
|
8
|
Nath D, Li X, Mondragon C, Post D, Chen M, White JR, Hryniewicz-Jankowska A, Caza T, Kuznetsov VA, Hehnly H, Jamaspishvili T, Berman DM, Zhang F, Kung SHY, Fazli L, Gleave ME, Bratslavsky G, Pandolfi PP, Kotula L. Abi1 loss drives prostate tumorigenesis through activation of EMT and non-canonical WNT signaling. Cell Commun Signal 2019; 17:120. [PMID: 31530281 PMCID: PMC6749699 DOI: 10.1186/s12964-019-0410-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 08/01/2019] [Indexed: 12/29/2022] Open
Abstract
Background Prostate cancer development involves various mechanisms, which are poorly understood but pointing to epithelial mesenchymal transition (EMT) as the key mechanism in progression to metastatic disease. ABI1, a member of WAVE complex and actin cytoskeleton regulator and adaptor protein, acts as tumor suppressor in prostate cancer but the role of ABI1 in EMT is not clear. Methods To investigate the molecular mechanism by which loss of ABI1 contributes to tumor progression, we disrupted the ABI1 gene in the benign prostate epithelial RWPE-1 cell line and determined its phenotype. Levels of ABI1 expression in prostate organoid tumor cell lines was evaluated by Western blotting and RNA sequencing. ABI1 expression and its association with prostate tumor grade was evaluated in a TMA cohort of 505 patients and metastatic cell lines. Results Low ABI1 expression is associated with biochemical recurrence, metastasis and death (p = 0.038). Moreover, ABI1 expression was significantly decreased in Gleason pattern 5 vs. pattern 4 (p = 0.0025) and 3 (p = 0.0012), indicating an association between low ABI1 expression and highly invasive prostate tumors. Disruption of ABI1 gene in RWPE-1 cell line resulted in gain of an invasive phenotype, which was characterized by a loss of cell-cell adhesion markers and increased migratory ability of RWPE-1 spheroids. Through RNA sequencing and protein expression analysis, we discovered that ABI1 loss leads to activation of non-canonical WNT signaling and EMT pathways, which are rescued by re-expression of ABI1. Furthermore, an increase in STAT3 phosphorylation upon ABI1 inactivation and the evidence of a high-affinity interaction between the FYN SH2 domain and ABI1 pY421 support a model in which ABI1 acts as a gatekeeper of non-canonical WNT-EMT pathway activation downstream of the FZD2 receptor. Conclusions ABI1 controls prostate tumor progression and epithelial plasticity through regulation of EMT-WNT pathway. Here we discovered that ABI1 inhibits EMT through suppressing FYN-STAT3 activation downstream from non-canonical WNT signaling thus providing a novel mechanism of prostate tumor suppression. Electronic supplementary material The online version of this article (10.1186/s12964-019-0410-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Disharee Nath
- Department of Urology, Upstate Cancer Center, SUNY Upstate Medical University, 750 East Adams Street, Syracuse, New York, 13210, USA.,Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY, 13210, USA
| | - Xiang Li
- Department of Urology, Upstate Cancer Center, SUNY Upstate Medical University, 750 East Adams Street, Syracuse, New York, 13210, USA.,Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY, 13210, USA
| | - Claudia Mondragon
- Department of Urology, Upstate Cancer Center, SUNY Upstate Medical University, 750 East Adams Street, Syracuse, New York, 13210, USA
| | - Dawn Post
- Department of Urology, Upstate Cancer Center, SUNY Upstate Medical University, 750 East Adams Street, Syracuse, New York, 13210, USA
| | - Ming Chen
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Department of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA.,Present address: Department of Pathology, Duke University School of Medicine, Durham, NC, 27710, USA.,Duke Cancer Institute, Duke University, Durham, NC, 27710, USA
| | - Julie R White
- Laboratory of Comparative Pathology, Memorial Sloan-Kettering Cancer Center, New York, NY, 10065, USA
| | - Anita Hryniewicz-Jankowska
- Department of Urology, Upstate Cancer Center, SUNY Upstate Medical University, 750 East Adams Street, Syracuse, New York, 13210, USA.,Department of Cytobiochemistry, Faculty of Biotechnology, University of Wroclaw, ul. F. Joliot-Curie 14a, 50-383, Wroclaw, Poland
| | - Tiffany Caza
- Department of Pathology and Medicine, SUNY Upstate Medical University, Syracuse, NY, 13210, USA
| | - Vladimir A Kuznetsov
- Department of Urology, Upstate Cancer Center, SUNY Upstate Medical University, 750 East Adams Street, Syracuse, New York, 13210, USA.,Bioinformatics Institute, A-STAR, Singapore, 138671, Singapore
| | - Heidi Hehnly
- Department of Cell and Developmental Biology, SUNY Upstate Medical University, Syracuse, NY, 13210, USA
| | - Tamara Jamaspishvili
- Department of Pathology and Molecular Medicine and Division of Cancer Biology & Genetics, Queen's Cancer Research Institute, Queen's University, 10 Stuart St, Kingston, ON, K7L 3N6, Canada
| | - David M Berman
- Department of Pathology and Molecular Medicine and Division of Cancer Biology & Genetics, Queen's Cancer Research Institute, Queen's University, 10 Stuart St, Kingston, ON, K7L 3N6, Canada
| | - Fan Zhang
- Department of Urologic Sciences, Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, V6H 3Z6, Canada
| | - Sonia H Y Kung
- Department of Urologic Sciences, Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, V6H 3Z6, Canada
| | - Ladan Fazli
- Department of Urologic Sciences, Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, V6H 3Z6, Canada
| | - Martin E Gleave
- Department of Urologic Sciences, Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, V6H 3Z6, Canada
| | - Gennady Bratslavsky
- Department of Urology, Upstate Cancer Center, SUNY Upstate Medical University, 750 East Adams Street, Syracuse, New York, 13210, USA
| | - Pier Paolo Pandolfi
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Department of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Leszek Kotula
- Department of Urology, Upstate Cancer Center, SUNY Upstate Medical University, 750 East Adams Street, Syracuse, New York, 13210, USA. .,Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY, 13210, USA.
| |
Collapse
|
9
|
Machnicka B, Grochowalska R, Bogusławska DM, Sikorski AF. The role of spectrin in cell adhesion and cell-cell contact. Exp Biol Med (Maywood) 2019; 244:1303-1312. [PMID: 31226892 DOI: 10.1177/1535370219859003] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Spectrins are proteins that are responsible for many aspects of cell function and adaptation to changing environments. Primarily the spectrin-based membrane skeleton maintains cell membrane integrity and its mechanical properties, together with the cytoskeletal network a support cell shape. The occurrence of a variety of spectrin isoforms in diverse cellular environments indicates that it is a multifunctional protein involved in numerous physiological pathways. Participation of spectrin in cell–cell and cell–extracellular matrix adhesion and formation of dynamic plasma membrane protrusions and associated signaling events is a subject of interest for researchers in the fields of cell biology and molecular medicine. In this mini-review, we focus on data concerning the role of spectrins in cell surface activities such as adhesion, cell–cell contact, and invadosome formation. We discuss data on different adhesion proteins that directly or indirectly interact with spectrin repeats. New findings support the involvement of spectrin in cell adhesion and spreading, formation of lamellipodia, and also the participation in morphogenetic processes, such as eye development, oogenesis, and angiogenesis. Here, we review the role of spectrin in cell adhesion and cell–cell contact.Impact statementThis article reviews properties of spectrins as a group of proteins involved in cell surface activities such as, adhesion and cell–cell contact, and their contribution to morphogenesis. We show a new area of research and discuss the involvement of spectrin in regulation of cell–cell contact leading to immunological synapse formation and in shaping synapse architecture during myoblast fusion. Data indicate involvement of spectrins in adhesion and cell–cell or cell–extracellular matrix interactions and therefore in signaling pathways. There is evidence of spectrin’s contribution to the processes of morphogenesis which are connected to its interactions with adhesion molecules, membrane proteins (and perhaps lipids), and actin. Our aim was to highlight the essential role of spectrin in cell–cell contact and cell adhesion.
Collapse
Affiliation(s)
- Beata Machnicka
- Department of Biochemistry and Bioinformatics, Faculty of Biological Sciences, University of Zielona Góra, Zielona Góra 65-516, Poland
| | - Renata Grochowalska
- Department of Biochemistry and Bioinformatics, Faculty of Biological Sciences, University of Zielona Góra, Zielona Góra 65-516, Poland
| | - Dżamila M Bogusławska
- Department of Biochemistry and Bioinformatics, Faculty of Biological Sciences, University of Zielona Góra, Zielona Góra 65-516, Poland
| | - Aleksander F Sikorski
- Department of Cytobiochemistry, Faculty of Biotechnology, University of Wrocław, Wrocław 50-383, Poland
| |
Collapse
|
10
|
The Role of Nonerythroid Spectrin αII in Cancer. JOURNAL OF ONCOLOGY 2019; 2019:7079604. [PMID: 31186638 PMCID: PMC6521328 DOI: 10.1155/2019/7079604] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 04/05/2019] [Accepted: 04/09/2019] [Indexed: 12/18/2022]
Abstract
Nonerythroid spectrin αII (SPTAN1) is an important cytoskeletal protein that ensures vital cellular properties including polarity and cell stabilization. In addition, it is involved in cell adhesion, cell-cell contact, and apoptosis. The detection of altered expression of SPTAN1 in tumors indicates that SPTAN1 might be involved in the development and progression of cancer. SPTAN1 has been described in cancer and therapy response and proposed as a potential marker protein for neoplasia, tumor aggressiveness, and therapeutic efficiency. On one hand, the existing data suggest that overexpression of SPTAN1 in tumor cells reflects neoplastic and tumor promoting activity. On the other hand, nuclear SPTAN1 can have tumor suppressing effects by enabling DNA repair through interaction with DNA repair proteins. Moreover, SPTAN1 cleavage products occur during apoptosis and could serve as markers for the efficacy of cancer therapy. Due to SPTAN1's multifaceted functions and its role in adhesion and migration, SPTAN1 can influence tumor growth and progression in both positive and negative directions depending on its specific regulation. This review summarizes the current knowledge on SPTAN1 in cancer and depicts several mechanisms by which SPTAN1 could impact tumor development and aggressiveness.
Collapse
|
11
|
Wang Y, Ji T, Nelson AD, Glanowska K, Murphy GG, Jenkins PM, Parent JM. Critical roles of αII spectrin in brain development and epileptic encephalopathy. J Clin Invest 2018; 128:760-773. [PMID: 29337302 DOI: 10.1172/jci95743] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 11/28/2017] [Indexed: 12/26/2022] Open
Abstract
The nonerythrocytic α-spectrin-1 (SPTAN1) gene encodes the cytoskeletal protein αII spectrin. Mutations in SPTAN1 cause early infantile epileptic encephalopathy type 5 (EIEE5); however, the role of αII spectrin in neurodevelopment and EIEE5 pathogenesis is unknown. Prior work suggests that αII spectrin is absent in the axon initial segment (AIS) and contributes to a diffusion barrier in the distal axon. Here, we have shown that αII spectrin is expressed ubiquitously in rodent and human somatodendritic and axonal domains. CRISPR-mediated deletion of Sptan1 in embryonic rat forebrain by in utero electroporation caused altered dendritic and axonal development, loss of the AIS, and decreased inhibitory innervation. Overexpression of human EIEE5 mutant SPTAN1 in embryonic rat forebrain and mouse hippocampal neurons led to similar developmental defects that were also observed in EIEE5 patient-derived neurons. Additionally, patient-derived neurons displayed aggregation of spectrin complexes. Taken together, these findings implicate αII spectrin in critical aspects of dendritic and axonal development and synaptogenesis, and support a dominant-negative mechanism of SPTAN1 mutations in EIEE5.
Collapse
Affiliation(s)
| | | | | | | | - Geoffrey G Murphy
- Molecular and Behavioral Neuroscience Institute.,Department of Molecular and Integrative Physiology, and
| | - Paul M Jenkins
- Department of Pharmacology.,Department of Psychiatry, University of Michigan, Ann Arbor, Michigan, USA
| | - Jack M Parent
- Department of Neurology.,Ann Arbor VA Healthcare System, Ann Arbor, Michigan, USA
| |
Collapse
|
12
|
Phosphorylation of SOS1 on tyrosine 1196 promotes its RAC GEF activity and contributes to BCR-ABL leukemogenesis. Leukemia 2017; 32:820-827. [PMID: 28819285 PMCID: PMC5739283 DOI: 10.1038/leu.2017.267] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 08/07/2017] [Accepted: 08/11/2017] [Indexed: 02/01/2023]
Abstract
Son of Sevenless 1 (SOS1) is a dual guanine nucleotide exchange factor (GEF) that activates the small GTPases RAC and RAS. Although the molecular mechanisms of RAS GEF catalysis have been unveiled, how SOS1 acquires RAC GEF activity and what is the physio-pathological relevance of this activity is much less understood. Here we show that SOS1 is tyrosine phosphorylated on Y1196 by ABL. Phosphorylation of Y1196 controls SOS1 inter-molecular interaction, is required to promote the exchange of nucleotides on RAC in vitro and for platelet-derived growth factor (PDGF) activation of RAC- and RAC-dependent actin remodeling and cell migration. SOS1 is also phosphorylated on Y1196 by BCR-ABL in chronic myelogenous leukemic cells. Importantly, in these cells, SOS1 is required for BCR-ABL-mediated activation of RAC, cell proliferation and transformation in vitro and in a xenograft mouse model. Finally, genetic removal of Sos1 in the bone marrow-derived cells (BMDCs) from Sos1fl/fl mice and infected with BCR-ABL causes a significant delay in the onset of leukemogenesis once BMDCs are injected into recipient, lethally irradiated mice. Thus, SOS1 is required for full transformation and critically contribute to the leukemogenic potential of BCR-ABL.
Collapse
|
13
|
Kumar S, Lu B, Dixit U, Hossain S, Liu Y, Li J, Hornbeck P, Zheng W, Sowalsky AG, Kotula L, Birge RB. Reciprocal regulation of Abl kinase by Crk Y251 and Abi1 controls invasive phenotypes in glioblastoma. Oncotarget 2016; 6:37792-807. [PMID: 26473374 PMCID: PMC4741966 DOI: 10.18632/oncotarget.6096] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 09/29/2015] [Indexed: 11/25/2022] Open
Abstract
Crk is the prototypical member of a class of Src homology 2 (SH2) and Src homology 3 (SH3) domain-containing adaptor proteins that positively regulate cell motility via the activation of Rac1 and, in certain tumor types such as GBM, can promote cell invasion and metastasis by mechanisms that are not well understood. Here we demonstrate that Crk, via its phosphorylation at Tyr251, promotes invasive behavior of tumor cells, is a prominent feature in GBM, and correlating with aggressive glioma grade IV staging and overall poor survival outcomes. At the molecular level, Tyr251 phosphorylation of Crk is negatively regulated by Abi1, which competes for Crk binding to Abl and attenuates Abl transactivation. Together, these results show that Crk and Abi1 have reciprocal biological effects and act as a molecular rheostat to control Abl activation and cell invasion. Finally, these data suggest that Crk Tyr251 phosphorylation regulate invasive cell phenotypes and may serve as a biomarker for aggressive GBM.
Collapse
Affiliation(s)
- Sushil Kumar
- Department of Microbiology, Biochemistry and Molecular Genetics, Cancer Center, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Bin Lu
- Institute of Biophysics, School of Life Sciences, Wenzhou Medical University, Wenzhou, China.,Attardi Institute of Mitochondrial Biomedicine, School of Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Updesh Dixit
- Department of Microbiology, Biochemistry and Molecular Genetics, Cancer Center, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Sajjad Hossain
- Departments of Urology and Biochemistry and Molecular Biology, SUNY Upstate Medical University, New York, NY, USA
| | - Yongzhang Liu
- Institute of Biophysics, School of Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Jing Li
- Cell Signaling Technology, Danvers, MA, USA
| | | | - Weiming Zheng
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Adam G Sowalsky
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Leszek Kotula
- Departments of Urology and Biochemistry and Molecular Biology, SUNY Upstate Medical University, New York, NY, USA
| | - Raymond B Birge
- Department of Microbiology, Biochemistry and Molecular Genetics, Cancer Center, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| |
Collapse
|
14
|
Lambert MW. Nuclear alpha spectrin: Critical roles in DNA interstrand cross-link repair and genomic stability. Exp Biol Med (Maywood) 2016; 241:1621-38. [PMID: 27480253 PMCID: PMC4999628 DOI: 10.1177/1535370216662714] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Non-erythroid alpha spectrin (αIISp) is a structural protein which we have shown is present in the nucleus of human cells. It interacts with a number of nuclear proteins such as actin, lamin, emerin, chromatin remodeling factors, and DNA repair proteins. αIISp's interaction with DNA repair proteins has been extensively studied. We have demonstrated that nuclear αIISp is critical in DNA interstrand cross-link (ICL) repair in S phase, in both genomic (non-telomeric) and telomeric DNA, and in maintenance of genomic stability following ICL damage to DNA. We have proposed that αIISp acts as a scaffold aiding to recruit repair proteins to sites of damage. This involvement of αIISp in ICL repair and telomere maintenance after ICL damage represents new and critical functions for αIISp. These studies have led to development of a model for the role of αIISp in DNA ICL repair. They have been aided by examination of cells from patients with Fanconi anemia (FA), a repair-deficient genetic disorder in which a deficiency in αIISp leads to defective ICL repair in genomic and telomeric DNA, telomere dysfunction, and chromosome instability following DNA ICL damage. We have shown that loss of αIISp in FA cells is due to increased breakdown by the protease, µ-calpain. Importantly, we have demonstrated that this deficiency can be corrected by knockdown of µ-calpain and restoring αIISp levels to normal. This corrects a number of the phenotypic deficiencies in FA after ICL damage. These studies suggest a new and unexplored direction for therapeutically restoring genomic stability in FA cells and for correcting numerous phenotypic deficiencies occurring after ICL damage. Developing a more in-depth understanding of the importance of the interaction of αIISp with other nuclear proteins could significantly enhance our knowledge of the consequences of loss of αIISp on critical nuclear processes.
Collapse
Affiliation(s)
- Muriel W Lambert
- Department of Pathology and Laboratory Medicine, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
| |
Collapse
|
15
|
Ponceau A, Albigès-Rizo C, Colin-Aronovicz Y, Destaing O, Lecomte MC. αII-spectrin regulates invadosome stability and extracellular matrix degradation. PLoS One 2015; 10:e0120781. [PMID: 25830635 PMCID: PMC4382279 DOI: 10.1371/journal.pone.0120781] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Accepted: 01/27/2015] [Indexed: 01/07/2023] Open
Abstract
Invadosomes are actin-rich adhesion structures involved in tissue invasion and extracellular matrix (ECM) remodelling. αII-Spectrin, an ubiquitous scaffolding component of the membrane skeleton and a partner of actin regulators (ABI1, VASP and WASL), accumulates highly and specifically in the invadosomes of multiple cell types, such as mouse embryonic fibroblasts (MEFs) expressing SrcY527F, the constitutively active form of Src or activated HMEC-1 endothelial cells. FRAP and live-imaging analysis revealed that αII-spectrin is a highly dynamic component of invadosomes as actin present in the structures core. Knockdown of αII-spectrin expression destabilizes invadosomes and reduces the ability of the remaining invadosomes to digest the ECM and to promote invasion. The ECM degradation defect observed in spectrin-depleted-cells is associated with highly dynamic and unstable invadosome rings. Moreover, FRAP measurement showed the specific involvement of αII-spectrin in the regulation of the mobile/immobile β3-integrin ratio in invadosomes. Our findings suggest that spectrin could regulate invadosome function and maturation by modulating integrin mobility in the membrane, allowing the normal processes of adhesion, invasion and matrix degradation. Altogether, these data highlight a new function for spectrins in the stability of invadosomes and the coupling between actin regulation and ECM degradation.
Collapse
Affiliation(s)
- Aurélie Ponceau
- Institut National de la Transfusion Sanguine, INSERM UMR-S 665, Paris, France, Université Paris 7/Denis Diderot, Paris, France
| | - Corinne Albigès-Rizo
- Institut Albert Bonniot, Université Joseph Fourier, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale-Université Joseph Fourier U823 Site Santé, Grenoble, France
| | - Yves Colin-Aronovicz
- Institut National de la Transfusion Sanguine, INSERM UMR-S 665, Paris, France, Université Paris 7/Denis Diderot, Paris, France
| | - Olivier Destaing
- Institut Albert Bonniot, Université Joseph Fourier, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale-Université Joseph Fourier U823 Site Santé, Grenoble, France
| | - Marie Christine Lecomte
- Institut National de la Transfusion Sanguine, INSERM UMR-S 665, Paris, France, Université Paris 7/Denis Diderot, Paris, France
- * E-mail:
| |
Collapse
|
16
|
Patra M, Mukhopadhyay C, Chakrabarti A. Malachite green interacts with the membrane skeletal protein, spectrin. RSC Adv 2015. [DOI: 10.1039/c5ra15488j] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Energy minimized complex of MG with the self association domain of spectrin.
Collapse
Affiliation(s)
- Malay Patra
- Chemistry Department
- University of Calcutta
- Kolkata 700009
- India
| | | | - Abhijit Chakrabarti
- Crystallography & Molecular Biology Division
- Saha Institute of Nuclear Physics
- Kolkata 700064
- India
| |
Collapse
|
17
|
AlphaII-spectrin participates in the surface expression of cell adhesion molecule L1 and neurite outgrowth. Exp Cell Res 2014; 322:365-80. [DOI: 10.1016/j.yexcr.2014.01.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Revised: 01/11/2014] [Accepted: 01/13/2014] [Indexed: 11/22/2022]
|
18
|
Machnicka B, Czogalla A, Hryniewicz-Jankowska A, Bogusławska DM, Grochowalska R, Heger E, Sikorski AF. Spectrins: a structural platform for stabilization and activation of membrane channels, receptors and transporters. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2013; 1838:620-34. [PMID: 23673272 DOI: 10.1016/j.bbamem.2013.05.002] [Citation(s) in RCA: 146] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Revised: 04/25/2013] [Accepted: 05/06/2013] [Indexed: 12/22/2022]
Abstract
This review focuses on structure and functions of spectrin as a major component of the membrane skeleton. Recent advances on spectrin function as an interface for signal transduction mediation and a number of data concerning interaction of spectrin with membrane channels, adhesion molecules, receptors and transporters draw a picture of multifaceted protein. Here, we attempted to show the current depiction of multitask role of spectrin in cell physiology. This article is part of a Special Issue entitled: Reciprocal influences between cell cytoskeleton and membrane channels, receptors and transporters. Guest Editor: Jean Claude Hervé.
Collapse
Affiliation(s)
- Beata Machnicka
- University of Zielona Góra, Faculty of Biological Sciences, Poland
| | | | | | | | | | - Elżbieta Heger
- University of Zielona Góra, Faculty of Biological Sciences, Poland
| | | |
Collapse
|
19
|
Singh A, Winterbottom EF, Ji YJ, Hwang YS, Daar IO. Abelson interactor 1 (ABI1) and its interaction with Wiskott-Aldrich syndrome protein (wasp) are critical for proper eye formation in Xenopus embryos. J Biol Chem 2013; 288:14135-14146. [PMID: 23558677 DOI: 10.1074/jbc.m112.445643] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Abl interactor 1 (Abi1) is a scaffold protein that plays a central role in the regulation of actin cytoskeleton dynamics as a constituent of several key protein complexes, and homozygous loss of this protein leads to embryonic lethality in mice. Because this scaffold protein has been shown in cultured cells to be a critical component of pathways controlling cell migration and actin regulation at cell-cell contacts, we were interested to investigate the in vivo role of Abi1 in morphogenesis during the development of Xenopus embryos. Using morpholino-mediated translation inhibition, we demonstrate that knockdown of Abi1 in the whole embryo, or specifically in eye field progenitor cells, leads to disruption of eye morphogenesis. Moreover, signaling through the Src homology 3 domain of Abi1 is critical for proper movement of retinal progenitor cells into the eye field and their appropriate differentiation, and this process is dependent upon an interaction with the nucleation-promoting factor Wasp (Wiskott-Aldrich syndrome protein). Collectively, our data demonstrate that the Abi1 scaffold protein is an essential regulator of cell movement processes required for normal eye development in Xenopus embryos and specifically requires an Src homology 3 domain-dependent interaction with Wasp to regulate this complex morphogenetic process.
Collapse
Affiliation(s)
- Arvinder Singh
- Laboratory of Cell and Developmental Signaling, NCI-Frederick, National Institutes of Health, Frederick, Maryland 21702
| | - Emily F Winterbottom
- Laboratory of Cell and Developmental Signaling, NCI-Frederick, National Institutes of Health, Frederick, Maryland 21702
| | - Yon Ju Ji
- Laboratory of Cell and Developmental Signaling, NCI-Frederick, National Institutes of Health, Frederick, Maryland 21702
| | - Yoo-Seok Hwang
- Laboratory of Cell and Developmental Signaling, NCI-Frederick, National Institutes of Health, Frederick, Maryland 21702
| | - Ira O Daar
- Laboratory of Cell and Developmental Signaling, NCI-Frederick, National Institutes of Health, Frederick, Maryland 21702.
| |
Collapse
|
20
|
Hossain S, Dubielecka PM, Sikorski AF, Birge RB, Kotula L. Crk and ABI1: binary molecular switches that regulate abl tyrosine kinase and signaling to the cytoskeleton. Genes Cancer 2012; 3:402-13. [PMID: 23226578 DOI: 10.1177/1947601912460051] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The nonreceptor tyrosine kinases Abl and Arg are among the most well-characterized tyrosine kinases in the human genome. The activation of Abl by N-terminal fusions with Bcr (Bcr-Abl) or Gag (v-Abl) is responsible for chronic myeloid leukemia or Ph+ acute lymphoblastic leukemia and mouse leukemia virus, respectively. In addition, aberrant Abl and Arg activation downstream of several oncogenic growth factor receptors contributes to the development and progression of a variety of human cancers, often associated with poor clinical outcome, drug resistance, and tumor invasion and metastasis. Abl activation can occur by a variety of mechanisms that include domain interactions involving structural remodeling of autoinhibited conformations as well as direct phosphorylation by upstream kinases and phosphatases. Constitutive activation of Abl plays a significant role in regulating the actin cytoskeleton by modulating cell adhesion, motility, and invadopodia. This review addresses the role of Abl and Arg in tumor progression with particular emphasis on the roles of Crk and Abi1 adapter proteins as distinct molecular switches for Abl transactivation. These insights, combined with new insights into the structure of these kinases, provide the rationale to envision that Crk and Abi1 fine-tune Abl regulation to control signaling to the cytoskeleton.
Collapse
Affiliation(s)
- Sajjad Hossain
- New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA ; Current address: Stony Brook University, Stony Brook, NY, USA
| | | | | | | | | |
Collapse
|
21
|
Xiong X, Chorzalska A, Dubielecka PM, White JR, Vedvyas Y, Hedvat CV, Haimovitz-Friedman A, Koutcher JA, Reimand J, Bader GD, Sawicki JA, Kotula L. Disruption of Abi1/Hssh3bp1 expression induces prostatic intraepithelial neoplasia in the conditional Abi1/Hssh3bp1 KO mice. Oncogenesis 2012; 1:e26. [PMID: 23552839 PMCID: PMC3503296 DOI: 10.1038/oncsis.2012.28] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2012] [Revised: 07/10/2012] [Accepted: 07/31/2012] [Indexed: 12/26/2022] Open
Abstract
Prostate cancer is one of the leading causes of cancer-related deaths in the United States and a leading diagnosed non-skin cancer in American men. Genetic mutations underlying prostate tumorigenesis include alterations of tumor suppressor genes. We tested the tumor suppressor hypothesis for ABI1/hSSH3BP1 by searching for gene mutations in primary prostate tumors from patients, and by analyzing the consequences of prostate-specific disruption of the mouse Abi1/Hssh3bp1 ortholog. We sequenced the ABI1/hSSH3BP1 gene and identified recurring mutations in 6 out of 35 prostate tumors. Moreover, complementation and anchorage-independent growth, proliferation, cellular adhesion and xenograft assays using the LNCaP cell line, which contains a loss-of-function Abi1 mutation, and a stably expressed wild-type or mutated ABI gene, were consistent with the tumor suppressor hypothesis. To test the hypothesis further, we disrupted the gene in the mouse prostate by breeding the Abi1 floxed strain with the probasin promoter-driven Cre recombinase strain. Histopathological evaluation of mice indicated development of prostatic intraepithelial neoplasia (PIN) in Abi1/Hssh3bp1 knockout mouse as early as the eighth month, but no progression beyond PIN was observed in mice as old as 12 months. Observed decreased levels of E-cadherin, β-catenin and WAVE2 in mouse prostate suggest abnormal cellular adhesion as the mechanism underlying PIN development owing to Abi1 disruption. Analysis of syngeneic cell lines point to the possibility that upregulation of phospho-Akt underlies the enhanced cellular proliferation phenotype of cells lacking Abi1. This study provides proof-of-concept for the hypothesis that Abi1 downregulation has a role in the development of prostate cancer.
Collapse
Affiliation(s)
- X Xiong
- Laboratory of Cell Signaling, New York Blood Center, New York, NY, USA
| | - A Chorzalska
- Laboratory of Cell Signaling, New York Blood Center, New York, NY, USA
| | - P M Dubielecka
- Laboratory of Cell Signaling, New York Blood Center, New York, NY, USA
| | - J R White
- Laboratory of Comparative Pathology, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Y Vedvyas
- Laboratory of Cell Signaling, New York Blood Center, New York, NY, USA
| | - C V Hedvat
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - A Haimovitz-Friedman
- Department of Radiation Oncology, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - J A Koutcher
- Department of Medical Physics, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - J Reimand
- The Donnelly Center for Cellular and Biomolecular Research, Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - G D Bader
- The Donnelly Center for Cellular and Biomolecular Research, Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - J A Sawicki
- Lankenau Institute for Medical Research, Wynnewood, PA, USA
| | - L Kotula
- Laboratory of Cell Signaling, New York Blood Center, New York, NY, USA
| |
Collapse
|
22
|
Ding K, Shameer K, Jouni H, Masys DR, Jarvik GP, Kho AN, Ritchie MD, McCarty CA, Chute CG, Manolio TA, Kullo IJ. Genetic Loci implicated in erythroid differentiation and cell cycle regulation are associated with red blood cell traits. Mayo Clin Proc 2012; 87:461-74. [PMID: 22560525 PMCID: PMC3538470 DOI: 10.1016/j.mayocp.2012.01.016] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2011] [Revised: 01/10/2012] [Accepted: 01/19/2012] [Indexed: 01/16/2023]
Abstract
OBJECTIVE To identify common genetic variants influencing red blood cell (RBC) traits. PATIENTS AND METHODS We performed a genomewide association study from June 2008 through July 2011 of hemoglobin, hematocrit, RBC count, mean corpuscular volume, mean corpuscular hemoglobin, and mean corpuscular hemoglobin concentration in 12,486 patients of European ancestry from the electronic MEdical Records and Genomics (eMERGE) network. We developed an electronic medical record-based algorithm that included individuals who had RBC measurements obtained for clinical care and excluded values measured in the setting of hematopoietic disorders, comorbid conditions, or medications known to affect RBC production or a recent history of blood loss. RESULTS We identified 4 new genetic loci and replicated 11 loci previously reported to be associated with one or more RBC traits in individuals of European ancestry. Notably, genes present in 3 of the 4 newly identified loci (THRB, PTPLAD1, CDT1) and in 6 of the 11 replicated loci (KLF1, ALDH8A1, CCND3, SPTA1, FBXO7, TFR2/EPO) are implicated in erythroid differentiation and regulation of cell cycle in hematopoietic stem cells. CONCLUSION Genes in the erythroid differentiation and cell cycle regulation pathways influence interindividual variation in RBC indices. Our results provide insights into the molecular basis underlying variation in RBC traits.
Collapse
Key Words
- emerge, electronic medical records and genomics
- emmax, mixed-model association-expedited
- emr, electronic medical record
- eqtl, expression quantitative trait locus
- ghc, group health cooperative--university of washington
- gwas, genomewide association study
- hct, hematocrit
- hgb, hemoglobin
- ibs, identity-by-state
- ld, linkage disequilibrium
- mc, marshfield clinic
- mch, mean corpuscular hemoglobin
- mchc, mean corpuscular hemoglobin concentration
- mcv, mean corpuscular volume
- mim, mendelian inheritance of man
- nu, northwestern university
- rbc, red blood cell
- snp, single-nucleotide polymorphism
- vumc, vanderbilt university medical center
Collapse
Affiliation(s)
- Keyue Ding
- Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN
| | - Khader Shameer
- Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN
| | - Hayan Jouni
- Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN
| | - Daniel R. Masys
- Division of Biomedical and Health Informatics, Department of Medical Education and Biomedical Informatics, University of Washington, Seattle
| | - Gail P. Jarvik
- Department of Medicine (Medical Genetics) and Department of Genome Sciences, University of Washington, Seattle
| | - Abel N. Kho
- Department of Medicine, Northwestern University, Chicago, IL
| | - Marylyn D. Ritchie
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park
| | | | | | - Teri A. Manolio
- Office of Population Genomics, National Human Genome Research Institute, Bethesda, MD
| | - Iftikhar J. Kullo
- Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN
- Correspondence: Address to Iftikhar J. Kullo, MD, Mayo Clinic, 200 First St SW, Rochester, MN 55905
| |
Collapse
|
23
|
Stankewich MC, Cianci CD, Stabach PR, Ji L, Nath A, Morrow JS. Cell organization, growth, and neural and cardiac development require αII-spectrin. J Cell Sci 2011; 124:3956-66. [PMID: 22159418 DOI: 10.1242/jcs.080374] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Spectrin α2 (αII-spectrin) is a scaffolding protein encoded by the Spna2 gene and constitutively expressed in most tissues. Exon trapping of Spna2 in C57BL/6 mice allowed targeted disruption of αII-spectrin. Heterozygous animals displayed no phenotype by 2 years of age. Homozygous deletion of Spna2 was embryonic lethal at embryonic day 12.5 to 16.5 with retarded intrauterine growth, and craniofacial, neural tube and cardiac anomalies. The loss of αII-spectrin did not alter the levels of αI- or βI-spectrin, or the transcriptional levels of any β-spectrin or any ankyrin, but secondarily reduced by about 80% the steady state protein levels of βII- and βIII-spectrin. Residual βII- and βIII-spectrin and ankyrins B and G were concentrated at the apical membrane of bronchial and renal epithelial cells, without impacting cell morphology. Neuroepithelial cells in the developing brain were more concentrated and more proliferative in the ventricular zone than normal; axon formation was also impaired. Embryonic fibroblasts cultured on fibronectin from E14.5 (Spna2(-/-)) animals displayed impaired growth and spreading, a spiky morphology, and sparse lamellipodia without cortical actin. These data indicate that the spectrin-ankyrin scaffold is crucial in vertebrates for cell spreading, tissue patterning and organ development, particularly in the developing brain and heart, but is not required for cell viability.
Collapse
Affiliation(s)
- Michael C Stankewich
- Department of Pathology, Yale University School of Medicine, 310 Cedar St. BML 150, New Haven, CT 06520, USA.
| | | | | | | | | | | |
Collapse
|
24
|
Machnicka B, Grochowalska R, Bogusławska DM, Sikorski AF, Lecomte MC. Spectrin-based skeleton as an actor in cell signaling. Cell Mol Life Sci 2011; 69:191-201. [PMID: 21877118 PMCID: PMC3249148 DOI: 10.1007/s00018-011-0804-5] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2011] [Revised: 08/08/2011] [Accepted: 08/10/2011] [Indexed: 01/12/2023]
Abstract
This review focuses on the recent advances in functions of spectrins in non-erythroid cells. We discuss new data concerning the commonly known role of the spectrin-based skeleton in control of membrane organization, stability and shape, and tethering protein mosaics to the cellular motors and to all major filament systems. Particular effort has been undertaken to highlight recent advances linking spectrin to cell signaling phenomena and its participation in signal transduction pathways in many cell types.
Collapse
Affiliation(s)
- B Machnicka
- University of Zielona Góra, Zielona Góra, Poland
| | | | | | | | | |
Collapse
|
25
|
Essential role for Abi1 in embryonic survival and WAVE2 complex integrity. Proc Natl Acad Sci U S A 2011; 108:7022-7. [PMID: 21482783 DOI: 10.1073/pnas.1016811108] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Abl interactor 1 (Abi1) plays a critical function in actin cytoskeleton dynamics through participation in the WAVE2 complex. To gain a better understanding of the specific role of Abi1, we generated a conditional Abi1-KO mouse model and MEFs lacking Abi1 expression. Abi1-KO cells displayed defective regulation of the actin cytoskeleton, and this dysregulation was ascribed to altered activity of the WAVE2 complex. Changes in motility of Abi1-KO cells were manifested by a decreased migration rate and distance but increased directional persistence. Although these phenotypes did not correlate with peripheral ruffling, which was unaffected, Abi1-KO cells exhibited decreased dorsal ruffling. Western blotting analysis of Abi1-KO cell lysates indicated reduced levels of the WAVE complex components WAVE1 and WAVE2, Nap1, and Sra-1/PIR121. Although relative Abi2 levels were more than doubled in Abi1-KO cells, the absolute Abi2 expression in these cells amounted only to a fifth of Abi1 levels in the control cell line. This finding suggests that the presence of Abi1 is critical for the integrity and stability of WAVE complex and that Abi2 levels are not sufficiently increased to compensate fully for the loss of Abi1 in KO cells and to restore the integrity and function of the WAVE complex. The essential function of Abi1 in WAVE complexes and their regulation might explain the observed embryonic lethality of Abi1-deficient embryos, which survived until approximately embryonic day 11.5 and displayed malformations in the developing heart and brain. Cells lacking Abi1 and the conditional Abi1-KO mouse will serve as critical models for defining Abi1 function.
Collapse
|
26
|
Yuditskaya S, Suffredini AF, Kato GJ. The proteome of sickle cell disease: insights from exploratory proteomic profiling. Expert Rev Proteomics 2010; 7:833-48. [PMID: 21142886 PMCID: PMC3068560 DOI: 10.1586/epr.10.88] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
The expanding realm of exploratory proteomics has added a unique dimension to the study of the complex pathophysiology involved in sickle cell disease. A review of proteomic studies published on sickle cell erythrocytes and plasma shows trends of upregulation of antioxidant proteins, an increase in cytoskeletal defects, an increase in protein repair and turnover components, a decrease in lipid raft proteins and apolipoprotein dysregulation. Many of these findings are consistent with the pathophysiology of sickle cell disease, including high oxidant burden, resulting in damage to cytoskeletal and other proteins, and erythrocyte rigidity. More unexpected findings, such as a decrease in lipid raft components and apolipoprotein dysregulation, offer previously unexplored targets for future investigation and potential therapeutic intervention. Exploratory proteomic profiling is a valuable source of hypothesis generation for the cellular and molecular pathophysiology of sickle cell disease.
Collapse
Affiliation(s)
- Susan Yuditskaya
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | |
Collapse
|
27
|
Baines AJ. The spectrin-ankyrin-4.1-adducin membrane skeleton: adapting eukaryotic cells to the demands of animal life. PROTOPLASMA 2010; 244:99-131. [PMID: 20668894 DOI: 10.1007/s00709-010-0181-1] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2010] [Accepted: 07/05/2010] [Indexed: 05/29/2023]
Abstract
The cells in animals face unique demands beyond those encountered by their unicellular eukaryotic ancestors. For example, the forces engendered by the movement of animals places stresses on membranes of a different nature than those confronting free-living cells. The integration of cells into tissues, as well as the integration of tissue function into whole animal physiology, requires specialisation of membrane domains and the formation of signalling complexes. With the evolution of mammals, the specialisation of cell types has been taken to an extreme with the advent of the non-nucleated mammalian red blood cell. These and other adaptations to animal life seem to require four proteins--spectrin, ankyrin, 4.1 and adducin--which emerged during eumetazoan evolution. Spectrin, an actin cross-linking protein, was probably the earliest of these, with ankyrin, adducin and 4.1 only appearing as tissues evolved. The interaction of spectrin with ankyrin is probably a prerequisite for the formation of tissues; only with the advent of vertebrates did 4.1 acquires the ability to bind spectrin and actin. The latter activity seems to allow the spectrin complex to regulate the cell surface accumulation of a wide variety of proteins. Functionally, the spectrin-ankyrin-4.1-adducin complex is implicated in the formation of apical and basolateral domains, in aspects of membrane trafficking, in assembly of certain signalling and cell adhesion complexes and in providing stability to otherwise mechanically fragile cell membranes. Defects in this complex are manifest in a variety of hereditary diseases, including deafness, cardiac arrhythmia, spinocerebellar ataxia, as well as hereditary haemolytic anaemias. Some of these proteins also function as tumor suppressors. The spectrin-ankyrin-4.1-adducin complex represents a remarkable system that underpins animal life; it has been adapted to many different functions at different times during animal evolution.
Collapse
Affiliation(s)
- Anthony J Baines
- School of Biosciences and Centre for Biomedical Informatics, University of Kent, Canterbury, CT2 7NJ, UK.
| |
Collapse
|
28
|
Dubielecka PM, Machida K, Xiong X, Hossain S, Ogiue-Ikeda M, Carrera AC, Mayer BJ, Kotula L. Abi1/Hssh3bp1 pY213 links Abl kinase signaling to p85 regulatory subunit of PI-3 kinase in regulation of macropinocytosis in LNCaP cells. FEBS Lett 2010; 584:3279-86. [PMID: 20598684 DOI: 10.1016/j.febslet.2010.06.029] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2010] [Revised: 06/09/2010] [Accepted: 06/13/2010] [Indexed: 12/25/2022]
Abstract
Macropinocytosis is regulated by Abl kinase via an unknown mechanism. We previously demonstrated that Abl kinase activity is, itself, regulated by Abi1 subsequent to Abl kinase phosphorylation of Abi1 tyrosine 213 (pY213) [1]. Here we show that blocking phosphorylation of Y213 abrogated the ability of Abl to regulate macropinocytosis, implicating Abi1 pY213 as a key regulator of macropinocytosis. Results from screening the human SH2 domain library and mapping the interaction site between Abi1 and the p85 regulatory domain of PI-3 kinase, coupled with data from cells transfected with loss-of-function p85 mutants, support the hypothesis that macropinocytosis is regulated by interactions between Abi1 pY213 and the C-terminal SH2 domain of p85-thereby linking Abl kinase signaling to p85-dependent regulation of macropinocytosis.
Collapse
|
29
|
Derivery E, Gautreau A. Generation of branched actin networks: assembly and regulation of the N-WASP and WAVE molecular machines. Bioessays 2010; 32:119-31. [PMID: 20091750 DOI: 10.1002/bies.200900123] [Citation(s) in RCA: 109] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The Arp2/3 complex is a molecular machine that generates branched actin networks responsible for membrane remodeling during cell migration, endocytosis, and other morphogenetic events. This machine requires activators, which themselves are multiprotein complexes. This review focuses on recent advances concerning the assembly of stable complexes containing the most-studied activators, N-WASP and WAVE proteins, and the level of regulation that is provided by these complexes. N-WASP is the paradigmatic auto-inhibited protein, which is activated by a conformational opening. Even though this regulation has been successfully reconstituted in vitro with isolated N-WASP, the native dimeric complex with a WIP family protein has unique additional properties. WAVE proteins are part of a pentameric complex, whose basal state and activated state when bound to the Rac GTPase were recently clarified. Moreover, this review attempts to put together diverse observations concerning the WAVE complex in the conceptual frame of an in vivo assembly pathway that has gained support from the recent identification of a precursor.
Collapse
Affiliation(s)
- Emmanuel Derivery
- CNRS UPR3082, Laboratoire d'Enzymologie et de Biochimie Structurales, Bât. 34, Avenue de la Terrasse, 91198 Gif-sur-Yvette Cedex, France
| | | |
Collapse
|
30
|
Dubielecka PM, Cui P, Xiong X, Hossain S, Heck S, Angelov L, Kotula L. Differential regulation of macropinocytosis by Abi1/Hssh3bp1 isoforms. PLoS One 2010; 5:e10430. [PMID: 20479892 PMCID: PMC2866655 DOI: 10.1371/journal.pone.0010430] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2010] [Accepted: 04/07/2010] [Indexed: 12/17/2022] Open
Abstract
Background Macropinocytosis, which is a constitutive cellular process of fluid and macromolecule uptake, is regulated by actin cytoskeleton rearrangements near the plasma membrane. Activation of Rac1, which is proposed to act upstream of the actin polymerization regulatory Wave 2 complex, has been found to correlate with enhanced macropinocytosis. One of the components of the Wave 2 complex is Abi1. Multiple, alternatively spliced isoforms of Abi1 are expressed in mammalian cells, but the functional significance of the various isoforms is unknown. Principal Findings Here, using flow cytometric assay analysis for Alexa Fluor 647, we demonstrate that Abi1 isoforms 2 and 3 differentially regulate macropinocytosis. LNCaP cells expressing isoform 3 had increased macropinocytic uptake that correlated with enhanced cell spreading and higher Rac1 activation in comparison to cells expressing isoform 2. Isoform 2 expressing cells had decreased macropinocytic uptake, but demonstrated greater sensitivity to Rac1 activation. Moreover, more isoform 2 was localized within the cytoplasm in comparison to isoform 3, which was more associated with the plasma membrane. Activated Rac1 was found to specifically bind to a site in exon 10 of isoform 2 in vitro. Because of alternative mRNA splicing, exon 10 is absent from isoform 3, precluding similar binding of activated Rac1. Both isoforms, however, bound to inactive Rac1 through the same non-exon 10 site. Thus, Abi1 isoform 3-containing Wave 2 complex exhibited a differential binding to activated vs. inactive Rac1, whereas isoform 2-containing Wave 2 complex bound activated or inactive Rac1 comparably. Conclusion Based on these observations, we postulate that Abi1 isoforms differentially regulate macropinocytosis as a consequence of their different relative affinities for activated Rac1 in Wave 2 complex. These findings also raise the possibility that isoform-specific roles occur in other Abi1 functions.
Collapse
Affiliation(s)
- Patrycja M. Dubielecka
- Laboratory of Cell Signaling, New York Blood Center, New York, New York, United States of America
| | - Ping Cui
- Laboratory of Cell Signaling, New York Blood Center, New York, New York, United States of America
| | - Xiaoling Xiong
- Laboratory of Cell Signaling, New York Blood Center, New York, New York, United States of America
| | - Sajjad Hossain
- Laboratory of Cell Signaling, New York Blood Center, New York, New York, United States of America
| | - Susanne Heck
- Flow Cytometry Core, New York Blood Center, New York, New York, United States of America
| | - Lyudmil Angelov
- Confocal Microscopy Laboratory, New York Blood Center, New York, New York, United States of America
| | - Leszek Kotula
- Laboratory of Cell Signaling, New York Blood Center, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
31
|
DCVs Exocytosis is Damaged in The Dominant Allele of β-G Spectrin Mutant in <I>C. elegans</I>*. PROG BIOCHEM BIOPHYS 2010. [DOI: 10.3724/sp.j.1206.2009.00365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
32
|
Derivery E, Lombard B, Loew D, Gautreau A. The Wave complex is intrinsically inactive. ACTA ACUST UNITED AC 2009; 66:777-90. [PMID: 19206172 DOI: 10.1002/cm.20342] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The Wave proteins activate the Arp2/3 complex at the leading edge of migrating cells. The resulting actin polymerization powers the projection of the plasma membrane in lamellipodia and membrane ruffles. The Wave proteins are always found associated with partner proteins. The canonical Wave complex is a stable complex containing five subunits. Even though it is well admitted that this complex plays an essential regulatory role on Wave function, the mechanisms by which Wave proteins are regulated within the complex are still elusive. Even the constitutive activity or inactivity of the complex is controversial. The major difficulty of these assays resides in the long and difficult purification of the Wave complex by a combination of several chromatography steps, which gives an overall low yield and increases the chance of Wave complex denaturation. Here we report a greatly simplified approach to purify the human Wave complex using a stable cell line expressing a tagged subunit and affinity chromatography. This protocol provided us with sufficient amount of pure Wave complex for functional assays. These assays unambiguously established that the Wave complex in its native conformation is intrinsically inactive, indicating that, like WASP proteins, Wave proteins have a masked C-terminal Arp2/3 binding site at resting state. As a consequence, the Wave complex has to be recruited and activated at the plasma membrane to project migration structures. Importantly, the approach we describe here for multiprotein complex purification is likely applicable to a wide range of human multiprotein complexes.
Collapse
Affiliation(s)
- Emmanuel Derivery
- Institut Curie, Centre de Recherche, Laboratory of Cell Morphogenesis and Intracellular Signaling, 75248 Paris Cedex 05, France
| | | | | | | |
Collapse
|
33
|
Abstract
Spectrin is a cytoskeletal protein thought to have descended from an alpha-actinin-like ancestor. It emerged during evolution of animals to promote integration of cells into tissues by assembling signalling and cell adhesion complexes, by enhancing the mechanical stability of membranes and by promoting assembly of specialized membrane domains. Spectrin functions as an (alphabeta([H]))(2) tetramer that cross-links transmembrane proteins, membrane lipids and the actin cytoskeleton, either directly or via adaptor proteins such as ankyrin and 4.1. In the present paper, I review recent findings on the origins and adaptations in this system. (i) The genome of the choanoflagellate Monosiga brevicollis encodes alpha-, beta- and beta(Heavy)-spectrin, indicating that spectrins evolved in the immediate unicellular precursors of animals. (ii) Ankyrin and 4.1 are not encoded in that genome, indicating that spectrin gained function during subsequent animal evolution. (iii) Protein 4.1 gained a spectrin-binding activity in the evolution of vertebrates. (iv) Interaction of chicken or mammal beta-spectrin with PtdInsP(2) can be regulated by differential mRNA splicing, which can eliminate the PH (pleckstrin homology) domain in betaI- or betaII-spectrins; in the case of mammalian betaII-spectrin, the alternative C-terminal region encodes a phosphorylation site that regulates interaction with alpha-spectrin. (v) In mammalian evolution, the single pre-existing alpha-spectrin gene was duplicated, and one of the resulting pair (alphaI) neo-functionalized for rapid make-and-break of tetramers. I hypothesize that the elasticity of mammalian non-nucleated erythrocytes depends on the dynamic rearrangement of spectrin dimers/tetramers under the shearing forces experienced in circulation.
Collapse
|
34
|
Pasini EM, Lutz HU, Mann M, Thomas AW. Red blood cell (RBC) membrane proteomics--Part I: Proteomics and RBC physiology. J Proteomics 2009; 73:403-20. [PMID: 19540949 DOI: 10.1016/j.jprot.2009.06.005] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2009] [Revised: 05/29/2009] [Accepted: 06/04/2009] [Indexed: 10/20/2022]
Abstract
Membrane proteomics is concerned with accurately and sensitively identifying molecules involved in cell compartmentalisation, including those controlling the interface between the cell and the outside world. The high lipid content of the environment in which these proteins are found often causes a particular set of problems that must be overcome when isolating the required material before effective HPLC-MS approaches can be performed. The membrane is an unusually dynamic cellular structure since it interacts with an ever changing environment. A full understanding of this critical cell component will ultimately require, in addition to proteomics, lipidomics, glycomics, interactomics and study of post-translational modifications. Devoid of nucleus and organelles in mammalian species other than camelids, and constantly in motion in the blood stream, red blood cells (RBCs) are the sole mammalian oxygen transporter. The fact that mature mammalian RBCs have no internal membrane-bound organelles, somewhat simplifies proteomics analysis of the plasma membrane and the fact that it has no nucleus disqualifies microarray based methods. Proteomics has the potential to provide a better understanding of this critical interface, and thereby assist in identifying new approaches to diseases.
Collapse
Affiliation(s)
- Erica M Pasini
- Biomedical Primate Research Centre, Lange Kleiweg 139, 2288 GJ Rijswijk, The Netherlands
| | | | | | | |
Collapse
|
35
|
Lefferts JA, Wang C, Sridharan D, Baralt M, Lambert MW. The SH3 domain of alphaII spectrin is a target for the Fanconi anemia protein, FANCG. Biochemistry 2009; 48:254-63. [PMID: 19102630 DOI: 10.1021/bi801483u] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The structural protein nonerythroid alpha spectrin (alphaIISp) plays a role in the repair of DNA interstrand cross-links and is deficient in cells from patients with Fanconi anemia (FA), in which there is a defect in ability to repair such cross-links. We have proposed a model in which alphaIISp, whose stability is dependent on FA proteins, acts as a scaffold to aid in recruitment of repair proteins to sites of damage. In order to get a clearer understanding of the proposed role of FA proteins in maintaining stability of alphaIISp, yeast two-hybrid analysis was carried out to determine whether FA proteins directly interact with alphaIISp and, if so, to map the sites of interaction. Four overlapping regions of alphaIISp were constructed. FANCG interacted with one of these regions and specifically with the SH3 domain in this region of alphaIISp. The site of interaction in FANCG was mapped to a motif that binds to SH3 domains and contains a consensus sequence with preference for the SH3 domain of alphaIISp. This site of interaction was confirmed using site-directed mutagenesis. Two FA proteins that did not contain motifs that bind to SH3 domains, FANCC and FANCF, did not interact with the SH3 domain of alphaIISp. These results demonstrate that one of the FA proteins, FANCG, contains a motif that interacts directly with the SH3 domain of alphaIISp. We propose that this binding of FANCG to alphaIISp may be important for the stability of alphaIISp in cells and the role alphaIISp plays in the DNA repair process.
Collapse
Affiliation(s)
- Joel A Lefferts
- Department of Pathology and Laboratory Medicine, UMDNJNew Jersey Medical School and Graduate School of Biomedical Sciences, Newark, New Jersey 07103, USA
| | | | | | | | | |
Collapse
|
36
|
Yu W, Sun X, Clough N, Cobos E, Tao Y, Dai Z. Abi1 gene silencing by short hairpin RNA impairs Bcr-Abl-induced cell adhesion and migration in vitro and leukemogenesis in vivo. Carcinogenesis 2008; 29:1717-1724. [PMID: 18453543 PMCID: PMC2527646 DOI: 10.1093/carcin/bgn098] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2007] [Revised: 04/03/2008] [Accepted: 04/08/2008] [Indexed: 12/28/2022] Open
Abstract
Abl interactor (Abi) 1 was first identified as the downstream target of Abl tyrosine kinases and was found to be dysregulated in leukemic cells expressing oncogenic Bcr-Abl and v-Abl. Although the accumulating evidence supports a role of Abi1 in actin cytoskeleton remodeling and growth factor/receptor signaling, it is not clear how it contributes to Bcr-Abl-induced leukemogenesis. We show here that Abi1 gene silencing by short hairpin RNA attenuated the Bcr-Abl-induced abnormal actin remodeling, membrane-type 1 metalloproteinase clustering and inhibited cell adhesion and migration on fibronectin-coated surfaces. Although the knock down of Abi1 expression did not affect growth factor-independent growth of Bcr-Abl-transformed Ba/F3 cells in vitro, it impeded competitive expansion of these cells in non obese diabetic (NOD)/ severe combined immuno-deficiency (SCID) mice. Remarkably, the knock down of Abi1 expression in Bcr-Abl-transformed Ba/F3 cells impaired the leukemogenic potential of these cells in NOD/SCID mice. Abi1 contributes to Bcr-Abl-induced leukemogenesis in part through Src family kinases, as the knock down of Abi1 expression attenuates Bcr-Abl-stimulated activation of Lyn. Together, these data provide for the first time the direct evidence that supports a critical role of Abi1 pathway in the pathogenesis of Bcr-Abl-induced leukemia.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing/antagonists & inhibitors
- Adaptor Proteins, Signal Transducing/genetics
- Adaptor Proteins, Signal Transducing/metabolism
- Animals
- Blotting, Western
- Cell Adhesion/physiology
- Cell Movement/physiology
- Cell Transformation, Neoplastic
- Cytoskeletal Proteins/antagonists & inhibitors
- Cytoskeletal Proteins/genetics
- Cytoskeletal Proteins/metabolism
- Female
- Flow Cytometry
- Fusion Proteins, bcr-abl/physiology
- Gene Silencing
- Immunoprecipitation
- Leukemia/genetics
- Leukemia/pathology
- Matrix Metalloproteinase 14/metabolism
- Mice
- Mice, Inbred NOD
- Mice, SCID
- Microscopy, Fluorescence
- Phosphorylation
- Precursor Cells, B-Lymphoid/pathology
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- RNA, Small Interfering/pharmacology
- Retroviridae/genetics
- Reverse Transcriptase Polymerase Chain Reaction
- Survival Rate
- Tyrosine/metabolism
- src-Family Kinases/metabolism
Collapse
Affiliation(s)
- Weidong Yu
- Department of Internal Medicine, Texas Tech University Health Sciences Center, 1400 Wallace Boulevard, Amarillo, TX 79106, USA
- Institute of Clinical Molecular Biology, People's Hospital, Peking University, Beijing 100044, People's Republic of China
| | - Xiaolin Sun
- Department of Internal Medicine, Texas Tech University Health Sciences Center, 1400 Wallace Boulevard, Amarillo, TX 79106, USA
| | - Nancy Clough
- Division of Medical Oncology, University of Colorado at Denver and Health Sciences Center, Aurora, CO 80010, USA
| | - Everardo Cobos
- Department of Internal Medicine, Texas Tech University Health Sciences Center, 1400 Wallace Boulevard, Amarillo, TX 79106, USA
- Stem Cell Transplant Program, Texas Tech University Health Sciences Center, 1400 Wallace Boulevard, Amarillo, TX 79106, USA
| | - Yunxia Tao
- Department of Internal Medicine, Texas Tech University Health Sciences Center, 1400 Wallace Boulevard, Amarillo, TX 79106, USA
| | - Zonghan Dai
- Department of Internal Medicine, Texas Tech University Health Sciences Center, 1400 Wallace Boulevard, Amarillo, TX 79106, USA
- Stem Cell Transplant Program, Texas Tech University Health Sciences Center, 1400 Wallace Boulevard, Amarillo, TX 79106, USA
| |
Collapse
|
37
|
Xiong X, Cui P, Hossain S, Xu R, Warner B, Guo X, An X, Debnath AK, Cowburn D, Kotula L. Allosteric inhibition of the nonMyristoylated c-Abl tyrosine kinase by phosphopeptides derived from Abi1/Hssh3bp1. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2008; 1783:737-47. [PMID: 18328268 DOI: 10.1016/j.bbamcr.2008.01.028] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2007] [Revised: 01/09/2008] [Accepted: 01/28/2008] [Indexed: 11/25/2022]
Abstract
Here we report c-Abl kinase inhibition mediated by a phosphotyrosine located in trans in the c-Abl substrate, Abi1. The mechanism, which is pertinent to the nonmyristoylated c-Abl kinase, involves high affinity concurrent binding of the phosphotyrosine pY213 to the Abl SH2 domain and binding of a proximal PXXP motif to the Abl SH3 domain. Abi1 regulation of c-Abl in vivo appears to play a critical role, as demonstrated by inhibition of pY412 phosphorylation of the nonmyristoylated Abl by coexpression of Abi1. Pervanadate-induced c-Abl kinase activity was also reduced upon expression of the wild type Abi1 but not by expression of the Y213 to F213 mutant Abi1 in LNCaP cells, which are naturally deficient in the regulatory pY213. Our findings suggest a novel mechanism by which Abl kinase is regulated in cells.
Collapse
Affiliation(s)
- Xiaoling Xiong
- Laboratory of Cell Signaling, New York Blood Center, New York, NY 10065, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Campa F, Machuy N, Klein A, Rudel T. A new interaction between Abi-1 and betaPIX involved in PDGF-activated actin cytoskeleton reorganisation. Cell Res 2008; 16:759-70. [PMID: 16940963 DOI: 10.1038/sj.cr.7310091] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Members of the Rho family of GTPases are key regulators of the actin cytoskeleton. In particular, activated Rac1 stimulates membrane dorsal ruffle formation in response to platelet-derived growth factor (PDGF). Abl-interactor (Abi)-1 and betaPIX, a guanine nucleotide exchange factor for Rac1, localise at these Rac1-induced actin structures and play important roles in the induction of membrane dorsal ruffling in response to PDGF in fibroblasts. Here, we demonstrate a novel interaction between Abi-1 and betaPIX using the yeast two-hybrid system, in vitro pull-down assays, and in vivo co-immunoprecipitation experiments. In vitro, the C-terminal fragment of betaPIX interacted with Abi-1, while in vivo the N-terminal fragment of betaPIX interacted with Abi-1. The biological function of this interaction was investigated in mouse fibroblasts in response to PDGF stimulation. Abi-1 and betaPIX co-localised in the cytoplasm and to membrane dorsal ruffles after PDGF treatment. We show that the co-expression of Abi-1 and truncated forms of betaPIX in mouse fibroblasts blocked PDGF-induced membrane dorsal ruffles. Together, these results show that the interaction between Abi-1 and betaPIX is involved in the formation of growth factor-induced membrane dorsal ruffles.
Collapse
Affiliation(s)
- Fanny Campa
- Max Planck Institute for Infection Biology, Department of Molecular Biology, Campus Charité Mitte, Berlin, Germany
| | | | | | | |
Collapse
|
39
|
Machuy N, Campa F, Thieck O, Rudel T. c-Abl-binding protein interacts with p21-activated kinase 2 (PAK-2) to regulate PDGF-induced membrane ruffles. J Mol Biol 2007; 370:620-32. [PMID: 17543336 DOI: 10.1016/j.jmb.2007.04.080] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2006] [Revised: 03/21/2007] [Accepted: 04/25/2007] [Indexed: 11/25/2022]
Abstract
p21-Activated kinases (PAKs) are serine/threonine kinases involved in multiple cellular functions including cytoskeleton regulation, proliferation and apoptosis. We performed a screen for proteins interacting with PAK-2, a ubiquitously expressed kinase involved in apoptotic signaling. Among the PAK-2 interacting proteins were different members of the Abl-binding protein family. Abl-binding proteins bound to a proline-rich region of PAK-2 located in the regulatory N terminus. Moreover, active PAK-2 phosphorylated Abl-binding proteins in vitro. Interestingly, we show that PAK-2 also interacted with c-Abl but via a different domain than with the Abl-binding proteins. PAK-2 and Abi-1 co-localized in the cytoplasm and to membrane dorsal ruffles induced by PDGF treatment. Expression of mutant PAK-2 deficient in binding to Abl-binding proteins or silencing of PAK-2 expression prevented the formation of membrane dorsal ruffles in response to PDGF. Our findings define a new class of PAK-interacting proteins, which play an important role in actin cytoskeletal reorganization.
Collapse
Affiliation(s)
- Nikolaus Machuy
- Max Planck Institute for Infection Biology, Department of Molecular Biology, Charitéplatz 1, D-10117 Berlin, Germany
| | | | | | | |
Collapse
|
40
|
Li Y, Clough N, Sun X, Yu W, Abbott BL, Hogan CJ, Dai Z. Bcr-Abl induces abnormal cytoskeleton remodeling, beta1 integrin clustering and increased cell adhesion to fibronectin through the Abl interactor 1 pathway. J Cell Sci 2007; 120:1436-1446. [PMID: 17389688 PMCID: PMC1950936 DOI: 10.1242/jcs.03430] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Hematopoietic cells isolated from patients with Bcr-Abl-positive leukemia exhibit multiple abnormalities of cytoskeletal and integrin function. These abnormalities are thought to play a role in the pathogenesis of leukemia; however, the molecular events leading to these abnormalities are not fully understood. We show here that the Abi1 pathway is required for Bcr-Abl to stimulate actin cytoskeleton remodeling, integrin clustering and cell adhesion. Expression of Bcr-Abl induces tyrosine phosphorylation of Abi1. This is accompanied by a subcellular translocation of Abi1/WAVE2 to a site adjacent to membrane, where an F-actin-enriched structure containing the adhesion molecules such as beta1-integrin, paxillin and vinculin is assembled. Bcr-Abl-induced membrane translocation of Abi1/WAVE2 requires direct interaction between Abi1 and Bcr-Abl, but is independent of the phosphoinositide 3-kinase pathway. Formation of the F-actin-rich complex correlates with an increased cell adhesion to fibronectin. More importantly, disruption of the interaction between Bcr-Abl and Abi1 by mutations either in Bcr-Abl or Abi1 not only abolished tyrosine phosphorylation of Abi1 and membrane translocation of Abi1/WAVE2, but also inhibited Bcr-Abl-stimulated actin cytoskeleton remodeling, integrin clustering and cell adhesion to fibronectin. Together, these data define Abi1/WAVE2 as a downstream pathway that contributes to Bcr-Abl-induced abnormalities of cytoskeletal and integrin function.
Collapse
Affiliation(s)
- Yingzhu Li
- Department of Medicine, University of Colorado at Denver and Health Sciences Center, Aurora, CO 80045
| | - Nancy Clough
- Department of Medicine, University of Colorado at Denver and Health Sciences Center, Aurora, CO 80045
| | - Xiaolin Sun
- Department of Medicine, University of Colorado at Denver and Health Sciences Center, Aurora, CO 80045
| | - Weidong Yu
- Department of Medicine, University of Colorado at Denver and Health Sciences Center, Aurora, CO 80045
| | - Brian L. Abbott
- Department of Medicine, University of Colorado at Denver and Health Sciences Center, Aurora, CO 80045
| | - Christopher J. Hogan
- Department of Medicine, University of Colorado at Denver and Health Sciences Center, Aurora, CO 80045
- University of Colorado Cancer Center, University of Colorado at Denver and Health Sciences Center, Aurora, CO 80045
| | - Zonghan Dai
- Department of Medicine, University of Colorado at Denver and Health Sciences Center, Aurora, CO 80045
- Cell and Developmental Biology, University of Colorado at Denver and Health Sciences Center, Aurora, CO 80045
- University of Colorado Cancer Center, University of Colorado at Denver and Health Sciences Center, Aurora, CO 80045
| |
Collapse
|
41
|
Jefferson JJ, Ciatto C, Shapiro L, Liem RKH. Structural analysis of the plakin domain of bullous pemphigoid antigen1 (BPAG1) suggests that plakins are members of the spectrin superfamily. J Mol Biol 2006; 366:244-57. [PMID: 17161423 PMCID: PMC1850962 DOI: 10.1016/j.jmb.2006.11.036] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2006] [Revised: 11/03/2006] [Indexed: 11/21/2022]
Abstract
Bullous pemphigoid antigen 1 (BPAG1) is a member of the plakin family of proteins. The plakins are multi-domain proteins that have been shown to interact with microtubules, actin filaments and intermediate filaments, as well as proteins found in cellular junctions. These interactions are mediated through different domains on the plakins. The interactions between plakins and components of specialized cell junctions such as desmosomes and hemidesmosomes are mediated through the so-called plakin domain, which is a common feature of the plakins. We report the crystal structure of a stable fragment from BPAG1, residues 226-448, defined by limited proteolysis of the whole plakin domain. The structure, determined by single-wavelength anomalous diffraction phasing from a selenomethionine-substituted crystal at 3.0 A resolution, reveals a tandem pair of triple helical bundles closely related to spectrin repeats. Based on this structure and analysis of sequence conservation, we propose that the architecture of plakin domains is defined by two pairs of spectrin repeats interrupted by a putative Src-Homology 3 (SH3) domain.
Collapse
Affiliation(s)
- Julius J Jefferson
- Department of Pathology and Cell Biology, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA
| | | | | | | |
Collapse
|
42
|
Bournier O, Kroviarski Y, Rotter B, Nicolas G, Lecomte MC, Dhermy D. Spectrin interacts with EVL (Enabled/vasodilator-stimulated phosphoprotein-like protein), a protein involved in actin polymerization. Biol Cell 2006; 98:279-93. [PMID: 16336193 DOI: 10.1042/bc20050024] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
BACKGROUND INFORMATION The alpha- and beta-spectrin chains constitute the filaments of the spectrin-based skeleton, which was first identified in erythrocytes. The discovery of analogous structures at plasma membranes of eukaryotic cells has led to investigations of the role of this spectrin skeleton in many cellular processes. The alphaII-spectrin chain expressed in nucleated cells harbours in its central region several functional motifs, including an SH3 (Src homology 3) domain. RESULTS Using yeast two-hybrid screening, we have identified EVL [Enabled/VASP (vasodilator-stimulated phosphoprotein)-like protein] as a new potential partner of the alphaII-spectrin SH3 domain. In the present study, we investigated the interaction of the alphaII-spectrin SH3 domain with EVL and compared this with other proteins related to EVL [Mena (mammalian Enabled) and VASP]. We confirmed the in vitro interaction between EVL and the alphaII-spectrin SH3 domain by GST (glutathione S-transferase) pull-down assays, and showed that the co-expression of EVL with the alphaII-spectrin SH3 domain in COS-7 cells resulted in the partial delocalization of the SH3 domain from cytoplasm to filopodia and lamellipodia, where it was co-localized with EVL. In kidney epithelial and COS-7 cells, we demonstrated the co-immunoprecipitation of the alphaII-spectrin chain with over-expressed EVL. Immunofluorescence studies showed that the over-expression of EVL in COS-7 cells promoted the formation of filopodia and lamellipodia, and the expressed EVL was detected in filopodial tips and the leading edge of lamellipodia. In these cells over-expressing EVL, the alphaII-spectrin membrane labelling lagged behind EVL staining in lamellipodia and filopodia, with co-localization of these two stains in the contact area. In kidney epithelial cell lines, focused co-localization of spectrin with expressed EVL was observed in the membrane of the lateral domain, where the cell-cell contacts are reinforced. CONCLUSIONS The possible link between the spectrin-based skeleton and actin via the EVL protein suggests a new way of integrating the spectrin-based skeleton in areas of dynamic actin reorganization.
Collapse
|
43
|
Luo X, Levens E, Williams RS, Chegini N. The expression of Abl interactor 2 in leiomyoma and myometrium and regulation by GnRH analogue and transforming growth factor-β. Hum Reprod 2006; 21:1380-6. [PMID: 16488906 DOI: 10.1093/humrep/del011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Abelson (Abl) interactor 2 (Abi-2) has been considered as a key regulator of cell/tissue structural organization and is differentially expressed in leiomyomas. The objective of this study was to evaluate the expression of Abi-2 in leiomyoma/myometrium during the menstrual cycle and following GnRH analogue (GnRHa) therapy, as well as regulation by transforming growth factor (TGF)-beta1 in leiomyoma and myometrial smooth muscle cells (LSMC and MSMC). METHODS We used real-time PCR, Western blotting and immunohistochemistry to determine the expression of Abi-2 in paired leiomyoma and myometrium (n = 27) from proliferative (n = 8) and secretory (n = 12) phases of the menstrual cycle and from patients who received GnRHa therapy (n = 7). Time-dependent action of TGF-beta1 (2.5 ng/ml) and GnRHa (0.1 microM) on Abi-2 expression was determined in LSMC and MSMC. RESULTS Leiomyomas express elevated levels of Abi-2 as compared with myometrium from the proliferative but not the secretory phase of the menstrual cycle, with a significant reduction following GnRHa therapy (P < 0.05). Western blotting showed a similar trend in Abi-2 protein expression in leiomyoma/myometrial tissue extracts, which was immunolocalized in LSMC and MSMC, connective tissue fibroblasts and arterial walls. The expression of Abi-2 in LSMC and MSMC was increased by TGF-beta1 (2.5 ng/ml) and was inhibited by GnRHa (0.1 microM) in a time- and cell-dependent manner, and pretreatment with Smad3 SiRNA and U0126, an MEK-1/2 inhibitor, respectively, reversed their actions. CONCLUSION Based on the menstrual cycle-dependent expression, the influence of GnRHa therapy, and regulation by TGF-beta in LSMC/MSMC, we conclude that Abi-2 may have a key regulatory function in leiomyomas cellular/tissue structural organization during growth and regression.
Collapse
Affiliation(s)
- Xiaoping Luo
- Department of Obstetrics and Gynecology, University of Florida, Gainesville, FL, USA
| | | | | | | |
Collapse
|
44
|
Abstract
Transforming growth factor beta (TGF-beta) signaling leads to a number of biological end points involving cell growth, differentiation, and morphogenesis. Typically, the cellular effect accompanies an induction of mesodermal cell fate and inhibition of neural cell differentiation. However, during pathological conditions, these defined effects of TGF-beta can be reversed; for example, the growth-inhibitory effect is replaced with its tumor promoting ability. A multitude of factors and cross-signaling pathways have been reported to be involved in modulating the dual effects of TGF-beta. In this review, we focus on the potential role of TGF-beta signal transduction during development of neural progenitor cells and its relation to glioblastoma development from neural stem cells.
Collapse
Affiliation(s)
- Nady Golestaneh
- School of Medicine, Georgetown University, 3900 Reservoir Road NW, Washington, DC 20007, USA
| | | |
Collapse
|
45
|
Tsujio I, Zaidi T, Xu J, Kotula L, Grundke-Iqbal I, Iqbal K. Inhibitors of protein phosphatase-2A from human brain structures, immunocytological localization and activities towards dephosphorylation of the Alzheimer type hyperphosphorylated tau. FEBS Lett 2005; 579:363-72. [PMID: 15642345 DOI: 10.1016/j.febslet.2004.11.097] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2004] [Revised: 11/10/2004] [Accepted: 11/30/2004] [Indexed: 10/26/2022]
Abstract
Protein phosphatase (PP)-2A, which regulates the phosphorylation of tau, is regulated by two endogenous inhibitor proteins, I(1)(PP2A) and I(2)(PP2A), in mammalian tissues. Here, we report the cloning of I(1)(PP2A) and I(2)(PP2A) from human brain, and show that in PC12 cells and in I(1)(PP2A)-GFP or I(2)(PP2A)-GFP transfected NIH3T3 and human neural progenitor cells, I(1)(PP2A) is localized mostly in the cell cytoplasm and I(2)(PP2A) mostly in the nucleus. The recombinant I(1)(PP-2A) and I(2)(PP-2A) inhibit PP-2A activity towards hyperphosphorylated tau in vitro; the dephosphorylation of the hyperphosphorylated tau at specific sites is selectively inhibited. Overexpression of I(1)(PP2A) as well as I(2)(PP2A) results in tau hyperphosphorylation and degeneration of PC 12 cells.
Collapse
Affiliation(s)
- Ichiro Tsujio
- Department of Neurochemistry, New York State Institute for Basic Research in Developmental Disabilities, 1050 Forest Hill Road, Staten Island, NY 10314-6399, USA
| | | | | | | | | | | |
Collapse
|
46
|
Inclusion of Scar/WAVE3 in a similar complex to Scar/WAVE1 and 2. BMC Cell Biol 2005; 6:11. [PMID: 15752430 PMCID: PMC555569 DOI: 10.1186/1471-2121-6-11] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2004] [Accepted: 03/07/2005] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The Scar/WAVE family of proteins mediates signals to actin assembly by direct activation of the Arp2/3 complex. These proteins have been characterised as major regulators of lamellipodia formation downstream of Rac activation and as members of large protein complexes. RESULTS We have investigated the interactions of the three human Scar/WAVE isoforms with several previously described binding partners for Scar/WAVE 1 or 2. We find that all three Scar/WAVE isoforms behave similarly and are likely to participate in the same kinds of protein complexes that regulate actin assembly. CONCLUSION Differences between Scar/WAVE proteins are therefore likely to be at the level of tissue distribution or subtle differences in the affinity for specific binding partners.
Collapse
|
47
|
Dubielecka PM, Jaźwiec B, Potoczek S, Wróbel T, Miłoszewska J, Haus O, Kuliczkowski K, Sikorski AF. Changes in spectrin organisation in leukaemic and lymphoid cells upon chemotherapy. Biochem Pharmacol 2005; 69:73-85. [PMID: 15588716 DOI: 10.1016/j.bcp.2004.08.031] [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: 03/05/2004] [Accepted: 08/31/2004] [Indexed: 12/13/2022]
Abstract
The aim of the present study was to investigate changes in spectrin and protein kinase C theta; (PKC theta;) organisation in human lymphoid and leukaemic cells undergoing chemotherapeutically induced apoptosis. An analysis of spectrin arrangement in human peripheral lymphoid (non-Hodgkin lymphoma) and leukaemic (acute lymphoblastic leukaemia) cells before and after chemotherapy revealed radical differences in the distribution of this protein. By using immunofluorescent technique, in lymphocytes isolated before chemotherapy, we found spectrin evenly distributed in the cytoplasm and the plasma membrane, while after the therapy changes in spectrin organisation occurred. Moreover, in lymphocytes after chemotherapy, extraction with buffer containing non-ionic detergent (Triton X-100) revealed presence of an insoluble fraction of spectrin. In normal or malignant cells before chemotherapy spectrin was totally soluble, however it should be mentioned that in total cell extracts and supernatants (but not in pellets) apoptotic fragments of spectrin (in addition to intact alpha and beta chains) were also found. In malignant cells after chemotherapy changes in PKC theta; organisation, similar to this observed in the case of spectrin, were shown by the immunofluorescence technique. In contrast, no differences in the distribution of other isoforms of protein kinase C: betaI and betaII, before and after chemotherapy, were found. Apoptotic phosphatidyloserine (PS) externalisation, as well as cell shrinkage, membrane protrusions and blebbing were observed in lymphocytes after chemotherapy and treatment with cytostatics in vitro. The overall results may suggest that spectrin redistribution/aggregation is the phenomenon involved in programmed cell death (PCD) of normal and neoplastic lymphocytes and lymphoblasts, however molecular basis of this phenomenon should be further investigated.
Collapse
Affiliation(s)
- Patrycja M Dubielecka
- Laboratory of Cytobiochemistry, Institute of Biochemistry and Molecular Biology, University of Wrocław, S. Przybyszewskiego 63/77, 51-148 Wrocław, Poland
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Cestra G, Toomre D, Chang S, De Camilli P. The Abl/Arg substrate ArgBP2/nArgBP2 coordinates the function of multiple regulatory mechanisms converging on the actin cytoskeleton. Proc Natl Acad Sci U S A 2005; 102:1731-6. [PMID: 15659545 PMCID: PMC547834 DOI: 10.1073/pnas.0409376102] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
ArgBP2, and its brain-specific splice variant, nArgBP2, are interactors and substrates of Abl/Arg tyrosine kinases and of the ubiquitin ligase Cbl. They are members of a family of adaptor proteins that colocalize with actin on stress fibers and at cell-adhesion sites, including neuronal synapses. We show here that their NH2-terminal region, which contains a sorbin homology domain domain, interacts with spectrin, and we identify binding proteins for their COOH-terminal SH3 domains. All these binding partners participate in the regulation of the actin cytoskeleton. These include dynamin, synaptojanin, and WAVE isoforms, as well as WAVE regulatory proteins. At least two of the ArgBP2/nArgBP2 binding partners, synaptojanin 2B and WAVE2, undergo ubiquitination and Abl-dependent tyrosine phosphorylation. ArgBP2/nArgBP2 knockdown in astrocytes produces a redistribution of focal adhesion proteins and an increase in peripheral actin ruffles, whereas nArgBP2 overexpression produces a collapse of the actin cytoskeleton. Thus, ArgBP2/nArgBP2 is a scaffold protein that control the balance between adhesion and motility by coordinating the function of multiple signaling pathways converging on the actin cytoskeleton.
Collapse
Affiliation(s)
- Gianluca Cestra
- Department of Cell Biology and Howard Hughes Medical Institute, Yale University School of Medicine, 295 Congress Avenue, New Haven, CT 06510, USA
| | | | | | | |
Collapse
|
49
|
Grove M, Demyanenko G, Echarri A, Zipfel PA, Quiroz ME, Rodriguiz RM, Playford M, Martensen SA, Robinson MR, Wetsel WC, Maness PF, Pendergast AM. ABI2-deficient mice exhibit defective cell migration, aberrant dendritic spine morphogenesis, and deficits in learning and memory. Mol Cell Biol 2004; 24:10905-22. [PMID: 15572692 PMCID: PMC533973 DOI: 10.1128/mcb.24.24.10905-10922.2004] [Citation(s) in RCA: 119] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The Abl-interactor (Abi) family of adaptor proteins has been linked to signaling pathways involving the Abl tyrosine kinases and the Rac GTPase. Abi proteins localize to sites of actin polymerization in protrusive membrane structures and regulate actin dynamics in vitro. Here we demonstrate that Abi2 modulates cell morphogenesis and migration in vivo. Homozygous deletion of murine abi2 produced abnormal phenotypes in the eye and brain, the tissues with the highest Abi2 expression. In the absence of Abi2, secondary lens fiber orientation and migration were defective in the eye, without detectable defects in proliferation, differentiation, or apoptosis. These phenotypes were consistent with the localization of Abi2 at adherens junctions in the developing lens and at nascent epithelial cell adherens junctions in vitro. Downregulation of Abi expression by RNA interference impaired adherens junction formation and correlated with downregulation of the Wave actin-nucleation promoting factor. Loss of Abi2 also resulted in cell migration defects in the neocortex and hippocampus, abnormal dendritic spine morphology and density, and severe deficits in short- and long-term memory. These findings support a role for Abi2 in the regulation of cytoskeletal dynamics at adherens junctions and dendritic spines, which is critical for intercellular connectivity, cell morphogenesis, and cognitive functions.
Collapse
Affiliation(s)
- Matthew Grove
- Duke University Medical Center, Department of Pharmacology and Cancer Biology, Box 3813, Durham, NC 27710, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Chattopadhyay A, Rawat SS, Kelkar DA, Ray S, Chakrabarti A. Organization and dynamics of tryptophan residues in erythroid spectrin: novel structural features of denatured spectrin revealed by the wavelength-selective fluorescence approach. Protein Sci 2004; 12:2389-403. [PMID: 14573853 PMCID: PMC2366958 DOI: 10.1110/ps.03302003] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
We have investigated the organization and dynamics of the functionally important tryptophan residues of erythroid spectrin in native and denatured conditions utilizing the wavelength-selective fluorescence approach. We observed a red edge excitation shift (REES) of 4 nm for the tryptophans in the case of spectrin in its native state. This indicates that tryptophans in spectrin are localized in a microenvironment of restricted mobility, and that the regions surrounding the spectrin tryptophans offer considerable restriction to the reorientational motion of the water dipoles around the excited state tryptophans. Interestingly, spectrin exhibits a REES of 3 nm even when denatured in 8 M urea. This represents the first report of a denatured protein displaying REES. Observation of REES in the denatured state implies that some of the structural and dynamic features of this microenvironment around the spectrin tryptophans are retained even when the protein is denatured. Fluorescence quenching data of denatured spectrin support this conclusion. In addition, we have deduced the organization and dynamics of the hydrophobic binding site of the polarity-sensitive fluorescent probe PRODAN that binds erythroid spectrin with high affinity. When bound to spectrin, PRODAN exhibits a REES of 9 nm. Because PRODAN binds to a hydrophobic site in spectrin, such a result would directly imply that this region of spectrin offers considerable restriction to the reorientational motion of the solvent dipoles around the excited state fluorophore. The results of our study could provide vital insight into the role of tryptophans in the stability and folding of spectrin.
Collapse
|