1
|
Guo Z, Ma Y, Jia Z, Wang L, Lu X, Chen Y, Wang Y, Hao H, Yu S, Wang Z. Crosstalk between integrin/FAK and Crk/Vps25 governs invasion of bovine mammary epithelial cells by S. agalactiae. iScience 2023; 26:107884. [PMID: 37766995 PMCID: PMC10520442 DOI: 10.1016/j.isci.2023.107884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 07/26/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023] Open
Abstract
Streptococcus agalactiae (S. agalactiae) is a contagious obligate parasite of the udder in dairy cows. Here, we examined S. agalactiae-host interactions in bovine mammary epithelial cells (BMECs) in vitro. We found that S. agalactiae infected BMECs through laminin β2 and integrin. Crk, Vps25, and RhoA were differentially expressed in S. agalactiae-infected cells. S. agalactiae infection activated FAK and Crk. FAK deficiency decreased the number of intracellular S. agalactiae and Crk activation. Knockdown of Crk or Vps25 increased the level of intracellular S. agalactiae, whereas its overexpression had the opposite effect. RhoA expression and actin cytoskeleton were altered in S. agalactiae-infected BMECs. Crk and Vps25 interact in cells, and invaded S. agalactiae also activates Crk, allowing it to cooperate with Vps25 to defend against intracellular infection by S. agalactiae. This study provides insights into the mechanism by which intracellular infection by S. agalactiae is regulated in BMECs.
Collapse
Affiliation(s)
- Zhixin Guo
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot 010021, China
- School of Fisheries and Life Science, Dalian Ocean University, Dalian 116023, China
| | - Yuze Ma
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot 010021, China
| | - Zhibo Jia
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot 010021, China
| | - Liping Wang
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot 010021, China
| | - Xinyue Lu
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot 010021, China
| | - Yuhao Chen
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot 010021, China
- School of Life Sciences, Jining Normal University, Jining 012000, China
| | - Yanfeng Wang
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot 010021, China
| | - Huifang Hao
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot 010021, China
| | - Shuixing Yu
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot 010021, China
| | - Zhigang Wang
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot 010021, China
| |
Collapse
|
2
|
Gulen B, Rosselin M, Fauser J, Albers MF, Pett C, Krisp C, Pogenberg V, Schlüter H, Hedberg C, Itzen A. Identification of targets of AMPylating Fic enzymes by co-substrate-mediated covalent capture. Nat Chem 2020; 12:732-739. [PMID: 32632184 DOI: 10.1038/s41557-020-0484-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 05/07/2020] [Indexed: 11/09/2022]
Abstract
Various pathogenic bacteria use post-translational modifications to manipulate the central components of host cell functions. Many of the enzymes released by these bacteria belong to the large Fic family, which modify targets with nucleotide monophosphates. The lack of a generic method for identifying the cellular targets of Fic family enzymes hinders investigation of their role and the effect of the post-translational modification. Here, we establish an approach that uses reactive co-substrate-linked enzymes for proteome profiling. We combine synthetic thiol-reactive nucleotide derivatives with recombinantly produced Fic enzymes containing strategically placed cysteines in their active sites to yield reactive binary probes for covalent substrate capture. The binary complexes capture their targets from cell lysates and permit subsequent identification. Furthermore, we determined the structures of low-affinity ternary enzyme-nucleotide-substrate complexes by applying a covalent-linking strategy. This approach thus allows target identification of the Fic enzymes from both bacteria and eukarya.
Collapse
Affiliation(s)
- Burak Gulen
- Center for Integrated Protein Science Munich (CIPSM), Department of Chemistry, Technical University of Munich, Garching, Germany.,Department of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Marie Rosselin
- Chemical Biology Center (KBC), Institute of Chemistry, Umeå University, Umeå, Sweden
| | - Joel Fauser
- Center for Integrated Protein Science Munich (CIPSM), Department of Chemistry, Technical University of Munich, Garching, Germany.,Department of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Michael F Albers
- Chemical Biology Center (KBC), Institute of Chemistry, Umeå University, Umeå, Sweden
| | - Christian Pett
- Chemical Biology Center (KBC), Institute of Chemistry, Umeå University, Umeå, Sweden
| | - Christoph Krisp
- Clinical Chemistry and Laboratory Medicine, Mass Spectrometric Proteomics, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Vivian Pogenberg
- Department of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Hartmut Schlüter
- Clinical Chemistry and Laboratory Medicine, Mass Spectrometric Proteomics, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Christian Hedberg
- Chemical Biology Center (KBC), Institute of Chemistry, Umeå University, Umeå, Sweden.
| | - Aymelt Itzen
- Center for Integrated Protein Science Munich (CIPSM), Department of Chemistry, Technical University of Munich, Garching, Germany. .,Department of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany. .,Centre for Structural Systems Biology (CSSB), University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany.
| |
Collapse
|
3
|
Whelan R, McVicker G, Leo JC. Staying out or Going in? The Interplay between Type 3 and Type 5 Secretion Systems in Adhesion and Invasion of Enterobacterial Pathogens. Int J Mol Sci 2020; 21:E4102. [PMID: 32521829 PMCID: PMC7312957 DOI: 10.3390/ijms21114102] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 06/03/2020] [Accepted: 06/05/2020] [Indexed: 12/12/2022] Open
Abstract
Enteric pathogens rely on a variety of toxins, adhesins and other virulence factors to cause infections. Some of the best studied pathogens belong to the Enterobacterales order; these include enteropathogenic and enterohemorrhagic Escherichia coli, Shigella spp., and the enteropathogenic Yersiniae. The pathogenesis of these organisms involves two different secretion systems, a type 3 secretion system (T3SS) and type 5 secretion systems (T5SSs). The T3SS forms a syringe-like structure spanning both bacterial membranes and the host cell plasma membrane that translocates toxic effector proteins into the cytoplasm of the host cell. T5SSs are also known as autotransporters, and they export part of their own polypeptide to the bacterial cell surface where it exerts its function, such as adhesion to host cell receptors. During infection with these enteropathogens, the T3SS and T5SS act in concert to bring about rearrangements of the host cell cytoskeleton, either to invade the cell, confer intracellular motility, evade phagocytosis or produce novel structures to shelter the bacteria. Thus, in these bacteria, not only the T3SS effectors but also T5SS proteins could be considered "cytoskeletoxins" that bring about profound alterations in host cell cytoskeletal dynamics and lead to pathogenic outcomes.
Collapse
Affiliation(s)
| | | | - Jack C. Leo
- Antimicrobial Resistance, Omics and Microbiota Group, Department of Biosciences, Nottingham Trent University, Nottingham NG1 4FQ, UK; (R.W.); (G.M.)
| |
Collapse
|
4
|
Abstract
As a pathogen of plague, Yersinia pestis caused three massive pandemics in history that killed hundreds of millions of people. Yersinia pestis is highly invasive, causing severe septicemia which, if untreated, is usually fatal to its host. To survive in the host and maintain a persistent infection, Yersinia pestis uses several stratagems to evade the innate and the adaptive immune responses. For example, infections with this organism are biphasic, involving an initial "noninflammatory" phase where bacterial replication occurs initially with little inflammation and following by extensive phagocyte influx, inflammatory cytokine production, and considerable tissue destruction, which is called "proinflammatory" phase. In contrast, the host also utilizes its immune system to eliminate the invading bacteria. Neutrophil and macrophage are the first defense against Yersinia pestis invading through phagocytosis and killing. Other innate immune cells also play different roles, such as dendritic cells which help to generate more T helper cells. After several days post infection, the adaptive immune response begins to provide organism-specific protection and has a long-lasting immunological memory. Thus, with the cooperation and collaboration of innate and acquired immunity, the bacterium may be eliminated from the host. The research of Yersinia pestis and host immune systems provides an important topic to understand pathogen-host interaction and consequently develop effective countermeasures.
Collapse
Affiliation(s)
- Yujing Bi
- Beijing Institute of Microbiology and Epidemiology, No. Dongdajie, Fengtai, Beijing, 100071, China.
| |
Collapse
|
5
|
Luo LY, Hahn WC. Oncogenic Signaling Adaptor Proteins. J Genet Genomics 2015; 42:521-529. [PMID: 26554907 PMCID: PMC4643408 DOI: 10.1016/j.jgg.2015.09.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 08/31/2015] [Accepted: 09/02/2015] [Indexed: 02/08/2023]
Abstract
Signal transduction pathways activated by receptor tyrosine kinases (RTK) play a critical role in many aspects of cell function. Adaptor proteins serve an important scaffolding function that facilitates key signaling transduction events downstream of RTKs. Recent work integrating both structural and functional genomic approaches has identified several adaptor proteins as new oncogenes. In this review, we focus on the discovery, structure and function, and therapeutic implication of three of these adaptor oncogenes, CRKL, GAB2, and FRS2. Each of the three genes is recurrently amplified in lung adenocarcinoma or ovarian cancer, and is essential to cancer cell lines that harbor such amplification. Overexpression of each gene is able to transform immortalized human cell lines in in vitro or in vivo models. These observations identify adaptor protein as a distinct class of oncogenes and potential therapeutic targets.
Collapse
Affiliation(s)
- Leo Y Luo
- Health Sciences and Technology Program, Harvard Medical School, Boston, MA 02115, USA
| | - William C Hahn
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA 02142, USA; Center for Cancer Genome Discovery, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Medicine, Brigham and Women's Hospital, Boston, MA 02215, USA.
| |
Collapse
|
6
|
Deneka A, Korobeynikov V, Golemis EA. Embryonal Fyn-associated substrate (EFS) and CASS4: The lesser-known CAS protein family members. Gene 2015; 570:25-35. [PMID: 26119091 DOI: 10.1016/j.gene.2015.06.062] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 06/23/2015] [Indexed: 01/15/2023]
Abstract
The CAS (Crk-associated substrate) adaptor protein family consists of four members: CASS1/BCAR1/p130Cas, CASS2/NEDD9/HEF1/Cas-L, CASS3/EFS/Sin and CASS4/HEPL. While CAS proteins lack enzymatic activity, they contain specific recognition and binding sites for assembly of larger signaling complexes that are essential for cell proliferation, survival, migration, and other processes. All family members are intermediates in integrin-dependent signaling pathways mediated at focal adhesions, and associate with FAK and SRC family kinases to activate downstream effectors regulating the actin cytoskeleton. Most studies of CAS proteins to date have been focused on the first two members, BCAR1 and NEDD9, with altered expression of these proteins now appreciated as influencing disease development and prognosis for cancer and other serious pathological conditions. For these family members, additional mechanisms of action have been defined in receptor tyrosine kinase (RTK) signaling, estrogen receptor signaling or cell cycle progression, involving discrete partner proteins such as SHC, NSP proteins, or AURKA. By contrast, EFS and CASS4 have been less studied, although structure-function analyses indicate they conserve many elements with the better-known family members. Intriguingly, a number of recent studies have implicated these proteins in immune system function, and the pathogenesis of developmental disorders, autoimmune disorders including Crohn's disease, Alzheimer's disease, cancer and other diseases. In this review, we summarize the current understanding of EFS and CASS4 protein function in the context of the larger CAS family group.
Collapse
Affiliation(s)
- Alexander Deneka
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA, 19111, United States; Kazan Federal University, 420000, Kazan, Russian Federation
| | - Vladislav Korobeynikov
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA, 19111, United States; Novosibirsk State University, Medical Department, 630090, Novosibirsk, Russian Federation
| | - Erica A Golemis
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA, 19111, United States.
| |
Collapse
|
7
|
Camacho Leal MDP, Sciortino M, Tornillo G, Colombo S, Defilippi P, Cabodi S. p130Cas/BCAR1 scaffold protein in tissue homeostasis and pathogenesis. Gene 2015; 562:1-7. [PMID: 25727852 DOI: 10.1016/j.gene.2015.02.027] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Accepted: 02/01/2015] [Indexed: 12/11/2022]
Abstract
BCAR1 (also known as p130Cas/BCAR1) is an adaptor protein that belongs to the CAS family of scaffold proteins. In the past years, increasing evidence has demonstrated the ability of p130Cas/BCAR1 to activate signaling originating from mechanical stimuli, cell-extracellular matrix (ECM) adhesion and growth factor stimulation cascades during normal development and disease in various biological models. In this review we will specifically discuss the more recent data on the contribution of p130Cas/BCAR1 in the regulation of tissue homeostasis and its potential implications in pathological conditions.
Collapse
Affiliation(s)
| | - Marianna Sciortino
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Italy
| | - Giusy Tornillo
- European Cancer Stem Cell Research Institute and Cardiff School of Biosciences, Cardiff University, Cardiff, UK
| | - Shana Colombo
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Italy
| | - Paola Defilippi
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Italy
| | - Sara Cabodi
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Italy.
| |
Collapse
|
8
|
Contribution of Crk adaptor proteins to host cell and bacteria interactions. BIOMED RESEARCH INTERNATIONAL 2014; 2014:372901. [PMID: 25506591 PMCID: PMC4260429 DOI: 10.1155/2014/372901] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Accepted: 09/14/2014] [Indexed: 12/27/2022]
Abstract
The Crk adaptor family of proteins comprises the alternatively spliced CrkI and CrkII isoforms, as well as the paralog Crk-like (CrkL) protein, which is encoded by a different gene. Initially thought to be involved in signaling during apoptosis and cell adhesion, this ubiquitously expressed family of proteins is now known to play essential roles in integrating signals from a wide range of stimuli. In this review, we describe the structure and function of the different Crk proteins. We then focus on the emerging roles of Crk adaptors during Enterobacteriaceae pathogenesis, with special emphasis on the important human pathogens Salmonella, Shigella, Yersinia, and enteropathogenic Escherichia coli. Throughout, we remark on opportunities for future research into this intriguing family of proteins.
Collapse
|
9
|
Thinwa J, Segovia JA, Bose S, Dube PH. Integrin-mediated first signal for inflammasome activation in intestinal epithelial cells. THE JOURNAL OF IMMUNOLOGY 2014; 193:1373-82. [PMID: 24965773 DOI: 10.4049/jimmunol.1400145] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
How intestinal epithelial cells (IECs) recognize pathogens and activate inflammasomes at intestinal surfaces is poorly understood. We hypothesized that IECs use integrin receptors to recognize pathogens and initiate inflammation within the intestinal tract. We find that IECs infected with Yersinia enterocolitica, an enteric pathogen, use β1 integrins as pathogen recognition receptors detecting the bacterial adhesin invasin (Inv). The Inv-integrin interaction provides the first signal for NLRP3 inflammasome activation with the type three secretion system translocon providing the second signal for inflammasome activation, resulting in release of IL-18. During infection, Yersinia employs two virulence factors, YopE and YopH, to counteract Inv-mediated integrin-dependent inflammasome activation. Furthermore, NLRP3 inflammasome activation in epithelial cells requires components of the focal adhesion complex signaling pathway, focal adhesion kinase, and rac1. The binding of Inv to β1 integrins rapidly induces IL-18 mRNA expression, suggesting integrins provide a first signal for NLRP3 inflammasome activation. These data suggest integrins function as pathogen recognition receptors on IECs to rapidly induce inflammasome-derived IL-18-mediated responses.
Collapse
Affiliation(s)
- Josephine Thinwa
- Department of Microbiology and Immunology, University of Texas Health Science Center, San Antonio, TX 78229; and
| | - Jesus A Segovia
- Department of Microbiology and Immunology, University of Texas Health Science Center, San Antonio, TX 78229; and Center for Airway Inflammation Research, University of Texas Health Science Center, San Antonio, TX 78229
| | - Santanu Bose
- Department of Microbiology and Immunology, University of Texas Health Science Center, San Antonio, TX 78229; and Center for Airway Inflammation Research, University of Texas Health Science Center, San Antonio, TX 78229
| | - Peter H Dube
- Department of Microbiology and Immunology, University of Texas Health Science Center, San Antonio, TX 78229; and Center for Airway Inflammation Research, University of Texas Health Science Center, San Antonio, TX 78229
| |
Collapse
|
10
|
Subversion of trafficking, apoptosis, and innate immunity by type III secretion system effectors. Trends Microbiol 2013; 21:430-41. [DOI: 10.1016/j.tim.2013.06.008] [Citation(s) in RCA: 100] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2013] [Revised: 05/08/2013] [Accepted: 06/18/2013] [Indexed: 11/17/2022]
|
11
|
Barrett A, Pellet-Many C, Zachary IC, Evans IM, Frankel P. p130Cas: a key signalling node in health and disease. Cell Signal 2012; 25:766-77. [PMID: 23277200 DOI: 10.1016/j.cellsig.2012.12.019] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Accepted: 12/21/2012] [Indexed: 01/08/2023]
Abstract
p130Cas/breast cancer anti-oestrogen resistance 1 (BCAR1) is a member of the Cas (Crk-associated substrate) family of adaptor proteins, which have emerged as key signalling nodes capable of interactions with multiple proteins, with important regulatory roles in normal and pathological cell function. The Cas family of proteins is characterised by the presence of multiple conserved motifs for protein-protein interactions, and by extensive tyrosine and serine phosphorylations. Recent studies show that p130Cas contributes to migration, cell cycle control and apoptosis. p130Cas is essential during early embryogenesis, with a critical role in cardiovascular development. Furthermore, p130Cas has been reported to be involved in the development and progression of several human cancers. p130Cas is able to perform roles in multiple processes due to its capacity to regulate a diverse array of signalling pathways, transducing signals from growth factor receptor tyrosine kinases, non-receptor tyrosine kinases, and integrins. In this review we summarise the current understanding of the structure, function, and regulation of p130Cas, and discuss the importance of p130Cas in both physiological and pathophysiological settings, with a focus on the cardiovascular system and cancer.
Collapse
Affiliation(s)
- Angela Barrett
- Centre for Cardiovascular Biology and Medicine, Division of Medicine, University College London, London WC1E 6JJ, United Kingdom.
| | | | | | | | | |
Collapse
|
12
|
Ceelen L, Haesebrouck F, Vanhaecke T, Rogiers V, Vinken M. Modulation of connexin signaling by bacterial pathogens and their toxins. Cell Mol Life Sci 2011; 68:3047-64. [PMID: 21656255 PMCID: PMC11115019 DOI: 10.1007/s00018-011-0737-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2011] [Revised: 05/12/2011] [Accepted: 05/17/2011] [Indexed: 02/07/2023]
Abstract
Inherent to their pivotal tasks in the maintenance of cellular homeostasis, gap junctions, connexin hemichannels, and pannexin hemichannels are frequently involved in the dysregulation of this critical balance. The present paper specifically focuses on their roles in bacterial infection and disease. In particular, the reported biological outcome of clinically important bacteria including Escherichia coli, Shigella flexneri, Yersinia enterocolitica, Helicobacter pylori, Bordetella pertussis, Aggregatibacter actinomycetemcomitans, Pseudomonas aeruginosa, Citrobacter rodentium, Clostridium species, Streptococcus pneumoniae, and Staphylococcus aureus and their toxic products on connexin- and pannexin-related signaling in host cells is reviewed. Particular attention is paid to the underlying molecular mechanisms of these effects as well as to the actual biological relevance of these findings.
Collapse
Affiliation(s)
- Liesbeth Ceelen
- Department of Toxicology, Centre for Pharmaceutical Research, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium.
| | | | | | | | | |
Collapse
|
13
|
Tikhmyanova N, Little JL, Golemis EA. CAS proteins in normal and pathological cell growth control. Cell Mol Life Sci 2010; 67:1025-48. [PMID: 19937461 PMCID: PMC2836406 DOI: 10.1007/s00018-009-0213-1] [Citation(s) in RCA: 150] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2009] [Revised: 11/03/2009] [Accepted: 11/09/2009] [Indexed: 12/20/2022]
Abstract
Proteins of the CAS (Crk-associated substrate) family (BCAR1/p130Cas, NEDD9/HEF1/Cas-L, EFS/SIN and CASS4/HEPL) are integral players in normal and pathological cell biology. CAS proteins act as scaffolds to regulate protein complexes controlling migration and chemotaxis, apoptosis, cell cycle, and differentiation, and have more recently been linked to a role in progenitor cell function. Reflecting these complex functions, over-expression of CAS proteins has now been strongly linked to poor prognosis and increased metastasis in cancer, as well as resistance to first-line chemotherapeutics in multiple tumor types including breast and lung cancers, glioblastoma, and melanoma. Further, CAS proteins have also been linked to additional pathological conditions including inflammatory disorders, Alzheimer's and Parkinson's disease, as well as developmental defects. This review will explore the roles of the CAS proteins in normal and pathological states in the context of the many mechanistic insights into CAS protein function that have emerged in the past decade.
Collapse
Affiliation(s)
- Nadezhda Tikhmyanova
- Fox Chase Cancer Center, 333 Cottman Ave., Philadelphia, PA 19111 USA
- Department of Biochemistry, Drexel University Medical School, Philadelphia, PA 19102 USA
| | - Joy L. Little
- Fox Chase Cancer Center, 333 Cottman Ave., Philadelphia, PA 19111 USA
| | - Erica A. Golemis
- Fox Chase Cancer Center, 333 Cottman Ave., Philadelphia, PA 19111 USA
| |
Collapse
|
14
|
Uliczka F, Kornprobst T, Eitel J, Schneider D, Dersch P. Cell invasion of Yersinia pseudotuberculosis by invasin and YadA requires protein kinase C, phospholipase C-gamma1 and Akt kinase. Cell Microbiol 2009; 11:1782-801. [PMID: 19681907 DOI: 10.1111/j.1462-5822.2009.01371.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The outer membrane proteins YadA and invasin of Yersinia pseudotuberculosis promote invasion into mammalian cells through beta(1)-integrins and trigger the production of interleukin (IL)-8. FAK, c-Src and the PI3 kinase were previously found to be important for both YadA- and invasin-promoted uptake. Here, we demonstrate that two different downstream effectors of PI3 kinase, Akt and phospholipase Cgamma1 are required for efficient cell invasion. Inhibition of Akt or phospholipase C-gamma (PLC-gamma)1 by pharmaceutical agents as well as reduced expression of the isoforms Akt1 and Akt2, and of PLC-gamma1 by RNA interference decreased entry of YadA- and Inv-expressing bacteria significantly. In addition, we report that the conventional protein kinases C (PKC)alpha and -beta, positioned downstream of PLC-gamma1, are activated upon Inv- or YadA-promoted cell entry. They colocalize with intracellular bacteria and their depletion by siRNA treatment also resulted in a strong reduction of cell entry. In contrast, neither Akt nor PLC-gamma1, and the PKCs are essential for YadA- and Inv-mediated IL-8 synthesis and release. We conclude that YadA and invasin of Y. pseudotuberculosis both trigger similar signal transduction pathways during integrin-mediated phagocytosis into epithelial cells, which lead to the activation of Akt, PLC-gamma1, PKCalpha and -beta downstream of PI3 kinase, separate from the MAPK-dependent pathway that triggers IL-8 production.
Collapse
Affiliation(s)
- Frank Uliczka
- Institut für Mikrobiologie, Technische Universität Braunschweig, 38106 Braunschweig, Germany
| | | | | | | | | |
Collapse
|
15
|
Birge RB, Kalodimos C, Inagaki F, Tanaka S. Crk and CrkL adaptor proteins: networks for physiological and pathological signaling. Cell Commun Signal 2009; 7:13. [PMID: 19426560 PMCID: PMC2689226 DOI: 10.1186/1478-811x-7-13] [Citation(s) in RCA: 216] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2009] [Accepted: 05/10/2009] [Indexed: 01/24/2023] Open
Abstract
The Crk adaptor proteins (Crk and CrkL) constitute an integral part of a network of essential signal transduction pathways in humans and other organisms that act as major convergence points in tyrosine kinase signaling. Crk proteins integrate signals from a wide variety of sources, including growth factors, extracellular matrix molecules, bacterial pathogens, and apoptotic cells. Mounting evidence indicates that dysregulation of Crk proteins is associated with human diseases, including cancer and susceptibility to pathogen infections. Recent structural work has identified new and unusual insights into the regulation of Crk proteins, providing a rationale for how Crk can sense diverse signals and produce a myriad of biological responses.
Collapse
Affiliation(s)
- Raymond B Birge
- Department of Biochemistry & Molecular Biology, UMDNJ-New Jersey Medical School, 185 South Orange Ave, Newark, NJ 07103, USA.
| | | | | | | |
Collapse
|
16
|
Hallé M, Gomez MA, Stuible M, Shimizu H, McMaster WR, Olivier M, Tremblay ML. The Leishmania surface protease GP63 cleaves multiple intracellular proteins and actively participates in p38 mitogen-activated protein kinase inactivation. J Biol Chem 2008; 284:6893-908. [PMID: 19064994 DOI: 10.1074/jbc.m805861200] [Citation(s) in RCA: 102] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The Leishmania parasite is a widespread disease threat in tropical areas, causing symptoms ranging from skin lesions to death. Leishmania parasites typically invade macrophages but are also capable of infecting fibroblasts, which may serve as a reservoir for recurrent infection. Invasion by intracellular pathogens often involves exploitation of the host cell cytoskeletal and signaling machinery. Here we have observed a dramatic rearrangement of the actin cytoskeleton and marked modifications in the profile of protein tyrosine phosphorylation in fibroblasts infected with Leishmania major. Correspondingly, exposure to L. major resulted in degradation of the phosphorylated adaptor protein p130Cas and the protein-tyrosine phosphatase-PEST. Cellular and in vitro assays using pharmacological protease inhibitors, recombinant enzyme, and genetically modified strains of L. major identified the parasite protease GP63 as the principal catalyst of proteolysis during infection. A number of additional signaling proteins were screened for degradation during L. major infection as follows: a small subset was cleaved, including cortactin, T-cell protein-tyrosine phosphatase, and caspase-3, but the majority remained unaffected. Protein degradation occurred in cells incubated with Leishmania extracts in the absence of intact parasites, suggesting a mechanism permitting transfer of functional GP63 into the intracellular space. Finally, we evaluated the impact of Leishmania on MAPK signaling; unlike p44/42 and JNK, p38 was inactivated upon infection in a GP63- and protein degradation-dependent manner, which likely involves cleavage of the upstream adaptor TAB1. Our results establish that GP63 plays a central role in a number of hostcell molecular events that likely contribute to the infectivity of Leishmania.
Collapse
Affiliation(s)
- Maxime Hallé
- Rosalind and Morris Goodman Cancer Centre, Department of Biochemistry, McGill University, Montréal, Québec H3A 1A3, Canada
| | | | | | | | | | | | | |
Collapse
|
17
|
Abelson tyrosine kinase facilitates Salmonella enterica serovar Typhimurium entry into epithelial cells. Infect Immun 2008; 77:60-9. [PMID: 18936177 DOI: 10.1128/iai.00639-08] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The intracellular gram-negative bacterial pathogen Salmonella enterica serovar Typhimurium gains entry into nonphagocytic cells by manipulating the assembly of the host actin cytoskeleton. S. enterica serovar Typhimurium entry requires a functional type III secretion system, a conduit through which bacterial effector proteins are directly translocated into the host cytosol. We and others have previously reported the enhancement of tyrosine kinase activities during Salmonella serovar Typhimurium infection; however, neither specific kinases nor their targets have been well characterized. In this study, we investigated the roles of the cellular Abelson tyrosine kinase (c-Abl) and the related protein Arg in the context of serovar Typhimurium infection. We found that bacterial internalization was inhibited by more than 70% in cells lacking both c-Abl and Arg and that treatment of wild-type cells with a pharmaceutical inhibitor of the c-Abl kinase, STI571 (imatinib), reduced serovar Typhimurium invasion efficiency to a similar extent. Bacterial infection led to enhanced phosphorylation of two previously identified c-Abl substrates, the adaptor protein CT10 regulator of kinase (CrkII) and the Abelson-interacting protein Abi1, a component of the WAVE2 complex. Furthermore, overexpression of the nonphosphorylatable form of CrkII resulted in decreased invasion. Taken together, these findings indicate that c-Abl is activated during S. enterica serovar Typhimurium infection and that its phosphorylation of multiple downstream targets is functionally important in bacterial internalization.
Collapse
|
18
|
Velasquez Almonacid LA, Tafuri S, Dipineto L, Matteoli G, Fiorillo E, Della Morte R, Fioretti A, Menna LF, Staiano N. Role of connexin-43 hemichannels in the pathogenesis of Yersinia enterocolitica. Vet J 2008; 182:452-7. [PMID: 18824377 DOI: 10.1016/j.tvjl.2008.08.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2008] [Revised: 07/10/2008] [Accepted: 08/07/2008] [Indexed: 01/09/2023]
Abstract
Connexin (Cx) channels are sites of cytoplasmic communication between contacting cells. Evidence indicates that the opening of hemichannels occurs under both physiological and pathological conditions. In this paper, the involvement of Cx-43 hemichannels is demonstrated in the pathogenesis of Yersinia. Parental HeLa cells and transfected HeLa cells stably expressing Cx-43 (HCx43) were infected with Yersiniaenterocolitica, and bacterial uptake was measured by the colony-forming unit method. Bacterial uptake was higher in HCx43 cells than in parental cells and was inhibited by the Cx channel blocker, 18-alpha-glycyrrhetinic acid (AGA). The inhibitory effect of AGA was more pronounced on the Y. enterocolitica uptake by HCx43 cells than by parental cells. The ability of HCx43 cells to incorporate the permeable fluorescent tracer Lucifer Yellow (LY) was assessed. Dye incorporation was inhibited by AGA, whereas Y. enterocolitica infection of HCx43 cells increased LY incorporation. Western blotting analysis demonstrated that Y. enterocolitica infection of HCx43 cells induced tyrosine phosphorylation of Cx-43, thus supporting a critical role for Cx-43 in the strategies exploited by bacterial pathogens to invade non-phagocytic cells.
Collapse
Affiliation(s)
- L A Velasquez Almonacid
- Dipartimento di Patologia e Sanità Animale, Università di Napoli Federico II, via F. Delpino 1, 80137 Napoli, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Abstract
Bacterial pathogens utilize toxins to modify or kill host cells. The bacterial ADP-ribosyltransferases are a family of protein toxins that covalently transfer the ADP-ribose portion of NAD to host proteins. Each bacterial ADP-ribosyltransferase toxin modifies a specific host protein(s) that yields a unique pathology. These toxins possess the capacity to enter a host cell or to use a bacterial Type III apparatus for delivery into the host cell. Advances in our understanding of bacterial toxin action parallel the development of biophysical and structural biology as well as our understanding of the mammalian cell. Bacterial toxins have been utilized as vaccines, as tools to dissect host cell physiology, and more recently for the development of novel therapies to treat human disease.
Collapse
Affiliation(s)
- Qing Deng
- Department of Microbiology and Molecular Genetics, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, USA.
| | | |
Collapse
|
20
|
Abstract
Cell adhesion, migration and the maintenance of cell polarity are all processes that depend on the correct targeting of integrins and the dynamic remodelling of integrin-containing adhesion sites. The importance of the endo/exocytic cycle of integrins as a key regulator of these functions is increasingly recognized. Several recent publications have provided mechanistic insight into how integrin traffic is regulated in cells. Increasing evidence suggests that small GTPases such as Arf6 and members of the Rab family control integrin internalization and recycling back to the plasma membrane along microtubules. The fine tuning of these trafficking events seems to be mediated by specific guanine-nucleotide-exchange factors (GEFs) and GTPase-activating proteins (GAPs). In addition, several kinases regulate integrin traffic. The identification of their substrates has demonstrated how these kinases regulate integrin traffic by controlling small GTPases or stabilizing cytoskeletal tracks that are crucial for efficient traffic of integrins to the plasma membrane.
Collapse
|
21
|
Wong KW, Mohammadi S, Isberg RR. Disruption of RhoGDI and RhoA regulation by a Rac1 specificity switch mutant. J Biol Chem 2006; 281:40379-88. [PMID: 17074770 DOI: 10.1074/jbc.m605387200] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Rho family GTPases are important regulators of the actin cytoskeleton. Activation of these proteins can be promoted by guanine nucleotide exchange factors containing Dbl and Pleckstrin homology domains resulting in membrane insertion of a Rho family member, whereas the inactive GDP-bound form is sequestered primarily in the cytoplasm, bound to the guanosine dissociation inhibitor RhoGDI. Dominant interfering variants of Rac1, but not Cdc42, inhibit beta1 integrin-promoted uptake of Yersinia pseudotuberculosis. Unexpectedly, we found that the Rac1(W56F) guanine nucleotide exchange factors specificity switch mutant blocked invasin-promoted uptake as well as Cdc42-dependent uptake of enteropathogenic Escherichia coli. Fluorescence resonance energy transfer experiments demonstrated that Rac1(W56F) retained the ability to be loaded with GTP, bind a downstream effector, and interact with RhoGDI. Mutational analyses of intragenic suppressors and coexpression studies demonstrated that binding of the Rac1(W56F) mutant to RhoGDI appeared to play a role in the inhibition of uptake. As RhoGDI inhibits RhoA, overactivation of RhoA may account for the uptake interference caused by Rac1(W56F). Consistent with this model, a dominant interfering form of RhoA restored significant uptake in the presence of the Rac1(W56F) mutant but had no effect on another interfering Rac1 form. Furthermore, the cellular GTP-RhoA level was elevated by the presence of Rac1(W56F) mutant protein. These data are consistent with the proposition that Rac1(W56F) blocks invasin-promoted uptake by preventing RhoGDI from inactivating RhoA. We conclude that RhoGDI allows cross-talk between Rho family members that promote potentially antagonistic processes, and disruption of this cross-talk can interfere with invasin-promoted uptake.
Collapse
Affiliation(s)
- Ka-Wing Wong
- Howard Hughes Medical Institute, Department of Molecular Biology and Microbiology, Tufts University Medical School, Boston, Massachusetts 02111, USA
| | | | | |
Collapse
|
22
|
Owen KA, Thomas KS, Bouton AH. The differential expression of Yersinia pseudotuberculosis adhesins determines the requirement for FAK and/or Pyk2 during bacterial phagocytosis by macrophages. Cell Microbiol 2006; 9:596-609. [PMID: 16987330 DOI: 10.1111/j.1462-5822.2006.00811.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Phagocytosis of Yersinia pseudotuberculosis by macrophages is initiated by interactions between host cell integrin receptors and the bacterial adhesins, invasin and YadA. Two non-receptor protein tyrosine kinases, FAK and Pyk2, have been implicated in this process. In this study, we investigated the mechanisms of activation and functional requirements for these kinases during phagocytosis. A panel of Yersinia strains that differentially express invasin and YadA were used to infect cells in which FAK and/or Pyk2 expression was reduced by RNA interference. Bacterial strains that simultaneously express invasin and YadA activated FAK and Pyk2 signalling pathways that perform non-redundant functions required for Yersinia internalization. In contrast, FAK activation was found to be sufficient for phagocytosis of bacteria expressing invasin alone, and Pyk2 activation was sufficient when YadA was expressed in the absence of invasin. Based on these data, we suggest that the activation states of FAK and Pyk2, as well as the subsequent signalling events that lead to phagocytosis, are differentially regulated through the unique mechanisms of integrin engagement utilized by invasin and YadA. These findings lend insight into the molecular events that control bacterial phagocytosis as well as other integrin-based processes such as cell adhesion and migration.
Collapse
Affiliation(s)
- Katherine A Owen
- Department of Microbiology, Box 800734, University of Virginia Health System, Charlottesville, VA 22908-0734, USA
| | | | | |
Collapse
|
23
|
Shi J, Casanova JE. Invasion of host cells by Salmonella typhimurium requires focal adhesion kinase and p130Cas. Mol Biol Cell 2006; 17:4698-708. [PMID: 16914515 PMCID: PMC1635395 DOI: 10.1091/mbc.e06-06-0492] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Salmonella typhimurium colonizes the intestinal epithelium by injecting an array of effector proteins into host cells that induces phagocytic uptake of attached bacteria. However, the host molecules targeted by these effectors remain poorly defined. Here, we demonstrate that S. typhimurium induces formation of focal adhesion-like complexes at sites of bacterial attachment and that both focal adhesion kinase (FAK) and the scaffolding protein p130Cas are required for Salmonella uptake. Entry of Salmonella into FAK(-/-) cells is dramatically impaired and can be restored to control levels by expression of wild-type FAK. Surprisingly, reconstitution of bacterial internalization requires neither the kinase domain of FAK nor activation of c-Src, but does require a C-terminal PXXP motif through which FAK interacts with Cas. Infection of Cas(-/-) cells is also impaired, and reconstitution of invasiveness requires the central Cas YXXP repeat domain. The invasion defect in Cas(-/-) cells can be suppressed by overexpression of FAK, suggesting a functional link between FAK and Cas in the regulation of Salmonella invasion. Together, these findings reveal a novel role for focal adhesion proteins in the invasion of host cells by Salmonella.
Collapse
Affiliation(s)
- Jing Shi
- Department of Cell Biology, University of Virginia Health Sciences Center, Charlottesville, VA 22908-0732
| | - James E. Casanova
- Department of Cell Biology, University of Virginia Health Sciences Center, Charlottesville, VA 22908-0732
| |
Collapse
|
24
|
Schornberg K, Matsuyama S, Kabsch K, Delos S, Bouton A, White J. Role of endosomal cathepsins in entry mediated by the Ebola virus glycoprotein. J Virol 2006; 80:4174-8. [PMID: 16571833 PMCID: PMC1440424 DOI: 10.1128/jvi.80.8.4174-4178.2006] [Citation(s) in RCA: 354] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Using chemical inhibitors and small interfering RNA (siRNA), we have confirmed roles for cathepsin B (CatB) and cathepsin L (CatL) in Ebola virus glycoprotein (GP)-mediated infection. Treatment of Ebola virus GP pseudovirions with CatB and CatL converts GP1 from a 130-kDa to a 19-kDa species. Virus with 19-kDa GP1 displays significantly enhanced infection and is largely resistant to the effects of the CatB inhibitor and siRNA, but it still requires a low-pH-dependent endosomal/lysosomal function. These and other results support a model in which CatB and CatL prime GP by generating a 19-kDa intermediate that can be acted upon by an as yet unidentified endosomal/lysosomal enzyme to trigger fusion.
Collapse
Affiliation(s)
- Kathryn Schornberg
- Department of Microbiology, University of Virginia, 1300 Jefferson Park Ave., Charlottesville, Virginia 22908-0734, USA
| | | | | | | | | | | |
Collapse
|
25
|
Viboud GI, Bliska JB. Yersinia outer proteins: role in modulation of host cell signaling responses and pathogenesis. Annu Rev Microbiol 2006; 59:69-89. [PMID: 15847602 DOI: 10.1146/annurev.micro.59.030804.121320] [Citation(s) in RCA: 459] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
A type III secretion system (TTSS) is encoded on a virulence plasmid that is common to three pathogenic Yersinia species: Y. enterocolitica, Y. pseudotuberculosis, and Y. pestis. Pathogenic Yersinia species require this TTSS to survive and replicate within lymphoid tissues of their animal or human hosts. A set of pathogenicity factors, including those known as Yersinia outer proteins (Yops), is exported by this system upon bacterial infection of host cells. Two translocator Yops (YopB and YopD) insert into the host plasma membrane and function to transport six effector Yops (YopO, YopH, YopM, YopT, YopJ, and YopE) into the cytosol of the host cell. Effector Yops function to counteract multiple signaling responses in the infected host cell. The signaling responses counteracted by Yops are initiated by phagocytic receptors, Toll-like receptors, translocator Yops, and additional mechanisms. Innate and adaptive immune responses are thwarted as a consequence of Yop activities. A biochemical function for each effector Yop has been established, and the importance of these proteins for the pathogenesis process is being elucidated. This review focuses on the biochemical functions of Yops, the signaling pathways they modulate, and the role of these proteins in Yersinia virulence.
Collapse
Affiliation(s)
- Gloria I Viboud
- Department of Molecular Genetics and Microbiology, Center for Infectious Diseases, SUNY Stony Brook, Stony Brook, New York 11794-5222, USA.
| | | |
Collapse
|
26
|
Abstract
Exoenzyme T (ExoT) is a bifunctional type III cytotoxin of Pseudomonas aeruginosa that possesses both Rho GTPase-activating protein and ADP-ribosyltransferase activities. The ADP-ribosyltransferase activity of ExoT stimulated depolymerization of the actin cytoskeleton independent of Rho GTPase-activating protein function, and ExoT was subsequently shown to ADP-ribosylate Crk (CT10 regulator of kinase)-I and Crk-II. Crk proteins are eukaryotic adaptor proteins comprising SH2 and SH3 domains that are components of the integrin signaling pathway leading to Rac1 and Rap1 functions. Mass spectroscopic analysis identified Arg20 as the site of ADP-ribosylation by ExoT. Arg20 is a conserved residue located within the SH2 domain that is required for interactions with upstream signaling molecules such as paxillin and p130cas. Glutathione S-transferase pull-down and far Western assays showed that ADP-ribosylated Crk-I or Crk-I(R20K) failed to bind p130cas or paxillin. This indicates that ADP-ribosylation inhibited the direct interaction of Crk with these focal adhesion proteins. Overexpression of wild-type Crk-I reduced cell rounding by ExoT, whereas expression of dominant-active Rac1 interfered with the ability of ExoT to round cells. Thus, the ADP-ribosylation of Crk uncouples integrin signaling by direct inhibition of the binding of Crk to focal adhesion proteins.
Collapse
Affiliation(s)
- Qing Deng
- Department of Microbiology and Molecular Genetics, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, USA
| | | | | |
Collapse
|
27
|
Hudson KJ, Bliska JB, Bouton AH. Distinct mechanisms of integrin binding by Yersinia pseudotuberculosis adhesins determine the phagocytic response of host macrophages. Cell Microbiol 2005; 7:1474-89. [PMID: 16153246 DOI: 10.1111/j.1462-5822.2005.00571.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The enteropathogenic yersiniae express two outer membrane adhesins, invasin and YadA, that contribute to pathogenesis. While invasin binds directly to beta1 integrin receptors with high affinity, YadA binds indirectly through extracellular matrix (ECM) components. In this study, Yersinia pseudotuberculosis inv and yadA mutants were used to investigate how these distinct binding mechanisms compare and potentially compete in activating signalling pathways and promoting bacterial uptake by host macrophages. The efficiency of adhesin-mediated phagocytic responses was found to be dependent on the relative expression of invasin and YadA on the bacterial surface as well as the expression of ECM proteins in the extracellular milieu. Under conditions of low concentrations of ECM, invasin was found to be the dominant adhesin, promoting high levels of phagocytosis coincident with robust and sustained activation of the protein tyrosine kinases Fak and Pyk2, phosphorylation of the adaptor molecule Cas and activation of the small GTPase Rac1. In the presence of higher concentrations of ECM, YadA became the dominant functional adhesin through its ability to engage integrin receptors via an ECM bridge. We propose a model whereby invasin promotes robust and prolonged activation of phagocytic signalling cascades by inducing a 'high-affinity' integrin conformation as well as integrin clustering. We postulate that YadA-ECM promotes phagocytosis through a more transient activation of signalling cascades that arises from integrin clustering in the context of a cross-linked fibrillar ECM network.
Collapse
Affiliation(s)
- Krischan J Hudson
- Department of Microbiology, University of Virginia Health System, Charlottesville, 22908-0734, USA
| | | | | |
Collapse
|
28
|
Abstract
Molecular machines orchestrate the translocation and entry of pathogens through host cell membranes, in addition to the uptake and release of molecules during endocytosis and exocytosis. Viral cell entry requires a family of glycoproteins, and the structural organization and function of these viral glycoproteins are similar to the SNARE proteins, which are known to be involved in intracellular vesicle fusion, endocytosis and exocytosis. Here, we propose that a family of bacterial membrane proteins that are responsible for cell-mediated adherence and entry resembles the structural architecture of both viral fusion proteins and eukaryotic SNAREs and might therefore share similar, but distinct, mechanisms of cell membrane translocation. Furthermore, we propose that the recurrence of these molecular machines across species indicates that these architectural motifs were evolutionarily selected because they provided the best solution to ensure the survival of pathogens within a particular environment.
Collapse
Affiliation(s)
- Michèle A. Barocchi
- Department of Bioinformatics and Cellular Microbiology, Chiron Vaccines, Via Fiorentina 1, Siena, 53100 Italy
| | - Vega Masignani
- Department of Bioinformatics and Cellular Microbiology, Chiron Vaccines, Via Fiorentina 1, Siena, 53100 Italy
| | - Rino Rappuoli
- Department of Bioinformatics and Cellular Microbiology, Chiron Vaccines, Via Fiorentina 1, Siena, 53100 Italy
| |
Collapse
|
29
|
Mogemark L, McGee K, Yuan M, Deleuil F, Fällman M. Disruption of target cell adhesion structures by the Yersinia effector YopH requires interaction with the substrate domain of p130Cas. Eur J Cell Biol 2005; 84:477-89. [PMID: 15900707 DOI: 10.1016/j.ejcb.2004.11.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The docking protein p130Cas has, together with FAK, been found as a target of the Yersinia virulence effector YopH. YopH is a protein tyrosine phosphatase that is delivered into host cells via the bacterial type III secretion machinery, and the outcome of its activity is inhibition of host cell phagocytosis. In the present study using p130Cas-/- cells, and p130Cas-/- cells expressing variants of GFPp130Cas, we show that this docking protein, via its substrate domain, is responsible for subcellular targeting of YopH in eukaryotic cells. Since YopH inhibits phagocytosis, p130Cas was expected to be critical for signalling mediating bacterial internalization. However, p130Cas-/- cells did not exhibit reduced capacity to internalize Yersinia. On the other hand, when a dominant negative variant of p130Cas was expressed in these cells, the phagocytic capacity was severely impaired. Moreover, the p130Cas-/- cells displayed a marked reduced sensitivity towards YopH-mediated detachment compared to wild-type cells. Transfecting these cells with full-length p130Cas rendered cells hypersensitive to both mechanical and Yersinia-mediated detachment. This hypersensitivity was not seen upon transfection with the dominant negative substrate domain-deleted variant of p130Cas. This implicates p130Cas as a prominent regulator of cell adhesion, where its substrate-binding domain has a significant function.
Collapse
Affiliation(s)
- Lena Mogemark
- Department of Molecular Biology, Umeå University, S-901 87 Umeå, Sweden
| | | | | | | | | |
Collapse
|
30
|
Yuan M, Mogemark L, Fällman M. Fyn binding protein, Fyb, interacts with mammalian actin binding protein, mAbp1. FEBS Lett 2005; 579:2339-47. [PMID: 15848169 DOI: 10.1016/j.febslet.2005.03.031] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2004] [Revised: 03/07/2005] [Accepted: 03/09/2005] [Indexed: 11/20/2022]
Abstract
The immune cell specific protein Fyn-T binding protein (Fyb) has been identified as a target of the Yersinia antiphagocytic effector Yersinia outer protein H (YopH), but its role in macrophages is unknown. By using Fyb domains as bait to screen a mouse lymphoma cDNA library, we identified a novel interaction partner, mammalian actin binding protein 1 (mAbp1). We show that mAbp1 binds the Fyb N-terminal via its C-terminally located src homology 3 domain. The interaction between Fyb and mAbp1 is detected in macrophage lysates and the proteins co-localize with F-actin in the leading edge. Hence, mAbp1 is likely to constitute a downstream effector of Fyb involved in F-actin dynamics.
Collapse
Affiliation(s)
- Ming Yuan
- Department of Molecular Biology, Umeå University, Sweden
| | | | | |
Collapse
|
31
|
Fällman M, Gustavsson A. Cellular mechanisms of bacterial internalization counteracted by Yersinia. INTERNATIONAL REVIEW OF CYTOLOGY 2005; 246:135-88. [PMID: 16164968 DOI: 10.1016/s0074-7696(05)46004-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Upon host-cell contact, human pathogenic Yersinia species inject Yop virulence effectors into the host through a Type III secretion-and-translocation system. These virulence effectors cause a block in phagocytosis (YopE, YopT, YpkA, and YopH) and suppression of inflammatory mediators (YopJ). The Yops that block phagocytosis either interfere with the host cell actin regulation of Rho GTPases (YopE, YopT, and YpkA) or specifically and rapidly inactivate host proteins involved in signaling from the receptor to actin (YopH). The block in uptake has been shown to be activated following binding to Fc, Complement, and beta1-integrin receptors in virtually any kind of host cell. Thus, the use of Yersinia as a model system to study Yersinia-host cell interactions provides a good tool to explore signaling pathways involved in phagocytosis.
Collapse
Affiliation(s)
- Maria Fällman
- Department of Molecular Biology, Umeå University, SE-90187 Umeå, Sweden
| | | |
Collapse
|
32
|
Rottner K, Lommel S, Wehland J, Stradal TEB. Pathogen-induced actin filament rearrangement in infectious diseases. J Pathol 2004; 204:396-406. [PMID: 15495265 DOI: 10.1002/path.1638] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Host defence mechanisms involve the establishment and maintenance of numerous barriers to infectious microbes, including skin and mucosal surfaces, connective tissues, and a sophisticated immune system to detect and destroy invaders. Defeating these defence mechanisms and breaching the cell membrane barrier is the ultimate challenge for most pathogens. By invading the host and, moreover, by penetrating into individual host cells, pathogens gain access to a protective niche, not only to avoid immune clearance, but also to replicate and to disseminate from cell to cell within the infected host. Many pathogens are accomplishing these challenges by exploiting the actin cytoskeleton in a highly sophisticated manner as a result of having evolved common as well as unique strategies.
Collapse
Affiliation(s)
- Klemens Rottner
- Cytoskeleton Dynamics Group, German Research Centre for Biotechnology (GBF), Mascheroder Weg 1, D-38124 Braunschweig, Germany
| | | | | | | |
Collapse
|
33
|
Abstract
Cell adhesion molecules, such as integrins, cadherins, the immunoglobulin superfamily of cell adhesion molecules and selectins, play important structural roles and are involved in various signal transduction processes. As an initial step in the infectious process, many bacterial pathogens adhere to cell adhesion molecules as a means of exploiting the underlying signaling pathways, entering into host cells or establishing extracellular persistence. Often, bacteria are able to bind to cell adhesion molecules by mimicking or acting in place of host cell receptors or their ligands. Recent studies have contributed to our understanding of bacterial adherence mechanisms and the consequences of receptor engagement; they have also highlighted alternative functions of cell adhesion molecules.
Collapse
Affiliation(s)
- Erin C Boyle
- Department of Microbiology and Immunology, Biotechnology Laboratory, Wesbrook Building, Room 237, 6174 University Boulevard, University of British Columbia, Vancouver, British Columbia, Canada V6T 1Z3.
| | | |
Collapse
|
34
|
Sundin C, Hallberg B, Forsberg Ã. ADP-ribosylation by exoenzyme T ofPseudomonas aeruginosainduces an irreversible effect on the host cell cytoskeleton in vivo. FEMS Microbiol Lett 2004. [DOI: 10.1111/j.1574-6968.2004.tb09517.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
35
|
Forsberg M, Druid P, Zheng L, Stendahl O, Särndahl E. Activation of Rac2 and Cdc42 on Fc and complement receptor ligation in human neutrophils. J Leukoc Biol 2003; 74:611-9. [PMID: 12960248 DOI: 10.1189/jlb.1102525] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Phagocytosis is a complex process engaging a concerted action of signal-transduction cascades that leads to ingestion, subsequent phagolysosome fusion, and oxidative activation. We have previously shown that in human neutrophils, C3bi-mediated phagocytosis elicits a significant oxidative response, suggesting that activation of the small GTPase Rac is involved in this process. This is contradictory to macrophages, where only Fc receptor for immunoglobulin G (FcgammaR)-mediated activation is Rac-dependent. The present study shows that engagement of the complement receptor 3 (CR3) and FcgammaR and CR3- and FcgammaR-mediated phagocytosis activates Rac, as well as Cdc42. Furthermore, following receptor-engagement of the CR3 or FcgammaRs, a downstream target of these small GTPases, p21-activated kinase, becomes phosphorylated, and Rac2 is translocated to the membrane fraction. Using the methyltransferase inhibitors N-acetyl-S-farnesyl-L-cysteine and N-acetyl-S-geranylgeranyl-L-cysteine, we found that the phagocytic uptake of bacteria was not Rac2- or Cdc42-dependent, whereas the oxidative activation was decreased. In conclusion, our results indicate that in neutrophils, Rac2 and Cdc42 are involved in FcR- and CR3-induced activation and for properly functioning signal transduction involved in the generation of oxygen radicals.
Collapse
Affiliation(s)
- Maria Forsberg
- Department of Cell Biology, Faculty of Health Sciences, Linköping University, Sweden
| | | | | | | | | |
Collapse
|
36
|
Sun J, Barbieri JT. Pseudomonas aeruginosa ExoT ADP-ribosylates CT10 regulator of kinase (Crk) proteins. J Biol Chem 2003; 278:32794-800. [PMID: 12807879 DOI: 10.1074/jbc.m304290200] [Citation(s) in RCA: 123] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Pseudomonas aeruginosa ExoT is a type III cytotoxin that functions as an anti-internalization factor with an N-terminal RhoGAP domain and a C-terminal ADP-ribosyltransferase domain. Although ExoT RhoGAP stimulates actin reorganization through the inactivation of Rho, Rac, and Cdc42, the function of the ADP-ribosylation domain is unknown. The present study characterized the mammalian proteins that are ADP-ribosylated by ExoT, using two-dimensional SDS-PAGE and matrix-assisted laser desorption ionization/time of flight (MALDI-TOF) analysis. ExoT ADP-ribosylated two cytosolic proteins in cell lysates upon type III delivery into cultured HeLa cells. MALDI-TOF mass spectrometry analysis identified the two proteins as Crk-I and Crk-II that are Src homology 2-3 domains containing adaptor proteins, which mediate signal pathways involving focal adhesion and phagocytosis. ExoT ADP-ribosylated recombinant Crk-I at a rate similar to the ADP-ribosylation of soybean trypsin inhibitor by ExoS. ExoS did not ADP-ribosylate Crk-I. ADP-ribosylation of Crk-I may be responsible for the anti-phagocytosis phenotype elicited by ExoT in mammalian cells.
Collapse
Affiliation(s)
- Jianjun Sun
- Department of Microbiology and Molecular Genetics, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, USA
| | | |
Collapse
|
37
|
McGee K, Holmfeldt P, Fällman M. Microtubule-dependent regulation of Rho GTPases during internalisation of Yersinia pseudotuberculosis. FEBS Lett 2003; 533:35-41. [PMID: 12505155 DOI: 10.1016/s0014-5793(02)03745-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Internalisation of the human pathogen Yersinia pseudotuberculosis via interaction of bacterial invasin with host beta1 integrins depends on the actin cytoskeleton and involves Src family kinases, focal adhesion kinase, p130Crk-associated substrate, proline-rich tyrosine kinase 2, Rac, Arp 2/3 complex and WASP family members. We show here that Rho GTPases are regulated by the microtubule system during bacterial uptake. Interfering with microtubule organisation using nocodazole or paclitaxel suppressed uptake by HeLa cells. The nocodazole effect on microtubule depolymerisation was partially inhibited through overexpression of Rac, Cdc42, RhoG or RhoA and completely prevented by expression of Vav2. This suggests that microtubules influence Rho GTPases during invasin-mediated phagocytosis and in the absence of functional microtubules Vav2 can mimic their effect on one, or more, of the Rho family GTPases. Lastly, overexpression of p50 dynamitin partially inhibited bacterial uptake and this effect was also blocked by co-expression of Vav2, thus further implicating this guanine nucleotide exchange factor in activating Rho GTPases for internalisation during loss of microtubule function.
Collapse
Affiliation(s)
- Karen McGee
- Department of Molecular Biology, Umeå University, 901 87, Umeå, Sweden
| | | | | |
Collapse
|
38
|
Evans DJ, Kuo TC, Kwong M, Van R, Fleiszig SMJ. Mutation of csk, encoding the C-terminal Src kinase, reduces Pseudomonas aeruginosa internalization by mammalian cells and enhances bacterial cytotoxicity. Microb Pathog 2002; 33:135-43. [PMID: 12220990 DOI: 10.1006/mpat.2002.0521] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Clinical isolates of Pseudomonas aeruginosa are either invasive or cytotoxic towards mammalian epithelial cells, endothelial cells, and macrophages. Invasion requires host cell actin cytoskeleton function, and ExsA-regulated proteins of P. aeruginosa that inhibit invasion (ExoS and ExoT) can disrupt the cytoskeleton. Another ExsA regulated protein, ExoU, is involved in the cytotoxic activity of cytotoxic strains. Src-family kinases are thought to participate in the regulation of cytoskeleton function. Recent studies have suggested that Src-family tyrosine kinases, p60-Src and p59-Fyn, are activated during P. aeruginosa invasion. Using fibroblasts homozygous for mutation of csk (-/-), we tested the hypothesis that mutation of csk, encoding a negative regulator of Src-family tyrosine kinases, would be important in P. aeruginosa invasion and cytotoxicity. Mutation of csk was found to reduce invasion by approximately 8-fold, without reducing bacterial adherence to cells (P=0.0001). Conversely, csk (-/-) cells were approximately 5-fold more susceptible to ExoU-dependent cytotoxicity (P=0.024), which was accompanied by a small increase in ExsA-regulated adherence. ExoT-dependent invasion inhibitory activity of cytotoxic P. aeruginosa was attenuated in csk (-/-) cells as compared to normal fibroblasts. These data show that fibroblasts, like epithelial cells, are susceptible to P. aeruginosa invasion and cytotoxicity. They also show a role for Csk in P. aeruginosa invasion, while providing further evidence that actin cytoskeleton disruption contributes to ExsA-regulated P. aeruginosa cytotoxicity and invasion inhibition.
Collapse
Affiliation(s)
- David J Evans
- Morton D. Sarver Laboratory for Cornea and Contact Lens Research, School of Optometry, University of California at Berkeley, Berkeley, California 94720, USA
| | | | | | | | | |
Collapse
|
39
|
Bruce-Staskal PJ, Weidow CL, Gibson JJ, Bouton AH. Cas, Fak and Pyk2 function in diverse signaling cascades to promote Yersinia uptake. J Cell Sci 2002; 115:2689-700. [PMID: 12077360 DOI: 10.1242/jcs.115.13.2689] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The interplay between pathogen-encoded virulence factors and host cell signaling networks is critical for both the establishment and clearance of microbial infections. Yersinia uptake into host cells serves as an in vitro model for exploring how host cells respond to Yersinia adherence. In this study, we provide insight into the molecular nature and regulation of signaling networks that contribute to the uptake process. Using a reconstitution approach in Fak-/- fibroblasts, we have been able to specifically address the interplay between Fak, Cas and Pyk2 in this process. We show that both Fak and Cas play roles in the Yersinia uptake process and that Cas can function in a novel pathway that is independent of Fak. Fak-dependent Yersinia uptake does not appear to involve Cas-Crk signaling. By contrast, Cas-mediated uptake in the absence of Fak requires Crk as well as the protein tyrosine kinases Pyk2 and Src. In spite of these differences, the requirement for Rac1 activity is a common feature of both pathways. Furthermore, blocking the function of either Fak or Cas induces similar morphological defects in Yersinia internalization, which are manifested by incomplete membrane protrusive activity that is consistent with an inhibition of Rac1 activity. Pyk2 also functions in Yersinia uptake by macrophages, which are physiologically important for clearing Yersinia infections. Taken together, these data provide new insight into the host cellular signaling networks that are initiated upon infection with Y. pseudotuberculosis. Importantly, these findings also contribute to a better understanding of other cellular processes that involve actin remodeling, including the host response to other microbial pathogens, cell adhesion and migration.
Collapse
Affiliation(s)
- Pamela J Bruce-Staskal
- Department of Microbiology and Cancer Center, University of Virginia Health System, Charlottesville, VA 22908-0734, USA
| | | | | | | |
Collapse
|
40
|
Abstract
Phagocytosis constitutes the primary line of host innate and adaptive defence against incoming microbial pathogens, providing an efficient means for their removal and destruction. However, several virulent bacteria that do not function as intracellular pathogens have evolved mechanisms to avoid and prevent phagocytosis that constitute an essential part of their pathogenic capacity. Some of these mechanisms include preventing recognition by phagocytic receptors or blocking uptake by professional phagocytes. Recently, the molecular mechanisms of such antiphagocytic properties have been elucidated for some pathogens. Such mechanisms illustrate the diversity of mechanisms bacterial pathogens use to avoid phagocytic uptake.
Collapse
Affiliation(s)
- Jean Celli
- Biotechnology Laboratory, University of British Columbia, Room 237, 6174 University Boulevard, Vancouver, BC, Canada V6T 1Z3.
| | | |
Collapse
|
41
|
Abstract
Enteropathogenic species of the genus Yersinia penetrate the intestinal epithelium and then spread to the lymphatic system, where they proliferate extracellularly. At this location, most other bacteria are effectively ingested and destroyed by the resident phagocytes. Yersinia, on the other hand binds to receptors on the external surface of phagocytes, and from this location it blocks the capacity of these cells to exert their phagocytic function via different receptors. The mechanism behind the resistance to phagocytosis involves the essential virulence factor YopH, a protein tyrosine phosphatase that is translocated into interacting target cells via a type III secretion machinery. YopH disrupts peripheral focal complexes of host cells, seen as a rounding up of infected cells. The focal complex proteins that are dephosphorylated by YopH are focal adhesion kinase and Crk-associated substrate, the latter of which is a common substrate in both professional and non-professional phagocytes. In macrophages additional substrates have been found, the Fyn-binding/SLP-76-associated protein and SKAP-HOM. Phagocytosis is a rapid process that is activated when the bacterium interacts with the phagocyte. Consequently, the effect exerted by a microbe to block this process has to be rapid and precise. This review deals with the mechanisms involved in impeding uptake as well as with the role of the YopH substrates and focal complex structures in normal cell function.
Collapse
Affiliation(s)
- Maria Fällman
- Department of Molecular Biology, Umeå University, Sweden.
| | | | | |
Collapse
|
42
|
Lin M, Zhu MX, Rikihisa Y. Rapid activation of protein tyrosine kinase and phospholipase C-gamma2 and increase in cytosolic free calcium are required by Ehrlichia chaffeensis for internalization and growth in THP-1 cells. Infect Immun 2002; 70:889-98. [PMID: 11796624 PMCID: PMC127685 DOI: 10.1128/iai.70.2.889-898.2002] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Ehrlichia chaffeensis, a bacterium that cannot survive outside the eukaryotic cell, proliferates exclusively in human monocytes and macrophages. In this study, signaling events required for ehrlichial infection of human monocytic cell line THP-1 were characterized. Entry and proliferation of E. chaffeensis in THP-1 cells were significantly blocked by various inhibitors that can regulate calcium signaling, including 8-(diethylamino)octyl-3,4,5-trimethoxybenzoate and 2-aminoethoxydiphenyl borate (intracellular calcium mobilization inhibitors), verapamil and 1-[beta-[3-(4-methoxyphenyl)propyl]-4-methoxyphenethyl]-1H-imidazole (SKF-96365) (calcium channel inhibitors), neomycin and 1-(6-[[17beta-3-methoxyestra-1,3,5(10)-trien-17-yl]amino]hexyl)-1H-pyrrole-2,5-dione (U-73122) (phospholipase C [PLC] inhibitors), monodansylcadaverine (a transglutaminase [TGase] inhibitor), and genistein (a protein tyrosine kinase [PTK] inhibitor). Addition of E. chaffeensis resulted in rapid increases in the level of inositol 1,4,5-trisphosphate (IP(3)) and the level of cytosolic free calcium ([Ca(2+)](i)) in THP-1 cells, which were prevented by pretreatment of THP-1 cells with inhibitors of TGase, PTK, and PLC. E. chaffeensis induced rapid tyrosine phosphorylation of PLC-gamma2, and the presence of a PLC-gamma2 antisense oligonucleotide in THP-1 cells significantly blocked ehrlichial infection. Furthermore, tyrosine-phosphorylated proteins and PLC-gamma2 were colocalized with ehrlichial inclusions, as determined by double-immunofluorescence labeling. The heat-sensitive component of viable E. chaffeensis cells was essential for these signaling events. E. chaffeensis, therefore, can recruit interacting signal-transducing molecules and induce the following signaling events required for the establishment of infection in host cells: protein cross-linking by TGase, tyrosine phosphorylation, PLC-gamma2 activation, IP(3) production, and an increase in [Ca(2+)](i).
Collapse
Affiliation(s)
- Mingqun Lin
- Department of Veterinary Biosciences. Neurobiotechnology Center, The Ohio State University, Columbus, Ohio 43210, USA
| | | | | |
Collapse
|
43
|
Martinez JJ, Hultgren SJ. Requirement of Rho-family GTPases in the invasion of Type 1-piliated uropathogenic Escherichia coli. Cell Microbiol 2002; 4:19-28. [PMID: 11856170 DOI: 10.1046/j.1462-5822.2002.00166.x] [Citation(s) in RCA: 110] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Bladder infections caused by uropathogenic Escherichia coli (UPEC) depends on the ability of E. coli to express type 1 pili. The adhesive component of the pilus, FimH, mediates the invasion of E. coli into the bladder epithelium, a mechanism that facilitates the survival and persistence of E. coli in the bladder. The invasion mechanism requires actin polymerization, focal adhesion kinase phosphorylation and PI 3-kinase activation as well as the formation of FAK/PI 3-kinase and downstream vinculin/alpha-actinin complexes. In this study, we report a role for Rho-GTPase family members, namely RhoA, Cdc42 and Rac1, in the invasion process. Internalization of type 1-piliated E. coli (fimH+) and FimH-coated micro-spheres was inhibited by compactin, a pan-Rho-GTPase inhibitor and dominant negative isoforms of Rac1 and Cdc42. Expression of active Rac1 induced an internalization of E. coli that was insensitive to wortmannin and genistein. Expression of constitutively active Cdc42 induced the formation of FAK/PI 3-kinase and vinculin/alpha-actinin complexes whereas active Rac1 induced only a vinculin/alpha-actinin complex. Taken together, these data suggest that FimH-mediated invasion is dependent on GTP-binding protein activity that involves Cdc42 and PI 3-kinase activation probably upstream of Rac1.
Collapse
Affiliation(s)
- Juan J Martinez
- Department of Molecular Microbiology and Microbial Pathogenesis, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | |
Collapse
|
44
|
Abstract
Phagocytosis of Yersinia pseudotuberculosis occurs through interaction of the bacterial protein invasin with beta1-integrins. Here we report that N-WASP plays a role in internalisation of an invasin-expressing, avirulent strain of Y. pseudotuberculosis. Ectopic expression of N-WASP mutants, which affect recruitment of the Arp2/3 complex to the phagosome, reduces uptake of Yersinia. In addition, expression of the Cdc42/Rac-binding (CRIB) region of N-WASP has an inhibitory effect on uptake. Using GFP-tagged Rho GTPase mutants, we provide evidence that Rac1, but not Cdc42, is important for internalisation. Furthermore, activated Rac1 rescues Toxin B, CRIB and Src family kinase inhibitor PP2-mediated impairment of uptake. Our observations indicate that invasin-mediated phagocytosis occurs via a Src and WASP family-dependent mechanism(s), involving the Arp2/3 complex and Rac, but does not require Cdc42.
Collapse
Affiliation(s)
- K McGee
- Department of Microbiology, Umeå University, Sweden
| | | | | | | |
Collapse
|
45
|
Ruest PJ, Shin NY, Polte TR, Zhang X, Hanks SK. Mechanisms of CAS substrate domain tyrosine phosphorylation by FAK and Src. Mol Cell Biol 2001; 21:7641-52. [PMID: 11604500 PMCID: PMC99935 DOI: 10.1128/mcb.21.22.7641-7652.2001] [Citation(s) in RCA: 127] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Tyrosine phosphorylation of CAS (Crk-associated substrate, p130(Cas)) has been implicated as a key signaling step in integrin control of normal cellular behaviors, including motility, proliferation, and survival. Aberrant CAS tyrosine phosphorylation may contribute to cell transformation by certain oncoproteins, including v-Crk and v-Src, and to tumor growth and metastasis. The CAS substrate domain (SD) contains 15 Tyr-X-X-Pro motifs, which are thought to represent the major tyrosine phosphorylation sites and to function by recruiting downstream signaling effectors, including c-Crk and Nck. CAS makes multiple interactions, direct and indirect, with the tyrosine kinases Src and focal adhesion kinase (FAK), and as a result of this complexity, several plausible models have been proposed for the mechanism of CAS-SD phosphorylation. The objective of this study was to provide experimental tests of these models in order to determine the most likely mechanism(s) of CAS-SD tyrosine phosphorylation by FAK and Src. In vitro kinase assays indicated that FAK has a very poor capacity to phosphorylate CAS-SD, relative to Src. However, FAK expression along with Src was found to be important for achieving high levels of CAS tyrosine phosphorylation in COS-7 cells, as well as recovery of CAS-associated Src activity toward the SD. Structure-functional studies for both FAK and CAS further indicated that FAK plays a major role in regulating CAS-SD phosphorylation by acting as a docking or scaffolding protein to recruit Src to phosphorylate CAS, while a secondary FAK-independent mechanism involves Src directly bound to the CAS Src-binding domain (SBD). Our results do not support models in which FAK either phosphorylates CAS-SD directly or phosphorylates CAS-SBD to promote Src binding to this site.
Collapse
Affiliation(s)
- P J Ruest
- Department of Cell Biology, Vanderbilt University School of Medicine, Nahville, Tennessee 37232, USA
| | | | | | | | | |
Collapse
|
46
|
Abstract
Crk family adaptors are widely expressed and mediate the timely formation of signal transduction protein complexes upon a variety of extracellular stimuli, including various growth and differentiation factors. Selective formation of multi-protein complexes by the Crk and Crk-like (CRKL) proteins depends on specific motifs recognized by their SH2 and SH3 domains. In the case of the first SH3 domains [SH3(1)] a P-x-x-P-x-K motif is crucial for highly selective binding, while the SH2 domains prefer motifs which conform to the consensus pY-x-x-P. Crk family proteins are involved in the relocalization and activation of several different effector proteins which include guanine nucleotide releasing proteins like C3G, protein kinases of the Abl- and GCK-families and small GTPases like Rap1 and Rac. Crk-type proteins have been found not only in vertebrates but also in flies and nematodes. Major insight into the function of Crk within organisms came from the genetic model organism C. elegans, where the Crk-homologue CED-2 regulates cell engulfment and phagocytosis. Other biological outcomes of the Crk-activated signal transduction cascades include the modulation of cell adhesion, cell migration and immune cell responses. Crk family adaptors also appear to play a role in mediating the action of human oncogenes like the leukaemia-inducing Bcr-Abl protein. This review summarizes some key findings and highlights recent insights and open questions.
Collapse
Affiliation(s)
- S M Feller
- Cell Signalling Laboratory, Imperial Cancer Research Fund, University of Oxford, Institute of Molecular Medicine, John Radcliffe Hospital, Headington, Oxford OX3 9DS, UK.
| |
Collapse
|
47
|
Bouton AH, Riggins RB, Bruce-Staskal PJ. Functions of the adapter protein Cas: signal convergence and the determination of cellular responses. Oncogene 2001; 20:6448-58. [PMID: 11607844 DOI: 10.1038/sj.onc.1204785] [Citation(s) in RCA: 157] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Since Cas was first identified as a highly phosphorylated 130 kilodalton protein that associated with the v-Src and v-Crk-oncoproteins, considerable effort has been made to determine its function. Its predicted role as a scaffolding molecule based on its domain structure has been largely confirmed. Through its ability to undergo rapid changes in phosphorylation, subcellular localization and association with heterologous proteins, Cas may spatially and temporally regulate the function of its binding partners. Numerous proteins have been identified that bind to Cas in vitro and/or in vivo, but in only a few cases is there an understanding of how Cas may function in these protein complexes. To date, Cas-Crk and Cas-Src complexes have been most frequently implicated in Cas function, particularly in regards to processes involving regulation of the actin cytoskeleton and proliferation. These and other Cas protein complexes contribute to the critical role of Cas in cell adhesion, migration, proliferation and survival of normal cycling cells. However, under conditions in which these processes are deregulated, Cas appears to play a role in oncogenic transformation and perhaps metastasis. Therefore, in its capacity as an adapter protein, Cas serves as a point of convergence for many distinct signaling inputs, ultimately contributing to the generation of specific cellular responses.
Collapse
Affiliation(s)
- A H Bouton
- Department of Microbiology, University of Virginia School of Medicine, Box 800734, Charlottesville, Virginia VA 22908, USA.
| | | | | |
Collapse
|
48
|
Aepfelbacher M, Heesemann J. Modulation of Rho GTPases and the actin cytoskeleton by Yersinia outer proteins (Yops). Int J Med Microbiol 2001; 291:269-76. [PMID: 11680787 DOI: 10.1078/1438-4221-00130] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Pathogenic species of the genus Yersinia employ a type III secretion apparatus to inject up to six effector proteins (Yersinia outer proteins; Yops) into host cells. Thereby yersiniae disarm the immune cell system of the host to proliferate extracellularly. At least four of the Yop effectors (YopE, YpkA/YopO, YopT and YopH) are involved in the rearrangement of the actin cytoskeleton: YopE, YopT and YpkA/YopO modulate the activity of actin-regulating Rho GTP-binding proteins, whereas YopH dephosphorylates phospho-tyrosine residues in focal adhesion proteins. In this review we will focus on recent evidence implicating Rho GTPases and the actin cytoskeleton as major targets of Yersinia Yops.
Collapse
Affiliation(s)
- M Aepfelbacher
- Max-von-Pettenkofer Institut für Hygiene und Medizinische Mikrobiologie, München, Germany.
| | | |
Collapse
|
49
|
Knodler LA, Celli J, Finlay BB. Pathogenic trickery: deception of host cell processes. Nat Rev Mol Cell Biol 2001; 2:578-88. [PMID: 11483991 DOI: 10.1038/35085062] [Citation(s) in RCA: 129] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Microbial pathogens cause a spectrum of diseases in humans. Although the disease mechanisms vary considerably, most pathogens have developed virulence factors that interact with host molecules, often usurping normal cellular processes, including cytoskeletal dynamics and vesicle targeting. These virulence factors often mimic host molecules, and mediate events as diverse as bacterial invasion, antiphagocytosis, and intracellular parastism.
Collapse
Affiliation(s)
- L A Knodler
- Biotechnology Laboratory, Room 237-6174 University Boulevard, University of British Columbia, Vancouver, British Columbia, Canada, V6T 1Z3
| | | | | |
Collapse
|
50
|
Bruce-Staskal PJ, Bouton AH. PKC-dependent activation of FAK and src induces tyrosine phosphorylation of Cas and formation of Cas-Crk complexes. Exp Cell Res 2001; 264:296-306. [PMID: 11262186 DOI: 10.1006/excr.2000.5137] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
SH-SY5Y neuroblastoma cells are a well-characterized model for studying the induction of neuronal differentiation. TPA treatment of these cells induces cytoskeletal rearrangements that ultimately result in neurite extension. However, the signaling pathways that precede these changes are poorly understood. Other investigators have shown that TPA treatment of SH-SY5Y cells results in increased tyrosine phosphorylation of cytoskeletal-associated proteins, including the adapter protein Cas. In this report, we examine the events upstream and downstream of Cas phosphorylation. We show that TPA treatment induces the PKC-dependent association of tyrosine-phosphorylated Cas with Crk. The activity of two protein tyrosine kinases, Src and FAK, was shown to be necessary and sufficient for TPA-induced Cas phosphorylation. We propose that the PKC-dependent phosphorylation of Cas by Src and FAK promotes the establishment of Cas-Crk complexes and that these interactions may play an important role in regulating the actin cytoskeleton during neuronal differentiation.
Collapse
Affiliation(s)
- P J Bruce-Staskal
- Department of Microbiology, University of Virginia School of Medicine, Charlottesville, Virginia, 22908-0734, USA
| | | |
Collapse
|