1
|
Zhao H, Zhang X, Zhang N, Zhu L, Lian H. The interplay between Salmonella and host: Mechanisms and strategies for bacterial survival. CELL INSIGHT 2025; 4:100237. [PMID: 40177681 PMCID: PMC11964643 DOI: 10.1016/j.cellin.2025.100237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 01/20/2025] [Accepted: 01/21/2025] [Indexed: 04/05/2025]
Abstract
Salmonella, an intracellular pathogen, infects both humans and animals, causing diverse diseases such as gastroenteritis and enteric fever. The Salmonella type III secretion system (T3SS), encoded within its pathogenicity islands (SPIs), is critical for bacterial virulence by directly delivering multiple effectors into eukaryotic host cells. Salmonella utilizes these effectors to facilitate its survival and replication within the host through modulating cytoskeletal dynamics, inflammatory responses, the biogenesis of Salmonella-containing vacuole (SCV), and host cell survival. Moreover, these effectors also interfere with immune responses via inhibiting innate immunity or antigen presentation. In this review, we summarize the current progress in the survival strategies employed by Salmonella and the molecular mechanisms underlying its interactions with the host. Understanding the interplay between Salmonella and host can enhance our knowledge of the bacterium's pathogenic processes and provide new insights into how it manipulates host cellular physiological activities to ensure its survival.
Collapse
Affiliation(s)
- Hongyu Zhao
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Medical Research Institute, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430071, Hubei, China
| | - Xinyue Zhang
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Medical Research Institute, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430071, Hubei, China
| | - Ningning Zhang
- Yale Stem Cell Center, New Haven, CT, 06520, USA
- Department of Genetics, Yale University School of Medicine, New Haven, CT, 06510, USA
- Yale Cooperative Center of Excellence in Hematology, New Haven, CT, 12208, USA
| | - Li Zhu
- Department of Internal Medicine, Section of Infectious Diseases, Yale University School of Medicine, New Haven, CT, 06511, USA
| | - Huan Lian
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Medical Research Institute, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430071, Hubei, China
| |
Collapse
|
2
|
Wen X, Li D, Wang H, Zhang D, Song J, Zhou Z, Huang W, Xia X, Hu X, Liu W, Gonzales J, Via LE, Zhang L, Wang D. IQGAP1 domesticates macrophages to favor mycobacteria survival via modulating NF-κB signal and augmenting VEGF secretion. Int Immunopharmacol 2024; 138:112549. [PMID: 38944950 DOI: 10.1016/j.intimp.2024.112549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 06/18/2024] [Accepted: 06/20/2024] [Indexed: 07/02/2024]
Abstract
Tuberculosis, caused by Mycobacterium tuberculosis (Mtb), still ranks among the leading causes of annual human death by infectious disease. Mtb has developed several strategies to survive for years at a time within the host despite the presence of a robust immune response, including manipulating the progression of the inflammatory response and forming granulomatous lesions. Here we demonstrate that IQGAP1, a highly conserved scaffolding protein, compartmentalizes and coordinates multiple signaling pathways in macrophages infected with Mycobacterium marinum (Mm or M.marinum), the closest relative of Mtb. Upregulated IQGAP1 ultimately suppresses TNF-α production by repressing the MKK3 signal and reducing NF-κBp65 translocation, deactivating the p38MAPK pathway. Accordingly, IQGAP1 silencing and overexpression significantly alter p38MAPK activity by modulating the production of phosphorylated MKK3 during mycobacterial infection. Pharmacological inhibition of IQGAP1-associated microtubule assembly not only alleviates tissue damage caused by M.marinum infection but also significantly decreases the production of VEGF-a critical player for granuloma-associated angiogenesis during pathogenic mycobacterial infection. Similarly, IQGAP1 silencing in Mm-infected macrophages diminishes VEGF production, while IQGAP1 overexpression upregulates VEGF. Our data indicate that mycobacteria induce IQGAP1 to hijack NF-κBp65 activation, preventing the expression of proinflammatory cytokines as well as promoting VEGF production during infection and granuloma formation. Thus, therapies targeting host IQGAP1 may be a promising strategy for treating tuberculosis, particularly in drug-resistant diseases.
Collapse
Affiliation(s)
- Xin Wen
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Sciences, China Three Gorges University, Yichang 443002, PR China; Yichang Key Laboratory of Institute of Infection and Inflammation, College of Basic Medical Sciences, China Three Gorges University, Yichang 443002, PR China
| | - Dan Li
- Department of Tuberculosis, The Third People's Hospital of Yichang, Yichang 443003, PR China
| | - Hankun Wang
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Sciences, China Three Gorges University, Yichang 443002, PR China; Yichang Key Laboratory of Institute of Infection and Inflammation, College of Basic Medical Sciences, China Three Gorges University, Yichang 443002, PR China
| | - Ding Zhang
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Sciences, China Three Gorges University, Yichang 443002, PR China; Yichang Key Laboratory of Institute of Infection and Inflammation, College of Basic Medical Sciences, China Three Gorges University, Yichang 443002, PR China
| | - Jingrui Song
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Sciences, China Three Gorges University, Yichang 443002, PR China; Yichang Key Laboratory of Institute of Infection and Inflammation, College of Basic Medical Sciences, China Three Gorges University, Yichang 443002, PR China
| | - Ziwei Zhou
- State Key Laboratory of Genetic Engineering, Institute of Genetics, MOE Engineering Research Center of Gene Technology, School of Life Science, Fudan University, Shanghai 200433, PR China
| | - Weifeng Huang
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Sciences, China Three Gorges University, Yichang 443002, PR China; Yichang Key Laboratory of Institute of Infection and Inflammation, College of Basic Medical Sciences, China Three Gorges University, Yichang 443002, PR China
| | - Xuan Xia
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Sciences, China Three Gorges University, Yichang 443002, PR China; Yichang Key Laboratory of Institute of Infection and Inflammation, College of Basic Medical Sciences, China Three Gorges University, Yichang 443002, PR China
| | - Xiaohong Hu
- Department of Tuberculosis, The Third People's Hospital of Yichang, Yichang 443003, PR China
| | - Wei Liu
- The First College of Clinical Medical Science, China Three Gorges University, Yichang, PR China; Institute of Digestive Disease, China Three Gorges University, Yichang, PR China; Department of Gastroenterology, Yichang Central People's Hospital, Yichang, PR China
| | - Jacqueline Gonzales
- Tuberculosis Research Section, Laboratory of Clinical Infectious Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda 20982, MD, USA
| | - Laura E Via
- Tuberculosis Research Section, Laboratory of Clinical Infectious Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda 20982, MD, USA
| | - Lu Zhang
- State Key Laboratory of Genetic Engineering, Institute of Genetics, MOE Engineering Research Center of Gene Technology, School of Life Science, Fudan University, Shanghai 200433, PR China.
| | - Decheng Wang
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Sciences, China Three Gorges University, Yichang 443002, PR China; Yichang Key Laboratory of Institute of Infection and Inflammation, College of Basic Medical Sciences, China Three Gorges University, Yichang 443002, PR China.
| |
Collapse
|
3
|
Bandyopadhyay S, Zhang X, Ascura A, Edelblum KL, Bonder EM, Gao N. Salmonella engages CDC42 effector protein 1 for intracellular invasion. J Cell Physiol 2024; 239:36-50. [PMID: 37877586 PMCID: PMC11730249 DOI: 10.1002/jcp.31142] [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: 03/01/2023] [Revised: 08/25/2023] [Accepted: 10/03/2023] [Indexed: 10/26/2023]
Abstract
Human enterocytes are primary targets of infection by invasive bacterium Salmonella Typhimurium, and studies using nonintestinal epithelial cells established that S. Typhimurium activates Rho family GTPases, primarily CDC42, to modulate the actin cytoskeletal network for invasion. The host intracellular protein network that engages CDC42 and influences the pathogen's invasive capacity are relatively unclear. Here, proteomic analyses of canonical and variant CDC42 interactomes identified a poorly characterized CDC42 interacting protein, CDC42EP1, whose intracellular localization is rapidly redistributed and aggregated around the invading bacteria. CDC42EP1 associates with SEPTIN-7 and Villin, and its relocalization and bacterial engagement depend on host CDC42 and S. Typhimurium's capability of activating CDC42. Unlike CDC42, CDC42EP1 is not required for S. Typhimurium's initial cellular entry but is found to associate with Salmonella-containing vacuoles after long-term infections, indicating a contribution to the pathogen's intracellular growth and replication. These results uncover a new host regulator of enteric Salmonella infections, which may be targeted to restrict bacterial load at the primary site of infection to prevent systemic spread.
Collapse
Affiliation(s)
| | - Xiao Zhang
- Department of Biological Sciences, Rutgers University, Newark, New Jersey, USA
| | - Andrea Ascura
- Department of Biological Sciences, Rutgers University, Newark, New Jersey, USA
| | - Karen L. Edelblum
- Department of Pathology, Immunology, and Laboratory Medicine, Center for Immunity and Inflammation, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Edward M. Bonder
- Department of Biological Sciences, Rutgers University, Newark, New Jersey, USA
| | - Nan Gao
- Department of Biological Sciences, Rutgers University, Newark, New Jersey, USA
| |
Collapse
|
4
|
Chatterjee R, Chowdhury AR, Mukherjee D, Chakravortty D. From Eberthella typhi to Salmonella Typhi: The Fascinating Journey of the Virulence and Pathogenicity of Salmonella Typhi. ACS OMEGA 2023; 8:25674-25697. [PMID: 37521659 PMCID: PMC10373206 DOI: 10.1021/acsomega.3c02386] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Accepted: 06/30/2023] [Indexed: 08/01/2023]
Abstract
Salmonella Typhi (S. Typhi), the invasive typhoidal serovar of Salmonella enterica that causes typhoid fever in humans, is a severe threat to global health. It is one of the major causes of high morbidity and mortality in developing countries. According to recent WHO estimates, approximately 11-21 million typhoid fever illnesses occur annually worldwide, accounting for 0.12-0.16 million deaths. Salmonella infection can spread to healthy individuals by the consumption of contaminated food and water. Typhoid fever in humans sometimes is accompanied by several other critical extraintestinal complications related to the central nervous system, cardiovascular system, pulmonary system, and hepatobiliary system. Salmonella Pathogenicity Island-1 and Salmonella Pathogenicity Island-2 are the two genomic segments containing genes encoding virulent factors that regulate its invasion and systemic pathogenesis. This Review aims to shed light on a comparative analysis of the virulence and pathogenesis of the typhoidal and nontyphoidal serovars of S. enterica.
Collapse
Affiliation(s)
- Ritika Chatterjee
- Department
of Microbiology and Cell Biology, Division of Biological Sciences, Indian Institute of Science, Bangalore, Karnataka 560012, India
| | - Atish Roy Chowdhury
- Department
of Microbiology and Cell Biology, Division of Biological Sciences, Indian Institute of Science, Bangalore, Karnataka 560012, India
| | - Debapriya Mukherjee
- Department
of Microbiology and Cell Biology, Division of Biological Sciences, Indian Institute of Science, Bangalore, Karnataka 560012, India
| | - Dipshikha Chakravortty
- Department
of Microbiology and Cell Biology, Division of Biological Sciences, Indian Institute of Science, Bangalore, Karnataka 560012, India
- Centre
for Biosystems Science and Engineering, Indian Institute of Science, Bangalore, Karnataka 560012, India
| |
Collapse
|
5
|
Sayedyahossein S, Smith J, Barnaeva E, Li Z, Choe J, Ronzetti M, Dextras C, Hu X, Marugan J, Southall N, Baljinnyam B, Thines L, Tran AD, Ferrer M, Sacks DB. Discovery of small molecule inhibitors that effectively disrupt IQGAP1-Cdc42 interaction in breast cancer cells. Sci Rep 2022; 12:17372. [PMID: 36253497 PMCID: PMC9576799 DOI: 10.1038/s41598-022-21342-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 09/26/2022] [Indexed: 01/10/2023] Open
Abstract
The small GTPase Cdc42 is an integral component of the cytoskeleton, and its dysregulation leads to pathophysiological conditions, such as cancer. Binding of Cdc42 to the scaffold protein IQGAP1 stabilizes Cdc42 in its active form. The interaction between Cdc42 and IQGAP1 enhances migration and invasion of cancer cells. Disrupting this association could impair neoplastic progression and metastasis; however, no effective means to achieve this has been described. Here, we screened 78,500 compounds using a homogeneous time resolved fluorescence-based assay to identify small molecules that disrupt the binding of Cdc42 to IQGAP1. From the combined results of the validation assay and counter-screens, we selected 44 potent compounds for cell-based experiments. Immunoprecipitation and cell viability analysis rendered four lead compounds, namely NCGC00131308, NCGC00098561, MLS000332963 and NCGC00138812, three of which inhibited proliferation and migration of breast carcinoma cells. Microscale thermophoresis revealed that two compounds bind directly to Cdc42. One compound reduced the amount of active Cdc42 in cells and effectively impaired filopodia formation. Docking analysis provided plausible models of the compounds binding to the hydrophobic pocket adjacent to the GTP binding site of Cdc42. In conclusion, we identified small molecules that inhibit binding between Cdc42 and IQGAP1, which could potentially yield chemotherapeutic agents.
Collapse
Affiliation(s)
- Samar Sayedyahossein
- grid.94365.3d0000 0001 2297 5165Department of Laboratory Medicine, National Institutes of Health, Bethesda, MD 20892 USA ,grid.39381.300000 0004 1936 8884Present Address: Department of Physiology and Pharmacology, University of Western Ontario, London, ON Canada
| | - Jessica Smith
- grid.94365.3d0000 0001 2297 5165Department of Laboratory Medicine, National Institutes of Health, Bethesda, MD 20892 USA ,grid.94365.3d0000 0001 2297 5165Present Address: Center for Scientific Review, National Institutes of Health, Bethesda, MD USA
| | - Elena Barnaeva
- grid.94365.3d0000 0001 2297 5165National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD USA
| | - Zhigang Li
- grid.94365.3d0000 0001 2297 5165Department of Laboratory Medicine, National Institutes of Health, Bethesda, MD 20892 USA
| | - Jun Choe
- grid.94365.3d0000 0001 2297 5165National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD USA
| | - Michael Ronzetti
- grid.94365.3d0000 0001 2297 5165National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD USA
| | - Christopher Dextras
- grid.94365.3d0000 0001 2297 5165National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD USA
| | - Xin Hu
- grid.94365.3d0000 0001 2297 5165National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD USA
| | - Juan Marugan
- grid.94365.3d0000 0001 2297 5165National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD USA
| | - Noel Southall
- grid.94365.3d0000 0001 2297 5165National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD USA
| | - Bolormaa Baljinnyam
- grid.94365.3d0000 0001 2297 5165National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD USA
| | - Louise Thines
- grid.94365.3d0000 0001 2297 5165Department of Laboratory Medicine, National Institutes of Health, Bethesda, MD 20892 USA
| | - Andy D. Tran
- grid.48336.3a0000 0004 1936 8075Confocal Microscopy Core Facility, Laboratory of Cancer Biology and Genetics, NCI, National Institutes of Health, Rockville, MD USA
| | - Marc Ferrer
- grid.94365.3d0000 0001 2297 5165National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD USA
| | - David B. Sacks
- grid.94365.3d0000 0001 2297 5165Department of Laboratory Medicine, National Institutes of Health, Bethesda, MD 20892 USA
| |
Collapse
|
6
|
Zhang W, Zhou H, Jiang Y, He J, Yao Y, Wang J, Liu X, Leptihn S, Hua X, Yu Y. Acinetobacter baumannii Outer Membrane Protein A Induces Pulmonary Epithelial Barrier Dysfunction and Bacterial Translocation Through The TLR2/IQGAP1 Axis. Front Immunol 2022; 13:927955. [PMID: 35844614 PMCID: PMC9280087 DOI: 10.3389/fimmu.2022.927955] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 06/02/2022] [Indexed: 01/15/2023] Open
Abstract
Pulmonary epithelial barrier dysfunction is a critical pathophysiological process in pneumonia and associated invasive infections, such as those caused by Acinetobacter baumannii. However, the mechanisms underlying A. baumannii-induced pulmonary epithelial barrier dysfunction and bacterial translocation remain unclear. In this study, lungs of mice and A549 human epithelial cell monolayers were challenged with the A. baumannii wild-type strain and an outer membrane protein A (ompA) deletion strain. In addition, epithelial cells in culture were treated with purified OmpA protein or transfected with a eukaryotic expression vector encoding ompA (pCMV-ompA). Bacterial translocation across cell monolayers and intrapulmonary burden were measured, barrier function was evaluated in vivo and in vitro; cell migration ability was determined. The specific inhibitors C29 and JSH-23 were used to suppress the activity of Toll-like receptor 2 (TLR2) and of NF-κB, respectively. IQ-GTPase-activating protein 1 (IQGAP1) small interfering RNA was used to knock down endogenous IQGAP1 expression. In this work, we show that OmpA from A. baumannii increased the production of pro-inflammatory cytokines, remodeled the cytoskeleton, and internalized intercellular adherens junctions (AJs); these changes eventually induced pulmonary epithelial barrier dysfunction to promote bacterial translocation. IQGAP1-targeting small interfering RNA and chemical inhibition of TLR2 or NF-κB prevented high permeability of the pulmonary epithelial barrier. TLR2/NF-κB signaling was involved in OmpA-induced inflammation, IQGAP1-mediated OmpA-induced opening of the pulmonary epithelial barrier via cytoskeleton dynamic remodeling, and cellular redistribution of the major AJ protein, E-cadherin. These observations indicate that A. baumannii uses OmpA to overcome epithelial defences and cross the pulmonary epithelial barrier.
Collapse
Affiliation(s)
- Wang Zhang
- Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Regional Medical Center for National Institute of Respiratory Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, China
| | - Hua Zhou
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yan Jiang
- Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Regional Medical Center for National Institute of Respiratory Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, China
| | - Jintao He
- Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Regional Medical Center for National Institute of Respiratory Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, China
| | - Yue Yao
- Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Regional Medical Center for National Institute of Respiratory Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, China
| | - Jianfeng Wang
- Department of Respiratory and Critical Care Medicine, Zhejiang Provincial Hospital of Chinese Medicine, Hangzhou, China
| | - Xiaochen Liu
- Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Regional Medical Center for National Institute of Respiratory Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, China
| | - Sebastian Leptihn
- Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang University-University of Edinburgh Institute, Zhejiang University, Haining, China
- University of Edinburgh Medical School, Biomedical Sciences, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, United Kingdom
| | - Xiaoting Hua
- Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Regional Medical Center for National Institute of Respiratory Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, China
- *Correspondence: Yunsong Yu, ; Xiaoting Hua,
| | - Yunsong Yu
- Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Regional Medical Center for National Institute of Respiratory Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, China
- *Correspondence: Yunsong Yu, ; Xiaoting Hua,
| |
Collapse
|
7
|
Abstract
Salmonella is an intracellular pathogen of a substantial global health concern. In order to identify key players involved in Salmonella infection, we performed a global host phosphoproteome analysis subsequent to bacterial infection. Thereby, we identified the kinase SIK2 as a central component of the host defense machinery upon Salmonella infection. SIK2 depletion favors the escape of bacteria from the Salmonella-containing vacuole (SCV) and impairs Xenophagy, resulting in a hyperproliferative phenotype. Mechanistically, SIK2 associates with actin filaments under basal conditions; however, during bacterial infection, SIK2 is recruited to the SCV together with the elements of the actin polymerization machinery (Arp2/3 complex and Formins). Notably, SIK2 depletion results in a severe pathological cellular actin nucleation and polymerization defect upon Salmonella infection. We propose that SIK2 controls the formation of a protective SCV actin shield shortly after invasion and orchestrates the actin cytoskeleton architecture in its entirety to control an acute Salmonella infection after bacterial invasion.
Collapse
|
8
|
Negretti NM, Gourley CR, Talukdar PK, Clair G, Klappenbach CM, Lauritsen CJ, Adkins JN, Konkel ME. The Campylobacter jejuni CiaD effector co-opts the host cell protein IQGAP1 to promote cell entry. Nat Commun 2021; 12:1339. [PMID: 33637714 PMCID: PMC7910587 DOI: 10.1038/s41467-021-21579-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 01/27/2021] [Indexed: 02/07/2023] Open
Abstract
Campylobacter jejuni is a foodborne pathogen that binds to and invades the epithelial cells lining the human intestinal tract. Maximal invasion of host cells by C. jejuni requires cell binding as well as delivery of the Cia proteins (Campylobacter invasion antigens) to the host cell cytosol via the flagellum. Here, we show that CiaD binds to the host cell protein IQGAP1 (a Ras GTPase-activating-like protein), thus displacing RacGAP1 from the IQGAP1 complex. This, in turn, leads to the unconstrained activity of the small GTPase Rac1, which is known to have roles in actin reorganization and internalization of C. jejuni. Our results represent the identification of a host cell protein targeted by a flagellar secreted effector protein and demonstrate that C. jejuni-stimulated Rac signaling is dependent on IQGAP1.
Collapse
Affiliation(s)
- Nicholas M Negretti
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| | - Christopher R Gourley
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| | - Prabhat K Talukdar
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| | - Geremy Clair
- Integrative Omics, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Courtney M Klappenbach
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| | - Cody J Lauritsen
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| | - Joshua N Adkins
- Integrative Omics, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Michael E Konkel
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA, USA.
| |
Collapse
|
9
|
den Hartog G, Butcher LD, Ablack AL, Pace LA, Ablack JNG, Xiong R, Das S, Stappenbeck TS, Eckmann L, Ernst PB, Crowe SE. Apurinic/Apyrimidinic Endonuclease 1 Restricts the Internalization of Bacteria Into Human Intestinal Epithelial Cells Through the Inhibition of Rac1. Front Immunol 2021; 11:553994. [PMID: 33603730 PMCID: PMC7884313 DOI: 10.3389/fimmu.2020.553994] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 12/14/2020] [Indexed: 12/12/2022] Open
Abstract
Pathogenic intestinal bacteria lead to significant disease in humans. Here we investigated the role of the multifunctional protein, Apurinic/apyrimidinic endonuclease 1 (APE1), in regulating the internalization of bacteria into the intestinal epithelium. Intestinal tumor-cell lines and primary human epithelial cells were infected with Salmonella enterica serovar Typhimurium or adherent-invasive Escherichia coli. The effects of APE1 inhibition on bacterial internalization, the regulation of Rho GTPase Rac1 as well as the epithelial cell barrier function were assessed. Increased numbers of bacteria were present in APE1-deficient colonic tumor cell lines and primary epithelial cells. Activation of Rac1 was augmented following infection but negatively regulated by APE1. Pharmacological inhibition of Rac1 reversed the increase in intracellular bacteria in APE1-deficient cells whereas overexpression of constitutively active Rac1 augmented the numbers in APE1-competent cells. Enhanced numbers of intracellular bacteria resulted in the loss of barrier function and a delay in its recovery. Our data demonstrate that APE1 inhibits the internalization of invasive bacteria into human intestinal epithelial cells through its ability to negatively regulate Rac1. This activity also protects epithelial cell barrier function.
Collapse
Affiliation(s)
- Gerco den Hartog
- Department of Medicine, Division of Gastroenterology, University of California San Diego, La Jolla, CA, United States
| | - Lindsay D Butcher
- Department of Medicine, Division of Gastroenterology, University of California San Diego, La Jolla, CA, United States
| | - Amber L Ablack
- Department of Medicine, Division of Gastroenterology, University of California San Diego, La Jolla, CA, United States
| | - Laura A Pace
- Department of Medicine, Division of Gastroenterology, University of California San Diego, La Jolla, CA, United States
| | - Jailal N G Ablack
- Department of Medicine, Division of Rheumatology, University of California San Diego, La Jolla, CA, United States
| | - Richard Xiong
- Department of Medicine, Division of Gastroenterology, University of California San Diego, La Jolla, CA, United States
| | - Soumita Das
- Division of Comparative Pathology and Medicine, Department of Pathology, University of California San Diego, La Jolla, CA, United States
| | | | - Lars Eckmann
- Department of Medicine, Division of Gastroenterology, University of California San Diego, La Jolla, CA, United States
| | - Peter B Ernst
- Division of Comparative Pathology and Medicine, Department of Pathology, University of California San Diego, La Jolla, CA, United States.,Center for Mucosal Immunology, Allergy and Vaccine Development, Department of Pathology, University of California San Diego, La Jolla, CA, United States.,Department of Immunology, Chiba University, Chiba, Japan
| | - Sheila E Crowe
- Department of Medicine, Division of Gastroenterology, University of California San Diego, La Jolla, CA, United States.,Division of ImmunoBiology, Washington University, St. Louis, MO, United States
| |
Collapse
|
10
|
Lim C, Berk JM, Blaise A, Bircher J, Koleske AJ, Hochstrasser M, Xiong Y. Crystal structure of a guanine nucleotide exchange factor encoded by the scrub typhus pathogen Orientia tsutsugamushi. Proc Natl Acad Sci U S A 2020; 117:30380-30390. [PMID: 33184172 PMCID: PMC7720168 DOI: 10.1073/pnas.2018163117] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Rho family GTPases regulate an array of cellular processes and are often modulated by pathogens to promote infection. Here, we identify a cryptic guanine nucleotide exchange factor (GEF) domain in the OtDUB protein encoded by the pathogenic bacterium Orientia tsutsugamushi A proteomics-based OtDUB interaction screen identified numerous potential host interactors, including the Rho GTPases Rac1 and Cdc42. We discovered a domain in OtDUB with Rac1/Cdc42 GEF activity (OtDUBGEF), with higher activity toward Rac1 in vitro. While this GEF bears no obvious sequence similarity to known GEFs, crystal structures of OtDUBGEF alone (3.0 Å) and complexed with Rac1 (1.7 Å) reveal striking convergent evolution, with a unique topology, on a V-shaped bacterial GEF fold shared with other bacterial GEF domains. Structure-guided mutational analyses identified residues critical for activity and a mechanism for nucleotide displacement. Ectopic expression of OtDUB activates Rac1 preferentially in cells, and expression of the OtDUBGEF alone alters cell morphology. Cumulatively, this work reveals a bacterial GEF within the multifunctional OtDUB that co-opts host Rac1 signaling to induce changes in cytoskeletal structure.
Collapse
Affiliation(s)
- Christopher Lim
- Department of Molecular Biophysics & Biochemistry, Yale University, New Haven, CT 06520
| | - Jason M Berk
- Department of Molecular Biophysics & Biochemistry, Yale University, New Haven, CT 06520
| | - Alyssa Blaise
- Department of Molecular Biophysics & Biochemistry, Yale University, New Haven, CT 06520
| | - Josie Bircher
- Department of Molecular Biophysics & Biochemistry, Yale University, New Haven, CT 06520
| | - Anthony J Koleske
- Department of Molecular Biophysics & Biochemistry, Yale University, New Haven, CT 06520
| | - Mark Hochstrasser
- Department of Molecular Biophysics & Biochemistry, Yale University, New Haven, CT 06520
| | - Yong Xiong
- Department of Molecular Biophysics & Biochemistry, Yale University, New Haven, CT 06520
| |
Collapse
|
11
|
Shen EP, Chen MR, Chen WL, Chu HS, Chen KL, Hu FR. Knockdown of IQGAP-1 Enhances Tight Junctions and Prevents P. aeruginosa Invasion of Human Corneal Epithelial Cells. Ocul Immunol Inflamm 2020; 28:876-883. [PMID: 31621455 DOI: 10.1080/09273948.2019.1642494] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
PURPOSE To determine the role of IQ-domain GTPase-activating protein1 (IQGAP-1) in tight junctions of human corneal epithelial cells (HCECs) and its effect against P. aeruginosa (PAK) invasion. MATERIAL AND METHODS Primary human corneal epithelial cells (HCECs), immortalized HCECs, and IQGAP-1 RNA knockdown HCECs (siHCECs) were used. Confocal microscopy, transepithelial electrical resistance (TER), trypan blue exclusion assay and gentamicin invasion assay were done. RESULTS In primary and immortalized HCECs, IQGAP-1 co-localized with zonular occludin-1 (ZO-1) and actin. Enhanced actin and ZO-1 aggregation were seen in siHCECs. IQGAP-1 knockdown significantly increased TER of immortalized HCECs (P < .0001). Cell viability after PAK infection increased for siHCECs for up to 4 h after infection. PAK intracellular invasion was significantly lowered by 50% in siHCECs at 1 h post-infection. CONCLUSION IQGAP-1 knockdown increased the strength and integrity of tight junctions and may provide an early protective effect against P. aeruginosa invasion.
Collapse
Affiliation(s)
- Elizabeth P Shen
- Department of Ophthalmology, Taipei Tzu Chi Hospital, The Buddhist Tzu Chi Medical Foundation , Taipei, Taiwan.,School of Medicine, Tzu Chi University , Hua-Liang, Taiwan.,Department of Ophthalmology, National Taiwan University Hospital, College of Medicine, National Taiwan University , Taipei, Taiwan
| | - Mei-Ru Chen
- Graduate Institute and Department of Microbiology, College of Medicine, National Taiwan University , Taipei, Taiwan
| | - Wei-Li Chen
- Department of Ophthalmology, National Taiwan University Hospital, College of Medicine, National Taiwan University , Taipei, Taiwan
| | - Hsiao-Sang Chu
- Department of Ophthalmology, National Taiwan University Hospital, College of Medicine, National Taiwan University , Taipei, Taiwan
| | - Kai-Li Chen
- Department of Ophthalmology, National Taiwan University Hospital, College of Medicine, National Taiwan University , Taipei, Taiwan
| | - Fung-Rong Hu
- Department of Ophthalmology, National Taiwan University Hospital, College of Medicine, National Taiwan University , Taipei, Taiwan
| |
Collapse
|
12
|
Mo CF, Li J, Yang SX, Guo HJ, Liu Y, Luo XY, Wang YT, Li MH, Li JY, Zou Q. IQGAP1 promotes anoikis resistance and metastasis through Rac1-dependent ROS accumulation and activation of Src/FAK signalling in hepatocellular carcinoma. Br J Cancer 2020; 123:1154-1163. [PMID: 32632148 PMCID: PMC7525663 DOI: 10.1038/s41416-020-0970-z] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 04/01/2020] [Accepted: 06/18/2020] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Hepatitis B virus (HBV) has a crucial role in the progression of hepatocellular carcinoma (HCC). Tumour cells must develop anoikis resistance in order to survive before metastasis. This study aimed to investigate the mechanism of IQGAP1 in HBV-mediated anoikis evasion and metastasis in HCC cells. METHODS IQGAP1 expression was detected by immunohistochemistry, real-time PCR and immunoblot analysis. Lentiviral-mediated stable upregulation or knockdown of IGAQP1, immunoprecipitation, etc. were used in function and mechanism study. RESULTS IQGAP1 was markedly upregulated in HBV-positive compared with HBV-negative HCC cells and tissues. IQGAP1 was positively correlated to poor prognosis of HBV-associated HCC patients. IQGAP1 overexpression significantly enhanced the anchorage-independent growth and metastasis, whereas IQGAP1-deficient HCC cells are more sensitive to anoikis. Mechanistically, we found that HBV-induced ROS enhanced the association of IQGAP1 and Rac1 that activated Rac1, leading to phosphorylation of Src/FAK pathway. Antioxidants efficiently inhibited IQGAP1-mediated anoikis resistance and metastasis. CONCLUSIONS Our study indicated an important mechanism by which upregulated IQGAP1 by HBV promoted anoikis resistance, migration and invasion of HCC cells through Rac1-dependent ROS accumulation and activation of Src/FAK signalling, suggesting IQGAP1 as a prognostic indicator and a novel therapeutic target in HCC patients with HBV infection.
Collapse
Affiliation(s)
- Chun-Fen Mo
- Department of Immunology, School of Basic Medical Sciences, Chengdu Medical College, Chengdu, China.
| | - Jun Li
- Department of Gastroenterology, The first affiliated hospital of Chengdu medical college, Chengdu, China
| | - Shu-Xia Yang
- Department of Immunology, School of Basic Medical Sciences, Chengdu Medical College, Chengdu, China
| | - Hui-Jie Guo
- Department of Immunology, School of Basic Medical Sciences, Chengdu Medical College, Chengdu, China
| | - Yang Liu
- Department of Immunology, School of Basic Medical Sciences, Chengdu Medical College, Chengdu, China
| | - Xing-Yan Luo
- Department of Immunology, School of Basic Medical Sciences, Chengdu Medical College, Chengdu, China
| | - Yan-Tang Wang
- Department of Immunology, School of Basic Medical Sciences, Chengdu Medical College, Chengdu, China
| | - Min-Hui Li
- Department of Immunology, School of Basic Medical Sciences, Chengdu Medical College, Chengdu, China
| | - Jing-Yi Li
- Department of Urology, The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, China. .,School of Biological Sciences and Technology, Chengdu Medical College, Chengdu, China.
| | - Qiang Zou
- Department of Immunology, School of Basic Medical Sciences, Chengdu Medical College, Chengdu, China.
| |
Collapse
|
13
|
Pleiotropic Roles of Calmodulin in the Regulation of KRas and Rac1 GTPases: Functional Diversity in Health and Disease. Int J Mol Sci 2020; 21:ijms21103680. [PMID: 32456244 PMCID: PMC7279331 DOI: 10.3390/ijms21103680] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 05/18/2020] [Accepted: 05/21/2020] [Indexed: 12/21/2022] Open
Abstract
Calmodulin is a ubiquitous signalling protein that controls many biological processes due to its capacity to interact and/or regulate a large number of cellular proteins and pathways, mostly in a Ca2+-dependent manner. This complex interactome of calmodulin can have pleiotropic molecular consequences, which over the years has made it often difficult to clearly define the contribution of calmodulin in the signal output of specific pathways and overall biological response. Most relevant for this review, the ability of calmodulin to influence the spatiotemporal signalling of several small GTPases, in particular KRas and Rac1, can modulate fundamental biological outcomes such as proliferation and migration. First, direct interaction of calmodulin with these GTPases can alter their subcellular localization and activation state, induce post-translational modifications as well as their ability to interact with effectors. Second, through interaction with a set of calmodulin binding proteins (CaMBPs), calmodulin can control the capacity of several guanine nucleotide exchange factors (GEFs) to promote the switch of inactive KRas and Rac1 to an active conformation. Moreover, Rac1 is also an effector of KRas and both proteins are interconnected as highlighted by the requirement for Rac1 activation in KRas-driven tumourigenesis. In this review, we attempt to summarize the multiple layers how calmodulin can regulate KRas and Rac1 GTPases in a variety of cellular events, with biological consequences and potential for therapeutic opportunities in disease settings, such as cancer.
Collapse
|
14
|
Gorisse L, Li Z, Wagner CD, Worthylake DK, Zappacosta F, Hedman AC, Annan RS, Sacks DB. Ubiquitination of the scaffold protein IQGAP1 diminishes its interaction with and activation of the Rho GTPase CDC42. J Biol Chem 2020; 295:4822-4835. [PMID: 32094223 DOI: 10.1074/jbc.ra119.011491] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 02/19/2020] [Indexed: 12/12/2022] Open
Abstract
IQ motif-containing GTPase-activating protein 1 (IQGAP1) is a scaffold protein that interacts with numerous binding partners and thereby regulates fundamental biological processes. The functions of IQGAP1 are modulated by several mechanisms, including protein binding, self-association, subcellular localization, and phosphorylation. Proteome-wide screens have indicated that IQGAP1 is ubiquitinated, but the possible effects of this post-translational modification on its function are unknown. Here we characterized and evaluated the function of IQGAP1 ubiquitination. Using MS-based analysis in HEK293 cells, we identified six lysine residues (Lys-556, -1155, -1230, -1465, -1475, and -1528) as ubiquitination sites in IQGAP1. To elucidate the biological consequences of IQGAP1 ubiquitination, we converted each of these lysines to arginine and found that replacing two of these residues, Lys-1155 and Lys-1230, in the GAP-related domain of IQGAP1 (termed IQGAP1 GRD-2K) reduces its ubiquitination. Moreover, IQGAP1 GRD-2K bound a significantly greater proportion of the two Rho GTPases cell division cycle 42 (CDC42) and Rac family small GTPase 1 (RAC1) than did WT IQGAP1. Consistent with this observation, reconstitution of IQGAP1-null cells with IQGAP1 GRD-2K significantly increased the amount of active CDC42 and enhanced cell migration significantly more than WT IQGAP1. Our results reveal that ubiquitination of the CDC42 regulator IQGAP1 alters its ability to bind to and activate this GTPase, leading to physiological effects. Collectively, these findings expand our view of the role of ubiquitination in cell signaling and provide additional insight into CDC42 regulation.
Collapse
Affiliation(s)
- Laëtitia Gorisse
- Department of Laboratory Medicine, National Institutes of Health, Bethesda, Maryland 20892
| | - Zhigang Li
- Department of Laboratory Medicine, National Institutes of Health, Bethesda, Maryland 20892
| | - Craig D Wagner
- Discovery Analytical, GlaxoSmithKline, Collegeville, Pennsylvania 19426
| | - David K Worthylake
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences, New Orleans, Louisiana 70112
| | | | - Andrew C Hedman
- Department of Laboratory Medicine, National Institutes of Health, Bethesda, Maryland 20892
| | - Roland S Annan
- Discovery Analytical, GlaxoSmithKline, Collegeville, Pennsylvania 19426
| | - David B Sacks
- Department of Laboratory Medicine, National Institutes of Health, Bethesda, Maryland 20892
| |
Collapse
|
15
|
Hume PJ, Singh V, Davidson AC, Koronakis V. Swiss Army Pathogen: The Salmonella Entry Toolkit. Front Cell Infect Microbiol 2017; 7:348. [PMID: 28848711 PMCID: PMC5552672 DOI: 10.3389/fcimb.2017.00348] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 07/21/2017] [Indexed: 02/04/2023] Open
Abstract
Salmonella causes disease in humans and animals ranging from mild self-limiting gastroenteritis to potentially life-threatening typhoid fever. Salmonellosis remains a considerable cause of morbidity and mortality globally, and hence imposes a huge socio-economic burden worldwide. A key property of all pathogenic Salmonella strains is the ability to invade non-phagocytic host cells. The major determinant of this invasiveness is a Type 3 Secretion System (T3SS), a molecular syringe that injects virulence effector proteins directly into target host cells. These effectors cooperatively manipulate multiple host cell signaling pathways to drive pathogen internalization. Salmonella does not only rely on these injected effectors, but also uses several other T3SS-independent mechanisms to gain entry into host cells. This review summarizes our current understanding of the methods used by Salmonella for cell invasion, with a focus on the host signaling networks that must be coordinately exploited for the pathogen to achieve its goal.
Collapse
Affiliation(s)
- Peter J Hume
- Department of Pathology, University of CambridgeCambridge, United Kingdom
| | - Vikash Singh
- Department of Pathology, University of CambridgeCambridge, United Kingdom
| | - Anthony C Davidson
- Department of Pathology, University of CambridgeCambridge, United Kingdom
| | - Vassilis Koronakis
- Department of Pathology, University of CambridgeCambridge, United Kingdom
| |
Collapse
|
16
|
Hochstrasser R, Hilbi H. Intra-Species and Inter-Kingdom Signaling of Legionella pneumophila. Front Microbiol 2017; 8:79. [PMID: 28217110 PMCID: PMC5289986 DOI: 10.3389/fmicb.2017.00079] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 01/11/2017] [Indexed: 12/24/2022] Open
Abstract
The ubiquitous Gram-negative bacterium Legionella pneumophila parasitizes environ mental amoebae and, upon inhalation, replicates in alveolar macrophages, thus causing a life-threatening pneumonia called “Legionnaires’ disease.” The opportunistic pathogen employs a bi-phasic life cycle, alternating between a replicative, non-virulent phase and a stationary, transmissive/virulent phase. L. pneumophila employs the Lqs (Legionella quorum sensing) system as a major regulator of the growth phase switch. The Lqs system comprises the autoinducer synthase LqsA, the homologous sensor kinases LqsS and LqsT, as well as a prototypic response regulator termed LqsR. These components produce, detect, and respond to the α-hydroxyketone signaling molecule LAI-1 (Legionella autoinducer-1, 3-hydroxypentadecane-4-one). LAI-1-mediated signal transduction through the sensor kinases converges on LqsR, which dimerizes upon phosphorylation. The Lqs system regulates the bacterial growth phase switch, pathogen-host cell interactions, motility, natural competence, filament production, and expression of a chromosomal “fitness island.” Yet, LAI-1 not only mediates bacterial intra-species signaling, but also modulates the motility of eukaryotic cells through the small GTPase Cdc42 and thus promotes inter-kingdom signaling. Taken together, the low molecular weight compound LAI-1 produced by L. pneumophila and sensed by the bacteria as well as by eukaryotic cells plays a major role in pathogen-host cell interactions.
Collapse
Affiliation(s)
- Ramon Hochstrasser
- Department of Medicine, Institute of Medical Microbiology, University of Zürich Zürich, Switzerland
| | - Hubert Hilbi
- Department of Medicine, Institute of Medical Microbiology, University of Zürich Zürich, Switzerland
| |
Collapse
|
17
|
Chawla B, Hedman AC, Sayedyahossein S, Erdemir HH, Li Z, Sacks DB. Absence of IQGAP1 Protein Leads to Insulin Resistance. J Biol Chem 2017; 292:3273-3289. [PMID: 28082684 DOI: 10.1074/jbc.m116.752642] [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: 08/08/2016] [Revised: 01/10/2017] [Indexed: 11/06/2022] Open
Abstract
Insulin binds to the insulin receptor (IR) and induces tyrosine phosphorylation of the receptor and insulin receptor substrate-1 (IRS-1), leading to activation of the PKB/Akt and MAPK/ERK pathways. IQGAP1 is a scaffold protein that interacts with multiple binding partners and integrates diverse signaling cascades. Here we show that IQGAP1 associates with both IR and IRS-1 and influences insulin action. In vitro analysis with pure proteins revealed that the IQ region of IQGAP1 binds directly to the intracellular domain of IR. Similarly, the phosphotyrosine-binding domain of IRS-1 mediates a direct interaction with the C-terminal tail of IQGAP1. Consistent with these observations, both IR and IRS-1 co-immunoprecipitated with IQGAP1 from cells. Investigation of the functional effects of the interactions revealed that in the absence of IQGAP1, insulin-stimulated phosphorylation of Akt and ERK, as well as the association of phosphatidylinositol 3-kinase with IRS-1, were significantly decreased. Importantly, loss of IQGAP1 results in impaired insulin signaling and glucose homeostasis in vivo Collectively, these data reveal that IQGAP1 is a scaffold for IR and IRS-1 and implicate IQGAP1 as a participant in insulin signaling.
Collapse
Affiliation(s)
- Bhavna Chawla
- Department of Laboratory Medicine, National Institutes of Health, Bethesda, Maryland 20892
| | - Andrew C Hedman
- Department of Laboratory Medicine, National Institutes of Health, Bethesda, Maryland 20892
| | - Samar Sayedyahossein
- Department of Laboratory Medicine, National Institutes of Health, Bethesda, Maryland 20892
| | - Huseyin H Erdemir
- Department of Laboratory Medicine, National Institutes of Health, Bethesda, Maryland 20892
| | - Zhigang Li
- Department of Laboratory Medicine, National Institutes of Health, Bethesda, Maryland 20892
| | - David B Sacks
- Department of Laboratory Medicine, National Institutes of Health, Bethesda, Maryland 20892.
| |
Collapse
|
18
|
Nouri K, Fansa EK, Amin E, Dvorsky R, Gremer L, Willbold D, Schmitt L, Timson DJ, Ahmadian MR. IQGAP1 Interaction with RHO Family Proteins Revisited: KINETIC AND EQUILIBRIUM EVIDENCE FOR MULTIPLE DISTINCT BINDING SITES. J Biol Chem 2016; 291:26364-26376. [PMID: 27815503 PMCID: PMC5159498 DOI: 10.1074/jbc.m116.752121] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 10/27/2016] [Indexed: 11/16/2022] Open
Abstract
IQ motif-containing GTPase activating protein 1 (IQGAP1) plays a central role in the physical assembly of relevant signaling networks that are responsible for various cellular processes, including cell adhesion, polarity, and transmigration. The RHO family proteins CDC42 and RAC1 have been shown to mainly interact with the GAP-related domain (GRD) of IQGAP1. However, the role of its RASGAP C-terminal (RGCT) and C-terminal domains in the interactions with RHO proteins has remained obscure. Here, we demonstrate that IQGAP1 interactions with RHO proteins underlie a multiple-step binding mechanism: (i) a high affinity, GTP-dependent binding of RGCT to the switch regions of CDC42 or RAC1 and (ii) a very low affinity binding of GRD and a C terminus adjacent to the switch regions. These data were confirmed by phosphomimetic mutation of serine 1443 to glutamate within RGCT, which led to a significant reduction of IQGAP1 affinity for CDC42 and RAC1, clearly disclosing the critical role of RGCT for these interactions. Unlike CDC42, an extremely low affinity was determined for the RAC1-GRD interaction, suggesting that the molecular nature of IQGAP1 interaction with CDC42 partially differs from that of RAC1. Our study provides new insights into the interaction characteristics of IQGAP1 with RHO family proteins and highlights the complementary importance of kinetic and equilibrium analyses. We propose that the ability of IQGAP1 to interact with RHO proteins is based on a multiple-step binding process, which is a prerequisite for the dynamic functions of IQGAP1 as a scaffolding protein and a critical mechanism in temporal regulation and integration of IQGAP1-mediated cellular responses.
Collapse
Affiliation(s)
- Kazem Nouri
- From the Institute of Biochemistry and Molecular Biology II, Medical Faculty of the Heinrich-Heine University, 40225 Düsseldorf, Germany
| | - Eyad K Fansa
- From the Institute of Biochemistry and Molecular Biology II, Medical Faculty of the Heinrich-Heine University, 40225 Düsseldorf, Germany
| | - Ehsan Amin
- From the Institute of Biochemistry and Molecular Biology II, Medical Faculty of the Heinrich-Heine University, 40225 Düsseldorf, Germany
| | - Radovan Dvorsky
- From the Institute of Biochemistry and Molecular Biology II, Medical Faculty of the Heinrich-Heine University, 40225 Düsseldorf, Germany
| | - Lothar Gremer
- the Institute of Physical Biology, Heinrich-Heine University, 40225 Düsseldorf, Germany.,Forschungszentrum Jülich, ICS-6, 52428 Jülich, Germany
| | - Dieter Willbold
- the Institute of Physical Biology, Heinrich-Heine University, 40225 Düsseldorf, Germany.,Forschungszentrum Jülich, ICS-6, 52428 Jülich, Germany
| | - Lutz Schmitt
- the Institute of Biochemistry, Heinrich-Heine University, 40225 Düsseldorf, Germany, and
| | - David J Timson
- the School of Pharmacy and Biomolecular Sciences, University of Brighton, Huxley Building, Lewes Road, Brighton BN2 4GJ, United Kingdom
| | - Mohammad R Ahmadian
- From the Institute of Biochemistry and Molecular Biology II, Medical Faculty of the Heinrich-Heine University, 40225 Düsseldorf, Germany,
| |
Collapse
|
19
|
Bamidele AO, Kremer KN, Hirsova P, Clift IC, Gores GJ, Billadeau DD, Hedin KE. IQGAP1 promotes CXCR4 chemokine receptor function and trafficking via EEA-1+ endosomes. J Cell Biol 2016. [PMID: 26195666 PMCID: PMC4508899 DOI: 10.1083/jcb.201411045] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
IQGAP1 mediates CXCR4 cell surface expression and signaling by regulating EEA-1+ endosome interactions with microtubules during CXCR4 trafficking and recycling. IQ motif–containing GTPase-activating protein 1 (IQGAP1) is a cytoskeleton-interacting scaffold protein. CXCR4 is a chemokine receptor that binds stromal cell–derived factor-1 (SDF-1; also known as CXCL12). Both IQGAP1 and CXCR4 are overexpressed in cancer cell types, yet it was unclear whether these molecules functionally interact. Here, we show that depleting IQGAP1 in Jurkat T leukemic cells reduced CXCR4 expression, disrupted trafficking of endocytosed CXCR4 via EEA-1+ endosomes, and decreased efficiency of CXCR4 recycling. SDF-1–induced cell migration and activation of extracellular signal-regulated kinases 1 and 2 (ERK) MAPK were strongly inhibited, even when forced overexpression restored CXCR4 levels. Similar results were seen in KMBC and HEK293 cells. Exploring the mechanism, we found that SDF-1 treatment induced IQGAP1 binding to α-tubulin and localization to CXCR4-containing endosomes and that CXCR4-containing EEA-1+ endosomes were abnormally located distal from the microtubule (MT)-organizing center (MTOC) in IQGAP1-deficient cells. Thus, IQGAP1 critically mediates CXCR4 cell surface expression and signaling, evidently by regulating EEA-1+ endosome interactions with MTs during CXCR4 trafficking and recycling. IQGAP1 may similarly promote CXCR4 functions in other cancer cell types.
Collapse
Affiliation(s)
- Adebowale O Bamidele
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905 Department of Immunology, Mayo Clinic, Rochester, MN 55905
| | | | - Petra Hirsova
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905
| | - Ian C Clift
- Department of Immunology, Mayo Clinic, Rochester, MN 55905 Neurobiology of Disease Research Program, Mayo Clinic, Rochester, MN 55905
| | - Gregory J Gores
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905
| | - Daniel D Billadeau
- Department of Immunology, Mayo Clinic, Rochester, MN 55905 Division of Oncology Research, Mayo Clinic, Rochester, MN 55905
| | - Karen E Hedin
- Department of Immunology, Mayo Clinic, Rochester, MN 55905
| |
Collapse
|
20
|
Inter-kingdom Signaling by the Legionella Quorum Sensing Molecule LAI-1 Modulates Cell Migration through an IQGAP1-Cdc42-ARHGEF9-Dependent Pathway. PLoS Pathog 2015; 11:e1005307. [PMID: 26633832 PMCID: PMC4669118 DOI: 10.1371/journal.ppat.1005307] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 11/03/2015] [Indexed: 11/19/2022] Open
Abstract
Small molecule signaling promotes the communication between bacteria as well as between bacteria and eukaryotes. The opportunistic pathogenic bacterium Legionella pneumophila employs LAI-1 (3-hydroxypentadecane-4-one) for bacterial cell-cell communication. LAI-1 is produced and detected by the Lqs (Legionella quorum sensing) system, which regulates a variety of processes including natural competence for DNA uptake and pathogen-host cell interactions. In this study, we analyze the role of LAI-1 in inter-kingdom signaling. L. pneumophila lacking the autoinducer synthase LqsA no longer impeded the migration of infected cells, and the defect was complemented by plasmid-borne lqsA. Synthetic LAI-1 dose-dependently inhibited cell migration, without affecting bacterial uptake or cytotoxicity. The forward migration index but not the velocity of LAI-1-treated cells was reduced, and the cell cytoskeleton appeared destabilized. LAI-1-dependent inhibition of cell migration involved the scaffold protein IQGAP1, the small GTPase Cdc42 as well as the Cdc42-specific guanine nucleotide exchange factor ARHGEF9, but not other modulators of Cdc42, or RhoA, Rac1 or Ran GTPase. Upon treatment with LAI-1, Cdc42 was inactivated and IQGAP1 redistributed to the cell cortex regardless of whether Cdc42 was present or not. Furthermore, LAI-1 reversed the inhibition of cell migration by L. pneumophila, suggesting that the compound and the bacteria antagonistically target host signaling pathway(s). Collectively, the results indicate that the L. pneumophila quorum sensing compound LAI-1 modulates migration of eukaryotic cells through a signaling pathway involving IQGAP1, Cdc42 and ARHGEF9.
Collapse
|
21
|
Cao D, Su Z, Wang W, Wu H, Liu X, Akram S, Qin B, Zhou J, Zhuang X, Adams G, Jin C, Wang X, Liu L, Hill DL, Wang D, Ding X, Yao X. Signaling Scaffold Protein IQGAP1 Interacts with Microtubule Plus-end Tracking Protein SKAP and Links Dynamic Microtubule Plus-end to Steer Cell Migration. J Biol Chem 2015; 290:23766-80. [PMID: 26242911 DOI: 10.1074/jbc.m115.673517] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2015] [Indexed: 12/27/2022] Open
Abstract
Cell migration is orchestrated by dynamic interaction of microtubules with the plasma membrane cortex. However, the regulatory mechanisms underlying the cortical actin cytoskeleton and microtubule dynamics are less characterized. Our earlier study showed that small GTPase-activating proteins, IQGAPs, regulate polarized secretion in epithelial cells (1). Here, we show that IQGAP1 links dynamic microtubules to steer cell migration via interacting with the plus-end tracking protein, SKAP. Biochemical characterizations revealed that IQGAP1 and SKAP form a cognate complex and that their binding interfaces map to the WWIQ motif and the C-terminal of SKAP, respectively. The WWIQ peptide disrupts the biochemical interaction between IQGAP1 and SKAP in vitro, and perturbation of the IQGAP1-SKAP interaction in vivo using a membrane-permeable TAT-WWIQ peptide results in inhibition of directional cell migration elicited by EGF. Mechanistically, the N-terminal of SKAP binds to EB1, and its C terminus binds to IQGAP1 in migrating cells. Thus, we reason that a novel IQGAP1 complex orchestrates directional cell migration via coupling dynamic microtubule plus-ends to the cell cortex.
Collapse
Affiliation(s)
- Dan Cao
- From the BUCM-USTC Joint Program for Cellular Dynamics & Chemical Biology, University of Science & Technology of China, Hefei, China 230027, the Beijing University of Chinese Medicine, Beijing, China 100029, the Morehouse School of Medicine, Atlanta, Georgia 30310
| | - Zeqi Su
- From the BUCM-USTC Joint Program for Cellular Dynamics & Chemical Biology, University of Science & Technology of China, Hefei, China 230027, the Beijing University of Chinese Medicine, Beijing, China 100029
| | - Wenwen Wang
- From the BUCM-USTC Joint Program for Cellular Dynamics & Chemical Biology, University of Science & Technology of China, Hefei, China 230027, the Beijing University of Chinese Medicine, Beijing, China 100029, the Morehouse School of Medicine, Atlanta, Georgia 30310
| | - Huihui Wu
- From the BUCM-USTC Joint Program for Cellular Dynamics & Chemical Biology, University of Science & Technology of China, Hefei, China 230027, the Morehouse School of Medicine, Atlanta, Georgia 30310, the Airforce General Hospital, Beijing, China 100036, and
| | - Xing Liu
- From the BUCM-USTC Joint Program for Cellular Dynamics & Chemical Biology, University of Science & Technology of China, Hefei, China 230027, the Beijing University of Chinese Medicine, Beijing, China 100029, the Morehouse School of Medicine, Atlanta, Georgia 30310,
| | - Saima Akram
- From the BUCM-USTC Joint Program for Cellular Dynamics & Chemical Biology, University of Science & Technology of China, Hefei, China 230027
| | - Bo Qin
- From the BUCM-USTC Joint Program for Cellular Dynamics & Chemical Biology, University of Science & Technology of China, Hefei, China 230027, the Morehouse School of Medicine, Atlanta, Georgia 30310, the Airforce General Hospital, Beijing, China 100036, and
| | - Jiajia Zhou
- From the BUCM-USTC Joint Program for Cellular Dynamics & Chemical Biology, University of Science & Technology of China, Hefei, China 230027, the Airforce General Hospital, Beijing, China 100036, and
| | - Xiaoxuan Zhuang
- From the BUCM-USTC Joint Program for Cellular Dynamics & Chemical Biology, University of Science & Technology of China, Hefei, China 230027
| | - Gregory Adams
- From the BUCM-USTC Joint Program for Cellular Dynamics & Chemical Biology, University of Science & Technology of China, Hefei, China 230027, the Morehouse School of Medicine, Atlanta, Georgia 30310
| | - Changjiang Jin
- From the BUCM-USTC Joint Program for Cellular Dynamics & Chemical Biology, University of Science & Technology of China, Hefei, China 230027
| | - Xiwei Wang
- From the BUCM-USTC Joint Program for Cellular Dynamics & Chemical Biology, University of Science & Technology of China, Hefei, China 230027, the Morehouse School of Medicine, Atlanta, Georgia 30310
| | - Lifang Liu
- the Airforce General Hospital, Beijing, China 100036, and
| | - Donald L Hill
- the Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama 35294
| | - Dongmei Wang
- From the BUCM-USTC Joint Program for Cellular Dynamics & Chemical Biology, University of Science & Technology of China, Hefei, China 230027,
| | - Xia Ding
- From the BUCM-USTC Joint Program for Cellular Dynamics & Chemical Biology, University of Science & Technology of China, Hefei, China 230027, the Beijing University of Chinese Medicine, Beijing, China 100029,
| | - Xuebiao Yao
- From the BUCM-USTC Joint Program for Cellular Dynamics & Chemical Biology, University of Science & Technology of China, Hefei, China 230027
| |
Collapse
|
22
|
Budak G, Eren Ozsoy O, Aydin Son Y, Can T, Tuncbag N. Reconstruction of the temporal signaling network in Salmonella-infected human cells. Front Microbiol 2015; 6:730. [PMID: 26257716 PMCID: PMC4507143 DOI: 10.3389/fmicb.2015.00730] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 07/03/2015] [Indexed: 12/02/2022] Open
Abstract
Salmonella enterica is a bacterial pathogen that usually infects its host through food sources. Translocation of the pathogen proteins into the host cells leads to changes in the signaling mechanism either by activating or inhibiting the host proteins. Given that the bacterial infection modifies the response network of the host, a more coherent view of the underlying biological processes and the signaling networks can be obtained by using a network modeling approach based on the reverse engineering principles. In this work, we have used a published temporal phosphoproteomic dataset of Salmonella-infected human cells and reconstructed the temporal signaling network of the human host by integrating the interactome and the phosphoproteomic dataset. We have combined two well-established network modeling frameworks, the Prize-collecting Steiner Forest (PCSF) approach and the Integer Linear Programming (ILP) based edge inference approach. The resulting network conserves the information on temporality, direction of interactions, while revealing hidden entities in the signaling, such as the SNARE binding, mTOR signaling, immune response, cytoskeleton organization, and apoptosis pathways. Targets of the Salmonella effectors in the host cells such as CDC42, RHOA, 14-3-3δ, Syntaxin family, Oxysterol-binding proteins were included in the reconstructed signaling network although they were not present in the initial phosphoproteomic data. We believe that integrated approaches, such as the one presented here, have a high potential for the identification of clinical targets in infectious diseases, especially in the Salmonella infections.
Collapse
Affiliation(s)
- Gungor Budak
- Department of Health Informatics, Graduate School of Informatics, Middle East Technical University Ankara, Turkey
| | - Oyku Eren Ozsoy
- Department of Health Informatics, Graduate School of Informatics, Middle East Technical University Ankara, Turkey
| | - Yesim Aydin Son
- Department of Health Informatics, Graduate School of Informatics, Middle East Technical University Ankara, Turkey
| | - Tolga Can
- Department of Computer Engineering, College of Engineering, Middle East Technical University Ankara, Turkey
| | - Nurcan Tuncbag
- Department of Health Informatics, Graduate School of Informatics, Middle East Technical University Ankara, Turkey
| |
Collapse
|
23
|
Rieger J, Janczyk P, Hünigen H, Plendl J. Enhancement of immunohistochemical detection of Salmonella in tissues of experimentally infected pigs. Eur J Histochem 2015; 59:2516. [PMID: 26428884 PMCID: PMC4598596 DOI: 10.4081/ejh.2015.2516] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Revised: 05/05/2015] [Accepted: 05/15/2015] [Indexed: 12/13/2022] Open
Abstract
Salmonella Typhimurium is one of the main pathogens compromising porcine and human health as well as food safety, because it is a prevailing source of foodborne infections due to contaminated pork. A prominent problem in the management of this bacteriosis is the number of subclinically infected carrier pigs. As very little is known concerning the mechanisms allowing Salmonella to persist in pigs, the objective of this study was to develop an immunohistochemical approach for the detection of salmonellae in tissue of pigs experimentally infected with Salmonella Typhimurium. Samples were obtained from a challenge trial in which piglets of the German Landrace were intragastrically infected with Salmonella enterica serovar Typhimurium DT104 (1.4-2.1x1010 CFU). Piglets were sacrificed on days 2 and 28 post infection. Tissue samples of jejunum, ileum, colon, ileocecal mesenteric lymph nodes (Lnn. ileocolici), and tonsils (Tonsilla veli palatini) were fixed in Zamboni's fixative and paraffin-embedded. Different immunohistochemical staining protocols were evaluated. Salmonella was detected in varying amounts in the tissues. Brown iron-containing pigments in the lymph nodes interfered with the identification of Salmonella if DAB was used as a staining reagent. Detergents like Triton X-100 or Saponin enhanced the sensitivity. It seems advisable not to use a detection system with brown staining for bacteria in an experimental setup involving intestinal damage including haemorrhage. The use of detergents appears to result in a higher sensitivity in the immunohistochemical detection of salmonellae.
Collapse
|
24
|
Abel AM, Schuldt KM, Rajasekaran K, Hwang D, Riese MJ, Rao S, Thakar MS, Malarkannan S. IQGAP1: insights into the function of a molecular puppeteer. Mol Immunol 2015; 65:336-49. [PMID: 25733387 DOI: 10.1016/j.molimm.2015.02.012] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Revised: 02/09/2015] [Accepted: 02/09/2015] [Indexed: 02/06/2023]
Abstract
The intracellular spatiotemporal organization of signaling events is critical for normal cellular function. In response to environmental stimuli, cells utilize highly organized signaling pathways that are subject to multiple layers of regulation. However, the molecular mechanisms that coordinate these complex processes remain an enigma. Scaffolding proteins (scaffolins) have emerged as critical regulators of signaling pathways, many of which have well-described functions in immune cells. IQGAP1, a highly conserved cytoplasmic scaffold protein, is able to curb, compartmentalize, and coordinate multiple signaling pathways in a variety of cell types. IQGAP1 plays a central role in cell-cell interaction, cell adherence, and movement via actin/tubulin-based cytoskeletal reorganization. Evidence also implicates IQGAP1 as an essential regulator of the MAPK and Wnt/β-catenin signaling pathways. Here, we summarize the recent advances on the cellular and molecular biology of IQGAP1. We also describe how this pleiotropic scaffolin acts as a true molecular puppeteer, and highlight the significance of future research regarding the role of IQGAP1 in immune cells.
Collapse
Affiliation(s)
- Alex M Abel
- Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Department of Microbiology & Molecular Genetics, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Kristina M Schuldt
- Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Kamalakannan Rajasekaran
- Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - David Hwang
- Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Matthew J Riese
- Department of Microbiology & Molecular Genetics, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Sridhar Rao
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Monica S Thakar
- Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Subramaniam Malarkannan
- Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Department of Microbiology & Molecular Genetics, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| |
Collapse
|
25
|
Hedman AC, Smith JM, Sacks DB. The biology of IQGAP proteins: beyond the cytoskeleton. EMBO Rep 2015; 16:427-46. [PMID: 25722290 DOI: 10.15252/embr.201439834] [Citation(s) in RCA: 172] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Accepted: 01/07/2015] [Indexed: 01/02/2023] Open
Abstract
IQGAP scaffold proteins are evolutionarily conserved in eukaryotes and facilitate the formation of complexes that regulate cytoskeletal dynamics, intracellular signaling, and intercellular interactions. Fungal and mammalian IQGAPs are implicated in cytokinesis. IQGAP1, IQGAP2, and IQGAP3 have diverse roles in vertebrate physiology, operating in the kidney, nervous system, cardio-vascular system, pancreas, and lung. The functions of IQGAPs can be corrupted during oncogenesis and are usurped by microbial pathogens. Therefore, IQGAPs represent intriguing candidates for novel therapeutic agents. While modulation of the cytoskeletal architecture was initially thought to be the primary function of IQGAPs, it is now clear that they have roles beyond the cytoskeleton. This review describes contributions of IQGAPs to physiology at the organism level.
Collapse
Affiliation(s)
- Andrew C Hedman
- Department of Laboratory Medicine, National Institutes of Health, Bethesda, MD, USA
| | - Jessica M Smith
- Department of Laboratory Medicine, National Institutes of Health, Bethesda, MD, USA
| | - David B Sacks
- Department of Laboratory Medicine, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
26
|
Ramos-Morales F. Impact of Salmonella enterica Type III Secretion System Effectors on the Eukaryotic Host Cell. ACTA ACUST UNITED AC 2012. [DOI: 10.5402/2012/787934] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Type III secretion systems are molecular machines used by many Gram-negative bacterial pathogens to inject proteins, known as effectors, directly into eukaryotic host cells. These proteins manipulate host signal transduction pathways and cellular processes to the pathogen’s advantage. Salmonella enterica possesses two virulence-related type III secretion systems that deliver more than forty effectors. This paper reviews our current knowledge about the functions, biochemical activities, host targets, and impact on host cells of these effectors. First, the concerted action of effectors at the cellular level in relevant aspects of the interaction between Salmonella and its hosts is analyzed. Then, particular issues that will drive research in the field in the near future are discussed. Finally, detailed information about each individual effector is provided.
Collapse
Affiliation(s)
- Francisco Ramos-Morales
- Departamento de Genética, Facultad de Biología, Universidad de Sevilla, Avenida Reina Mercedes 6, 41012 Sevilla, Spain
| |
Collapse
|
27
|
Bhaskaran SS, Stebbins CE. Structure of the catalytic domain of the Salmonella virulence factor SseI. ACTA CRYSTALLOGRAPHICA. SECTION D, BIOLOGICAL CRYSTALLOGRAPHY 2012; 68:1613-21. [PMID: 23151626 PMCID: PMC3498931 DOI: 10.1107/s0907444912039042] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/17/2012] [Accepted: 09/12/2012] [Indexed: 11/15/2022]
Abstract
SseI is secreted into host cells by Salmonella and contributes to the establishment of systemic infections. The crystal structure of the C-terminal domain of SseI has been solved to 1.70 Å resolution, revealing it to be a member of the cysteine protease superfamily with a catalytic triad consisting of Cys178, His216 and Asp231 that is critical to its virulence activities. Structure-based analysis revealed that SseI is likely to possess either acyl hydrolase or acyltransferase activity, placing this virulence factor in the rapidly growing class of enzymes of this family utilized by bacterial pathogens inside eukaryotic cells.
Collapse
Affiliation(s)
- Shyam S. Bhaskaran
- Laboratory of Structural Microbiology, The Rockefeller University, New York, NY 10065, USA
| | - C. Erec Stebbins
- Laboratory of Structural Microbiology, The Rockefeller University, New York, NY 10065, USA
| |
Collapse
|
28
|
Karlsson T, Turkina MV, Yakymenko O, Magnusson KE, Vikström E. The Pseudomonas aeruginosa N-acylhomoserine lactone quorum sensing molecules target IQGAP1 and modulate epithelial cell migration. PLoS Pathog 2012; 8:e1002953. [PMID: 23071436 PMCID: PMC3469656 DOI: 10.1371/journal.ppat.1002953] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2011] [Accepted: 08/23/2012] [Indexed: 01/01/2023] Open
Abstract
Quorum sensing (QS) signaling allows bacteria to control gene expression once a critical population density is achieved. The Gram-negative human pathogen Pseudomonas aeruginosa uses N-acylhomoserine lactones (AHL) as QS signals, which coordinate the production of virulence factors and biofilms. These bacterial signals can also modulate human cell behavior. Little is known about the mechanisms of the action of AHL on their eukaryotic targets. Here, we found that N-3-oxo-dodecanoyl-L-homoserine lactone 3O-C12-HSL modulates human intestinal epithelial Caco-2 cell migration in a dose- and time-dependent manner. Using new 3O-C12-HSL biotin and fluorescently-tagged probes for LC-MS/MS and confocal imaging, respectively, we demonstrated for the first time that 3O-C12-HSL interacts and co-localizes with the IQ-motif-containing GTPase-activating protein IQGAP1 in Caco-2 cells. The interaction between IQGAP1 and 3O-C12-HSL was further confirmed by pull-down assay using a GST-tagged protein with subsequent Western blot of IQGAP1 and by identifying 3O-C12-HSL with a sensor bioassay. Moreover, 3O-C12-HSL induced changes in the phosphorylation status of Rac1 and Cdc42 and the localization of IQGAP1 as evidenced by confocal and STED microscopy and Western blots. Our findings suggest that the IQGAP1 is a novel partner for P.aeruginosa 3O-C12-HSL and likely the integrator of Rac1 and Cdc42- dependent altered cell migration. We propose that the targeting of IQGAP1 by 3O-C12-HSL can trigger essential changes in the cytoskeleton network and be an essential component in bacterial – human cell communication. The human pathogen Pseudomonas aeruginosa and other bacteria communicate with each other using quorum sensing (QS). This is important for their growth, virulence, motility and the formation of biofilms. Furthermore, eukaryotic cells “listen and respond” to QS signaling, but the exact mechanisms and receptors on mammalian cells have not been identified. We have previously shown that N-acylhomoserine lactones (AHL) alter epithelial barrier functions and increase chemotaxis in human neutrophils. We show here that 3O-C12-HSL modulates the migration of epithelial cells in a dose- and time-dependent manner. Using newly designed and validated biotin- and fluorescein-based 3O-C12-HSL probes we identified the IQ-motif-containing GTPase-activating protein IQGAP1 as a human target of 3O-C12-HSL. We propose that the interaction between IQGAP1 and 3O-C12-HSL provides a novel mechanism for its mode of action on eukaryotic cells, and by affecting the distribution of IQGAP1 and phosphorylation of Rac1 and Cdc42, upstream effectors of filamentous actin remodeling, also cell migration. We suggest that recognition of IQGAP1 by 3O-C12-HSL is a very early event in the communication between bacteria and human epithelial cells.
Collapse
Affiliation(s)
- Thommie Karlsson
- Division of Medical Microbiology, Department of Clinical and Experimental Medicine, Faculty of Health Sciences, Linköping University, Linköping, Sweden
| | - Maria V. Turkina
- Division of Cell Biology, Department of Clinical and Experimental Medicine, Faculty of Health Sciences, Linköping University, Linköping, Sweden
| | - Olena Yakymenko
- Division of Medical Microbiology, Department of Clinical and Experimental Medicine, Faculty of Health Sciences, Linköping University, Linköping, Sweden
| | - Karl-Eric Magnusson
- Division of Medical Microbiology, Department of Clinical and Experimental Medicine, Faculty of Health Sciences, Linköping University, Linköping, Sweden
| | - Elena Vikström
- Division of Medical Microbiology, Department of Clinical and Experimental Medicine, Faculty of Health Sciences, Linköping University, Linköping, Sweden
- * E-mail:
| |
Collapse
|
29
|
Zhang L, Li X, Zhang L, Wang B, Zhang T, Ye J. Chlamydophila (Chlamydia) pneumoniae infection promotes vascular smooth muscle cell adhesion and migration through IQ domain GTPase-activating protein 1. Microb Pathog 2012; 53:207-13. [PMID: 22835851 DOI: 10.1016/j.micpath.2012.07.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2012] [Revised: 07/14/2012] [Accepted: 07/17/2012] [Indexed: 01/16/2023]
Abstract
The mechanisms for Chlamydophila (Chlamydia) pneumoniae (C. pneumoniae) infection-induced atherosclerosis are still unclear. Cell adhesion has important roles in vascular smooth muscle cell (VSMC) migration required in the development of atherosclerosis. However, it is still unknown whether IQ domain GTPase-activating protein 1 (IQGAP1) plays pivotal roles in C. pneumoniae infection-induced the adhesion and migration of rat primary VSMCs. Accordingly, in this study, we demonstrated that rat primary VSMC adhesion (P < 0.001) and migration (P < 0.01) measured by cell adhesion assay and Transwell assay, respectively, were significantly enhanced after C. pneumoniae infection. Reverse transcription-polymerase chain reaction analysis revealed that the mRNA expression levels of IQGAP1 in the infected rat primary VSMCs were found to increase gradually to reach a peak and then decrease gradually to a level similar to the control. We further showed that the increases in rat primary VSMC adhesion to Matrigel (P < 0.001) and migration (P < 0.01) caused by C. pneumoniae infection were markedly inhibited after IQGAP1 knockdown by a pool of four short hairpin RNAs. Taken together, our results suggest that C. pneumoniae infection may promote the adhesion and migration of VSMCs possibly by upregulating the IQGAP1 expression.
Collapse
Affiliation(s)
- Lijun Zhang
- Department of Pathophysiology, School of Basic Medical Sciences, Tianjin Medical University, No. 22 Qixiangtai Road, Heping District, Tianjin 300070, China.
| | | | | | | | | | | |
Collapse
|
30
|
Taub N, Nairz M, Hilber D, Hess MW, Weiss G, Huber LA. The late endosomal adaptor p14 is a macrophage host-defense factor against Salmonella infection. J Cell Sci 2012; 125:2698-708. [PMID: 22427693 DOI: 10.1242/jcs.100073] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The outcome of an infection depends on the balance between host resistance and bacterial virulence. Here, we show that the late endosomal adaptor p14 (also known as LAMTOR2) is one of the components for cellular host defense against the intracellular pathogen Salmonella enterica serovar Typhimurium. During Salmonella infection, the complex of p14 and MP1 is required for the accurately timed transport of Salmonella through the endolysosomal system. Loss of p14 opens a time window that allows Salmonella to populate a replication niche, in which early and late antimicrobial effector systems, comprising NADPH phagocytic oxidase and inducible nitric oxide synthase, respectively, are inappropriately activated. Thus, p14 supports the accurate transport of Salmonella through the endolysosomal system, thereby limiting bacterial replication in both, professional phagocytes and in non-phagocytic cells in vitro, and helps mice to successfully battle Salmonella infection in vivo.
Collapse
Affiliation(s)
- Nicole Taub
- Biocenter, Division of Cell Biology, Innsbruck Medical University, Innsbruck 6020, Austria
| | | | | | | | | | | |
Collapse
|
31
|
Malarkannan S, Awasthi A, Rajasekaran K, Kumar P, Schuldt KM, Bartoszek A, Manoharan N, Goldner NK, Umhoefer CM, Thakar MS. IQGAP1: a regulator of intracellular spacetime relativity. THE JOURNAL OF IMMUNOLOGY 2012; 188:2057-63. [PMID: 22345702 DOI: 10.4049/jimmunol.1102439] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Activating and inhibiting receptors of lymphocytes collect valuable information about their mikròs kósmos. This information is essential to initiate or to turn off complex signaling pathways. Irrespective of these advances, our knowledge on how these intracellular activation cascades are coordinated in a spatiotemporal manner is far from complete. Among multiple explanations, the scaffolding proteins have emerged as a critical piece of this evolutionary tangram. Among many, IQGAP1 is one of the essential scaffolding proteins that coordinate multiple signaling pathways. IQGAP1 possesses multiple protein interaction motifs to achieve its scaffolding functions. Using these domains, IQGAP1 has been shown to regulate a number of essential cellular events. This includes actin polymerization, tubulin multimerization, microtubule organizing center formation, calcium/calmodulin signaling, Pak/Raf/Mek1/2-mediated Erk1/2 activation, formation of maestrosome, E-cadherin, and CD44-mediated signaling and glycogen synthase kinase-3/adenomatous polyposis coli-mediated β-catenin activation. In this review, we summarize the recent developments and exciting new findings of cellular functions of IQGAP1.
Collapse
Affiliation(s)
- Subramaniam Malarkannan
- Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, Milwaukee, WI 53226, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Krishnan S, Fernandez GE, Sacks DB, Prasadarao NV. IQGAP1 mediates the disruption of adherens junctions to promote Escherichia coli K1 invasion of brain endothelial cells. Cell Microbiol 2012; 14:1415-33. [PMID: 22519731 DOI: 10.1111/j.1462-5822.2012.01805.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2011] [Revised: 04/09/2012] [Accepted: 04/16/2012] [Indexed: 12/17/2022]
Abstract
The transcellular entry of Escherichia coli K1 through human brain microvascular endothelial cells (HBMEC) is responsible for tight junction disruption, leading to brain oedema in neonatal meningitis. Previous studies demonstrated that outer membrane protein A (OmpA) of E. coli K1 interacts with its receptor, Ecgp96, to induce PKC-α phosphorylation, adherens junction (AJ) disassembly (by dislodging β-catenin from VE-cadherin), and remodelling of actin in HBMEC. We report here that IQGAP1 mediates β-catenin dissociation from AJs to promote actin polymerization required for E. coli K1 invasion of HBMEC. Overexpression of C-terminal truncated IQGAP1 (IQΔC) that cannot bind β-catenin prevents both AJ disruption and E. coli K1 entry. Of note, phospho-PKC-α interacts with the C-terminal portion of Ecgp96 as well as with VE-cadherin after IQGAP1-mediated AJ disassembly. HBMEC overexpressing either C-terminal truncated Ecgp96 (Ecgp96Δ200) or IQΔC upon infection with E. coli showed no interaction of phospho-PKC-α with Ecgp96. These data indicate that the binding of OmpA to Ecgp96 induces PKC-α phosphorylation and association of phospho-PKC-α with Ecgp96, and then signals IQGAP1 to detach β-catenin from AJs. Subsequently, IQGAP1/β-catenin bound actin translocates to the site of E. coli K1 attachment to promote invasion.
Collapse
Affiliation(s)
- Subramanian Krishnan
- Division of Infectious Diseases, Department of Pediatrics, The Saban Research Institute, Children's Hospital Los Angeles, University of Southern California School of Medicine, Los Angeles, CA 90027, USA
| | | | | | | |
Collapse
|
33
|
Salmonella enterica serotype Typhimurium usurps the scaffold protein IQGAP1 to manipulate Rac1 and MAPK signalling. Biochem J 2012; 440:309-18. [PMID: 21851337 DOI: 10.1042/bj20110419] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Salmonella enterica serotype Typhimurium invades eukaryotic cells by re-arranging the host-cell cytoskeleton. However, the precise mechanisms by which Salmonella induces cytoskeletal changes remain undefined. IQGAP1 (IQ motif-containing GTPase-activating protein 1) is a scaffold protein that binds multiple proteins including actin, the Rho GTPases Rac1 and Cdc42 (cell division cycle 42), and components of the MAPK (mitogen-activated protein kinase) pathway. We have shown previously that optimal invasion of Salmonella into HeLa cells requires IQGAP1. In the present paper, we use IQGAP1-null MEFs (mouse embryonic fibroblasts) and selected well-characterized IQGAP1 mutant constructs to dissect the molecular determinants of Salmonella invasion. Knockout of IQGAP1 expression reduced Salmonella invasion into MEFs by 75%. Reconstituting IQGAP1-null MEFs with wild-type IQGAP1 completely rescued invasion. By contrast, reconstituting IQGAP1-null cells with mutant IQGAP1 constructs that specifically lack binding to either Cdc42 and Rac1 (termed IQGAP1ΔMK24), actin, MEK [MAPK/ERK (extracellular-signal-regulated kinase) kinase] or ERK only partially restored Salmonella entry. Cell-permeant inhibitors of Rac1 activation or MAPK signalling reduced Salmonella invasion into control cells by 50%, but had no effect on bacterial entry into IQGAP1-null MEFs. Importantly, the ability of IQGAP1ΔMK24 to promote Salmonella invasion into IQGAP1-null cells was abrogated by chemical inhibition of MAPK signalling. Collectively, these results imply that the scaffolding function of IQGAP1, which integrates Rac1 and MAPK signalling, is usurped by Salmonella to invade fibroblasts and suggest that IQGAP1 may be a potential therapeutic target for Salmonella pathogenesis.
Collapse
|
34
|
Rogers LD, Brown NF, Fang Y, Pelech S, Foster LJ. Phosphoproteomic analysis of Salmonella-infected cells identifies key kinase regulators and SopB-dependent host phosphorylation events. Sci Signal 2012; 4:rs9. [PMID: 21934108 DOI: 10.1126/scisignal.2001668] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Salmonella enterica is a bacterial pathogen that causes gastroenteritis and typhoid fever. Virulence is achieved by two type III secretion systems that translocate effector proteins into host cells, where they mimic or block host protein function. Effectors translocated into host cells by the first type III secretion system facilitate invasion and stimulate intracellular signaling cascades leading to inflammation. Here, we performed global temporal analysis of host signaling events induced during the initial stages of Salmonella infection and identified the dynamics of host protein phosphorylation as well as differences between growth factor-stimulated and bacteria-induced signaling. Informatics analysis predicted that sites with altered phosphorylation in infected cells were targeted by the serine-threonine kinases Akt, protein kinase C, and Pim and that up to half of the host phosphorylation events detected after Salmonella infection required the effector protein SopB. Our data reveal extensive manipulation of host phosphorylation cascades by this Salmonella effector and provide a detailed map of the events leading to intestinal inflammation, which is the crucial host response that enables Salmonella to proliferate in the intestine.
Collapse
Affiliation(s)
- Lindsay D Rogers
- Centre for High-Throughput Biology and Department of Biochemistry, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| | | | | | | | | |
Collapse
|
35
|
Malik-Kale P, Jolly CE, Lathrop S, Winfree S, Luterbach C, Steele-Mortimer O. Salmonella - at home in the host cell. Front Microbiol 2011; 2:125. [PMID: 21687432 PMCID: PMC3109617 DOI: 10.3389/fmicb.2011.00125] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2011] [Accepted: 05/19/2011] [Indexed: 11/16/2022] Open
Abstract
The Gram-negative bacterium Salmonella enterica has developed an array of sophisticated tools to manipulate the host cell and establish an intracellular niche, for successful propagation as a facultative intracellular pathogen. While Salmonella exerts diverse effects on its host cell, only the cell biology of the classic “trigger”-mediated invasion process and the subsequent development of the Salmonella-containing vacuole have been investigated extensively. These processes are dependent on cohorts of effector proteins translocated into host cells by two type III secretion systems (T3SS), although T3SS-independent mechanisms of entry may be important for invasion of certain host cell types. Recent studies into the intracellular lifestyle of Salmonella have provided new insights into the mechanisms used by this pathogen to modulate its intracellular environment. Here we discuss current knowledge of Salmonella-host interactions including invasion and establishment of an intracellular niche within the host.
Collapse
Affiliation(s)
- Preeti Malik-Kale
- Laboratory of Intracellular Parasites, Rocky Mountain Laboratories, National Institutes of Allergy and Infectious Disease, National Institute of Health Hamilton, MT, USA
| | | | | | | | | | | |
Collapse
|
36
|
Pathmanathan S, Hamilton E, Atcheson E, Timson DJ. The interaction of IQGAPs with calmodulin-like proteins. Biochem Soc Trans 2011; 39:694-9. [PMID: 21428964 DOI: 10.1042/bst0390694] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Since their identification over 15 years ago, the IQGAP (IQ-motif-containing GTPase-activating protein) family of proteins have been implicated in a wide range of cellular processes, including cytoskeletal reorganization, cell-cell adhesion, cytokinesis and apoptosis. These processes rely on protein-protein interactions, and understanding these (and how they influence one another) is critical in determining how the IQGAPs function. A key group of interactions is with calmodulin and the structurally related proteins myosin essential light chain and S100B. These interactions occur primarily through a series of IQ motifs, which are α-helical segments of the protein located towards the middle of the primary sequence. The three human IQGAP isoforms (IQGAP1, IQGAP2 and IQGAP3) all have four IQ motifs. However, these have different affinities for calmodulin, myosin light chain and S100B. Whereas all four IQ motifs of IQGAP1 interact with calmodulin in the presence of calcium, only the last two do so in the absence of calcium. IQ1 (the first IQ motif) interacts with the myosin essential light chain Mlc1sa and the first two undergo a calcium-dependent interaction with S100B. The significance of the interaction between Mlc1sa and IQGAP1 in mammals is unknown. However, a similar interaction involving the Saccharomyces cerevisiae IQGAP-like protein Iqg1p is involved in cytokinesis, leading to speculation that there may be a similar role in mammals.
Collapse
Affiliation(s)
- Sevvel Pathmanathan
- School of Biological Sciences, Queen's University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast BT9 7BL, UK
| | | | | | | |
Collapse
|
37
|
Kim H, White CD, Sacks DB. IQGAP1 in microbial pathogenesis: Targeting the actin cytoskeleton. FEBS Lett 2011; 585:723-9. [PMID: 21295032 PMCID: PMC3085995 DOI: 10.1016/j.febslet.2011.01.041] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2010] [Revised: 01/25/2011] [Accepted: 01/26/2011] [Indexed: 11/18/2022]
Abstract
Microbial pathogens cause widespread morbidity and mortality. Central to the pathogens' virulence is manipulation of the host cell's cytoskeleton, which facilitates microbial invasion, multiplication, and avoidance of the innate immune response. IQGAP1 is a ubiquitously expressed scaffold protein that integrates diverse signaling cascades. Research has shown that IQGAP1 binds to and modulates the activity of multiple proteins that participate in bacterial invasion. Here, we review data that support a role for IQGAP1 in infectious disease via its ability to regulate the actin cytoskeleton. In addition, we explore other mechanisms by which IQGAP1 may be exploited by microbial pathogens.
Collapse
Affiliation(s)
- Hugh Kim
- Department of Translational Medicine, Brigham and Women's Hospital and Harvard Medical School, 1 Blackfan Circle, Boston, MA 02115, USA
| | | | | |
Collapse
|
38
|
Dai Y, Dudek NL, Li Q, Muma NA. Phospholipase C, Ca2+, and calmodulin signaling are required for 5-HT2A receptor-mediated transamidation of Rac1 by transglutaminase. Psychopharmacology (Berl) 2011; 213:403-12. [PMID: 20717650 PMCID: PMC3033764 DOI: 10.1007/s00213-010-1984-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2010] [Accepted: 07/28/2010] [Indexed: 01/12/2023]
Abstract
RATIONALE Serotonin and especially serotonin 2A (5-HT(2A)) receptor signaling are important in the etiology and treatment of schizophrenia and affective disorders. We previously reported a novel 5-HT(2A) receptor effector, increased transglutaminase (TGase)-catalyzed transamidation, and activation of the small G protein Rac1 in A1A1v cells, a rat embryonic cortical cell line. OBJECTIVES In this study, we explore the signaling pathway involved in 5-HT(2A) receptor-mediated Rac1 transamidation. METHODS A1A1v cells were pretreated with pharmacological inhibitors of phospholipase C (PLC) or calmodulin (CaM), and then stimulated by the 5-HT(2A) receptor agonist, 2,5-dimethoxy-4-iodoamphetamine (DOI). Intracellular Ca(2+) concentration and TGase-modified Rac1 transamidation were monitored. The effect of manipulation of intracellular Ca(2+) by a Ca(2+) ionophore or a chelating agent on Rac1 transamidation was also evaluated. RESULTS In cells pretreated with a PLC inhibitor U73122, DOI-stimulated increases in the intracellular Ca(2+) concentration and TGase-modified Rac1 were significantly attenuated as compared to those pretreated with U73343, an inactive analog. The membrane-permeant Ca(2+) chelator, BAPTA-AM strongly reduced TGase-catalyzed Rac1 transamidation upon DOI stimulation. Conversely, the Ca(2+) ionophore ionomycin, at a concentration that induced an elevation of cytosolic Ca(2+) to a level comparable to cells treated with DOI, produced an increase in TGase-modified Rac1 without 5-HT(2A) receptor activation. Moreover, the CaM inhibitor W-7, significantly decreased Rac1 transamidation in a dose-dependent manner in DOI-treated cells. CONCLUSIONS These results indicate that 5-HT(2A) receptor-coupled PLC activation and subsequent Ca(2+) and CaM signaling are necessary for TGase-catalyzed Rac1 transamidation, and an increase in intracellular Ca(2+) is sufficient to induce Rac1 transamidation.
Collapse
Affiliation(s)
- Ying Dai
- Neuroscience Program, Loyola University, School of Medicine, Maywood, IL, USA; and Department of Pharmacology and Toxicology, University of Kansas, School of Pharmacy, 1251 Wescoe Hall Drive, Lawrence, KS 66045, USA
| | - Nichole L. Dudek
- Department of Pharmacology and Experimental Therapeutics, Loyola University Chicago, School of Medicine, Maywood, IL, USA
| | - Qian Li
- Department of Pharmacology and Toxicology, University of Kansas, School of Pharmacy, 1251 Wescoe Hall Drive, Lawrence, KS 66045, USA
| | - Nancy A. Muma
- Department of Pharmacology and Toxicology, University of Kansas, School of Pharmacy, 1251 Wescoe Hall Drive, Lawrence, KS 66045, USA
| |
Collapse
|
39
|
Gladue DP, Holinka LG, Fernandez-Sainz IJ, Prarat MV, O'Donnell V, Vepkhvadze NG, Lu Z, Risatti GR, Borca MV. Interaction between Core protein of classical swine fever virus with cellular IQGAP1 protein appears essential for virulence in swine. Virology 2011; 412:68-74. [PMID: 21262517 DOI: 10.1016/j.virol.2010.12.060] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2010] [Revised: 12/28/2010] [Accepted: 12/30/2010] [Indexed: 02/08/2023]
Abstract
Here we show that IQGAP1, a cellular protein that plays a pivotal role as a regulator of the cytoskeleton interacts with Classical Swine Fever Virus (CSFV) Core protein. Sequence analyses identified residues within CSFV Core protein (designated as areas I, II, III and IV) that maintain homology to regions within the matrix protein of Moloney Murine Leukemia Virus (MMLV) that mediate binding to IQGAP1 [EMBO J, 2006 25:2155]. Alanine-substitution within Core regions I, II, III and IV identified residues that specifically mediate the Core-IQGAP1 interaction. Recombinant CSFV viruses harboring alanine substitutions at residues (207)ATI(209) (I), (210)VVE(212) (II), (213)GVK(215) (III), or (232)GLYHN(236) (IV) have defective growth in primary swine macrophage cultures. In vivo, substitutions of residues in areas I and III yielded viruses that were completely attenuated in swine. These data shows that the interaction of Core with an integral component of cytoskeletal regulation plays a role in the CSFV cycle.
Collapse
Affiliation(s)
- D P Gladue
- Plum Island Animal Disease Center, Agricultural Research Service, U.S. Department of Agriculture, Greenport, NY 11944, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Lin YH, Lin YM, Kuo YC, Wang YY, Kuo PL. Identification and characterization of a novel Rab GTPase-activating protein in spermatids. ACTA ACUST UNITED AC 2010; 34:e358-67. [DOI: 10.1111/j.1365-2605.2010.01126.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
41
|
Dunn JD, Valdivia RH. Uncivil engineers: Chlamydia, Salmonella and Shigella alter cytoskeleton architecture to invade epithelial cells. Future Microbiol 2010; 5:1219-32. [DOI: 10.2217/fmb.10.77] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The obligate intracellular bacterial pathogen Chlamydia trachomatis is a major cause of blindness and sexually transmitted diseases. Like the enteric pathogens Salmonella and Shigella, Chlamydia injects effector proteins into epithelial cells to initiate extensive remodeling of the actin cytoskeleton at the bacterial attachment site, which culminates in the engulfment of the bacterium by plasma membrane extensions. Numerous Salmonella and Shigella effectors promote this remodeling by activating Rho GTPases and tyrosine kinase signaling cascades and by directly manipulating actin dynamics. Recent studies indicate that similar host-cell alterations occur during Chlamydia invasion, but few effectors are known. The identification of additional Chlamydia effectors and the elucidation of their modes of function are critical steps towards an understanding of how this clinically important pathogen breaches epithelial surfaces and causes infection.
Collapse
Affiliation(s)
- Joe Dan Dunn
- Department of Molecular Genetics & Microbiology & Center for Microbial Pathogenesis Duke University Medical Center, 272 Jones Building, Box 3580, Durham, NC 27710, USA
| | | |
Collapse
|
42
|
Salmonella enterica serovar typhimurium invades fibroblasts by multiple routes differing from the entry into epithelial cells. Infect Immun 2010; 78:2700-13. [PMID: 20368348 DOI: 10.1128/iai.01389-09] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Fibroblasts are ubiquitous cells essential to tissue homeostasis. Despite their nonphagocytic nature, fibroblasts restrain replication of intracellular bacterial pathogens such as Salmonella enterica serovar Typhimurium. The extent to which the entry route of the pathogen determines this intracellular response is unknown. Here, we analyzed S. Typhimurium invasion in fibroblasts obtained from diverse origins, including primary cultures and stable nontransformed cell lines derived from normal tissues. Features distinct to the invasion of epithelial cells were found in all fibroblasts tested. In some fibroblasts, bacteria lacking the type III secretion system encoded in the Salmonella pathogenicity island 1 displayed significant invasion rates and induced the formation of lamellipodia and filopodia at the fibroblast-bacteria contact site. Other bacterial invasion traits observed in fibroblasts were the requirement of phosphatidylinositol 3-kinase, mitogen-activated protein kinase MEK1, and both actin filaments and microtubules. RNA interference studies showed that different Rho family GTPases are targeted by S. Typhimurium to enter into distinct fibroblasts. Rac1 and Cdc42 knockdown affected invasion of normal rat kidney fibroblasts, whereas none of the GTPases tested (Rac1, Cdc42, RhoA, or RhoG) was essential for invasion of immortalized human foreskin fibroblasts. Collectively, these data reveal a marked diversity in the modes used by S. Typhimurium to enter into fibroblasts.
Collapse
|
43
|
McLaughlin LM, Govoni GR, Gerke C, Gopinath S, Peng K, Laidlaw G, Chien YH, Jeong HW, Li Z, Brown MD, Sacks DB, Monack D. The Salmonella SPI2 effector SseI mediates long-term systemic infection by modulating host cell migration. PLoS Pathog 2009; 5:e1000671. [PMID: 19956712 PMCID: PMC2777311 DOI: 10.1371/journal.ppat.1000671] [Citation(s) in RCA: 108] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2009] [Accepted: 10/28/2009] [Indexed: 11/24/2022] Open
Abstract
Host-adapted strains of Salmonella enterica cause systemic infections and have the ability to persist systemically for long periods of time despite the presence of a robust immune response. Chronically infected hosts are asymptomatic and transmit disease to naïve hosts via fecal shedding of bacteria, thereby serving as a critical reservoir for disease. We show that the bacterial effector protein SseI (also called SrfH), which is translocated into host cells by the Salmonella Pathogenicity Island 2 (SPI2) type III secretion system (T3SS), is required for Salmonella typhimurium to maintain a long-term chronic systemic infection in mice. SseI inhibits normal cell migration of primary macrophages and dendritic cells (DC) in vitro, and such inhibition requires the host factor IQ motif containing GTPase activating protein 1 (IQGAP1), an important regulator of cell migration. SseI binds directly to IQGAP1 and co-localizes with this factor at the cell periphery. The C-terminal domain of SseI is similar to PMT/ToxA, a bacterial toxin that contains a cysteine residue (C1165) that is critical for activity. Mutation of the corresponding residue in SseI (C178A) eliminates SseI function in vitro and in vivo, but not binding to IQGAP1. In addition, infection with wild-type (WT) S. typhimurium suppressed DC migration to the spleen in vivo in an SseI-dependent manner. Correspondingly, examination of spleens from mice infected with WT S. typhimurium revealed fewer DC and CD4+ T lymphocytes compared to mice infected with ΔsseI S. typhimurium. Taken together, our results demonstrate that SseI inhibits normal host cell migration, which ultimately counteracts the ability of the host to clear systemic bacteria. Bacteria belonging to the genus Salmonella are capable of causing long-term chronic systemic infections, and bacteria primarily reside within macrophages in lymphoid tissues and sporadically are shed in the feces. These persistently infected individuals serve as a significant reservoir for disease transmission. Despite the importance of Salmonella as a human pathogen, relatively little is known about the host immune response or virulence mechanisms of long-term systemic infections. Host-adapted Salmonella strains invade and manipulate host cells by releasing specialized bacterial effector proteins into the host cell. We show that one of these bacterial effector proteins, SseI (SrfH), is required for Salmonella to maintain a long-term chronic systemic infection in mice. SseI is able to block the migration of host immune cells and consequentially attenuate the host's ability to clear systemic bacteria. SseI accomplishes this inhibitory activity in part by associating with the host protein IQGAP1, an important regulator of cell migration. The amino acid sequence of SseI is similar to several other protein sequences of known bacterial pathogens, including PMT/ToxA, a toxin, indicating that these factors may function similarly to one another and may comprise a new family of bacterial effector proteins.
Collapse
Affiliation(s)
- Laura M. McLaughlin
- Department of Microbiology and Immunology, Stanford University Medical Center, Stanford, California, United States of America
| | - Gregory R. Govoni
- Department of Microbiology and Immunology, Stanford University Medical Center, Stanford, California, United States of America
| | - Christiane Gerke
- Department of Microbiology and Immunology, Stanford University Medical Center, Stanford, California, United States of America
| | - Smita Gopinath
- Department of Microbiology and Immunology, Stanford University Medical Center, Stanford, California, United States of America
| | - Kaitian Peng
- Department of Microbiology and Immunology, Stanford University Medical Center, Stanford, California, United States of America
| | - Grace Laidlaw
- Department of Microbiology and Immunology, Stanford University Medical Center, Stanford, California, United States of America
| | - Yueh-Hsiu Chien
- Department of Microbiology and Immunology, Stanford University Medical Center, Stanford, California, United States of America
| | - Ha-Won Jeong
- Department of Pathology, Harvard Medical School, Brigham and Women's Hospital, Boston, Massachusetts, United States of America
| | - Zhigang Li
- Department of Pathology, Harvard Medical School, Brigham and Women's Hospital, Boston, Massachusetts, United States of America
| | - Matthew D. Brown
- Department of Pathology, Harvard Medical School, Brigham and Women's Hospital, Boston, Massachusetts, United States of America
| | - David B. Sacks
- Department of Pathology, Harvard Medical School, Brigham and Women's Hospital, Boston, Massachusetts, United States of America
| | - Denise Monack
- Department of Microbiology and Immunology, Stanford University Medical Center, Stanford, California, United States of America
- * E-mail:
| |
Collapse
|
44
|
White CD, Brown MD, Sacks DB. IQGAPs in cancer: a family of scaffold proteins underlying tumorigenesis. FEBS Lett 2009; 583:1817-24. [PMID: 19433088 PMCID: PMC2743239 DOI: 10.1016/j.febslet.2009.05.007] [Citation(s) in RCA: 247] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2009] [Revised: 04/28/2009] [Accepted: 05/02/2009] [Indexed: 12/13/2022]
Abstract
The IQGAP family comprises three proteins in humans. The best characterized is IQGAP1, which participates in protein-protein interactions and integrates diverse signaling pathways. IQGAP2 and IQGAP3 harbor all the domains identified in IQGAP1, but their biological roles are poorly defined. Proteins that bind IQGAP1 include Cdc42 and Rac1, E-cadherin, beta-catenin, calmodulin and components of the mitogen-activated protein kinase pathway, all of which are involved in cancer. Here, we summarize the biological functions of IQGAPs that may contribute to neoplasia. Additionally, we review published data which implicate IQGAPs in cancer and tumorigenesis. The cumulative evidence suggests IQGAP1 is an oncogene while IQGAP2 may be a tumor suppressor.
Collapse
Affiliation(s)
- Colin D. White
- Brigham and Women's Hospital and Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| | - Matthew D. Brown
- Brigham and Women's Hospital and Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| | - David B. Sacks
- Brigham and Women's Hospital and Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| |
Collapse
|
45
|
Abstract
The mitogen-activated protein kinase (MAPK) pathway allows cells to interpret external signals and respond in an appropriate way. Diverse cellular functions, ranging from differentiation and proliferation to migration and inflammation, are regulated by MAPK signalling. Therefore, cells have developed mechanisms by which this single pathway modulates numerous cellular responses from a wide range of activating factors. This specificity is achieved by several mechanisms, including temporal and spatial control of MAPK signalling components. Key to this control are protein scaffolds, which are multidomain proteins that interact with components of the MAPK cascade in order to assemble signalling complexes. Studies conducted on different scaffolds, in different biological systems, have shown that scaffolds exert substantial control over MAPK signalling, influencing the signal intensity, time course and, importantly, the cellular responses. Protein scaffolds, therefore, are integral elements to the modulation of the MAPK network in fundamental physiological processes.
Collapse
Affiliation(s)
- Matthew D. Brown
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115
| | - David B. Sacks
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115
| |
Collapse
|
46
|
Dong PX, Jia N, Xu ZJ, Liu YT, Li DJ, Feng YJ. Silencing of IQGAP1 by shRNA inhibits the invasion of ovarian carcinoma HO-8910PM cells in vitro. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2008; 27:77. [PMID: 19036171 PMCID: PMC2626583 DOI: 10.1186/1756-9966-27-77] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/05/2008] [Accepted: 11/27/2008] [Indexed: 11/10/2022]
Abstract
BACKGROUND IQGAP1 is a scaffolding protein and overexpressed in many human tumors, including ovarian cancer. However, the contribution of IQGAP1 to invasive properties of ovarian cancer cells remains unknown. Here, we investigated the effect of IQGAP1-specific short hairpin RNA (shRNA) expressing plasmids on metastatic potential of ovarian cancer HO-8910PM cells. METHODS We used RT-PCR and Western blot analysis to characterize expression of IQGAP1 in three human ovarian cancer-derived cell lines SK-OV-3, HO-8910 and HO-8910PM. We then determined whether expression of endogenous IQGAP1 correlated with invasive and migratory ability by using an in vitro Matrigel assay and cell migration assay. We further knocked down IQGAP1 using shRNA expressing plasmids controlled by U1 promoter in HO-8910PM cells and examined the proliferation activity, invasive and migration potential of IQGAP1 shRNA transfectants using MTT assay, in vitro Matrigel-coated invasion assay and migration assay. RESULTS IQGAP1 expression level seemed to be closely associated with the enhanced invasion and migration in ovarian cancer cell lines. Levels of both IQGAP1 mRNA and protein were significantly reduced in HO-8910PM cells transfected with plasmid-based IQGAP1-specific shRNAs. RNAi-mediated knockdown of IQGAP1 expression in HO-8910PM cells resulted in a significant decrease in cell invasion and migration. CONCLUSION Our findings support the hypothesis that IQGAP1 promotes tumor progression and identify IQGAP1 as a potential therapeutic strategy for ovarian cancer and some other tumors with over-expression of the IQGAP1 gene.
Collapse
Affiliation(s)
- Pei-Xin Dong
- Hospital and Institute of Obstetrics and Gynecology, Fudan University Shanghai Medical, College, Shanghai, PR China.
| | | | | | | | | | | |
Collapse
|
47
|
Brown MD, Bry L, Li Z, Sacks DB. Actin pedestal formation by enteropathogenic Escherichia coli is regulated by IQGAP1, calcium, and calmodulin. J Biol Chem 2008; 283:35212-22. [PMID: 18809683 DOI: 10.1074/jbc.m803477200] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
During infection, enteropathogenic Escherichia coli (EPEC) injects effector proteins into the host cell to manipulate the actin cytoskeleton and promote formation of actin pedestals. IQGAP1 is a multidomain protein that participates in numerous cellular functions, including Rac1/Cdc42 and Ca(2+)/calmodulin signaling and actin polymerization. Here we report that IQGAP1, Ca(2+), and calmodulin modulate actin pedestal formation by EPEC. Infection with EPEC promotes both the interaction of IQGAP1 with calmodulin and the localization of IQGAP1 and calmodulin to actin pedestals while reducing the interaction of IQGAP1 with Rac1 and Cdc42. IQGAP1-null fibroblasts display a reduced polymerization of actin in response to EPEC. In addition, antagonism of calmodulin or chelation of intracellular Ca(2+) reduces EPEC-dependent actin polymerization. Furthermore, IQGAP1 specifically interacts with Tir in vitro and in cells. Together these data identify IQGAP1, Ca(2+), and calmodulin as a novel signaling complex regulating actin pedestal formation by EPEC.
Collapse
Affiliation(s)
- Matthew D Brown
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Thorn 530, Boston, Massachusetts 02115, USA
| | | | | | | |
Collapse
|
48
|
Jadeski L, Mataraza JM, Jeong HW, Li Z, Sacks DB. IQGAP1 stimulates proliferation and enhances tumorigenesis of human breast epithelial cells. J Biol Chem 2007; 283:1008-17. [PMID: 17981797 DOI: 10.1074/jbc.m708466200] [Citation(s) in RCA: 113] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The scaffold protein IQGAP1 integrates signaling pathways and participates in diverse cellular activities. IQGAP1 is overexpressed in a number of human solid neoplasms, but its functional role in tumorigenesis has not been previously evaluated. Here we report that IQGAP1 contributes to neoplastic transformation of human breast epithelial cells. The amount of IQGAP1 in breast carcinoma is greater than that in normal tissue, with highly metastatic breast epithelial cells expressing the highest levels. Overexpression of IQGAP1 enhances proliferation of MCF-7 breast epithelial cells. Reduction of endogenous IQGAP1 by RNA interference impairs both serum-dependent and anchorage-independent growth of MCF-7 cells. Consistent with these in vitro observations, immortalized MCF-7 cells overexpressing IQGAP1 form invasive tumors in immunocompromised mice, whereas tumors derived from MCF-7 cells with stable knockdown of IQGAP1 are smaller and less invasive. In vitro analysis with selected IQGAP1 mutant constructs and a chemical inhibitor suggests that actin, Cdc42/Rac1, and the mitogen-activated protein kinase pathway contribute to the mechanism by which IQGAP1 increases cell invasion. Collectively, our data reveal that IQGAP1 enhances mammary tumorigenesis, suggesting that it may be a target for therapeutic intervention.
Collapse
Affiliation(s)
- Lorraine Jadeski
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | | | | | |
Collapse
|