1
|
Kokusho R, Katsuma S. Baculoviruses remodel the cytoskeleton of insect hemocytes to breach the host basal lamina. Commun Biol 2025; 8:268. [PMID: 40011612 PMCID: PMC11865517 DOI: 10.1038/s42003-025-07579-x] [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: 01/08/2024] [Accepted: 01/20/2025] [Indexed: 02/28/2025] Open
Abstract
Many pathogens and endosymbionts hijack the host's cytoskeleton for efficient propagation and transfer within or between host cells. Once released into the host's circulatory system, however, they have to confront structural barriers without utilizing host cell functions. Many insect viruses and insect-borne viruses can re-enter from the hemolymph into insect tissues despite the barrier of the basal lamina (BL), but the molecular mechanism remains unclear in many cases. Here, we demonstrate that Bombyx mori nucleopolyhedrovirus (BmNPV) remodels host hemocytes to breach the BL. We found that the viral membrane protein actin rearrangement-inducing factor 1 (ARIF-1) induces filopodia-like protrusions and invadosome-like structures in hemocytes, which play a critical role in attaching to the tissue surface, penetrating the tracheal BL and thus facilitating the transport of viral nucleocapsids into host tissues. Our findings clearly show the role of hemocyte infection in viral systemic spread and its molecular basis.
Collapse
Affiliation(s)
- Ryuhei Kokusho
- Department of Agricultural and Environmental Biology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan.
| | - Susumu Katsuma
- Department of Agricultural and Environmental Biology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
2
|
Halpin JC, Keating AE. PairK: Pairwise k-mer alignment for quantifying protein motif conservation in disordered regions. Protein Sci 2025; 34:e70004. [PMID: 39720898 DOI: 10.1002/pro.70004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 11/19/2024] [Accepted: 12/05/2024] [Indexed: 12/26/2024]
Abstract
Protein-protein interactions are often mediated by a modular peptide recognition domain binding to a short linear motif (SLiM) in the disordered region of another protein. To understand the features of SLiMs that are important for binding and to identify motif instances that are important for biological function, it is useful to examine the evolutionary conservation of motifs across homologous proteins. However, the intrinsically disordered regions (IDRs) in which SLiMs reside evolve rapidly. Consequently, multiple sequence alignment (MSA) of IDRs often misaligns SLiMs and underestimates their conservation. We present PairK (pairwise k-mer alignment), an MSA-free method to align and quantify the relative local conservation of subsequences within an IDR. Lacking a ground truth for conservation, we tested PairK on the task of distinguishing biologically important motif instances from background motifs, under the assumption that biologically important motifs are more conserved. The method outperforms both standard MSA-based conservation scores and a modern LLM-based conservation score predictor. PairK can quantify conservation over wider phylogenetic distances than MSAs, indicating that some SLiMs are more conserved than MSA-based metrics imply. PairK is available as an open-source python package at https://github.com/jacksonh1/pairk. It is designed to be easily adapted for use with other SLiM tools and for diverse applications.
Collapse
Affiliation(s)
| | - Amy E Keating
- Department of Biology, MIT, Cambridge, Massachusetts, USA
- Department of Biological Engineering, MIT, Cambridge, Massachusetts, USA
- Koch Institute for Integrative Cancer Research, Cambridge, Massachusetts, USA
| |
Collapse
|
3
|
LaComb L, Ghosh A, Bonanno JB, Nilson DJ, Poppel AJ, Dada L, Cahill SM, Maianti JP, Kitamura S, Cowburn D, Almo SC. Insights into the Interaction Landscape of the EVH1 Domain of Mena. Biochemistry 2024; 63:2183-2195. [PMID: 39138154 DOI: 10.1021/acs.biochem.4c00331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/15/2024]
Abstract
The Enabled/VASP homology 1 (EVH1) domain is a small module that interacts with proline-rich stretches in its ligands and is found in various signaling and scaffolding proteins. Mena, the mammalian homologue of Ena, is involved in diverse actin-associated events, such as membrane dynamics, bacterial motility, and tumor intravasation and extravasation. Two-dimensional (2D) 1H-15N HSQC NMR was used to study Mena EVH1 binding properties, defining the amino acids involved in ligand recognition for the physiological ligands ActA and PCARE, and a synthetic polyproline-inspired small molecule (hereafter inhibitor 6c). Chemical shift perturbations indicated that proline-rich segments bind in the conserved EVH1 hydrophobic cleft. The PCARE-derived peptide elicited more perturbations compared to the ActA-derived peptide, consistent with a previous report of a structural alteration in the solvent-exposed β7-β8 loop. Unexpectedly, EVH1 and the proline-rich segment of PTP1B did not exhibit NMR chemical shift perturbations; however, the high-resolution crystal structure implicated the conserved EVH1 hydrophobic cleft in ligand recognition. Intrinsic steady-state fluorescence and fluorescence polarization assays indicate that residues outside the proline-rich segment enhance the ligand affinity for EVH1 (Kd = 3-8 μM). Inhibitor 6c displayed tighter binding (Kd ∼ 0.3 μM) and occupies the same EVH1 cleft as physiological ligands. These studies revealed that the EVH1 domain enhances ligand affinity through recognition of residues flanking the proline-rich segments. Additionally, a synthetic inhibitor binds more tightly to the EVH1 domain than natural ligands, occupying the same hydrophobic cleft.
Collapse
Affiliation(s)
- Lanette LaComb
- Department of Biochemistry, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, New York 10461, United States
| | - Agnidipta Ghosh
- Department of Biochemistry, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, New York 10461, United States
| | - Jeffrey B Bonanno
- Department of Biochemistry, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, New York 10461, United States
| | - Daniel J Nilson
- Department of Biochemistry, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, New York 10461, United States
| | - Alex J Poppel
- Department of Biochemistry, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, New York 10461, United States
| | - Lucas Dada
- Department of Biochemistry, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, New York 10461, United States
| | - Sean M Cahill
- Department of Biochemistry, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, New York 10461, United States
| | - Juan Pablo Maianti
- Department of Biochemistry, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, New York 10461, United States
| | - Seiya Kitamura
- Department of Biochemistry, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, New York 10461, United States
| | - David Cowburn
- Department of Biochemistry, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, New York 10461, United States
| | - Steven C Almo
- Department of Biochemistry, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, New York 10461, United States
| |
Collapse
|
4
|
Aydin F, Katkar HH, Morganthaler A, Harker AJ, Kovar DR, Voth GA. Prediction of the essential intermolecular contacts for side-binding of VASP on F-actin. Cytoskeleton (Hoboken) 2024; 81:382-392. [PMID: 38647032 PMCID: PMC11333183 DOI: 10.1002/cm.21864] [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: 12/13/2023] [Revised: 04/08/2024] [Accepted: 04/09/2024] [Indexed: 04/25/2024]
Abstract
Vasodilator-stimulated phosphoprotein (VASP) family proteins play a crucial role in mediating the actin network architecture in the cytoskeleton. The Ena/VASP homology 2 (EVH2) domain in each of the four identical arms of the tetrameric VASP consists of a loading poly-Pro region, a G-actin-binding domain (GAB), and an F-actin-binding domain (FAB). Together, the poly-Pro, GAB, and FAB domains allow VASP to bind to sides of actin filaments in a bundle, and recruit profilin-G-actin to processively elongate the filaments. The atomic resolution structure of the ternary complex, consisting of the loading poly-Pro region and GAB domain of VASP with profilin-actin, has been solved over a decade ago; however, a detailed structure of the FAB-F-actin complex has not been resolved to date. Experimental insights, based on homology of the FAB domain with the C region of WASP, have been used to hypothesize that the FAB domain binds to the cleft between subdomains 1 and 3 of F-actin. Here, in order to develop our understanding of the VASP-actin complex, we first augment known structural information about the GAB domain binding to actin with the missing FAB domain-actin structure, which we predict using homology modeling and docking simulations. In earlier work, we used mutagenesis and kinetic modeling to study the role of domain-level binding-unbinding kinetics of Ena/VASP on actin filaments in a bundle, specifically on the side of actin filaments. We further look at the nature of the side-binding of the FAB domain of VASP at the atomistic level using our predicted structure, and tabulate effective mutation sites on the FAB domain that would disrupt the VASP-actin complex. We test the binding affinity of Ena with mutated FAB domain using total internal reflection fluorescence microscopy experiments. The binding affinity of VASP is affected significantly for the mutant, providing additional support for our predicted structure.
Collapse
Affiliation(s)
- Fikret Aydin
- Department of Chemistry, The James Franck Institute, and Institute for Biophysical Dynamics, University of Chicago, Chicago, IL 60637
| | - Harshwardhan H. Katkar
- Department of Chemistry, The James Franck Institute, and Institute for Biophysical Dynamics, University of Chicago, Chicago, IL 60637
| | - Alisha Morganthaler
- Department of Biochemistry and Molecular Biology and Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL 60637
| | - Alyssa J. Harker
- Department of Biochemistry and Molecular Biology and Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL 60637
| | - David R. Kovar
- Department of Biochemistry and Molecular Biology and Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL 60637
| | - Gregory A. Voth
- Department of Chemistry, The James Franck Institute, and Institute for Biophysical Dynamics, University of Chicago, Chicago, IL 60637
| |
Collapse
|
5
|
Singer A, Ramos A, Keating AE. Elaboration of the Homer1 recognition landscape reveals incomplete divergence of paralogous EVH1 domains. Protein Sci 2024; 33:e5094. [PMID: 38989636 PMCID: PMC11237882 DOI: 10.1002/pro.5094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 06/11/2024] [Accepted: 06/16/2024] [Indexed: 07/12/2024]
Abstract
Short sequences that mediate interactions with modular binding domains are ubiquitous throughout eukaryotic proteomes. Networks of short linear motifs (SLiMs) and their corresponding binding domains orchestrate many cellular processes, and the low mutational barrier to evolving novel interactions provides a way for biological systems to rapidly sample selectable phenotypes. Mapping SLiM binding specificity and the rules that govern SLiM evolution is fundamental to uncovering the pathways regulated by these networks and developing the tools to manipulate them. We used high-throughput screening of the human proteome to identify sequences that bind to the Enabled/VASP homology 1 (EVH1) domain of the postsynaptic density scaffolding protein Homer1. This expanded our understanding of the determinants of Homer EVH1 binding preferences and defined a new motif that can facilitate the discovery of additional Homer-mediated interactions. Interestingly, the Homer1 EVH1 domain preferentially binds to sequences containing an N-terminally overlapping motif that is bound by the paralogous family of Ena/VASP actin polymerases, and many of these sequences can bind to EVH1 domains from both protein families. We provide evidence from orthologous EVH1 domains in pre-metazoan organisms that the overlap in human Ena/VASP and Homer binding preferences corresponds to an incomplete divergence from a common Ena/VASP ancestor. Given this overlap in binding profiles, promiscuous sequences that can be recognized by both families either achieve specificity through extrinsic regulatory strategies or may provide functional benefits via multi-specificity. This may explain why these paralogs incompletely diverged despite the accessibility of further diverged isoforms.
Collapse
Affiliation(s)
- Avinoam Singer
- Department of BiologyMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
| | - Alejandra Ramos
- Department of BiologyMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
| | - Amy E. Keating
- Department of BiologyMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
- Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
- Koch Institute for Integrative Cancer ResearchMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
| |
Collapse
|
6
|
Halpin JC, Keating AE. PairK: Pairwise k-mer alignment for quantifying protein motif conservation in disordered regions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.23.604860. [PMID: 39091826 PMCID: PMC11291154 DOI: 10.1101/2024.07.23.604860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
Protein-protein interactions are often mediated by a modular peptide recognition domain binding to a short linear motif (SLiM) in the disordered region of another protein. The ability to predict domain-SLiM interactions would allow researchers to map protein interaction networks, predict the effects of perturbations to those networks, and develop biologically meaningful hypotheses. Unfortunately, sequence database searches for SLiMs generally yield mostly biologically irrelevant motif matches or false positives. To improve the prediction of novel SLiM interactions, researchers employ filters to discriminate between biologically relevant and improbable motif matches. One promising criterion for identifying biologically relevant SLiMs is the sequence conservation of the motif, exploiting the fact that functional motifs are more likely to be conserved than spurious motif matches. However, the difficulty of aligning disordered regions has significantly hampered the utility of this approach. We present PairK (pairwise k-mer alignment), an MSA-free method to quantify motif conservation in disordered regions. PairK outperforms both standard MSA-based conservation scores and a modern LLM-based conservation score predictor on the task of identifying biologically important motif instances. PairK can quantify conservation over wider phylogenetic distances than MSAs, indicating that SLiMs may be more conserved than is implied by MSA-based metrics. PairK is available as open-source code at https://github.com/jacksonh1/pairk.
Collapse
Affiliation(s)
- Jackson C. Halpin
- MIT Department of Biology, 77 Massachusetts Ave., Cambridge, MA 02139
| | - Amy E. Keating
- MIT Department of Biology, 77 Massachusetts Ave., Cambridge, MA 02139
- MIT Department of Biological Engineering, 77 Massachusetts Ave., Cambridge, MA 02139
- Koch Institute for Integrative Cancer Research, 77 Massachusetts Ave., Cambridge, MA 02139
| |
Collapse
|
7
|
McCormick LE, Suarez C, Herring LE, Cannon KS, Kovar DR, Brown NG, Gupton SL. Multi-monoubiquitylation controls VASP-mediated actin dynamics. J Cell Sci 2024; 137:jcs261527. [PMID: 38277158 PMCID: PMC10917064 DOI: 10.1242/jcs.261527] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 12/13/2023] [Indexed: 01/13/2024] Open
Abstract
The actin cytoskeleton performs multiple cellular functions, and as such, actin polymerization must be tightly regulated. We previously demonstrated that reversible, non-degradative ubiquitylation regulates the function of the actin polymerase VASP in developing neurons. However, the underlying mechanism of how ubiquitylation impacts VASP activity was unknown. Here, we show that mimicking multi-monoubiquitylation of VASP at K240 and K286 negatively regulates VASP interactions with actin. Using in vitro biochemical assays, we demonstrate the reduced ability of multi-monoubiquitylated VASP to bind, bundle, and elongate actin filaments. However, multi-monoubiquitylated VASP maintained the ability to bind and protect barbed ends from capping protein. Finally, we demonstrate the electroporation of recombinant multi-monoubiquitylated VASP protein altered cell spreading morphology. Collectively, these results suggest a mechanism in which ubiquitylation controls VASP-mediated actin dynamics.
Collapse
Affiliation(s)
- Laura E. McCormick
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Cristian Suarez
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL 60637, USA
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL 60637, USA
| | - Laura E. Herring
- Michael Hooker Proteomics Core, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Kevin S. Cannon
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - David R. Kovar
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL 60637, USA
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL 60637, USA
| | - Nicholas G. Brown
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Stephanie L. Gupton
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
8
|
Gui J, Zhou H, Wan H, Yang D, Liu Q, Zhu L, Mi Y. The Role of Vasodilator-stimulated Phosphoproteins in the Development of Malignant Tumors. Curr Cancer Drug Targets 2024; 24:477-489. [PMID: 37962042 PMCID: PMC11092557 DOI: 10.2174/0115680096262439231023110106] [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: 05/15/2023] [Revised: 08/06/2023] [Accepted: 09/06/2023] [Indexed: 11/15/2023]
Abstract
Vasodilator-stimulated phosphoprotein (VASP) is an actin-binding protein that includes three structural domains: Enabled/VASP homolog1 (EVH1), EVH2, and proline-rich (PRR). VASP plays an important role in various cellular behaviors related to cytoskeletal regulation. More importantly, VASP plays a key role in the progression of several malignant tumors and is associated with malignant cell proliferation, invasion, and metastasis. Here, we have summarized current studies on the impact of VASP on the development of several malignant tumors and their mechanisms. This study provides a new theoretical basis for clinical molecular diagnosis and molecular targeted therapy.
Collapse
Affiliation(s)
- Jiandong Gui
- Wuxi School of Medicine, Jiangnan University, 1800 Lihudadao, Wuxi, 214122, Jiangsu Province, China
- Department of Urology, Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Wuxi, 214122, Jiangsu Province, China
| | - Hangsheng Zhou
- Wuxi School of Medicine, Jiangnan University, 1800 Lihudadao, Wuxi, 214122, Jiangsu Province, China
- Department of Urology, Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Wuxi, 214122, Jiangsu Province, China
| | - Hongyuan Wan
- Wuxi School of Medicine, Jiangnan University, 1800 Lihudadao, Wuxi, 214122, Jiangsu Province, China
- Department of Urology, Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Wuxi, 214122, Jiangsu Province, China
| | - Dongjie Yang
- Department of Urology, Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Wuxi, 214122, Jiangsu Province, China
| | - Qing Liu
- Department of Urology, Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Wuxi, 214122, Jiangsu Province, China
- Huadong Sanatorium, 67 Dajishan, Wuxi 214122, Jiangsu Province, China
| | - Lijie Zhu
- Department of Urology, Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Wuxi, 214122, Jiangsu Province, China
| | - Yuanyuan Mi
- Department of Urology, Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Wuxi, 214122, Jiangsu Province, China
| |
Collapse
|
9
|
Chu Z, Zhu M, Luo Y, Hu Y, Feng X, Wang H, Sunagawa M, Liu Y. PTBP1 plays an important role in the development of gastric cancer. Cancer Cell Int 2023; 23:195. [PMID: 37670313 PMCID: PMC10478210 DOI: 10.1186/s12935-023-03043-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 08/25/2023] [Indexed: 09/07/2023] Open
Abstract
BACKGROUND Polypyrimidine tract binding protein 1 (PTBP1) has been found to play an important role in the occurrence and development of various tumors. At present, the role of PTBP1 in gastric cancer (GC) is still unknown and worthy of further investigation. METHODS We used bioinformatics to analyze the expression of PTBP1 in patients with GC. Cell proliferation related experiments were used to detect cell proliferation after PTBP1 knockdown. Skeleton staining, scanning electron microscopy and transmission electron microscopy were used to observe the changes of actin skeleton. Proliferation and actin skeleton remodeling signaling pathways were detected by Western Blots. The relationship between PTBP1 and proliferation of gastric cancer cells was further detected by subcutaneous tumor transplantation. Finally, tissue microarray data from clinical samples were used to further explore the expression of PTBP1 in patients with gastric cancer and its correlation with prognosis. RESULTS Through bioinformatics studies, we found that PTBP1 was highly expressed in GC patients and correlated with poor prognosis. Cell proliferation and cycle analysis showed that PTBP1 down-regulation could significantly inhibit cell proliferation. The results of cell proliferation detection related experiments showed that PTBP1 down-regulation could inhibit the division and proliferation of GC cells. Furthermore, changes in the morphology of the actin skeleton of cells showed that PTBP1 down-regulation inhibited actin skeletal remodeling in GC cells. Western Blots showed that PTBP1 could regulate proliferation and actin skeleton remodeling signaling pathways. In addition, we constructed PTBP1 Cas9-KO mouse model and performed xenograft assays to further confirm that down-regulation of PTBP1 could inhibit the proliferation of GC cells. Finally, tissue microarray was used to further verify the close correlation between PTBP1 and poor prognosis in patients with GC. CONCLUSIONS Our study demonstrates for the first time that PTBP1 may affect the proliferation of GC cells by regulating actin skeleton remodeling. In addition, PTBP1 is closely related to actin skeleton remodeling and proliferation signaling pathways. We suppose that PTBP1 might be a potential target for the treatment of GC.
Collapse
Affiliation(s)
- Zewen Chu
- The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou, China
| | - Miao Zhu
- The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou, China
| | - Yuanyuan Luo
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou, China
| | - Yaqi Hu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou, China
| | - Xinyi Feng
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou, China
| | - Haibo Wang
- The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China.
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China.
- The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou, China.
| | - Masataka Sunagawa
- Department of physiology, School of Medicine, Showa University, Tokyo, Japan.
| | - Yanqing Liu
- The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China.
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China.
- The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou, China.
| |
Collapse
|
10
|
Benz PM, Frömel T, Laban H, Zink J, Ulrich L, Groneberg D, Boon RA, Poley P, Renne T, de Wit C, Fleming I. Cardiovascular Functions of Ena/VASP Proteins: Past, Present and Beyond. Cells 2023; 12:1740. [PMID: 37443774 PMCID: PMC10340426 DOI: 10.3390/cells12131740] [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: 04/26/2023] [Revised: 06/18/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
Actin binding proteins are of crucial importance for the spatiotemporal regulation of actin cytoskeletal dynamics, thereby mediating a tremendous range of cellular processes. Since their initial discovery more than 30 years ago, the enabled/vasodilator-stimulated phosphoprotein (Ena/VASP) family has evolved as one of the most fascinating and versatile family of actin regulating proteins. The proteins directly enhance actin filament assembly, but they also organize higher order actin networks and link kinase signaling pathways to actin filament assembly. Thereby, Ena/VASP proteins regulate dynamic cellular processes ranging from membrane protrusions and trafficking, and cell-cell and cell-matrix adhesions, to the generation of mechanical tension and contractile force. Important insights have been gained into the physiological functions of Ena/VASP proteins in platelets, leukocytes, endothelial cells, smooth muscle cells and cardiomyocytes. In this review, we summarize the unique and redundant functions of Ena/VASP proteins in cardiovascular cells and discuss the underlying molecular mechanisms.
Collapse
Affiliation(s)
- Peter M. Benz
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, 60596 Frankfurt am Main, Germany
- German Centre of Cardiovascular Research (DZHK), Partner Site Rhein-Main, 60596 Frankfurt am Main, Germany
| | - Timo Frömel
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, 60596 Frankfurt am Main, Germany
| | - Hebatullah Laban
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, 60596 Frankfurt am Main, Germany
| | - Joana Zink
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, 60596 Frankfurt am Main, Germany
| | - Lea Ulrich
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, 60596 Frankfurt am Main, Germany
| | - Dieter Groneberg
- Institute of Physiology I, University of Würzburg, 97070 Würzburg, Germany
| | - Reinier A. Boon
- German Centre of Cardiovascular Research (DZHK), Partner Site Rhein-Main, 60596 Frankfurt am Main, Germany
- Cardiopulmonary Institute, 60596 Frankfurt am Main, Germany
- Centre of Molecular Medicine, Institute of Cardiovascular Regeneration, Goethe-University, 60596 Frankfurt am Main, Germany
- Department of Physiology, Amsterdam Cardiovascular Sciences, VU University Medical Centre, 1081 HZ Amsterdam, The Netherlands
| | - Philip Poley
- Institut für Physiologie, Universität zu Lübeck, 23562 Lübeck, Germany
- German Centre of Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 23562 Lübeck, Germany
| | - Thomas Renne
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- Center for Thrombosis and Hemostasis (CTH), Johannes Gutenberg University Medical Center, 55131 Mainz, Germany
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, D02 VN51 Dublin, Ireland
| | - Cor de Wit
- Institut für Physiologie, Universität zu Lübeck, 23562 Lübeck, Germany
- German Centre of Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 23562 Lübeck, Germany
| | - Ingrid Fleming
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, 60596 Frankfurt am Main, Germany
- German Centre of Cardiovascular Research (DZHK), Partner Site Rhein-Main, 60596 Frankfurt am Main, Germany
- Cardiopulmonary Institute, 60596 Frankfurt am Main, Germany
| |
Collapse
|
11
|
Parker SS, Ly KT, Grant AD, Sweetland J, Wang AM, Parker JD, Roman MR, Saboda K, Roe DJ, Padi M, Wolgemuth CW, Langlais P, Mouneimne G. EVL and MIM/MTSS1 regulate actin cytoskeletal remodeling to promote dendritic filopodia in neurons. J Cell Biol 2023; 222:e202106081. [PMID: 36828364 PMCID: PMC9998662 DOI: 10.1083/jcb.202106081] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 11/22/2022] [Accepted: 01/20/2023] [Indexed: 02/25/2023] Open
Abstract
Dendritic spines are the postsynaptic compartment of a neuronal synapse and are critical for synaptic connectivity and plasticity. A developmental precursor to dendritic spines, dendritic filopodia (DF), facilitate synapse formation by sampling the environment for suitable axon partners during neurodevelopment and learning. Despite the significance of the actin cytoskeleton in driving these dynamic protrusions, the actin elongation factors involved are not well characterized. We identified the Ena/VASP protein EVL as uniquely required for the morphogenesis and dynamics of DF. Using a combination of genetic and optogenetic manipulations, we demonstrated that EVL promotes protrusive motility through membrane-direct actin polymerization at DF tips. EVL forms a complex at nascent protrusions and DF tips with MIM/MTSS1, an I-BAR protein important for the initiation of DF. We proposed a model in which EVL cooperates with MIM to coalesce and elongate branched actin filaments, establishing the dynamic lamellipodia-like architecture of DF.
Collapse
Affiliation(s)
- Sara S. Parker
- Department of Cellular and Molecular Medicine, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - Kenneth Tran Ly
- Department of Cellular and Molecular Medicine, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - Adam D. Grant
- Cancer Biology Program, University of Arizona Cancer Center, Tucson, AZ, USA
| | - Jillian Sweetland
- Department of Cellular and Molecular Medicine, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - Ashley M. Wang
- Department of Cellular and Molecular Medicine, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - James D. Parker
- Department of Cellular and Molecular Medicine, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - Mackenzie R. Roman
- Division of Endocrinology, Department of Medicine, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - Kathylynn Saboda
- University of Arizona Cancer Center and Mel and Enid Zuckerman College of Public Health, University of Arizona, Tucson, AZ, USA
| | - Denise J. Roe
- University of Arizona Cancer Center and Mel and Enid Zuckerman College of Public Health, University of Arizona, Tucson, AZ, USA
| | - Megha Padi
- Cancer Biology Program, University of Arizona Cancer Center, Tucson, AZ, USA
- Department of Molecular and Cellular Biology, College of Science, University of Arizona, Tucson, AZ, USA
| | - Charles W. Wolgemuth
- University of Arizona Cancer Center and Mel and Enid Zuckerman College of Public Health, University of Arizona, Tucson, AZ, USA
- Department of Molecular and Cellular Biology, College of Science, University of Arizona, Tucson, AZ, USA
- Department of Physics, College of Science, University of Arizona, Tucson, AZ, USA
- Johns Hopkins Physical Sciences-Oncology Center, Johns Hopkins University, Baltimore, MD, USA
| | - Paul Langlais
- Division of Endocrinology, Department of Medicine, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - Ghassan Mouneimne
- Department of Cellular and Molecular Medicine, College of Medicine, University of Arizona, Tucson, AZ, USA
- Cancer Biology Program, University of Arizona Cancer Center, Tucson, AZ, USA
| |
Collapse
|
12
|
Klein MT, Krause BM, Neudörfl JM, Kühne R, Schmalz HG. Design and synthesis of a tetracyclic tripeptide mimetic frozen in a polyproline type II (PP2) helix conformation. Org Biomol Chem 2022; 20:9368-9377. [PMID: 36385673 DOI: 10.1039/d2ob01857h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
A synthesis of the new tetracyclic scaffold ProM-19, which represents a XPP tripeptide unit frozen in a PPII helix conformation, was developed. As a key building block, N-Boc-protected ethyl (1S,3S,4R)-2-azabicyclo[2.2.1]hept-5-ene-2-carboxylate was prepared through a diastereoselective aza-Diels-Alder reaction and subsequent hydrogenolytic removal of the chiral N-1-phenylethyl substituent under temporary protection of the double bond through dihydroxylation and reconstitution by Corey-Winter olefination. The target compound Boc-[ProM-19]-OMe was then prepared via subsequent peptide coupling and Ru-catalyzed ring-closing metathesis steps employing (S)-N-Boc-allylgylcine and cis-5-vinyl-proline methyl ester as additional building blocks. In addition, Ac-[2-Cl-Phe]-[Pro]-[ProM-19]-OMe was prepared by solution phase peptide synthesis as a potential ligand for the ena-VASP EVH1 domain.
Collapse
Affiliation(s)
- Marco T Klein
- University of Cologne, Department of Chemistry, Greinstrasse 4, 50939 Köln, Germany.
| | - Bernhard M Krause
- University of Cologne, Department of Chemistry, Greinstrasse 4, 50939 Köln, Germany.
| | - Jörg-Martin Neudörfl
- University of Cologne, Department of Chemistry, Greinstrasse 4, 50939 Köln, Germany.
| | - Ronald Kühne
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Robert-Rössle-Strasse 10, 13125 Berlin, Germany
| | - Hans-Günther Schmalz
- University of Cologne, Department of Chemistry, Greinstrasse 4, 50939 Köln, Germany.
| |
Collapse
|
13
|
Molina-Pelayo C, Olguin P, Mlodzik M, Glavic A. The conserved Pelado/ZSWIM8 protein regulates actin dynamics by promoting linear actin filament polymerization. Life Sci Alliance 2022; 5:e202201484. [PMID: 35940847 PMCID: PMC9375228 DOI: 10.26508/lsa.202201484] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 07/15/2022] [Accepted: 07/18/2022] [Indexed: 11/24/2022] Open
Abstract
Actin filament polymerization can be branched or linear, which depends on the associated regulatory proteins. Competition for actin monomers occurs between proteins that induce branched or linear actin polymerization. Cell specialization requires the regulation of actin filaments to allow the formation of cell type-specific structures, like cuticular hairs in <i>Drosophila</i>, formed by linear actin filaments. Here, we report the functional analysis of CG34401/<i>pelado</i>, a gene encoding a SWIM domain-containing protein, conserved throughout the animal kingdom, called ZSWIM8 in mammals. Mutant <i>pelado</i> epithelial cells display actin hair elongation defects. This phenotype is reversed by increasing actin monomer levels or by either pushing linear actin polymerization or reducing branched actin polymerization. Similarly, in hemocytes, Pelado is essential to induce filopodia, a linear actin-based structure. We further show that this function of Pelado/ZSWIM8 is conserved in human cells, where Pelado inhibits branched actin polymerization in a cell migration context. In summary, our data indicate that the function of Pelado/ZSWIM8 in regulating actin cytoskeletal dynamics is conserved, favoring linear actin polymerization at the expense of branched filaments.
Collapse
Affiliation(s)
- Claudia Molina-Pelayo
- Department of Cell, Developmental, and Regenerative Biology, Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Departamento de Biología, Centro FONDAP de Regulación del Genoma, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - Patricio Olguin
- Department of Cell, Developmental, and Regenerative Biology, Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Departamento de Neurociencia, Programa de Genética Humana, Instituto de Ciencias Biomédicas, Instituto de Neurociencia Biomédica, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Marek Mlodzik
- Department of Cell, Developmental, and Regenerative Biology, Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alvaro Glavic
- Departamento de Biología, Centro FONDAP de Regulación del Genoma, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| |
Collapse
|
14
|
Faix J, Rottner K. Ena/VASP proteins in cell edge protrusion, migration and adhesion. J Cell Sci 2022; 135:274697. [DOI: 10.1242/jcs.259226] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
ABSTRACT
The tightly coordinated, spatiotemporal control of actin filament remodeling provides the basis of fundamental cellular processes, such as cell migration and adhesion. Specific protein assemblies, composed of various actin-binding proteins, are thought to operate in these processes to nucleate and elongate new filaments, arrange them into complex three-dimensional (3D) arrays and recycle them to replenish the actin monomer pool. Actin filament assembly is not only necessary to generate pushing forces against the leading edge membrane or to propel pathogens through the cytoplasm, but also coincides with the generation of stress fibers (SFs) and focal adhesions (FAs) that generate, transmit and sense mechanical tension. The only protein families known to date that directly enhance the elongation of actin filaments are formins and the family of Ena/VASP proteins. Their mechanisms of action, however, in enhancing processive filament elongation are distinct. The aim of this Review is to summarize our current knowledge on the molecular mechanisms of Ena/VASP-mediated actin filament assembly, and to discuss recent insights into the cell biological functions of Ena/VASP proteins in cell edge protrusion, migration and adhesion.
Collapse
Affiliation(s)
- Jan Faix
- Institute for Biophysical Chemistry, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| | - Klemens Rottner
- Division of Molecular Cell Biology, Zoological Institute, Technical University Braunschweig, Spielmannstrasse 7, 38106 Braunschweig, Germany
- Molecular Cell Biology Group, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| |
Collapse
|
15
|
Hwang T, Parker SS, Hill SM, Grant RA, Ilunga MW, Sivaraman V, Mouneimne G, Keating AE. Native proline-rich motifs exploit sequence context to target actin-remodeling Ena/VASP protein ENAH. eLife 2022; 11:70680. [PMID: 35076015 PMCID: PMC8789275 DOI: 10.7554/elife.70680] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 01/06/2022] [Indexed: 12/13/2022] Open
Abstract
The human proteome is replete with short linear motifs (SLiMs) of four to six residues that are critical for protein-protein interactions, yet the importance of the sequence surrounding such motifs is underexplored. We devised a proteomic screen to examine the influence of SLiM sequence context on protein-protein interactions. Focusing on the EVH1 domain of human ENAH, an actin regulator that is highly expressed in invasive cancers, we screened 36-residue proteome-derived peptides and discovered new interaction partners of ENAH and diverse mechanisms by which context influences binding. A pocket on the ENAH EVH1 domain that has diverged from other Ena/VASP paralogs recognizes extended SLiMs and favors motif-flanking proline residues. Many high-affinity ENAH binders that contain two proline-rich SLiMs use a noncanonical site on the EVH1 domain for binding and display a thermodynamic signature consistent with the two-motif chain engaging a single domain. We also found that photoreceptor cilium actin regulator (PCARE) uses an extended 23-residue region to obtain a higher affinity than any known ENAH EVH1-binding motif. Our screen provides a way to uncover the effects of proteomic context on motif-mediated binding, revealing diverse mechanisms of control over EVH1 interactions and establishing that SLiMs can’t be fully understood outside of their native context.
Collapse
Affiliation(s)
- Theresa Hwang
- Department of Biology, Massachusetts Institute of Technology
| | - Sara S Parker
- Department of Cellular & Molecular Medicine, University of Arizona
| | - Samantha M Hill
- Department of Cellular & Molecular Medicine, University of Arizona
| | - Robert A Grant
- Department of Biology, Massachusetts Institute of Technology
| | - Meucci W Ilunga
- Department of Biology, Massachusetts Institute of Technology
| | | | | | - Amy E Keating
- Department of Biology, Massachusetts Institute of Technology
- Department of Biological Engineering and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology
| |
Collapse
|
16
|
Hwang T, Parker SS, Hill SM, Ilunga MW, Grant RA, Mouneimne G, Keating AE. A distributed residue network permits conformational binding specificity in a conserved family of actin remodelers. eLife 2021; 10:e70601. [PMID: 34854809 PMCID: PMC8639148 DOI: 10.7554/elife.70601] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 11/08/2021] [Indexed: 11/23/2022] Open
Abstract
Metazoan proteomes contain many paralogous proteins that have evolved distinct functions. The Ena/VASP family of actin regulators consists of three members that share an EVH1 interaction domain with a 100 % conserved binding site. A proteome-wide screen revealed photoreceptor cilium actin regulator (PCARE) as a high-affinity ligand for ENAH EVH1. Here, we report the surprising observation that PCARE is ~100-fold specific for ENAH over paralogs VASP and EVL and can selectively bind ENAH and inhibit ENAH-dependent adhesion in cells. Specificity arises from a mechanism whereby PCARE stabilizes a conformation of the ENAH EVH1 domain that is inaccessible to family members VASP and EVL. Structure-based modeling rapidly identified seven residues distributed throughout EVL that are sufficient to differentiate binding by ENAH vs. EVL. By exploiting the ENAH-specific conformation, we rationally designed the tightest and most selective ENAH binder to date. Our work uncovers a conformational mechanism of interaction specificity that distinguishes highly similar paralogs and establishes tools for dissecting specific Ena/VASP functions in processes including cancer cell invasion.
Collapse
Affiliation(s)
- Theresa Hwang
- Department of Biology, Massachusetts Institute of TechnologyCambridgeUnited States
| | - Sara S Parker
- Department of Cellular and Molecular Medicine, University of Arizona Cancer Center, University of ArizonaTucsonUnited States
| | - Samantha M Hill
- Department of Cellular and Molecular Medicine, University of Arizona Cancer Center, University of ArizonaTucsonUnited States
| | - Meucci W Ilunga
- Department of Biology, Massachusetts Institute of TechnologyCambridgeUnited States
| | - Robert A Grant
- Department of Biology, Massachusetts Institute of TechnologyCambridgeUnited States
| | - Ghassan Mouneimne
- Department of Cellular and Molecular Medicine, University of Arizona Cancer Center, University of ArizonaTucsonUnited States
| | - Amy E Keating
- Department of Biology, Massachusetts Institute of TechnologyCambridgeUnited States
- Department of Biological Engineering and Koch Institue for Integrative Cancer Research, Massachusetts Institute of TechnologyCambridgeUnited States
| |
Collapse
|
17
|
Augustin V, Kins S. Fe65: A Scaffolding Protein of Actin Regulators. Cells 2021; 10:cells10071599. [PMID: 34202290 PMCID: PMC8304848 DOI: 10.3390/cells10071599] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 06/19/2021] [Accepted: 06/21/2021] [Indexed: 01/19/2023] Open
Abstract
The scaffolding protein family Fe65, composed of Fe65, Fe65L1, and Fe65L2, was identified as an interaction partner of the amyloid precursor protein (APP), which plays a key function in Alzheimer’s disease. All three Fe65 family members possess three highly conserved interaction domains, forming complexes with diverse binding partners that can be assigned to different cellular functions, such as transactivation of genes in the nucleus, modulation of calcium homeostasis and lipid metabolism, and regulation of the actin cytoskeleton. In this article, we rule out putative new intracellular signaling mechanisms of the APP-interacting protein Fe65 in the regulation of actin cytoskeleton dynamics in the context of various neuronal functions, such as cell migration, neurite outgrowth, and synaptic plasticity.
Collapse
|
18
|
D'Andréa ÉD, Retel JS, Diehl A, Schmieder P, Oschkinat H, Pires JR. NMR structure and dynamics of Q4DY78, a conserved kinetoplasid-specific protein from Trypanosoma cruzi. J Struct Biol 2021; 213:107715. [PMID: 33705979 DOI: 10.1016/j.jsb.2021.107715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 03/01/2021] [Accepted: 03/03/2021] [Indexed: 10/21/2022]
Abstract
The 106-residue protein Q4DY78 (UniProt accession number) from Trypanosoma cruzi is highly conserved in the related kinetoplastid pathogens Trypanosoma brucei and Leishmania major. Given the essentiality of its orthologue in T. brucei, the high sequence conservation with other trypanosomatid proteins, and the low sequence similarity with mammalian proteins, Q4DY78 is an attractive protein for structural characterization. Here, we solved the structure of Q4DY78 by solution NMR and evaluated its backbone dynamics. Q4DY78 is composed of five α -helices and a small, two-stranded antiparallel β-sheet. The backbone RMSD is 0.22 ± 0.05 Å for the representative ensemble of the 20 lowest-energy structures. Q4DY78 is overall rigid, except for N-terminal residues (V8 to I10), residues at loop 4 (K57 to G65) and residues at the C-terminus (F89 to F112). Q4DY78 has a short motif FPCAP that could potentially mediate interactions with the host cytoskeleton via interaction with EVH1 (Drosophila Enabled (Ena)/Vasodilator-stimulated phosphoprotein (VASP) homology 1) domains. Albeit Q4DY78 lacks calcium-binding motifs, its fold resembles that of eukaryotic calcium-binding proteins such as calcitracin, calmodulin, and polcacin Bet V4. We characterized this novel protein with a calcium binding fold without the capacity to bind calcium.
Collapse
Affiliation(s)
- Éverton Dias D'Andréa
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho, 373 - Bloco E, sala 32, Rio de Janeiro, RJ 21941-902, Brazil
| | - Joren Sebastian Retel
- Leibniz-Institut für Molekulare Pharmakologie, FMP, Robert-Rössle-Straβe 10, Berlin 13125, Germany
| | - Anne Diehl
- Leibniz-Institut für Molekulare Pharmakologie, FMP, Robert-Rössle-Straβe 10, Berlin 13125, Germany
| | - Peter Schmieder
- Leibniz-Institut für Molekulare Pharmakologie, FMP, Robert-Rössle-Straβe 10, Berlin 13125, Germany
| | - Hartmut Oschkinat
- Leibniz-Institut für Molekulare Pharmakologie, FMP, Robert-Rössle-Straβe 10, Berlin 13125, Germany; Freie Universität Berlin, Institut für Chemie und Biochemie, Takustrasse 3, Berlin 14195, Germany
| | - José Ricardo Pires
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho, 373 - Bloco E, sala 32, Rio de Janeiro, RJ 21941-902, Brazil.
| |
Collapse
|
19
|
Designed nanomolar small-molecule inhibitors of Ena/VASP EVH1 interaction impair invasion and extravasation of breast cancer cells. Proc Natl Acad Sci U S A 2020; 117:29684-29690. [PMID: 33184177 PMCID: PMC7703624 DOI: 10.1073/pnas.2007213117] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Protein–protein interactions mediated by proline-rich motifs are involved in regulation of many important signaling cascades. These motifs belong to the most abundant recognition motifs in the eukaryotic genome and preferentially adopt a left-handed polyproline helix II, a secondary structure element that has been notoriously difficult to mimic with small molecules. Here, we present a structure-guided design effort yielding a toolkit of chemical entities that enables rational construction of selective small molecule inhibitors for these protein domains. We succeeded in developing an inhibitor for the Ena/VASP protein family that is active in vivo and reduces extravasation of invasive breast cancer cells in a zebrafish model. Battling metastasis through inhibition of cell motility is considered a promising approach to support cancer therapies. In this context, Ena/VASP-depending signaling pathways, in particular interactions with their EVH1 domains, are promising targets for pharmaceutical intervention. However, protein–protein interactions involving proline-rich segments are notoriously difficult to address by small molecules. Hence, structure-based design efforts in combination with the chemical synthesis of additional molecular entities are required. Building on a previously developed nonpeptidic micromolar inhibitor, we determined 22 crystal structures of ENAH EVH1 in complex with inhibitors and rationally extended our library of conformationally defined proline-derived modules (ProMs) to succeed in developing a nanomolar inhibitor (Kd=120 nM,MW=734 Da). In contrast to the previous inhibitor, the optimized compounds reduced extravasation of invasive breast cancer cells in a zebrafish model. This study represents an example of successful, structure-guided development of low molecular weight inhibitors specifically and selectively addressing a proline-rich sequence-recognizing domain that is characterized by a shallow epitope lacking defined binding pockets. The evolved high-affinity inhibitor may now serve as a tool in validating the basic therapeutic concept, i.e., the suppression of cancer metastasis by inhibiting a crucial protein–protein interaction involved in actin filament processing and cell migration.
Collapse
|
20
|
Donhauser N, Socher E, Millen S, Heym S, Sticht H, Thoma-Kress AK. Transfer of HTLV-1 p8 and Gag to target T-cells depends on VASP, a novel interaction partner of p8. PLoS Pathog 2020; 16:e1008879. [PMID: 32997728 PMCID: PMC7526893 DOI: 10.1371/journal.ppat.1008879] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Accepted: 08/12/2020] [Indexed: 12/16/2022] Open
Abstract
The Human T-cell leukemia virus type 1 (HTLV-1) orf I-encoded accessory protein p8 is cleaved from its precursor p12, and both proteins contribute to viral persistence. p8 induces cellular protrusions, which are thought to facilitate transfer of p8 to target cells and virus transmission. Host factors interacting with p8 and mediating p8 transfer are unknown. Here, we report that vasodilator-stimulated phosphoprotein (VASP), which promotes actin filament elongation, is a novel interaction partner of p8 and important for p8 and HTLV-1 Gag cell-to-cell transfer. VASP contains an Ena/VASP homology 1 (EVH1) domain that targets the protein to focal adhesions. Bioinformatics identified a short stretch in p8 (amino acids (aa) 24–45) which may mediate interactions with the EVH1 domain of VASP. Co-immunoprecipitations confirmed interactions of VASP:p8 in 293T, Jurkat and HTLV-1-infected MT-2 cells. Co-precipitation of VASP:p8 could be significantly blocked by peptides mimicking aa 26–37 of p8. Mutational studies revealed that the EVH1-domain of VASP is necessary, but not sufficient for the interaction with p8. Further, deletion of the VASP G- and F-actin binding domains significantly diminished co-precipitation of p8. Imaging identified areas of partial co-localization of VASP with p8 at the plasma membrane and in protrusive structures, which was confirmed by proximity ligation assays. Co-culture experiments revealed that p8 is transferred between Jurkat T-cells via VASP-containing conduits. Imaging and flow cytometry revealed that repression of both endogenous and overexpressed VASP by RNA interference or by CRISPR/Cas9 reduced p8 transfer to the cell surface and to target Jurkat T-cells. Stable repression of VASP by RNA interference in chronically infected MT-2 cells impaired both p8 and HTLV-1 Gag transfer to target Jurkat T-cells, while virus release was unaffected. Thus, we identified VASP as a novel interaction partner of p8, which is important for transfer of HTLV-1 p8 and Gag to target T-cells. The delta-retrovirus Human T-cell leukemia virus type 1 encodes the accessory protein p8, which is generated by proteolytic cleavage from p12. Earlier work has shown that p8 enhances the formation of cellular conduits between T-cells, is transferred through these conduits to target T-cells and increases HTLV-1 transmission. It was suggested that p8 dampens T-cell responses in target T-cells, thus facilitating HTLV-1 infection. Our work sheds light on the mechanism of p8 transfer to target T-cells. We show that vasodilator-stimulated phosphoprotein (VASP), a novel interaction partner of p8, contributes to transfer of p8 to target T-cells. Mechanistically, VASP is crucial for recruitment of p8 to the cell surface. Since VASP is known to promote elongation of actin filaments by preventing them from capping, interactions of p8 with VASP are an elegant strategy to exploit the host cell machinery for being transported to the cell surface, and as a consequence, to other cells. Given that VASP is also important for cell-to-cell transfer of the HTLV-1 Gag protein, our work proposes that VASP is a new cellular target to counteract HTLV-1 cell-to-cell transmission.
Collapse
Affiliation(s)
- Norbert Donhauser
- Institute of Clinical and Molecular Virology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Eileen Socher
- Division of Bioinformatics, Institute of Biochemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Sebastian Millen
- Institute of Clinical and Molecular Virology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Stefanie Heym
- Institute of Clinical and Molecular Virology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Heinrich Sticht
- Division of Bioinformatics, Institute of Biochemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Andrea K. Thoma-Kress
- Institute of Clinical and Molecular Virology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- * E-mail:
| |
Collapse
|
21
|
Cheng KW, Mullins RD. Initiation and disassembly of filopodia tip complexes containing VASP and lamellipodin. Mol Biol Cell 2020; 31:2021-2034. [PMID: 32579429 PMCID: PMC7543071 DOI: 10.1091/mbc.e20-04-0270] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
The shapes of many eukaryotic cells depends on the actin cytoskeleton, and changes in actin assembly dynamics underlie many changes in cell shape. Ena/VASP-family actin polymerases, for example, modulate cell shape by accelerating actin filament assembly locally and slowing filament capping. When concentrated into discrete foci at the leading edge, VASP promotes filopodia assembly and forms part of a poorly understood molecular complex that remains associated with growing filopodia tips. Here we identify precursors of this filopodia tip complex in migrating B16F1 cells: small leading-edge clusters of the adaptor protein lamellipodin (Lpd) that subsequently recruit VASP and initiate filopodia formation. Dimerization, membrane association, and VASP binding are all required for lamellipodin to incorporate into filopodia tip complexes, and overexpression of monomeric, membrane-targeted lamellipodin mutants disrupts tip complex assembly. Once formed, tip complexes containing VASP and lamellipodin grow by fusing with each other, but their growth is limited by a size-dependent dynamic instability. Our results demonstrate that assembly and disassembly dynamics of filopodia tip complexes are determined, in part, by a network of multivalent interactions between Ena/VASP proteins, EVH1 ligands, and actin filaments.
Collapse
Affiliation(s)
- Karen W Cheng
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA 94143
| | - R Dyche Mullins
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA 94143.,Howard Hughes Medical Institute, University of California, San Francisco, CA 94143
| |
Collapse
|
22
|
Sámano-Sánchez H, Gibson TJ. Mimicry of Short Linear Motifs by Bacterial Pathogens: A Drugging Opportunity. Trends Biochem Sci 2020; 45:526-544. [PMID: 32413327 DOI: 10.1016/j.tibs.2020.03.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 02/25/2020] [Accepted: 03/03/2020] [Indexed: 12/11/2022]
Abstract
Bacterial pathogens have developed complex strategies to successfully survive and proliferate within their hosts. Throughout the infection cycle, direct interaction with host cells occurs. Many bacteria have been found to secrete proteins, such as effectors and toxins, directly into the host cell with the potential to interfere with cell regulatory processes, either enzymatically or through protein-protein interactions (PPIs). Short linear motifs (SLiMs) are abundant peptide modules in cell signaling proteins. Here, we cover the reported examples of eukaryotic-like SLiM mimicry being used by pathogenic bacteria to hijack host cell machinery and discuss how drugs targeting SLiM-regulated cell signaling networks are being evaluated for interference with bacterial infections. This emerging anti-infective opportunity may become an essential contributor to antibiotic replacement strategies.
Collapse
Affiliation(s)
- Hugo Sámano-Sánchez
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Meyerhofstrasse 1, 69117 Heidelberg, Germany; Collaboration for Joint PhD Degree between EMBL and Heidelberg University, Faculty of Biosciences, 69120 Heidelberg, Germany
| | - Toby J Gibson
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Meyerhofstrasse 1, 69117 Heidelberg, Germany.
| |
Collapse
|
23
|
The Wnt/β-catenin/VASP positive feedback loop drives cell proliferation and migration in breast cancer. Oncogene 2019; 39:2258-2274. [PMID: 31831834 DOI: 10.1038/s41388-019-1145-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Revised: 11/27/2019] [Accepted: 12/02/2019] [Indexed: 12/21/2022]
Abstract
Previous studies have shown that the main function of VASP is to regulate the cytoskeleton and play an important role in promoting tumor cell metastasis. In this study, we first reveal that VASP is located in the nucleus of breast cancer cells and elucidate a Wnt/β-catenin/VASP positive feedback loop. We identify that VASP is a target gene of Wnt/β-catenin signaling pathway, and activation of Wnt/β-catenin signaling pathway can significantly upregulate VASP protein expression, while upregulated VASP protein can in turn promote translocation of β-catenin and DVL3 proteins into the nucleus. In the nucleus, VASP, DVL3, β-catenin, and TCF4 can form VASP/DVL3/β-catenin/TCF4 protein complex, activating Wnt/β-catenin signaling pathway, and promoting the expression of target genes VASP, c-myc, and cyclin D1. Thus, our study reveals that there is a Wnt/β-catenin/VASP malignant positive feedback loop in breast cancer, which promotes the proliferation and migration of breast cancer cells, and breaking this positive feedback loop may provide new strategy for breast cancer treatment.
Collapse
|
24
|
A Consensus Binding Motif for the PP4 Protein Phosphatase. Mol Cell 2019; 76:953-964.e6. [PMID: 31585692 DOI: 10.1016/j.molcel.2019.08.029] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 07/08/2019] [Accepted: 08/28/2019] [Indexed: 12/18/2022]
Abstract
Dynamic protein phosphorylation constitutes a fundamental regulatory mechanism in all organisms. Phosphoprotein phosphatase 4 (PP4) is a conserved and essential nuclear serine and threonine phosphatase. Despite the importance of PP4, general principles of substrate selection are unknown, hampering the study of signal regulation by this phosphatase. Here, we identify and thoroughly characterize a general PP4 consensus-binding motif, the FxxP motif. X-ray crystallography studies reveal that FxxP motifs bind to a conserved pocket in the PP4 regulatory subunit PPP4R3. Systems-wide in silico searches integrated with proteomic analysis of PP4 interacting proteins allow us to identify numerous FxxP motifs in proteins controlling a range of fundamental cellular processes. We identify an FxxP motif in the cohesin release factor WAPL and show that this regulates WAPL phosphorylation status and is required for efficient cohesin release. Collectively our work uncovers basic principles of PP4 specificity with broad implications for understanding phosphorylation-mediated signaling in cells.
Collapse
|
25
|
Tseng CC, Zhang P, Lee AS. The COOH-Terminal Proline-Rich Region of GRP78 Is a Key Regulator of Its Cell Surface Expression and Viability of Tamoxifen-Resistant Breast Cancer Cells. Neoplasia 2019; 21:837-848. [PMID: 31306849 PMCID: PMC6629921 DOI: 10.1016/j.neo.2019.05.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 05/23/2019] [Accepted: 05/28/2019] [Indexed: 12/11/2022] Open
Abstract
Translocation of 78-kDa glucose-regulated protein (GRP78) from endoplasmic reticulum (ER) to plasma membrane represents a paradigm shift beyond its traditional function as an ER chaperone protein. Cell surface GRP78 (csGRP78) exerts novel signaling functions, and mechanisms underlying its cell surface expression are just emerging. Acquired tamoxifen resistance of breast cancer cells is accompanied with elevated level of csGRP78. Therefore, the tamoxifen-resistant MCF7 breast cancer cells (MCF7-LR) represents a clinically relevant model to study mechanisms of csGRP78 expression. We discovered that a proline-rich region (PRR) containing three consecutive prolines close to the COOH-terminus of GRP78 is important for its ability to form a complex with the partner protein, CD44v, as demonstrated by in vitro glutathione S-transferase pull-down assay. Proline to alanine mutations at the PRR compromised GRP78 expression level on the cell surface as evidenced by purification of biotinylated cell surface proteins. Reconstitution of MCF7-LR cells with the PRR mutant after knockdown of endogenous GRP78 diminished the capacity of GRP78 to stimulate STAT3 activation. The enforced expression of a short peptide bearing the PRR region of GRP78 led to reduction of CD44v and Cyclin D1 protein levels as well as cell viability, accompanied with increase in apoptotic signaling including cleaved Caspase-3 and PARP. These findings suggest that the COOH-terminal PRR of GRP78 is critical for its interaction with CD44v as well as its cell surface expression, and enforced expression of the short peptide bearing the PRR region may provide a new approach to lower the viability of tamoxifen-resistant breast cancer cells.
Collapse
Affiliation(s)
- Chun-Chih Tseng
- Department of Biochemistry and Molecular Medicine, University of Southern California, 1441 Eastlake Avenue, Los Angeles, California 90089, USA; USC Norris Comprehensive Cancer Center, University of Southern California, 1441 Eastlake Avenue, Los Angeles, California 90089, USA.
| | - Pu Zhang
- Department of Molecular Microbiology and Immunology, University of Southern California, 1441 Eastlake Avenue, Los Angeles, California 90089, USA; USC Norris Comprehensive Cancer Center, University of Southern California, 1441 Eastlake Avenue, Los Angeles, California 90089, USA.
| | - Amy S Lee
- Department of Biochemistry and Molecular Medicine, University of Southern California, 1441 Eastlake Avenue, Los Angeles, California 90089, USA; USC Norris Comprehensive Cancer Center, University of Southern California, 1441 Eastlake Avenue, Los Angeles, California 90089, USA.
| |
Collapse
|
26
|
Tateya S, Rizzo-De Leon N, Cheng AM, Dick BP, Lee WJ, Kim ML, O’Brien K, Morton GJ, Schwartz MW, Kim F. The role of vasodilator-stimulated phosphoprotein (VASP) in the control of hepatic gluconeogenic gene expression. PLoS One 2019; 14:e0215601. [PMID: 31017943 PMCID: PMC6481847 DOI: 10.1371/journal.pone.0215601] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 04/04/2019] [Indexed: 01/22/2023] Open
Abstract
During periods in which glucose absorption from the gastrointestinal (GI) tract is insufficient to meet body requirements, hepatic gluconeogenesis plays a key role to maintain normal blood glucose levels. The current studies investigated the role in this process played by vasodilatory-associated phosphoprotein (VASP), a protein that is phosphorylated in hepatocytes by cAMP/protein kinase A (PKA), a key mediator of the action of glucagon. We report that following stimulation of hepatocytes with 8Br-cAMP, phosphorylation of VASP preceded induction of genes encoding key gluconeogenic enzymes, glucose-6-phosphatase (G6p) and phosphoenolpyruvate carboxykinase (Pck1), and that VASP overexpression enhanced this gene induction. Conversely, hepatocytes from mice lacking VASP (Vasp-/-) displayed blunted induction of gluconeogenic enzymes in response to cAMP, and Vasp-/- mice exhibited both greater fasting hypoglycemia and blunted hepatic gluconeogenic enzyme gene expression in response to fasting in vivo. These effects of VASP deficiency were associated with reduced phosphorylation of both CREB (a key transcription factor for gluconeogenesis that lies downstream of PKA) and histone deacetylase 4 (HDAC4), a combination of effects that inhibit transcription of gluconeogenic genes. These data support a model in which VASP functions as a molecular bridge linking the two key signal transduction pathways governing hepatic gluconeogenic gene expression.
Collapse
Affiliation(s)
- Sanshiro Tateya
- Department of Medicine, University of Washington, Seattle, WA, United States of America
- University of Washington Medicine Diabetes Institute, University of Washington, Seattle, WA, United States of America
| | - Norma Rizzo-De Leon
- Department of Medicine, University of Washington, Seattle, WA, United States of America
- University of Washington Medicine Diabetes Institute, University of Washington, Seattle, WA, United States of America
| | - Andrew M. Cheng
- Department of Medicine, University of Washington, Seattle, WA, United States of America
- University of Washington Medicine Diabetes Institute, University of Washington, Seattle, WA, United States of America
| | - Brian P. Dick
- Department of Medicine, University of Washington, Seattle, WA, United States of America
- University of Washington Medicine Diabetes Institute, University of Washington, Seattle, WA, United States of America
| | - Woo Je Lee
- Department of Medicine, University of Washington, Seattle, WA, United States of America
- University of Washington Medicine Diabetes Institute, University of Washington, Seattle, WA, United States of America
| | - Madeleine L. Kim
- University of Washington Medicine Diabetes Institute, University of Washington, Seattle, WA, United States of America
| | - Kevin O’Brien
- Department of Medicine, University of Washington, Seattle, WA, United States of America
| | - Gregory J. Morton
- Department of Medicine, University of Washington, Seattle, WA, United States of America
- University of Washington Medicine Diabetes Institute, University of Washington, Seattle, WA, United States of America
| | - Michael W. Schwartz
- Department of Medicine, University of Washington, Seattle, WA, United States of America
- University of Washington Medicine Diabetes Institute, University of Washington, Seattle, WA, United States of America
| | - Francis Kim
- Department of Medicine, University of Washington, Seattle, WA, United States of America
- University of Washington Medicine Diabetes Institute, University of Washington, Seattle, WA, United States of America
- * E-mail:
| |
Collapse
|
27
|
Bitard‐Feildel T, Lamiable A, Mornon J, Callebaut I. Order in Disorder as Observed by the "Hydrophobic Cluster Analysis" of Protein Sequences. Proteomics 2018; 18:e1800054. [PMID: 30299594 PMCID: PMC7168002 DOI: 10.1002/pmic.201800054] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 08/29/2018] [Indexed: 12/17/2022]
Abstract
Hydrophobic cluster analysis (HCA) is an original approach for protein sequence analysis, which provides access to the foldable repertoire of the protein universe, including yet unannotated protein segments ("dark proteome"). Foldable segments correspond to ordered regions, as well as to intrinsically disordered regions (IDRs) undergoing disorder to order transitions. In this review, how HCA can be used to give insight into this last category of foldable segments is illustrated, with examples matching known 3D structures. After reviewing the HCA principles, examples of short foldable segments are given, which often contain short linear motifs, typically matching hydrophobic clusters. These segments become ordered upon contact with partners, with secondary structure preferences generally corresponding to those observed in the 3D structures within the complexes. Such small foldable segments are sometimes larger than the segments of known 3D structures, including flanking hydrophobic clusters that may be critical for interaction specificity or regulation, as well as intervening sequences allowing fuzziness. Cases of larger conditionally disordered domains are also presented, with lower density in hydrophobic clusters than well-folded globular domains or with exposed hydrophobic patches, which are stabilized by interaction with partners.
Collapse
Affiliation(s)
- Tristan Bitard‐Feildel
- Institut de Minéralogie, de Physique des Matériaux et de Cosmochimie (IMPMC)Institut de recherche pour le développement (IRD)UMR CNRS 7590Muséum National d'Histoire NaturelleSorbonne Université75005ParisFrance
- Laboratoire de Biologie Computationnelle et Quantitative (LCQB)Institute of Biology Paris‐Seine (IBPS)Centre national de la recherche scientifique (CNRS)Sorbonne Université75005ParisFrance
| | - Alexis Lamiable
- Institut de Minéralogie, de Physique des Matériaux et de Cosmochimie (IMPMC)Institut de recherche pour le développement (IRD)UMR CNRS 7590Muséum National d'Histoire NaturelleSorbonne Université75005ParisFrance
| | - Jean‐Paul Mornon
- Institut de Minéralogie, de Physique des Matériaux et de Cosmochimie (IMPMC)Institut de recherche pour le développement (IRD)UMR CNRS 7590Muséum National d'Histoire NaturelleSorbonne Université75005ParisFrance
| | - Isabelle Callebaut
- Institut de Minéralogie, de Physique des Matériaux et de Cosmochimie (IMPMC)Institut de recherche pour le développement (IRD)UMR CNRS 7590Muséum National d'Histoire NaturelleSorbonne Université75005ParisFrance
| |
Collapse
|
28
|
Laban H, Weigert A, Zink J, Elgheznawy A, Schürmann C, Günther L, Abdel Malik R, Bothur S, Wingert S, Bremer R, Rieger MA, Brüne B, Brandes RP, Fleming I, Benz PM. VASP regulates leukocyte infiltration, polarization, and vascular repair after ischemia. J Cell Biol 2018; 217:1503-1519. [PMID: 29507126 PMCID: PMC5881493 DOI: 10.1083/jcb.201702048] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 07/06/2017] [Accepted: 01/26/2018] [Indexed: 01/14/2023] Open
Abstract
In ischemic vascular diseases, leukocyte recruitment and polarization are crucial for revascularization and tissue repair. The study of Laban et al. provides evidence that VASP is a major regulator of leukocyte recruitment and polarization and vascular repair after ischemia. Mechanistically, the study supports a novel role of VASP in chemokine receptor trafficking. In ischemic vascular diseases, leukocyte recruitment and polarization are crucial for revascularization and tissue repair. We investigated the role of vasodilator-stimulated phosphoprotein (VASP) in vascular repair. After hindlimb ischemia induction, blood flow recovery, angiogenesis, arteriogenesis, and leukocyte infiltration into ischemic muscles in VASP−/− mice were accelerated. VASP deficiency also elevated the polarization of the macrophages through increased signal transducer and activator of transcription (STAT) signaling, which augmented the release of chemokines, cytokines, and growth factors to promote leukocyte recruitment and vascular repair. Importantly, VASP deletion in bone marrow–derived cells was sufficient to mimic the increased blood flow recovery of global VASP−/− mice. In chemotaxis experiments, VASP−/− neutrophils/monocytes were significantly more responsive to M1-related chemokines than wild-type controls. Mechanistically, VASP formed complexes with the chemokine receptor CCR2 and β-arrestin-2, and CCR2 receptor internalization was significantly reduced in VASP−/− leukocytes. Our data indicate that VASP is a major regulator of leukocyte recruitment and polarization in postischemic revascularization and support a novel role of VASP in chemokine receptor trafficking.
Collapse
Affiliation(s)
- Hebatullah Laban
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt am Main, Germany.,German Centre of Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, Germany
| | - Andreas Weigert
- Institute of Biochemistry I-Pathobiochemistry, Faculty of Medicine, Goethe University, Frankfurt am Main, Germany
| | - Joana Zink
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt am Main, Germany.,German Centre of Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, Germany
| | - Amro Elgheznawy
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt am Main, Germany.,German Centre of Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, Germany
| | - Christoph Schürmann
- German Centre of Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, Germany.,Institute for Cardiovascular Physiology, Goethe University, Frankfurt am Main, Germany
| | - Lea Günther
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt am Main, Germany.,German Centre of Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, Germany
| | - Randa Abdel Malik
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt am Main, Germany.,German Centre of Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, Germany
| | - Sabrina Bothur
- LOEWE Center for Cell and Gene Therapy and Department for Medicine, Hematology/Oncology, Goethe University, Frankfurt am Main, Germany
| | - Susanne Wingert
- LOEWE Center for Cell and Gene Therapy and Department for Medicine, Hematology/Oncology, Goethe University, Frankfurt am Main, Germany
| | - Rolf Bremer
- HBB Datenkommunikation and Abrechnungssysteme, Hannover, Germany
| | - Michael A Rieger
- LOEWE Center for Cell and Gene Therapy and Department for Medicine, Hematology/Oncology, Goethe University, Frankfurt am Main, Germany
| | - Bernhard Brüne
- Institute of Biochemistry I-Pathobiochemistry, Faculty of Medicine, Goethe University, Frankfurt am Main, Germany
| | - Ralf P Brandes
- German Centre of Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, Germany.,Institute for Cardiovascular Physiology, Goethe University, Frankfurt am Main, Germany
| | - Ingrid Fleming
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt am Main, Germany.,German Centre of Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, Germany
| | - Peter M Benz
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt am Main, Germany .,German Centre of Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, Germany
| |
Collapse
|
29
|
Chiha S, Soicke A, Barone M, Müller M, Bruns J, Opitz R, Neudörfl JM, Kühne R, Schmalz HG. Design and Synthesis of Building Blocks for PPII-Helix Secondary-Structure Mimetics: A Stereoselective Entry to 4-Substituted 5-Vinylprolines. European J Org Chem 2017. [DOI: 10.1002/ejoc.201701584] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Slim Chiha
- Department of Chemistry; University of Cologne; Greinstrasse 4 50939 Cologne Germany
| | - Arne Soicke
- Department of Chemistry; University of Cologne; Greinstrasse 4 50939 Cologne Germany
| | - Matthias Barone
- Campus BerlinBuch; Leibniz-Institut für Molekulare Pharmakologie (FMP); 13125 Berlin Germany
| | - Matthias Müller
- Campus BerlinBuch; Leibniz-Institut für Molekulare Pharmakologie (FMP); 13125 Berlin Germany
| | - Judith Bruns
- Department of Chemistry; University of Cologne; Greinstrasse 4 50939 Cologne Germany
| | - Robert Opitz
- Campus BerlinBuch; Leibniz-Institut für Molekulare Pharmakologie (FMP); 13125 Berlin Germany
| | - Jörg-Martin Neudörfl
- Department of Chemistry; University of Cologne; Greinstrasse 4 50939 Cologne Germany
| | - Ronald Kühne
- Campus BerlinBuch; Leibniz-Institut für Molekulare Pharmakologie (FMP); 13125 Berlin Germany
| | - Hans-Günther Schmalz
- Department of Chemistry; University of Cologne; Greinstrasse 4 50939 Cologne Germany
| |
Collapse
|
30
|
Frappier V, Duran M, Keating AE. PixelDB: Protein-peptide complexes annotated with structural conservation of the peptide binding mode. Protein Sci 2017; 27:276-285. [PMID: 29024246 DOI: 10.1002/pro.3320] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 10/09/2017] [Accepted: 10/09/2017] [Indexed: 11/08/2022]
Abstract
PixelDB, the Peptide Exosite Location Database, compiles 1966 non-redundant, high-resolution structures of protein-peptide complexes filtered to minimize the impact of crystal packing on peptide conformation. The database is organized to facilitate study of structurally conserved versus non-conserved elements of protein-peptide engagement. PixelDB clusters complexes based on the structural similarity of the peptide-binding protein, and by comparing complexes within a cluster highlights examples of domains that engage peptides using more than one binding mode. PixelDB also identifies conserved peptide core structural motifs characteristic of each binding mode. Peptide regions that flank core motifs often make non-structurally conserved interactions with the protein surface in regions we call exosites. Many examples establish that exosite contacts can be important for enhancing protein binding and interaction specificity. PixelDB provides a resource for computational and structural biologists to study, model, and predict core-motif and exosite-contacting peptide interactions. PixelDB is available to the community without restriction in a convenient flat-file format with accompanying visualization tools.
Collapse
Affiliation(s)
- Vincent Frappier
- MIT Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Madeleine Duran
- MIT Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Amy E Keating
- MIT Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts.,MIT Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts
| |
Collapse
|
31
|
Acevedo LA, Greenwood AI, Nicholson LK. A Noncanonical Binding Site in the EVH1 Domain of Vasodilator-Stimulated Phosphoprotein Regulates Its Interactions with the Proline Rich Region of Zyxin. Biochemistry 2017; 56:4626-4636. [PMID: 28783324 DOI: 10.1021/acs.biochem.7b00618] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Vasodilator-stimulated phosphoprotein (VASP) is a processive actin polymerase with roles in the control of cell shape and cell migration. Through interaction with the cytoskeletal adaptor protein Zyxin, VASP can localize to damaged stress fibers where it serves to repair and reinforce these structures. VASP localization is mediated by its N-terminal Ena/VASP homology (EVH1) domain, which binds to the (W/F)PxφP motif (most commonly occurring as FPPPP) found in cytoskeletal proteins such as vinculin, lamellipodin, and Zyxin. Sequentially close clusters of four or five of these motifs frequently occur, as in the proline rich region of Zyxin with four such motifs. This suggests that tetrameric VASP might bind very tightly to Zyxin through avidity, with all four EVH1 domains binding to a single Zyxin molecule. Here, quantitative nuclear magnetic resonance titration analysis reveals a dominant bivalent 1:1 (Zyxin:EVH1) interaction between the Zyxin proline rich region and the VASP EVH1 domain that utilizes the EVH1 canonical binding site and a novel secondary binding site on the opposite face of the EVH1 domain. We further show that binding to the secondary binding site is specifically inhibited by mutation of VASP EVH1 domain residue Y39 to E, which mimics Abl-induced phosphorylation of Y39. On the basis of these findings, we propose a model in which phosphorylation of Y39 acts as a stoichiometry switch that governs binding partner selection by the constitutive VASP tetramer. These results have broader implications for other multivalent VASP EVH1 domain binding partners and for furthering our understanding of the role of Y39 phosphorylation in regulating VASP localization and cellular function.
Collapse
Affiliation(s)
- Lucila Andrea Acevedo
- Department of Molecular Biology and Genetics, Cornell University , Ithaca, New York 14853, United States
| | - Alexander I Greenwood
- Department of Applied Science, College of William and Mary , Williamsburg, Virginia 23185, United States
| | - Linda K Nicholson
- Department of Molecular Biology and Genetics, Cornell University , Ithaca, New York 14853, United States
| |
Collapse
|
32
|
Distinct VASP tetramers synergize in the processive elongation of individual actin filaments from clustered arrays. Proc Natl Acad Sci U S A 2017; 114:E5815-E5824. [PMID: 28667124 DOI: 10.1073/pnas.1703145114] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Ena/VASP proteins act as actin polymerases that drive the processive elongation of filament barbed ends in membrane protrusions or at the surface of bacterial pathogens. Based on previous analyses of fast and slow elongating VASP proteins by in vitro total internal reflection fluorescence microscopy (TIRFM) and kinetic and thermodynamic measurements, we established a kinetic model of Ena/VASP-mediated actin filament elongation. At steady state, it entails that tetrameric VASP uses one of its arms to processively track growing filament barbed ends while three G-actin-binding sites (GABs) on other arms are available to recruit and deliver monomers to the filament tip, suggesting that VASP operates as a single tetramer in solution or when clustered on a surface, albeit processivity and resistance toward capping protein (CP) differ dramatically between both conditions. Here, we tested the model by variation of the oligomerization state and by increase of the number of GABs on individual polypeptide chains. In excellent agreement with model predictions, we show that in solution the rates of filament elongation directly correlate with the number of free GABs. Strikingly, however, irrespective of the oligomerization state or presence of additional GABs, filament elongation on a surface invariably proceeded with the same rate as with the VASP tetramer, demonstrating that adjacent VASP molecules synergize in the elongation of a single filament. Additionally, we reveal that actin ATP hydrolysis is not required for VASP-mediated filament assembly. Finally, we show evidence for the requirement of VASP to form tetramers and provide an amended model of processive VASP-mediated actin assembly in clustered arrays.
Collapse
|
33
|
Rajadurai CV, Havrylov S, Coelho PP, Ratcliffe CDH, Zaoui K, Huang BH, Monast A, Chughtai N, Sangwan V, Gertler FB, Siegel PM, Park M. 5'-Inositol phosphatase SHIP2 recruits Mena to stabilize invadopodia for cancer cell invasion. J Cell Biol 2016; 214:719-34. [PMID: 27597754 PMCID: PMC5021089 DOI: 10.1083/jcb.201501003] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2015] [Accepted: 08/05/2016] [Indexed: 12/11/2022] Open
Abstract
Invadopodia are membrane protrusions used by cancer cells to remodel and invade the extracellular matrix. Here, Rajadurai et al. show that the lipid phosphatase SHIP2 recruits the Ena/VASP-family actin regulatory protein Mena to stabilize invadopodia membrane protrusions and promote cell invasion. Invadopodia are specialized membrane protrusions that support degradation of extracellular matrix (ECM) by cancer cells, allowing invasion and metastatic spread. Although early stages of invadopodia assembly have been elucidated, little is known about maturation of invadopodia into structures competent for ECM proteolysis. The localized conversion of phosphatidylinositol(3,4,5)-triphosphate and accumulation of phosphatidylinositol(3,4)-bisphosphate at invadopodia is a key determinant for invadopodia maturation. Here we investigate the role of the 5′-inositol phosphatase, SHIP2, and reveal an unexpected scaffold function of SHIP2 as a prerequisite for invadopodia-mediated ECM degradation. Through biochemical and structure-function analyses, we identify specific interactions between SHIP2 and Mena, an Ena/VASP-family actin regulatory protein. We demonstrate that SHIP2 recruits Mena, but not VASP, to invadopodia and that disruption of SHIP2–Mena interaction in cancer cells leads to attenuated capacity for ECM degradation and invasion in vitro, as well as reduced metastasis in vivo. Together, these findings identify SHIP2 as a key modulator of carcinoma invasiveness and a target for metastatic disease.
Collapse
Affiliation(s)
- Charles V Rajadurai
- Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montréal, Québec H3A 1A1, Canada Department of Biochemistry, McGill University, Montréal, Québec H3A 1A1, Canada
| | - Serhiy Havrylov
- Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montréal, Québec H3A 1A1, Canada
| | - Paula P Coelho
- Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montréal, Québec H3A 1A1, Canada Department of Biochemistry, McGill University, Montréal, Québec H3A 1A1, Canada
| | - Colin D H Ratcliffe
- Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montréal, Québec H3A 1A1, Canada Department of Biochemistry, McGill University, Montréal, Québec H3A 1A1, Canada
| | - Kossay Zaoui
- Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montréal, Québec H3A 1A1, Canada
| | - Bruce H Huang
- Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montréal, Québec H3A 1A1, Canada Department of Biochemistry, McGill University, Montréal, Québec H3A 1A1, Canada
| | - Anie Monast
- Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montréal, Québec H3A 1A1, Canada
| | - Naila Chughtai
- Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montréal, Québec H3A 1A1, Canada
| | - Veena Sangwan
- Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montréal, Québec H3A 1A1, Canada Department of Oncology, McGill University, Montréal, Québec H3A 1A1, Canada
| | - Frank B Gertler
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139 Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Peter M Siegel
- Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montréal, Québec H3A 1A1, Canada Department of Biochemistry, McGill University, Montréal, Québec H3A 1A1, Canada Department of Medicine, McGill University, Montréal, Québec H3A 1A1, Canada
| | - Morag Park
- Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montréal, Québec H3A 1A1, Canada Department of Biochemistry, McGill University, Montréal, Québec H3A 1A1, Canada Department of Medicine, McGill University, Montréal, Québec H3A 1A1, Canada Department of Oncology, McGill University, Montréal, Québec H3A 1A1, Canada
| |
Collapse
|
34
|
Krol A, Henle SJ, Goodrich LV. Fat3 and Ena/VASP proteins influence the emergence of asymmetric cell morphology in the developing retina. Development 2016; 143:2172-82. [PMID: 27122175 DOI: 10.1242/dev.133678] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 04/14/2016] [Indexed: 12/27/2022]
Abstract
Neurons exhibit asymmetric morphologies throughout development - from migration to the elaboration of axons and dendrites - that are correctly oriented for the flow of information. For instance, retinal amacrine cells migrate towards the inner plexiform layer (IPL) and then retract their trailing processes, thereby acquiring a unipolar morphology with a single dendritic arbor restricted to the IPL. Here, we provide evidence that the Fat-like cadherin Fat3 acts during multiple stages of amacrine cell development in mice to orient overall changes in cell shape towards the IPL. Using a time-lapse imaging assay, we found that developing amacrine cells are less directed towards the IPL in the absence of Fat3, during both migration and retraction. Consistent with its predicted role as a cell-surface receptor, Fat3 functions cell-autonomously and is able to influence the cytoskeleton directly through its intracellular domain, which can bind and localize Ena/VASP family actin regulators. Indeed, a change in Ena/VASP protein distribution is sufficient to recapitulate the Fat3 mutant amacrine cell phenotype. Thus, Fat-like proteins might control the polarized development of tissues by sculpting the cytoskeleton of individual cells.
Collapse
Affiliation(s)
- Alexandra Krol
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Steven J Henle
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Lisa V Goodrich
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
35
|
Pillich H, Puri M, Chakraborty T. ActA of Listeria monocytogenes and Its Manifold Activities as an Important Listerial Virulence Factor. Curr Top Microbiol Immunol 2016; 399:113-132. [PMID: 27726006 DOI: 10.1007/82_2016_30] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Listeria monocytogenes is a ubiquitously occurring gram-positive bacterium in the environment that causes listeriosis, one of the deadliest foodborne infections known today. It is a versatile facultative intracellular pathogen capable of growth within the host's cytosolic compartment. Following entry into the host cell, L. monocytogenes escapes from vacuolar compartments to the cytosol, where the bacterium begins a remarkable journey within the host cytoplasm, culminating in bacterial spread from cell to cell, to deeper tissues and organs. This dissemination process depends on the ability of the bacterium to harness central components of the host cell actin cytoskeleton using the surface bound bacterial factor ActA (actin assembly inducing protein). Hence ActA plays a major role in listerial virulence, and its absence renders bacteria intracellularly immotile and essentially non-infectious. As the bacterium, moving by building a network of filamentous actin behind itself that is often referred to as its actin tail, encounters cell-cell contacts it forms double-vacuolar protrusions that allow it to enter the neighboring cell where the cycle then continues. Recent studies have now implicated ActA in other stages of the life cycle of L. monocytogenes. These include extracellular properties of aggregation and biofilm formation to mediate colonization of the gut lumen, promotion and enhancement of bacterial host cell entry, evasion of autophagy, vacuolar exit, as well as nuclear factor of kappa light polypeptide gene enhancer in B-cells (NF-κB) activation. These novel properties provide a new view of ActA and help explain its role as an essential virulence factor of L. monocytogenes.
Collapse
Affiliation(s)
- Helena Pillich
- Institute of Medical Microbiology, Justus-Liebig University Giessen, Schubertstrasse 81, 35392, Giessen, Germany
| | - Madhu Puri
- Institute of Medical Microbiology, Justus-Liebig University Giessen, Schubertstrasse 81, 35392, Giessen, Germany
| | - Trinad Chakraborty
- Institute of Medical Microbiology, Justus-Liebig University Giessen, Schubertstrasse 81, 35392, Giessen, Germany.
| |
Collapse
|
36
|
Zeng J, Tu WW, Lazar L, Chen DN, Zhao JS, Xu J. Hyperphosphorylation of microfilament-associated proteins is involved in microcystin-LR-induced toxicity in HL7702 cells. ENVIRONMENTAL TOXICOLOGY 2015; 30:981-988. [PMID: 24677693 DOI: 10.1002/tox.21974] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Revised: 02/05/2014] [Accepted: 02/09/2014] [Indexed: 06/03/2023]
Abstract
Microcystin-LR (MC-LR) has been regarded as a hepatotoxin, which can cause cytoskeletal reorganization, especially of the actin filaments. However, the underlying mechanisms remain unclear. In this study, whether MC-LR could induce microfilaments disruption was verified in the normal human liver cell line HL7702; and then the transcription, translation, and phosphorylation levels of major microfilament-associated proteins were measured; finally, the underlying mechanisms was investigated. After treatment with MC-LR, the actin filaments lost their characteristic filamentous organization in the cells, demonstrating increased actin depolymerization. The mRNA and protein levels of ezrin, vasodilator-stimulated phosphoprotein (VASP), actin-related protein2/3, and cofilin remained unchanged. However, the phosphorylation levels of ezrin and VASP were increased, when treated with 10 μM MC-LR. Moreover, P38 and ERK1/2 were involved in MC-LR-induced hyperphosphorylation of microfilament-associated proteins. In summary, this study demonstrates that MC-LR can cause disruption of actin filaments in HL7702 cells due to MC-LR-induced mitogen-activated protein kinase pathway activation and hyperphosphorylation of different types of microfilament-associated proteins.
Collapse
Affiliation(s)
- Jing Zeng
- Department of Preventive Medicine, School of Medicine, Ningbo University, Ningbo, 315211, China
| | - Wei-Wei Tu
- Department of Preventive Medicine, School of Medicine, Ningbo University, Ningbo, 315211, China
| | - Lissy Lazar
- Department of Preventive Medicine, School of Medicine, Ningbo University, Ningbo, 315211, China
| | - Dong-Ni Chen
- Department of Preventive Medicine, School of Medicine, Ningbo University, Ningbo, 315211, China
| | - Jin-Shun Zhao
- Department of Preventive Medicine, School of Medicine, Ningbo University, Ningbo, 315211, China
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, 315211, China
| | - Jin Xu
- Department of Preventive Medicine, School of Medicine, Ningbo University, Ningbo, 315211, China
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, 315211, China
| |
Collapse
|
37
|
Reuter C, Opitz R, Soicke A, Dohmen S, Barone M, Chiha S, Klein MT, Neudörfl JM, Kühne R, Schmalz HG. Design and Stereoselective Synthesis of ProM-2: A Spirocyclic Diproline Mimetic with Polyproline Type II (PPII) Helix Conformation. Chemistry 2015; 21:8464-70. [DOI: 10.1002/chem.201406493] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Revised: 03/11/2015] [Indexed: 11/08/2022]
|
38
|
A modular toolkit to inhibit proline-rich motif-mediated protein-protein interactions. Proc Natl Acad Sci U S A 2015; 112:5011-6. [PMID: 25848013 DOI: 10.1073/pnas.1422054112] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Small-molecule competitors of protein-protein interactions are urgently needed for functional analysis of large-scale genomics and proteomics data. Particularly abundant, yet so far undruggable, targets include domains specialized in recognizing proline-rich segments, including Src-homology 3 (SH3), WW, GYF, and Drosophila enabled (Ena)/vasodilator-stimulated phosphoprotein (VASP) homology 1 (EVH1) domains. Here, we present a modular strategy to obtain an extendable toolkit of chemical fragments (ProMs) designed to replace pairs of conserved prolines in recognition motifs. As proof-of-principle, we developed a small, selective, peptidomimetic inhibitor of Ena/VASP EVH1 domain interactions. Highly invasive MDA MB 231 breast-cancer cells treated with this ligand showed displacement of VASP from focal adhesions, as well as from the front of lamellipodia, and strongly reduced cell invasion. General applicability of our strategy is illustrated by the design of an ErbB4-derived ligand containing two ProM-1 fragments, targeting the yes-associated protein 1 (YAP1)-WW domain with a fivefold higher affinity.
Collapse
|
39
|
Ke Y, Tan Y, Wei N, Yang F, Yang H, Cao S, Wang X, Wang J, Han Y, Bi Y, Cui Y, Yan Y, Song Y, Yang X, Du Z, Yang R. Yersinia protein kinase A phosphorylates vasodilator-stimulated phosphoprotein to modify the host cytoskeleton. Cell Microbiol 2014; 17:473-85. [PMID: 25298072 DOI: 10.1111/cmi.12378] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 09/13/2014] [Accepted: 10/04/2014] [Indexed: 01/03/2023]
Abstract
Pathogenic Yersinia species evolved a type III secretion system that injects a set of effectors into the host cell cytosol to promote infection. One of these effectors, Yersinia protein kinase A (YpkA), is a multidomain effector that harbours a Ser/Thr kinase domain and a guanine dissociation inhibitor (GDI) domain. The intercellular targets of the kinase and GDI domains of YpkA were identified to be Gαq and the small GTPases RhoA and Rac1, respectively, which synergistically induce cytotoxic effects on infected cells. In this study, we demonstrate that vasodilator-stimulated phosphoprotein (VASP), which is critical for regulation of actin assembly, cell adhesion and motility, is a direct substrate of YpkA kinase activity. Ectopic co-expression of YpkA and VASP in HEK293T cells leads to the phosphorylation of VASP at S157, and YpkA kinase activity is essential for VASP phosphorylation at this site. Moreover, YpkA directly phosphorylates VASP in in vitro kinase assay. YpkA-mediated VASP phosphorylation significantly inhibits actin polymerization and promotes the disruption of actin cytoskeleton, which inhibits the phagocytosis. Taken together, our study found a novel molecular mechanism used by YpkA to disrupt cytoskeleton dynamics, thereby promoting the anti-phagocytosis ability of pathogenic Yersiniae.
Collapse
Affiliation(s)
- Yuehua Ke
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, 100071, China; Beijing Institute of Disease Control and Prevention, Beijing, 100071, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Wang H, Liu J, Lin S, Wang B, Xing M, Guo Z, Xu L. MCLR-induced PP2A inhibition and subsequent Rac1 inactivation and hyperphosphorylation of cytoskeleton-associated proteins are involved in cytoskeleton rearrangement in SMMC-7721 human liver cancer cell line. CHEMOSPHERE 2014; 112:141-153. [PMID: 25048900 DOI: 10.1016/j.chemosphere.2014.03.130] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Revised: 03/26/2014] [Accepted: 03/27/2014] [Indexed: 06/03/2023]
Abstract
Cyanobacteria-derived toxin microcystin-LR (MCLR) has been widely investigated in its effects on normal cells, there is little information concerning its effects on cancer cells. In the present study, the SMMC-7721 human liver cancer cell line treated with MCLR was used to investigate the change of PP2A, cytoskeleton rearrangement, phosphorylation levels of PP2A substrates that related with cytoskeleton stability and explored underlying mechanisms. Here, we confirmed that MCLR entered into SMMC-7721 cells, bound to PP2A/C subunit and inhibited the activity of PP2A. The upregulation of phosphorylation of the PP2A/C subunit and PP2A regulation protein α4, as well as the change in the association of PP2A/C with α4, were responsible for the decrease in PP2A activity. Another novel finding is that the rearrangement of filamentous actin and microtubules led by MCLR may attribute to the increased phosphorylation of HSP27, VASP and cofilin due to PP2A inhibition. As a result of weakened interactions with PP2A and alterations in its subcellular localization, Rac1 may contribute to the cytoskeletal rearrangement induced by MCLR in SMMC-7721 cells. The current paper presents the first report demonstrating the characteristic of PP2A in MCLR exposed cancer cells, which were more susceptible to MCLR compared with the normal cell lines we previously found, which may be owing to the absence of some type of compensatory mechanisms. The hyperphosphorylation of cytoskeleton-associated proteins and Rac1 inactivation which were induced by inhibition of PP2A are shown to be involved in cytoskeleton rearrangement.
Collapse
Affiliation(s)
- Hao Wang
- Department of Biochemistry, School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Jinghui Liu
- Department of Biochemistry, School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Shuyan Lin
- Department of Biochemistry, School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Beilei Wang
- Department of Biochemistry, School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Mingluan Xing
- Zhejiang Center for Disease Control and Prevention, Hangzhou 310051, China
| | - Zonglou Guo
- Department of Biosystem Engineering, College of Biosystem Engineering and Food Science, Zhejiang University, Hangzhou 310058, China.
| | - Lihong Xu
- Department of Biochemistry, School of Medicine, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
41
|
Chen XJ, Squarr AJ, Stephan R, Chen B, Higgins TE, Barry DJ, Martin MC, Rosen MK, Bogdan S, Way M. Ena/VASP proteins cooperate with the WAVE complex to regulate the actin cytoskeleton. Dev Cell 2014; 30:569-84. [PMID: 25203209 PMCID: PMC4165403 DOI: 10.1016/j.devcel.2014.08.001] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2013] [Revised: 07/21/2014] [Accepted: 08/01/2014] [Indexed: 01/25/2023]
Abstract
Ena/VASP proteins and the WAVE regulatory complex (WRC) regulate cell motility by virtue of their ability to independently promote actin polymerization. We demonstrate that Ena/VASP and the WRC control actin polymerization in a cooperative manner through the interaction of the Ena/VASP EVH1 domain with an extended proline rich motif in Abi. This interaction increases cell migration and enables VASP to cooperatively enhance WRC stimulation of Arp2/3 complex-mediated actin assembly in vitro in the presence of Rac. Loss of this interaction in Drosophila macrophages results in defects in lamellipodia formation, cell spreading, and redistribution of Ena to the tips of filopodia-like extensions. Rescue experiments of abi mutants also reveals a physiological requirement for the Abi:Ena interaction in photoreceptor axon targeting and oogenesis. Our data demonstrate that the activities of Ena/VASP and the WRC are intimately linked to ensure optimal control of actin polymerization during cell migration and development.
Collapse
Affiliation(s)
- Xing Judy Chen
- Cell Motility Laboratory, London Research Institute, Cancer Research UK, 44 Lincoln's Inn Fields, London WC2A 3LY, UK
| | - Anna Julia Squarr
- Institute of Neurobiology, University of Muenster, Badestrasse 9, 48149 Muenster, Germany
| | - Raiko Stephan
- Institute of Neurobiology, University of Muenster, Badestrasse 9, 48149 Muenster, Germany
| | - Baoyu Chen
- Howard Hughes Medical Institute and Department of Biophysics, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Theresa E Higgins
- Cell Motility Laboratory, London Research Institute, Cancer Research UK, 44 Lincoln's Inn Fields, London WC2A 3LY, UK
| | - David J Barry
- Cell Motility Laboratory, London Research Institute, Cancer Research UK, 44 Lincoln's Inn Fields, London WC2A 3LY, UK
| | - Morag C Martin
- Cell Motility Laboratory, London Research Institute, Cancer Research UK, 44 Lincoln's Inn Fields, London WC2A 3LY, UK
| | - Michael K Rosen
- Howard Hughes Medical Institute and Department of Biophysics, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Sven Bogdan
- Institute of Neurobiology, University of Muenster, Badestrasse 9, 48149 Muenster, Germany.
| | - Michael Way
- Cell Motility Laboratory, London Research Institute, Cancer Research UK, 44 Lincoln's Inn Fields, London WC2A 3LY, UK.
| |
Collapse
|
42
|
Barzik M, McClain LM, Gupton SL, Gertler FB. Ena/VASP regulates mDia2-initiated filopodial length, dynamics, and function. Mol Biol Cell 2014; 25:2604-19. [PMID: 24989797 PMCID: PMC4148250 DOI: 10.1091/mbc.e14-02-0712] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Filopodia are long plasma membrane extensions involved in the formation of adhesive, contractile, and protrusive actin-based structures in spreading and migrating cells. Whether filopodia formed by different molecular mechanisms equally support these cellular functions is unresolved. We used Enabled/vasodilator-stimulated phosphoprotein (Ena/VASP)-deficient MV(D7) fibroblasts, which are also devoid of endogenous mDia2, as a model system to investigate how these different actin regulatory proteins affect filopodia morphology and dynamics independently of one another. Filopodia initiated by either Ena/VASP or mDia2 contained similar molecular inventory but differed significantly in parameters such as number, length, F-actin organization, lifetime, and protrusive persistence. Moreover, in the absence of Ena/VASP, filopodia generated by mDia2 did not support initiation of integrin-dependent signaling cascades required for adhesion and subsequent lamellipodial extension, thereby causing a defect in early cell spreading. Coexpression of VASP with constitutively active mDia2(M/A) rescued these early adhesion defects. We conclude that Ena/VASP and mDia2 support the formation of filopodia with significantly distinct properties and that Ena/VASP regulates mDia2-initiated filopodial morphology, dynamics, and function.
Collapse
Affiliation(s)
- Melanie Barzik
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Leslie M McClain
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Stephanie L Gupton
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Frank B Gertler
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
| |
Collapse
|
43
|
Greenwood AI, Kwon J, Nicholson LK. Isomerase-catalyzed binding of interleukin-1 receptor-associated kinase 1 to the EVH1 domain of vasodilator-stimulated phosphoprotein. Biochemistry 2014; 53:3593-607. [PMID: 24857403 DOI: 10.1021/bi500031e] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Interleukin-1 receptor-associated kinase 1 (IRAK1) is a crucial signaling kinase in the immune system, involved in Toll-like receptor signaling. Vasodilator-stimulated phosphoprotein (VASP) is a central player in cell migration that regulates actin polymerization and connects signaling events to cytoskeletal remodeling. A VASP–IRAK1 interaction is thought to be important in controlling macrophage migration in response to protein kinase C-ε activation. We show that the monomeric VASP EVH1 domain directly binds to the 168WPPPP172 motif in the IRAK1 undefined domain (IRAK1-UD) with moderate affinity (KDApp = 203 ± 3 μM). We further show that this motif adopts distinct cis and trans isomers for the Trp168–Pro169 peptide bond with nearly equal populations, and that binding to the VASP EVH1 domain is specific for the trans isomer, coupling binding to isomerization. Nuclear magnetic resonance line shape analysis and tryptophan fluorescence experiments reveal the complete kinetics and thermodynamics of the binding reaction, showing diffusion-limited binding to the trans isomer followed by slow, isomerization-dependent binding. We further demonstrate that the peptidyl-prolyl isomerase cyclophilin A (CypA) catalyzes isomerization of the Trp168–Pro169 peptide bond and accelerates binding of the IRAK1-UD to the VASP EVH1 domain. We propose that binding of IRAK1 to tetrameric VASP is regulated by avidity through the assembly of IRAK1 onto receptor-anchored signaling complexes and that an isomerase such as CypA may modulate IRAK1 signaling in vivo. These studies demonstrate a direct interaction between IRAK1 and VASP and suggest a potential mechanism for how this interaction might be regulated by both assembly of IRAK1 onto an activated signaling complex and PPIase enzymes.
Collapse
Affiliation(s)
- Alexander I Greenwood
- Department of Molecular Biology and Genetics, Cornell University , Ithaca, New York 14853, United States
| | | | | |
Collapse
|
44
|
Bilancia CG, Winkelman JD, Tsygankov D, Nowotarski SH, Sees JA, Comber K, Evans I, Lakhani V, Wood W, Elston TC, Kovar DR, Peifer M. Enabled negatively regulates diaphanous-driven actin dynamics in vitro and in vivo. Dev Cell 2014; 28:394-408. [PMID: 24576424 PMCID: PMC3992947 DOI: 10.1016/j.devcel.2014.01.015] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Revised: 12/20/2013] [Accepted: 01/15/2014] [Indexed: 11/03/2022]
Abstract
Actin regulators facilitate cell migration by controlling cell protrusion architecture and dynamics. As the behavior of individual actin regulators becomes clear, we must address why cells require multiple regulators with similar functions and how they cooperate to create diverse protrusions. We characterized Diaphanous (Dia) and Enabled (Ena) as a model, using complementary approaches: cell culture, biophysical analysis, and Drosophila morphogenesis. We found that Dia and Ena have distinct biochemical properties that contribute to the different protrusion morphologies each induces. Dia is a more processive, faster elongator, paralleling the long, stable filopodia it induces in vivo, while Ena promotes filopodia with more dynamic changes in number, length, and lifetime. Acting together, Ena and Dia induce protrusions distinct from those induced by either alone, with Ena reducing Dia-driven protrusion length and number. Consistent with this, EnaEVH1 binds Dia directly and inhibits DiaFH1FH2-mediated nucleation in vitro. Finally, Ena rescues hemocyte migration defects caused by activated Dia. Dia and Ena differ biochemically, promoting distinct filopodia dynamics Dia and Ena colocalization negatively regulates filopodia Ena’s EVH1 binds Dia’s FH1 and reduces Dia-driven filopodia and actin nucleation Ena rescues DiaΔDAD inhibition of hemocyte migration speed to wounds in vivo
Collapse
Affiliation(s)
- Colleen G Bilancia
- Biology Department, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jonathan D Winkelman
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL 60637, USA
| | - Denis Tsygankov
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Stephanie H Nowotarski
- Biology Department, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jennifer A Sees
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL 60637, USA
| | - Kate Comber
- Department of Biology and Biochemistry, University of Bath, Bath BA2 7AY, UK
| | - Iwan Evans
- Department of Biology and Biochemistry, University of Bath, Bath BA2 7AY, UK
| | - Vinal Lakhani
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Will Wood
- Department of Biology and Biochemistry, University of Bath, Bath BA2 7AY, UK
| | - Timothy C Elston
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - David R Kovar
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL 60637, USA; Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL 60637, USA
| | - Mark Peifer
- Biology Department, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
45
|
Döppler H, Storz P. Regulation of VASP by phosphorylation: consequences for cell migration. Cell Adh Migr 2013; 7:482-6. [PMID: 24401601 DOI: 10.4161/cam.27351] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Phosphorylations control all aspects of vasodilator-stimulated phospho-protein (VASP) function. Mapped phosphorylation sites include Y39, S157, S239, T278, and S322, and multiple kinases have been shown to mediate their phosphorylation. Recently, Protein Kinase D1 (PKD1) as a direct kinase for S157 and S322 joined this group. While S157 phosphorylation generally seems to serve as a signal for membrane localization, phosphorylations at S322 or at S239 and T278 have opposite effects on F-actin accumulation. In migrating cells, S322 phosphorylation increases filopodia numbers and length, while S239/T278 phosphorylations decrease these and also disrupt formation of focal adhesions. Therefore, the kinases mediating these phosphorylations can be seen as switches needed to facilitate cell motility.
Collapse
Affiliation(s)
- Heike Döppler
- Department of Cancer Biology; Mayo Clinic Comprehensive Cancer Center; Mayo Clinic; Jacksonville, FL USA
| | - Peter Storz
- Department of Cancer Biology; Mayo Clinic Comprehensive Cancer Center; Mayo Clinic; Jacksonville, FL USA
| |
Collapse
|
46
|
Berisio R, Vitagliano L. Polyproline and triple helix motifs in host-pathogen recognition. Curr Protein Pept Sci 2013; 13:855-65. [PMID: 23305370 PMCID: PMC3707005 DOI: 10.2174/138920312804871157] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2012] [Revised: 08/10/2012] [Accepted: 08/30/2012] [Indexed: 01/18/2023]
Abstract
Secondary structure elements often mediate protein-protein interactions. Despite their low abundance in folded proteins, polyproline II (PPII) and its variant, the triple helix, are frequently involved in protein-protein interactions, likely due to their peculiar propensity to be solvent-exposed. We here review the role of PPII and triple helix in mediating host-pathogen interactions, with a particular emphasis to the structural aspects of these processes. After a brief description of the basic structural features of these elements, examples of host-pathogen interactions involving these motifs are illustrated. Literature data suggest that the role played by PPII motif in these processes is twofold. Indeed, PPII regions may directly mediate interactions between proteins of the host and the pathogen. Alternatively, PPII may act as structural spacers needed for the correct positioning of the elements needed for adhesion and infectivity. Recent investigations have highlighted that collagen triple helix is also a common target for bacterial adhesins. Although structural data on complexes between adhesins and collagen models are rather limited, experimental and theoretical studies have unveiled some interesting clues of the recognition process. Interestingly, very recent data show that not only is the triple helix used by pathogens as a target in the host-pathogen interaction but it may also act as a bait in these processes since bacterial proteins containing triple helix regions have been shown to interact with host proteins. As both PPII and triple helix expose several main chain non-satisfied hydrogen bond acceptors and donors, both elements are highly solvated. The preservation of the solvation state of both PPII and triple helix upon protein-protein interaction is an emerging aspect that will be here thoroughly discussed.
Collapse
Affiliation(s)
- Rita Berisio
- Istituto di Biostrutture e Bioimmagini, CNR via Mezzocannone 16, I-80134 Napoli, Italy
| | | |
Collapse
|
47
|
Döppler HR, Bastea LI, Lewis-Tuffin LJ, Anastasiadis PZ, Storz P. Protein kinase D1-mediated phosphorylations regulate vasodilator-stimulated phosphoprotein (VASP) localization and cell migration. J Biol Chem 2013; 288:24382-93. [PMID: 23846685 PMCID: PMC3750140 DOI: 10.1074/jbc.m113.474676] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2013] [Revised: 07/08/2013] [Indexed: 11/06/2022] Open
Abstract
Enabled/Vasodilator-stimulated phosphoprotein (Ena/VASP) protein family members link actin dynamics and cellular signaling pathways. VASP localizes to regions of dynamic actin reorganization such as the focal adhesion contacts, the leading edge or filopodia, where it contributes to F-actin filament elongation. Here we identify VASP as a novel substrate for protein kinase D1 (PKD1). We show that PKD1 directly phosphorylates VASP at two serine residues, Ser-157 and Ser-322. These phosphorylations occur in response to RhoA activation and mediate VASP re-localization from focal contacts to the leading edge region. The net result of this PKD1-mediated phosphorylation switch in VASP is increased filopodia formation and length at the leading edge. However, such signaling when persistent induced membrane ruffling and decreased cell motility.
Collapse
Affiliation(s)
- Heike R. Döppler
- From the Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Mayo Clinic, Jacksonville, Florida 32224
| | - Ligia I. Bastea
- From the Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Mayo Clinic, Jacksonville, Florida 32224
| | - Laura J. Lewis-Tuffin
- From the Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Mayo Clinic, Jacksonville, Florida 32224
| | - Panos Z. Anastasiadis
- From the Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Mayo Clinic, Jacksonville, Florida 32224
| | - Peter Storz
- From the Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Mayo Clinic, Jacksonville, Florida 32224
| |
Collapse
|
48
|
Tateya S, Rizzo-De Leon N, Handa P, Cheng AM, Morgan-Stevenson V, Ogimoto K, Kanter JE, Bornfeldt KE, Daum G, Clowes AW, Chait A, Kim F. VASP increases hepatic fatty acid oxidation by activating AMPK in mice. Diabetes 2013; 62:1913-22. [PMID: 23349495 PMCID: PMC3661609 DOI: 10.2337/db12-0325] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Activation of AMP-activated protein kinase (AMPK) signaling reduces hepatic steatosis and hepatic insulin resistance; however, its regulatory mechanisms are not fully understood. In this study, we sought to determine whether vasodilator-stimulated phosphoprotein (VASP) signaling improves lipid metabolism in the liver and, if so, whether VASP's effects are mediated by AMPK. We show that disruption of VASP results in significant hepatic steatosis as a result of significant impairment of fatty acid oxidation, VLDL-triglyceride (TG) secretion, and AMPK signaling. Overexpression of VASP in hepatocytes increased AMPK phosphorylation and fatty acid oxidation and reduced hepatocyte TG accumulation; however, these responses were suppressed in the presence of an AMPK inhibitor. Restoration of AMPK phosphorylation by administration of 5-aminoimidazole-4-carboxamide riboside in Vasp(-/-) mice reduced hepatic steatosis and normalized fatty acid oxidation and VLDL-TG secretion. Activation of VASP by the phosphodiesterase-5 inhibitor, sildenafil, in db/db mice reduced hepatic steatosis and increased phosphorylated (p-)AMPK and p-acetyl CoA carboxylase. In Vasp(-/-) mice, however, sildendafil treatment did not increase p-AMPK or reduce hepatic TG content. These studies identify a role of VASP to enhance hepatic fatty acid oxidation by activating AMPK and to promote VLDL-TG secretion from the liver.
Collapse
Affiliation(s)
- Sanshiro Tateya
- Department of Medicine, University of Washington, Seattle, Washington, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Braun JE, Truffault V, Boland A, Huntzinger E, Chang CT, Haas G, Weichenrieder O, Coles M, Izaurralde E. A direct interaction between DCP1 and XRN1 couples mRNA decapping to 5′ exonucleolytic degradation. Nat Struct Mol Biol 2012; 19:1324-31. [DOI: 10.1038/nsmb.2413] [Citation(s) in RCA: 115] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2012] [Accepted: 09/18/2012] [Indexed: 11/09/2022]
|
50
|
Mise N, Savai R, Yu H, Schwarz J, Kaminski N, Eickelberg O. Zyxin is a transforming growth factor-β (TGF-β)/Smad3 target gene that regulates lung cancer cell motility via integrin α5β1. J Biol Chem 2012; 287:31393-405. [PMID: 22778267 DOI: 10.1074/jbc.m112.357624] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Although TGF-β acts as a tumor suppressor in normal tissues and in early carcinogenesis, these tumor suppressor effects are lost in advanced malignancies. Single cell migration and epithelial-mesenchymal transition (EMT), both of which are regulated by TGF-β, are critical steps in mediating cancer progression. Here, we sought to identify novel direct targets of TGF-β signaling in lung cancer cells and have indentified the zyxin gene as a target of Smad3-mediated TGF-β1 signaling. Zyxin concentrates at focal adhesions and along the actin cytoskeleton; as such, we hypothesized that cytoskeletal organization, motility, and EMT in response to TGF-β1 might be regulated by zyxin expression. We show that TGF-β1 treatment of lung cancer cells caused rapid phospho-Smad3-dependent expression of zyxin. Zyxin expression was critical for the formation and integrity of cell adherens junctions. Silencing of zyxin decreased expression of the focal adhesion protein vasodilator-activated phospho-protein (VASP), although the formation and morphology of focal adhesions remained unchanged. Zyxin-depleted cells displayed significantly increased integrin α5β1 levels, accompanied by enhanced adhesion to fibronectin and acquisition of a mesenchymal phenotype in response to TGF-β1. Zyxin silencing led to elevated integrin α5β1-dependent single cell motility. Importantly, these features are mirrored in the K-ras-driven mouse model of lung cancer. Here, lung tumors revealed decreased levels of both zyxin and phospho-Smad3 when compared with normal tissues. Our data thus demonstrate that zyxin is a novel functional target and effector of TGF-β signaling in lung cancer. By regulating cell-cell junctions, integrin α5β1 expression, and cell-extracellular matrix adhesion, zyxin may regulate cancer cell motility and EMT during lung cancer development and progression.
Collapse
Affiliation(s)
- Nikica Mise
- Comprehensive Pneumology Center, University Hospital, Ludwig-Maximilians University and Helmholtz Zentrum München, 81377 Munich, Germany.
| | | | | | | | | | | |
Collapse
|