1
|
Wang J, Du LF, Zhang MZ, Wei W, Chen ZY, Zhang X, Xiong T, Wang ZF, Xia LY, Jiang JF, Li WJ, Zhu DY, Jia N, Cao WC. Stomach as the target organ of Rickettsia heilongjiangensis infection in C57BL/6 mice identified by click chemistry. Commun Biol 2024; 7:784. [PMID: 38951577 PMCID: PMC11217389 DOI: 10.1038/s42003-024-06468-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 06/19/2024] [Indexed: 07/03/2024] Open
Abstract
Spotted fever group rickettsiae (SFGR) are obligate intracellular bacteria that cause spotted fever. The limitations of gene manipulation pose great challenges to studying the infection mechanisms of Rickettsia. By combining bioorthogonal metabolism and click chemistry, we developed a method to label R. heilongjiangensis via azide moieties and achieved rapid pathogen localization without complex procedures. Moreover, we constructed a C57BL/6 mice infection model by simulating tick bites and discovered that the stomach is the target organ of R. heilongjiangensis infection through in vivo imaging systems, which explained the occurrence of gastrointestinal symptoms following R. heilongjiangensis infection in some cases. This study offers a unique perspective for subsequent investigations into the pathogenic mechanisms of SFGR and identifies a potential target organ for R. heilongjiangensis.
Collapse
Affiliation(s)
- Juan Wang
- School of Public Health and Health Management, Gannan Medical University, Ganzhou, 341000, Jiangxi, P. R. China
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, 100071, P.R. China
| | - Li-Feng Du
- Institute of EcoHealth, School of Public Health, Shandong University, 44 Wenhuaxi Street, Jinan, 250012, Shandong, P.R. China
| | - Ming-Zhu Zhang
- Institute of EcoHealth, School of Public Health, Shandong University, 44 Wenhuaxi Street, Jinan, 250012, Shandong, P.R. China
| | - Wei Wei
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, 100071, P.R. China
| | - Zi-Yun Chen
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, 100071, P.R. China
- School of Public Health, Fujian Medical University, Fuzhou, Fujian, 350122, China
| | - Xu Zhang
- Guangdong Key Laboratory of Nanomedicine CAS-HK Joint Lab of Biomaterials, Shenzhen Engineering Laboratory of Nanomedicine and Nanoformulations Shenzhen Institute of Advanced Technology (SIAT) Chinese Academy of Sciences, Shenzhen, 518055, P. R. China
| | - Tao Xiong
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, 100071, P.R. China
| | - Zhen-Fei Wang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Luo-Yuan Xia
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, 100071, P.R. China
- Institute of EcoHealth, School of Public Health, Shandong University, 44 Wenhuaxi Street, Jinan, 250012, Shandong, P.R. China
| | - Jia-Fu Jiang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, 100071, P.R. China.
| | - Wen-Jun Li
- Guangdong Key Laboratory of Nanomedicine CAS-HK Joint Lab of Biomaterials, Shenzhen Engineering Laboratory of Nanomedicine and Nanoformulations Shenzhen Institute of Advanced Technology (SIAT) Chinese Academy of Sciences, Shenzhen, 518055, P. R. China.
| | - Dai-Yun Zhu
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, 100071, P.R. China.
| | - Na Jia
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, 100071, P.R. China.
| | - Wu-Chun Cao
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, 100071, P.R. China.
| |
Collapse
|
2
|
Romero MD, Carabeo RA. Dynamin-dependent entry of Chlamydia trachomatis is sequentially regulated by the effectors TarP and TmeA. Nat Commun 2024; 15:4926. [PMID: 38858371 PMCID: PMC11164928 DOI: 10.1038/s41467-024-49350-6] [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: 09/21/2023] [Accepted: 05/30/2024] [Indexed: 06/12/2024] Open
Abstract
Chlamydia invasion of epithelial cells is a pathogen-driven process involving two functionally distinct effectors - TarP and TmeA. They collaborate to promote robust actin dynamics at sites of entry. Here, we extend studies on the molecular mechanism of invasion by implicating the host GTPase dynamin 2 (Dyn2) in the completion of pathogen uptake. Importantly, Dyn2 function is modulated by TarP and TmeA at the levels of recruitment and activation through oligomerization, respectively. TarP-dependent recruitment requires phosphatidylinositol 3-kinase and the small GTPase Rac1, while TmeA has a post-recruitment role related to Dyn2 oligomerization. This is based on the rescue of invasion duration and efficiency in the absence of TmeA by the Dyn2 oligomer-stabilizing small molecule activator Ryngo 1-23. Notably, Dyn2 also regulated turnover of TarP- and TmeA-associated actin networks, with disrupted Dyn2 function resulting in aberrant turnover dynamics, thus establishing the interdependent functional relationship between Dyn2 and the effectors TarP and TmeA.
Collapse
Affiliation(s)
- Matthew D Romero
- Department of Pathology, Microbiology, and Immunology, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Rey A Carabeo
- Department of Pathology, Microbiology, and Immunology, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
3
|
Lehman SS, Verhoeve VI, Driscoll TP, Beckmann JF, Gillespie JJ. Metagenome diversity illuminates the origins of pathogen effectors. mBio 2024; 15:e0075923. [PMID: 38564675 PMCID: PMC11077975 DOI: 10.1128/mbio.00759-23] [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: 03/27/2023] [Accepted: 02/12/2024] [Indexed: 04/04/2024] Open
Abstract
Recent metagenome-assembled genome (MAG) analyses have profoundly impacted Rickettsiology systematics. The discovery of basal lineages (novel families Mitibacteraceae and Athabascaceae) with predicted extracellular lifestyles exposed an evolutionary timepoint for the transition to host dependency, which seemingly occurred independent of mitochondrial evolution. Notably, these basal rickettsiae carry the Rickettsiales vir homolog (rvh) type IV secretion system and purportedly use rvh to kill congener microbes rather than parasitize host cells as described for later-evolving rickettsial pathogens. MAG analysis also substantially increased diversity for the genus Rickettsia and delineated a sister lineage (the novel genus Tisiphia) that stands to inform on the emergence of human pathogens from protist and invertebrate endosymbionts. Herein, we probed Rickettsiales MAG and genomic diversity for the distribution of Rickettsia rvh effectors to ascertain their origins. A sparse distribution of most Rickettsia rvh effectors outside of Rickettsiaceae lineages illuminates unique rvh evolution from basal extracellular species and other rickettsial families. Remarkably, nearly every effector was found in multiple divergent forms with variable architectures, indicating profound roles for gene duplication and recombination in shaping effector repertoires in Rickettsia pathogens. Lateral gene transfer plays a prominent role in shaping the rvh effector landscape, as evinced by the discovery of many effectors on plasmids and conjugative transposons, as well as pervasive effector gene exchange between Rickettsia and Legionella species. Our study exemplifies how MAGs can yield insight into pathogen effector origins, particularly how effector architectures might become tailored to the discrete host cell functions of different eukaryotic hosts.IMPORTANCEWhile rickettsioses are deadly vector-borne human diseases, factors distinguishing Rickettsia pathogens from the innumerable bevy of environmental rickettsial endosymbionts remain lacking. Recent metagenome-assembled genome (MAG) studies revealed evolutionary timepoints for rickettsial transitions to host dependency. The rvh type IV secretion system was likely repurposed from congener killing in basal extracellular species to parasitizing host cells in later-evolving pathogens. Our analysis of MAG diversity for over two dozen rvh effectors unearthed their presence in some non-pathogens. However, most effectors were found in multiple divergent forms with variable architectures, indicating gene duplication and recombination-fashioned effector repertoires of Rickettsia pathogens. Lateral gene transfer substantially shaped pathogen effector arsenals, evinced by the discovery of effectors on plasmids and conjugative transposons, as well as pervasive effector gene exchanges between Rickettsia and Legionella species. Our study exemplifies how MAGs yield insight into pathogen effector origins and evolutionary processes tailoring effectors to eukaryotic host cell biology.
Collapse
Affiliation(s)
- Stephanie S. Lehman
- Division of Molecular and Cellular Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - Victoria I. Verhoeve
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Timothy P. Driscoll
- Department of Biology, West Virginia University, Morgantown, West Virginia, USA
| | - John F. Beckmann
- Department of Microbiology and Immunology, University of South Alabama, Mobile, Alabama, USA
| | - Joseph J. Gillespie
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
4
|
Abstract
Our understanding of free-living bacterial models like Escherichia coli far outpaces that of obligate intracellular bacteria, which cannot be cultured axenically. All obligate intracellular bacteria are host-associated, and many cause serious human diseases. Their constant exposure to the distinct biochemical niche of the host has driven the evolution of numerous specialized bacteriological and genetic adaptations, as well as innovative molecular mechanisms of infection. Here, we review the history and use of pathogenic Rickettsia species, which cause an array of vector-borne vascular illnesses, as model systems to probe microbial biology. Although many challenges remain in our studies of these organisms, the rich pathogenic and biological diversity of Rickettsia spp. constitutes a unique backdrop to investigate how microbes survive and thrive in host and vector cells. We take a bacterial-focused perspective and highlight emerging insights that relate to new host-pathogen interactions, bacterial physiology, and evolution. The transformation of Rickettsia spp. from pathogens to models demonstrates how recalcitrant microbes may be leveraged in the lab to tap unmined bacterial diversity for new discoveries. Rickettsia spp. hold great promise as model systems not only to understand other obligate intracellular pathogens but also to discover new biology across and beyond bacteria.
Collapse
Affiliation(s)
- Brandon Sit
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Rebecca L. Lamason
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| |
Collapse
|
5
|
Romero MD, Carabeo RA. Dynamin-dependent entry of Chlamydia trachomatis is sequentially regulated by the effectors TarP and TmeA. RESEARCH SQUARE 2023:rs.3.rs-3376558. [PMID: 37841835 PMCID: PMC10571596 DOI: 10.21203/rs.3.rs-3376558/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/17/2023]
Abstract
Chlamydia invasion of epithelial cells is a pathogen-driven process involving two functionally distinct effectors - TarP and TmeA. They collaborate to promote robust actin dynamics at sites of entry. Here, we extend studies on the molecular mechanism of invasion by implicating the host GTPase dynamin 2 (Dyn2) in the completion of pathogen uptake. Importantly, Dyn2 function is modulated by TarP and TmeA at the levels of recruitment and activation through oligomerization, respectively. TarP-dependent recruitment requires phosphatidylinositol 3-kinase and the small GTPase Rac1, while TmeA has a post-recruitment role related to Dyn2 oligomerization. This is based on the rescue of invasion duration and efficiency in the absence of TmeA by the Dyn2 oligomer-stabilizing small molecule activator Ryngo 1-23. Notably, Dyn2 also regulated turnover of TarP- and TmeA-associated actin networks, with disrupted Dyn2 function resulting in aberrant turnover dynamics, thus establishing the interdependent functional relationship between Dyn2 and the effectors TarP and TmeA.
Collapse
Affiliation(s)
- Matthew D. Romero
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, Omaha, NE
| | - Rey A. Carabeo
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, Omaha, NE
| |
Collapse
|
6
|
Verhoeve VI, Lehman SS, Driscoll TP, Beckmann JF, Gillespie JJ. Metagenome diversity illuminates origins of pathogen effectors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.26.530123. [PMID: 36909625 PMCID: PMC10002696 DOI: 10.1101/2023.02.26.530123] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/02/2023]
Abstract
Recent metagenome assembled genome (MAG) analyses have profoundly impacted Rickettsiology systematics. Discovery of basal lineages (Mitibacteraceae and Athabascaceae) with predicted extracellular lifestyles reveals an evolutionary timepoint for the transition to host dependency, which occurred independent of mitochondrial evolution. Notably, these basal rickettsiae carry the Rickettsiales vir homolog (rvh) type IV secretion system (T4SS) and purportedly use rvh to kill congener microbes rather than parasitize host cells as described for derived rickettsial pathogens. MAG analysis also substantially increased diversity for genus Rickettsia and delineated a basal lineage (Tisiphia) that stands to inform on the rise of human pathogens from protist and invertebrate endosymbionts. Herein, we probed Rickettsiales MAG and genomic diversity for the distribution of Rickettsia rvh effectors to ascertain their origins. A sparse distribution of most Rickettsia rvh effectors outside of Rickettsiaceae lineages indicates unique rvh evolution from basal extracellular species and other rickettsial families. Remarkably, nearly every effector was found in multiple divergent forms with variable architectures, illuminating profound roles for gene duplication and recombination in shaping effector repertoires in Rickettsia pathogens. Lateral gene transfer plays a prominent role shaping the rvh effector landscape, as evinced by the discover of many effectors on plasmids and conjugative transposons, as well as pervasive effector gene exchange between Rickettsia and Legionella species. Our study exemplifies how MAGs can provide incredible insight on the origins of pathogen effectors and how their architectural modifications become tailored to eukaryotic host cell biology.
Collapse
Affiliation(s)
- Victoria I Verhoeve
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Stephanie S Lehman
- Division of Molecular and Cellular Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Timothy P Driscoll
- Department of Biology, West Virginia University, Morgantown, West Virginia, USA
| | - John F Beckmann
- Microbiology and Immunology, University of South Alabama, Mobile, AL, USA
| | - Joseph J Gillespie
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
7
|
Romero MD, Carabeo RA. Distinct roles of the Chlamydia trachomatis effectors TarP and TmeA in the regulation of formin and Arp2/3 during entry. J Cell Sci 2022; 135:jcs260185. [PMID: 36093837 PMCID: PMC9659389 DOI: 10.1242/jcs.260185] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 09/05/2022] [Indexed: 12/13/2022] Open
Abstract
The obligate intracellular pathogen Chlamydia trachomatis manipulates the host actin cytoskeleton to assemble actin-rich structures that drive pathogen entry. The recent discovery of TmeA, which, like TarP, is an invasion-associated type III effector implicated in actin remodeling, raised questions regarding the nature of their functional interaction. Quantitative live-cell imaging of actin remodeling at invasion sites revealed differences in recruitment and turnover kinetics associated with the TarP and TmeA pathways, with the former accounting for most of the robust actin dynamics at invasion sites. TarP-mediated recruitment of actin nucleators, i.e. formins and the Arp2/3 complex, was crucial for rapid actin kinetics, generating a collaborative positive feedback loop that enhanced their respective actin-nucleating activities within invasion sites. In contrast, the formin Fmn1 was not recruited to invasion sites and did not collaborate with Arp2/3 within the context of TmeA-associated actin recruitment. Although the TarP-Fmn1-Arp2/3 signaling axis is responsible for the majority of actin dynamics, its inhibition had similar effects as the deletion of TmeA on invasion efficiency, consistent with the proposed model that TarP and TmeA act on different stages of the same invasion pathway.
Collapse
Affiliation(s)
- Matthew D. Romero
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198-5900, USA
| | - Rey A. Carabeo
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198-5900, USA
| |
Collapse
|
8
|
Spernovasilis N, Markaki I, Papadakis M, Mazonakis N, Ierodiakonou D. Mediterranean Spotted Fever: Current Knowledge and Recent Advances. Trop Med Infect Dis 2021; 6:172. [PMID: 34698275 PMCID: PMC8544691 DOI: 10.3390/tropicalmed6040172] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 09/17/2021] [Accepted: 09/21/2021] [Indexed: 12/15/2022] Open
Abstract
Mediterranean spotted fever (MSF) is an emerging tick-borne rickettsiosis of the spotted fever group (SFG), endemic in the Mediterranean basin. By virtue of technological innovations in molecular genetics, it has been determined that the causative agent of MSF is Rickettsia conorii subspecies conorii. The arthropod vector of this bacterium is the brown dog tick Rhipicephalus sanguineus. The true nature of the reservoir of R. conorii conorii has not been completely deciphered yet, although many authors theorize that the canine population, other mammals, and the ticks themselves could potentially contribute as reservoirs. Typical symptoms of MSF include fever, maculopapular rash, and a characteristic eschar ("tache noire"). Atypical clinical features and severe multi-organ complications may also be present. All of these manifestations arise from the disseminated infection of the endothelium by R. conorii conorii. Several methods exist for the diagnosis of MSF. Serological tests are widely used and molecular techniques have become increasingly available. Doxycycline remains the treatment of choice, while preventive measures are focused on modification of human behavior and vector control strategies. The purpose of this review is to summarize the current knowledge on the epidemiology, pathogenesis, clinical features, diagnosis, and treatment of MSF.
Collapse
Affiliation(s)
- Nikolaos Spernovasilis
- School of Medicine, University of Crete, 71003 Heraklion, Greece; (N.S.); (M.P.); (N.M.)
| | - Ioulia Markaki
- “Trifyllio” General Hospital of Kythira, 80200 Kythira, Greece;
| | - Michail Papadakis
- School of Medicine, University of Crete, 71003 Heraklion, Greece; (N.S.); (M.P.); (N.M.)
| | - Nikolaos Mazonakis
- School of Medicine, University of Crete, 71003 Heraklion, Greece; (N.S.); (M.P.); (N.M.)
| | - Despo Ierodiakonou
- Department of Social Medicine, School of Medicine, University of Crete, 71003 Heraklion, Greece
- Department of Primary Care and Population Health, University of Nicosia Medical School, Nicosia 2417, Cyprus
| |
Collapse
|
9
|
McGinn J, Lamason RL. The enigmatic biology of rickettsiae: recent advances, open questions and outlook. Pathog Dis 2021; 79:ftab019. [PMID: 33784388 PMCID: PMC8035066 DOI: 10.1093/femspd/ftab019] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 03/26/2021] [Indexed: 02/05/2023] Open
Abstract
Rickettsiae are obligate intracellular bacteria that can cause life-threatening illnesses and are among the oldest known vector-borne pathogens. Members of this genus are extraordinarily diverse and exhibit a broad host range. To establish intracellular infection, Rickettsia species undergo complex, multistep life cycles that are encoded by heavily streamlined genomes. As a result of reductive genome evolution, rickettsiae are exquisitely tailored to their host cell environment but cannot survive extracellularly. This host-cell dependence makes for a compelling system to uncover novel host-pathogen biology, but it has also hindered experimental progress. Consequently, the molecular details of rickettsial biology and pathogenesis remain poorly understood. With recent advances in molecular biology and genetics, the field is poised to start unraveling the molecular mechanisms of these host-pathogen interactions. Here, we review recent discoveries that have shed light on key aspects of rickettsial biology. These studies have revealed that rickettsiae subvert host cells using mechanisms that are distinct from other better-studied pathogens, underscoring the great potential of the Rickettsia genus for revealing novel biology. We also highlight several open questions as promising areas for future study and discuss the path toward solving the fundamental mysteries of this neglected and emerging human pathogen.
Collapse
Affiliation(s)
- Jon McGinn
- Department of Biology, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, United States
| | - Rebecca L Lamason
- Department of Biology, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, United States
| |
Collapse
|
10
|
Kristof MN, Allen PE, Yutzy LD, Thibodaux B, Paddock CD, Martinez JJ. Significant Growth by Rickettsia Species within Human Macrophage-Like Cells Is a Phenotype Correlated with the Ability to Cause Disease in Mammals. Pathogens 2021; 10:pathogens10020228. [PMID: 33669499 PMCID: PMC7934685 DOI: 10.3390/pathogens10020228] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 02/09/2021] [Accepted: 02/16/2021] [Indexed: 11/16/2022] Open
Abstract
Rickettsia are significant sources of tick-borne diseases in humans worldwide. In North America, two species in the spotted fever group of Rickettsia have been conclusively associated with disease of humans: Rickettsia rickettsii, the causative agent of Rocky Mountain spotted fever, and Rickettsia parkeri, the cause of R. parkeri rickettsiosis. Previous work in our lab demonstrated non-endothelial parasitism by another pathogenic SFG Rickettsia species, Rickettsia conorii, within THP-1-derived macrophages, and we have hypothesized that this growth characteristic may be an underappreciated aspect of rickettsial pathogenesis in mammalian hosts. In this work, we demonstrated that multiple other recognized human pathogenic species of Rickettsia, including R. rickettsii, R. parkeri, Rickettsia africae, and Rickettsiaakari can grow within target endothelial cells as well as within PMA-differentiated THP-1 cells. In contrast, Rickettsia bellii, a Rickettsia species not associated with disease of humans, and R. rickettsii strain Iowa, an avirulent derivative of pathogenic R. rickettsii, could invade both cell types but proliferate only within endothelial cells. Further analysis revealed that similar to previous studies on R. conorii, other recognized pathogenic Rickettsia species could grow within the cytosol of THP-1-derived macrophages and avoided localization with two different markers of lysosomal compartments; LAMP-2 and cathepsin D. R. bellii, on the other hand, demonstrated significant co-localization with lysosomal compartments. Collectively, these findings suggest that the ability of pathogenic rickettsial species to establish a niche within macrophage-like cells could be an important factor in their ability to cause disease in mammals. These findings also suggest that analysis of growth within mammalian phagocytic cells may be useful to predict the pathogenic potential of newly isolated and identified Rickettsia species.
Collapse
Affiliation(s)
- M. Nathan Kristof
- Vector Borne Disease Laboratories, Department of Pathobiological Sciences, LSU School of Veterinary Medicine, Baton Rouge, LA 70803, USA; (M.N.K.); (P.E.A.); (L.D.Y.); (B.T.)
| | - Paige E. Allen
- Vector Borne Disease Laboratories, Department of Pathobiological Sciences, LSU School of Veterinary Medicine, Baton Rouge, LA 70803, USA; (M.N.K.); (P.E.A.); (L.D.Y.); (B.T.)
| | - Lane D. Yutzy
- Vector Borne Disease Laboratories, Department of Pathobiological Sciences, LSU School of Veterinary Medicine, Baton Rouge, LA 70803, USA; (M.N.K.); (P.E.A.); (L.D.Y.); (B.T.)
| | - Brandon Thibodaux
- Vector Borne Disease Laboratories, Department of Pathobiological Sciences, LSU School of Veterinary Medicine, Baton Rouge, LA 70803, USA; (M.N.K.); (P.E.A.); (L.D.Y.); (B.T.)
| | - Christopher D. Paddock
- Rickettsial Zoonoses Branch, Division of Vector-Borne Diseases, Centers for Disease Control and Prevention, U.S. Department of Health and Human Services, Atlanta, GA 30329, USA;
| | - Juan J. Martinez
- Vector Borne Disease Laboratories, Department of Pathobiological Sciences, LSU School of Veterinary Medicine, Baton Rouge, LA 70803, USA; (M.N.K.); (P.E.A.); (L.D.Y.); (B.T.)
- Correspondence: ; Tel.: +1-225-578-9297
| |
Collapse
|
11
|
Chamberlain NB, Mehari YT, Hayes BJ, Roden CM, Kidane DT, Swehla AJ, Lorenzana-DeWitt MA, Farone AL, Gunderson JH, Berk SG, Farone MB. Infection and nuclear interaction in mammalian cells by 'Candidatus Berkiella cookevillensis', a novel bacterium isolated from amoebae. BMC Microbiol 2019; 19:91. [PMID: 31072343 PMCID: PMC6507137 DOI: 10.1186/s12866-019-1457-z] [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: 09/10/2017] [Accepted: 04/15/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND 'Candidatus Berkiella cookevillensis' and 'Ca. Berkiella aquae' have previously been described as intranuclear bacteria of amoebae. Both bacteria were isolated from amoebae and were described as appearing within the nuclei of Acanthamoeba polyphaga and ultimately lysing their host cells within 4 days. Both bacteria are Gammaproteobacteria in the order Legionellales with the greatest similarity to Coxiella burnetii. Neither bacterium grows axenically in artificial culture media. In this study, we further characterized 'Ca. B. cookevillensis' by demonstrating association with nuclei of human phagocytic and nonphagocytic cell lines. RESULTS Transmission electron microscopy (TEM) and confocal microscopy were used to confirm nuclear co-localization of 'Ca. B. cookevillensis' in the amoeba host A. polyphaga with 100% of cells having bacteria co-localized with host nuclei by 48 h. TEM and confocal microscopy demonstrated that the bacterium was also observed to be closely associated with nuclei of human U937 and THP-1 differentiated macrophage cell lines and nonphagocytic HeLa human epithelial-like cells. Immunofluorescent staining revealed that the bacteria-containing vacuole invaginates the nuclear membranes and appears to cross from the cytoplasm into the nucleus as an intact vacuole. CONCLUSION Results of this study indicate that a novel coccoid bacterium isolated from amoebae can infect human cell lines by associating with the host cell nuclei, either by crossing the nuclear membranes or by deeply invaginating the nuclear membranes. When associated with the nuclei, the bacteria appear to be bound within a vacuole and replicate to high numbers by 48 h. We believe this is the first report of such a process involving bacteria and human cell lines.
Collapse
Affiliation(s)
- Nicholas B Chamberlain
- Molecular Biosciences Program, Middle Tennessee State University, 1301 E. Main St, Murfreesboro, TN, 37130, USA.,Department of Biology, Middle Tennessee State University, 1301 E. Main St, Murfreesboro, TN, 37130, USA
| | - Yohannes T Mehari
- Molecular Biosciences Program, Middle Tennessee State University, 1301 E. Main St, Murfreesboro, TN, 37130, USA.,Department of Biology, Middle Tennessee State University, 1301 E. Main St, Murfreesboro, TN, 37130, USA
| | - B Jason Hayes
- Department of Biology, Middle Tennessee State University, 1301 E. Main St, Murfreesboro, TN, 37130, USA
| | - Colleen M Roden
- Department of Biology, Middle Tennessee State University, 1301 E. Main St, Murfreesboro, TN, 37130, USA
| | - Destaalem T Kidane
- Molecular Biosciences Program, Middle Tennessee State University, 1301 E. Main St, Murfreesboro, TN, 37130, USA.,Department of Biology, Middle Tennessee State University, 1301 E. Main St, Murfreesboro, TN, 37130, USA
| | - Andrew J Swehla
- Department of Biology, Middle Tennessee State University, 1301 E. Main St, Murfreesboro, TN, 37130, USA
| | - Mario A Lorenzana-DeWitt
- Department of Biology, Middle Tennessee State University, 1301 E. Main St, Murfreesboro, TN, 37130, USA
| | - Anthony L Farone
- Molecular Biosciences Program, Middle Tennessee State University, 1301 E. Main St, Murfreesboro, TN, 37130, USA.,Department of Biology, Middle Tennessee State University, 1301 E. Main St, Murfreesboro, TN, 37130, USA
| | - John H Gunderson
- Department of Biology, Tennessee Technological University, 1 William L Jones Dr, Cookeville, TN, 38505, USA
| | - Sharon G Berk
- Department of Biology, Middle Tennessee State University, 1301 E. Main St, Murfreesboro, TN, 37130, USA
| | - Mary B Farone
- Molecular Biosciences Program, Middle Tennessee State University, 1301 E. Main St, Murfreesboro, TN, 37130, USA. .,Department of Biology, Middle Tennessee State University, 1301 E. Main St, Murfreesboro, TN, 37130, USA.
| |
Collapse
|
12
|
Rauch J, Eisermann P, Noack B, Mehlhoop U, Muntau B, Schäfer J, Tappe D. Typhus Group Rickettsiosis, Germany, 2010-2017 1. Emerg Infect Dis 2019; 24:1213-1220. [PMID: 29912688 PMCID: PMC6038764 DOI: 10.3201/eid2407.180093] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Typhus group rickettsiosis is caused by the vectorborne bacteria Rickettsia typhi and R. prowazekii. R. typhi, which causes murine typhus, the less severe endemic form of typhus, is transmitted by fleas; R. prowazekii, which causes the severe epidemic form of typhus, is transmitted by body lice. To examine the immunology of human infection with typhus group rickettsiae, we retrospectively reviewed clinical signs and symptoms, laboratory changes, and travel destinations of 28 patients who had typhus group rickettsiosis diagnosed by the German Reference Center for Tropical Pathogens, Hamburg, Germany, during 2010-2017. Immunofluorescence assays of follow-up serum samples indicated simultaneous seroconversion of IgM, IgA, and IgG or concurrence in the first serum sample. Cytokine levels peaked during the second week of infection, coinciding with organ dysfunction and seroconversion. For 3 patients, R. typhi was detected by species-specific nested quantitative PCR. For all 28 patients, R. typhi was the most likely causative pathogen.
Collapse
|
13
|
Cruz R, Pereira-Castro I, Almeida MT, Moreira A, Cabanes D, Sousa S. Epithelial Keratins Modulate cMet Expression and Signaling and Promote InlB-Mediated Listeria monocytogenes Infection of HeLa Cells. Front Cell Infect Microbiol 2018; 8:146. [PMID: 29868502 PMCID: PMC5960701 DOI: 10.3389/fcimb.2018.00146] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 04/20/2018] [Indexed: 12/11/2022] Open
Abstract
The host cytoskeleton is a major target for bacterial pathogens during infection. In particular, pathogens usurp the actin cytoskeleton function to strongly adhere to the host cell surface, to induce plasma membrane remodeling allowing invasion and to spread from cell to cell and disseminate to the whole organism. Keratins are cytoskeletal proteins that are the major components of intermediate filaments in epithelial cells however, their role in bacterial infection has been disregarded. Here we investigate the role of the major epithelial keratins, keratins 8 and 18 (K8 and K18), in the cellular infection by Listeria monocytogenes. We found that K8 and K18 are required for successful InlB/cMet-dependent L. monocytogenes infection, but are dispensable for InlA/E-cadherin-mediated invasion. Both K8 and K18 accumulate at InlB-mediated internalization sites following actin recruitment and modulate actin dynamics at those sites. We also reveal the key role of K8 and K18 in HGF-induced signaling which occurs downstream the activation of cMet. Strikingly, we show here that K18, and at a less extent K8, controls the expression of cMet and other surface receptors such TfR and integrin β1, by promoting the stability of their corresponding transcripts. Together, our results reveal novel functions for major epithelial keratins in the modulation of actin dynamics at the bacterial entry sites and in the control of surface receptors mRNA stability and expression.
Collapse
Affiliation(s)
- Rui Cruz
- Group of Molecular Microbiology, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Group of Molecular Microbiology, Institute for Molecular and Cell Biology, Porto, Portugal.,Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Isabel Pereira-Castro
- Group of Molecular Microbiology, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Gene Regulation Group, Institute for Molecular and Cell Biology, Porto, Portugal
| | - Maria T Almeida
- Group of Molecular Microbiology, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Group of Molecular Microbiology, Institute for Molecular and Cell Biology, Porto, Portugal
| | - Alexandra Moreira
- Group of Molecular Microbiology, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal.,Gene Regulation Group, Institute for Molecular and Cell Biology, Porto, Portugal
| | - Didier Cabanes
- Group of Molecular Microbiology, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Group of Molecular Microbiology, Institute for Molecular and Cell Biology, Porto, Portugal
| | - Sandra Sousa
- Group of Molecular Microbiology, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Group of Molecular Microbiology, Institute for Molecular and Cell Biology, Porto, Portugal
| |
Collapse
|
14
|
Sabaneyeva E, Castelli M, Szokoli F, Benken K, Lebedeva N, Salvetti A, Schweikert M, Fokin S, Petroni G. Host and symbiont intraspecific variability: The case of Paramecium calkinsi and "Candidatus Trichorickettsia mobilis". Eur J Protistol 2017; 62:79-94. [PMID: 29287245 DOI: 10.1016/j.ejop.2017.12.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2017] [Revised: 10/17/2017] [Accepted: 12/04/2017] [Indexed: 01/08/2023]
Abstract
Newly isolated strains of the ciliate Paramecium calkinsi and their cytoplasmic bacterial endosymbionts were characterized by a multidisciplinary approach, including live observation, ultrastructural investigation, and molecular analysis. Despite morphological resemblance, the characterized P. calkinsi strains showed a significant molecular divergence compared to conspecifics, possibly hinting for a cryptic speciation. The endosymbionts were clearly found to be affiliated to the species "Candidatus Trichorickettsia mobilis" (Rickettsiales, Rickettsiaceae), currently encompassing only bacteria retrieved in an obligate intracellular association with other ciliates. However, a relatively high degree of intraspecific divergence was observed as well, thus it was possible to split "Candidatus Trichorickettsia" into three subspecies, one of which represented so far only by the newly characterized endosymbionts of P. calkinsi. Other features distinguished the members of each different subspecies. In particular, the endosymbionts of P. calkinsi resided in the cytoplasm and possessed numerous peritrichous flagella, although no motility was evidenced, whereas their conspecifics in other hosts were either cytoplasmic and devoid of flagella, or macronuclear, displaying flagellar-driven motility. Moreover, contrarily to previously analyzed "Candidatus Trichorickettsia" hosts, infected P. calkinsi cells frequently became amicronucleate and demonstrated abnormal cell division, eventually leading to decline of the laboratory culture.
Collapse
Affiliation(s)
- E Sabaneyeva
- Department of Cytology and Histology, St. Petersburg State University, Russian Federation.
| | - M Castelli
- Department of Veterinary Medicine, University of Milan, Italy; Department of Biosciences, University of Milan, Italy
| | - F Szokoli
- Dipartimento di Biologia, Università di Pisa, Italy; Institut für Hydrobiologie, Technische Universität Dresden, Germany
| | - K Benken
- Core Facility Center for Microscopy and Microanalysis, St. Petersburg State University, Russian Federation
| | - N Lebedeva
- Core Facility Center for Cultivation of Microorganisms, St. Petersburg State University, Russian Federation
| | - A Salvetti
- Dipartimento di Medicina Clinica e Sperimentale, Università di Pisa, Italy
| | - M Schweikert
- Institut of Biomaterials and Biomolecular Systems, Stuttgart University, Germany
| | - S Fokin
- Dipartimento di Biologia, Università di Pisa, Italy; Department of Invertebrate Zoology, St. Petersburg State University, Russian Federation
| | - G Petroni
- Dipartimento di Biologia, Università di Pisa, Italy.
| |
Collapse
|
15
|
Rengarajan M, Hayer A, Theriot JA. Endothelial Cells Use a Formin-Dependent Phagocytosis-Like Process to Internalize the Bacterium Listeria monocytogenes. PLoS Pathog 2016; 12:e1005603. [PMID: 27152864 PMCID: PMC4859537 DOI: 10.1371/journal.ppat.1005603] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 04/06/2016] [Indexed: 01/11/2023] Open
Abstract
Vascular endothelial cells act as gatekeepers that protect underlying tissue from blood-borne toxins and pathogens. Nevertheless, endothelial cells are able to internalize large fibrin clots and apoptotic debris from the bloodstream, although the precise mechanism of such phagocytosis-like uptake is unknown. We show that cultured primary human endothelial cells (HUVEC) internalize both pathogenic and non-pathogenic Listeria bacteria comparably, in a phagocytosis-like process. In contrast with previously studied host cell types, including intestinal epithelial cells and hepatocytes, we find that endothelial internalization of Listeria is independent of all known pathogenic bacterial surface proteins. Consequently, we exploited the internalization and intracellular replication of L. monocytogenes to identify distinct host cell factors that regulate phagocytosis-like uptake in HUVEC. Using siRNA screening and subsequent genetic and pharmacologic perturbations, we determined that endothelial infectivity was modulated by cytoskeletal proteins that normally modulate global architectural changes, including phosphoinositide-3-kinase, focal adhesions, and the small GTPase Rho. We found that Rho kinase (ROCK) is acutely necessary for adhesion of Listeria to endothelial cells, whereas the actin-nucleating formins FHOD1 and FMNL3 specifically regulate internalization of bacteria as well as inert beads, demonstrating that formins regulate endothelial phagocytosis-like uptake independent of the specific cargo. Finally, we found that neither ROCK nor formins were required for macrophage phagocytosis of L. monocytogenes, suggesting that endothelial cells have distinct requirements for bacterial internalization from those of classical professional phagocytes. Our results identify a novel pathway for L. monocytogenes uptake by human host cells, indicating that this wily pathogen can invade a variety of tissues by using a surprisingly diverse suite of distinct uptake mechanisms that operate differentially in different host cell types.
Collapse
Affiliation(s)
- Michelle Rengarajan
- Department of Biochemistry, Stanford University School of Medicine, Stanford, California, United States of America
| | - Arnold Hayer
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, California, United States of America
| | - Julie A. Theriot
- Department of Biochemistry, Stanford University School of Medicine, Stanford, California, United States of America
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, United States of America
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, California, United States of America
- * E-mail:
| |
Collapse
|
16
|
Bugalhão JN, Mota LJ, Franco IS. Bacterial nucleators: actin' on actin. Pathog Dis 2015; 73:ftv078. [PMID: 26416078 DOI: 10.1093/femspd/ftv078] [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] [Accepted: 09/22/2015] [Indexed: 11/13/2022] Open
Abstract
The actin cytoskeleton is a key target of numerous microbial pathogens, including protozoa, fungi, bacteria and viruses. In particular, bacterial pathogens produce and deliver virulence effector proteins that hijack actin dynamics to enable bacterial invasion of host cells, allow movement within the host cytosol, facilitate intercellular spread or block phagocytosis. Many of these effector proteins directly or indirectly target the major eukaryotic actin nucleator, the Arp2/3 complex, by either mimicking nucleation promoting factors or activating upstream small GTPases. In contrast, this review is focused on a recently identified class of effector proteins from Gram-negative bacteria that function as direct actin nucleators. These effector proteins mimic functional activities of formins, WH2-nucleators and Ena/VASP assembly promoting factors demonstrating that bacteria have coopted the complete set of eukaryotic actin assembly pathways. Structural and functional analyses of these nucleators have revealed several motifs and/or mechanistic activities that are shared with eukaryotic actin nucleators. However, functional effects of these proteins during infection extend beyond plain actin polymerization leading to interference with other host cell functions such as vesicle trafficking, cell cycle progression and cell death. Therefore, their use as model systems could not only help in the understanding of the mechanistic details of actin polymerization but also provide novel insights into the connection between actin dynamics and other cellular pathways.
Collapse
Affiliation(s)
- Joana N Bugalhão
- UCIBIO-REQUIMTE, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, 2829-516 Caparica, Portugal
| | - Luís Jaime Mota
- UCIBIO-REQUIMTE, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, 2829-516 Caparica, Portugal
| | - Irina S Franco
- UCIBIO-REQUIMTE, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, 2829-516 Caparica, Portugal Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, 2780-157 Oeiras, Portugal
| |
Collapse
|
17
|
Rennoll-Bankert KE, Rahman MS, Gillespie JJ, Guillotte ML, Kaur SJ, Lehman SS, Beier-Sexton M, Azad AF. Which Way In? The RalF Arf-GEF Orchestrates Rickettsia Host Cell Invasion. PLoS Pathog 2015; 11:e1005115. [PMID: 26291822 PMCID: PMC4546372 DOI: 10.1371/journal.ppat.1005115] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Accepted: 07/28/2015] [Indexed: 01/09/2023] Open
Abstract
Bacterial Sec7-domain-containing proteins (RalF) are known only from species of Legionella and Rickettsia, which have facultative and obligate intracellular lifestyles, respectively. L. pneumophila RalF, a type IV secretion system (T4SS) effector, is a guanine nucleotide exchange factor (GEF) of ADP-ribosylation factors (Arfs), activating and recruiting host Arf1 to the Legionella-containing vacuole. In contrast, previous in vitro studies showed R. prowazekii (Typhus Group) RalF is a functional Arf-GEF that localizes to the host plasma membrane and interacts with the actin cytoskeleton via a unique C-terminal domain. As RalF is differentially encoded across Rickettsia species (e.g., pseudogenized in all Spotted Fever Group species), it may function in lineage-specific biology and pathogenicity. Herein, we demonstrate RalF of R. typhi (Typhus Group) interacts with the Rickettsia T4SS coupling protein (RvhD4) via its proximal C-terminal sequence. RalF is expressed early during infection, with its inactivation via antibody blocking significantly reducing R. typhi host cell invasion. For R. typhi and R. felis (Transitional Group), RalF ectopic expression revealed subcellular localization with the host plasma membrane and actin cytoskeleton. Remarkably, R. bellii (Ancestral Group) RalF showed perinuclear localization reminiscent of ectopically expressed Legionella RalF, for which it shares several structural features. For R. typhi, RalF co-localization with Arf6 and PI(4,5)P2 at entry foci on the host plasma membrane was determined to be critical for invasion. Thus, we propose recruitment of PI(4,5)P2 at entry foci, mediated by RalF activation of Arf6, initiates actin remodeling and ultimately facilitates bacterial invasion. Collectively, our characterization of RalF as an invasin suggests that, despite carrying a similar Arf-GEF unknown from other bacteria, different intracellular lifestyles across Rickettsia and Legionella species have driven divergent roles for RalF during infection. Furthermore, our identification of lineage-specific Arf-GEF utilization across some rickettsial species illustrates different pathogenicity factors that define diverse agents of rickettsial diseases. Phylogenomics analysis indicates divergent mechanisms for host cell invasion across diverse species of obligate intracellular Rickettsia. For instance, only some Rickettsia species carry RalF, the rare bacterial Arf-GEF effector utilized by Legionella pneumophila to facilitate fusion of ER-derived membranes with its host-derived vacuole. For R. prowazekii (Typhus Group, TG), prior in vitro studies suggested the Arf-GEF activity of RalF, which is absent from Spotted Fever Group species, might be spatially regulated at the host plasma membrane. Herein, we demonstrate RalF of R. typhi (TG) and R. felis (Transitional Group) localizes to the host plasma membrane, yet R. bellii (Ancestral Group) RalF shows perinuclear localization reminiscent of RalF-mediated recruitment of Arf1 by L. pneumophila to its vacuole. For R. typhi, RalF expression occurs early during infection, with RalF inactivation significantly reducing host cell invasion. Furthermore, RalF co-localization with Arf6 and the phosphoinositide PI(4,5)P2 at the host plasma membrane was determined to be critical for R. typhi invasion. Thus, our work illustrates that different intracellular lifestyles across species of Rickettsia and Legionella have driven divergent roles for RalF during host cell infection. Collectively, we identify lineage-specific Arf-GEF utilization across diverse rickettsial species, previously unappreciated mechanisms for host cell invasion and infection.
Collapse
Affiliation(s)
- Kristen E. Rennoll-Bankert
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - M. Sayeedur Rahman
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Joseph J. Gillespie
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Mark L. Guillotte
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Simran J. Kaur
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Stephanie S. Lehman
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Magda Beier-Sexton
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Abdu F. Azad
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
18
|
Zheng K, Kitazato K, Wang Y, He Z. Pathogenic microbes manipulate cofilin activity to subvert actin cytoskeleton. Crit Rev Microbiol 2015; 42:677-95. [PMID: 25853495 DOI: 10.3109/1040841x.2015.1010139] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Actin-depolymerizing factor (ADF)/cofilin proteins are key players in controlling the temporal and spatial extent of actin dynamics, which is crucial for mediating host-pathogen interactions. Pathogenic microbes have evolved molecular mechanisms to manipulate cofilin activity to subvert the actin cytoskeletal system in host cells, promoting their internalization into the target cells, modifying the replication niche and facilitating their intracellular and intercellular dissemination. The study of how these pathogens exploit cofilin pathways is crucial for understanding infectious disease and providing potential targets for drug therapies.
Collapse
Affiliation(s)
- Kai Zheng
- a Department of Pharmacy, School of Medicine , Shenzhen University , Shenzhen , Guangdong , People's Republic of China .,c Guangzhou Jinan Biomedicine Research and Development Center, National Engineering Research Center of Genetic Medicine, Jinan University , Guangzhou , China
| | - Kaio Kitazato
- b Division of Molecular Pharmacology of Infectious Agents, Department of Molecular Microbiology and Immunology , Nagasaki University , Nagasaki , Japan , and
| | - Yifei Wang
- c Guangzhou Jinan Biomedicine Research and Development Center, National Engineering Research Center of Genetic Medicine, Jinan University , Guangzhou , China
| | - Zhendan He
- a Department of Pharmacy, School of Medicine , Shenzhen University , Shenzhen , Guangdong , People's Republic of China
| |
Collapse
|
19
|
Gillespie JJ, Kaur SJ, Rahman MS, Rennoll-Bankert K, Sears KT, Beier-Sexton M, Azad AF. Secretome of obligate intracellular Rickettsia. FEMS Microbiol Rev 2014; 39:47-80. [PMID: 25168200 DOI: 10.1111/1574-6976.12084] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The genus Rickettsia (Alphaproteobacteria, Rickettsiales, Rickettsiaceae) is comprised of obligate intracellular parasites, with virulent species of interest both as causes of emerging infectious diseases and for their potential deployment as bioterrorism agents. Currently, there are no effective commercially available vaccines, with treatment limited primarily to tetracycline antibiotics, although others (e.g. josamycin, ciprofloxacin, chloramphenicol, and azithromycin) are also effective. Much of the recent research geared toward understanding mechanisms underlying rickettsial pathogenicity has centered on characterization of secreted proteins that directly engage eukaryotic cells. Herein, we review all aspects of the Rickettsia secretome, including six secretion systems, 19 characterized secretory proteins, and potential moonlighting proteins identified on surfaces of multiple Rickettsia species. Employing bioinformatics and phylogenomics, we present novel structural and functional insight on each secretion system. Unexpectedly, our investigation revealed that the majority of characterized secretory proteins have not been assigned to their cognate secretion pathways. Furthermore, for most secretion pathways, the requisite signal sequences mediating translocation are poorly understood. As a blueprint for all known routes of protein translocation into host cells, this resource will assist research aimed at uniting characterized secreted proteins with their apposite secretion pathways. Furthermore, our work will help in the identification of novel secreted proteins involved in rickettsial 'life on the inside'.
Collapse
Affiliation(s)
- Joseph J Gillespie
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Simran J Kaur
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - M Sayeedur Rahman
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Kristen Rennoll-Bankert
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Khandra T Sears
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Magda Beier-Sexton
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Abdu F Azad
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
20
|
Reed SCO, Lamason RL, Risca VI, Abernathy E, Welch MD. Rickettsia actin-based motility occurs in distinct phases mediated by different actin nucleators. Curr Biol 2013; 24:98-103. [PMID: 24361066 DOI: 10.1016/j.cub.2013.11.025] [Citation(s) in RCA: 88] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Revised: 10/21/2013] [Accepted: 11/12/2013] [Indexed: 11/16/2022]
Abstract
Many intracellular bacterial pathogens undergo actin-based motility to promote cell-cell spread during infection [1]. For each pathogen, motility was assumed to be driven by a single actin polymerization pathway. Curiously, spotted fever group Rickettsia differ from other pathogens in possessing two actin-polymerizing proteins. RickA, an activator of the host Arp2/3 complex, was initially proposed to drive motility [2, 3]. Sca2, a mimic of host formins [4, 5], was later shown to be required for motility [6]. Whether and how their activities are coordinated has remained unclear. Here, we show that each protein directs an independent mode of Rickettsia parkeri motility at different times during infection. Early after invasion, motility is slow and meandering, generating short, curved actin tails that are enriched with Arp2/3 complex and cofilin. Early motility requires RickA and Arp2/3 complex and is correlated with transient RickA localization to the bacterial pole. Later in infection, motility is faster and directionally persistent, resulting in long, straight actin tails. Late motility is independent of Arp2/3 complex and RickA and requires Sca2, which accumulates at the bacterial pole. Both motility pathways facilitate cell-to-cell spread. The ability to exploit two actin assembly pathways may allow Rickettsia to establish an intracellular niche and spread between diverse cells throughout a prolonged infection.
Collapse
Affiliation(s)
- Shawna C O Reed
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA; Microbiology Graduate Group, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Rebecca L Lamason
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Viviana I Risca
- Biophysics Graduate Group, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Emma Abernathy
- Microbiology Graduate Group, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Matthew D Welch
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA; Microbiology Graduate Group, University of California, Berkeley, Berkeley, CA 94720, USA.
| |
Collapse
|
21
|
Frickmann H, Schröpfer E, Dobler G. Actin assessment in addition to specific immuno-fluorescence staining to demonstrate rickettsial growth in cell culture. Eur J Microbiol Immunol (Bp) 2013; 3:198-203. [PMID: 24265939 DOI: 10.1556/eujmi.3.2013.3.8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2013] [Accepted: 06/08/2013] [Indexed: 11/19/2022] Open
Abstract
Rickettsiae are able to spread within infected cell mono-layers by modifying intra-cellular actin formations. The study analyzes whether a visualization of actin modifications in addition to specific immuno-fluorescence staining of rickettsiae might facilitate the proof of rickettsial growth in cell culture. Cell mono-layers of Vero E6 und BGM cells were infected with Rickettsia honei. Intra-cellular actin was fluorescence stained with TRITC-(tetra-methyl-5,6-isothiocyanate)-labeled phalloidin in addition to specific immuno-fluorescence staining of rickettsiae with FITC-(fluorescein-isothiocyanate)-labeled antibodies. DNA of bacteria and cells was counter-stained with DAPI (4´,6-diamino-2-phenyl-indole). Cell cultures infected with Vaccinia virus were used as positive controls, cell cultures infected with Coxiella burnetii as negative controls. High concentrations of R. honei are necessary to demonstrate characteristic modifications of the intra-cellular actin. This effect is more pronounced in Vero E6 cells than in BGM cells. Actin staining with phalloidin is not suited for an early proof of rickettsial growth in cell culture but may confirm unclear findings in specific immuno-fluorescence staining in case of sufficient bacterial density.
Collapse
|
22
|
Abstract
Tick-borne diseases are prevalent throughout the world and present a diagnostic challenge owing to their nonspecific clinical symptoms. Many tick-borne diseases involve the central and peripheral nervous systems. Early diagnosis or at least suspicion of a tick-borne cause is necessary to institute early empiric treatment. After a brief review of tick biology, we present the most common tick-borne diseases. A brief discussion of epidemiology, the transmission route, and pathogenesis is followed by a discussion of the clinical manifestations, diagnosis and treatment options when available. The review emphasizes the infectious causes with a significant neurological manifestation.
Collapse
|
23
|
Melnikow E, Xu S, Liu J, Bell AJ, Ghedin E, Unnasch TR, Lustigman S. A potential role for the interaction of Wolbachia surface proteins with the Brugia malayi glycolytic enzymes and cytoskeleton in maintenance of endosymbiosis. PLoS Negl Trop Dis 2013; 7:e2151. [PMID: 23593519 PMCID: PMC3617236 DOI: 10.1371/journal.pntd.0002151] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2012] [Accepted: 02/22/2013] [Indexed: 11/19/2022] Open
Abstract
The human filarial parasite Brugia malayi harbors an endosymbiotic bacterium of the genus Wolbachia. The Wolbachia represent an attractive target for the control of filarial induced disease as elimination of the bacteria affects molting, reproduction and survival of the worms. The molecular basis for the symbiotic relationship between Wolbachia and their filarial hosts has yet to be elucidated. To identify proteins involved in this process, we focused on the Wolbachia surface proteins (WSPs), which are known to be involved in bacteria-host interactions in other bacterial systems. Two WSP-like proteins (wBm0152 and wBm0432) were localized to various host tissues of the B. malayi female adult worms and are present in the excretory/secretory products of the worms. We provide evidence that both of these proteins bind specifically to B. malayi crude protein extracts and to individual filarial proteins to create functional complexes. The wBm0432 interacts with several key enzymes involved in the host glycolytic pathway, including aldolase and enolase. The wBm0152 interacts with the host cytoskeletal proteins actin and tubulin. We also show these interactions in vitro and have verified that wBm0432 and B. malayi aldolase, as well as wBm0152 and B. malayi actin, co-localize to the vacuole surrounding Wolbachia. We propose that both WSP protein complexes interact with each other via the aldolase-actin link and/or via the possible interaction between the host's enolase and the cytoskeleton, and play a role in Wolbachia distribution during worm growth and embryogenesis.
Collapse
Affiliation(s)
- Elena Melnikow
- Molecular Parasitology, Lindsley F. Kimball Research Institute, New York Blood Center, New York, New York, United States of America
| | - Shulin Xu
- Department of Global Health, University of South Florida, Tampa, Florida, United States of America
| | - Jing Liu
- Molecular Parasitology, Lindsley F. Kimball Research Institute, New York Blood Center, New York, New York, United States of America
| | - Aaron J. Bell
- Electron Microscopy, Lindsley F. Kimball Research Institute, New York Blood Center, New York, New York, United States of America
| | - Elodie Ghedin
- Department of Computational & Systems Biology, Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Thomas R. Unnasch
- Department of Global Health, University of South Florida, Tampa, Florida, United States of America
| | - Sara Lustigman
- Molecular Parasitology, Lindsley F. Kimball Research Institute, New York Blood Center, New York, New York, United States of America
| |
Collapse
|
24
|
Wood DO, Hines A, Tucker AM, Woodard A, Driskell LO, Burkhardt NY, Kurtti TJ, Baldridge GD, Munderloh UG. Establishment of a replicating plasmid in Rickettsia prowazekii. PLoS One 2012; 7:e34715. [PMID: 22529927 PMCID: PMC3328469 DOI: 10.1371/journal.pone.0034715] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2012] [Accepted: 03/08/2012] [Indexed: 11/18/2022] Open
Abstract
Rickettsia prowazekii, the causative agent of epidemic typhus, grows only within the cytosol of eukaryotic host cells. This obligate intracellular lifestyle has restricted the genetic analysis of this pathogen and critical tools, such as replicating plasmid vectors, have not been developed for this species. Although replicating plasmids have not been reported in R. prowazekii, the existence of well-characterized plasmids in several less pathogenic rickettsial species provides an opportunity to expand the genetic systems available for the study of this human pathogen. Competent R. prowazekii were transformed with pRAM18dRGA, a 10.3 kb vector derived from pRAM18 of R. amblyommii. A plasmid-containing population of R. prowazekii was obtained following growth under antibiotic selection, and the rickettsial plasmid was maintained extrachromosomally throughout multiple passages. The transformant population exhibited a generation time comparable to that of the wild type strain with a copy number of approximately 1 plasmid per rickettsia. These results demonstrate for the first time that a plasmid can be maintained in R. prowazekii, providing an important genetic tool for the study of this obligate intracellular pathogen.
Collapse
Affiliation(s)
- David O Wood
- Department of Microbiology and Immunology, Laboratory of Molecular Biology, University of South Alabama College of Medicine, Mobile, Alabama, United States of America.
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Sunyakumthorn P, Petchampai N, Kearney MT, Sonenshine DE, Macaluso KR. Molecular characterization and tissue-specific gene expression of Dermacentor variabilis α-catenin in response to rickettsial infection. INSECT MOLECULAR BIOLOGY 2012; 21:197-204. [PMID: 22221256 PMCID: PMC3299920 DOI: 10.1111/j.1365-2583.2011.01126.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Alpha catenin is a cytoskeleton protein that acts as a regulator of actin rearrangement by forming an E-cadherin adhesion complex. In Dermacentor variabilis, a putative α-catenin (Dvα-catenin) was previously identified as differentially regulated in ovaries of ticks chronically infected with Rickettsia montanensis. To begin characterizing the role(s) of Dvα-catenin during rickettsial infection, the full-length Dvα-catenin cDNA was cloned and analysed. Comparative sequence analysis demonstrates a 3069-bp cDNA with a 2718-bp open reading frame with a sequence similar to Ixodes scapularisα-catenin. A portion of Dvα-catenin is homologous to the vinculin-conserved domain containing a putative actin-binding region and β-catenin-binding and -dimerization regions. Quantitative reverse-transcription PCR analysis demonstrated that Dvα-catenin is predominantly expressed in tick ovaries and is responsive to tick feeding. The tissue-specific gene expression analysis of ticks exposed to Rickettsia demonstrates that Dvα-catenin expression was significantly downregulated 12 h after exposure to R. montanensis, but not in Rickettsia amblyommii-exposed ovaries, compared with Rickettsia-unexposed ticks. Studying tick-derived molecules associated with rickettsial infection will provide a better understanding of the transmission dynamics of tick-borne rickettsial diseases.
Collapse
Affiliation(s)
- Piyanate Sunyakumthorn
- Department of Pathobiological Sciences, Louisiana State University, School of Veterinary Medicine, Skip Bertman Drive, SVM-3213, Baton Rouge, Louisiana 70803, USA
| | - Natthida Petchampai
- Department of Pathobiological Sciences, Louisiana State University, School of Veterinary Medicine, Skip Bertman Drive, SVM-3213, Baton Rouge, Louisiana 70803, USA
| | - Michael T. Kearney
- Department of Pathobiological Sciences, Louisiana State University, School of Veterinary Medicine, Skip Bertman Drive, SVM-3213, Baton Rouge, Louisiana 70803, USA
| | - Daniel E. Sonenshine
- Department of Biological Sciences, Old Dominion University, Norfolk, Virginia 23529
| | - Kevin R. Macaluso
- Department of Pathobiological Sciences, Louisiana State University, School of Veterinary Medicine, Skip Bertman Drive, SVM-3213, Baton Rouge, Louisiana 70803, USA
| |
Collapse
|
26
|
|
27
|
Kang H, Perlmutter DS, Shenoy VB, Tang JX. Observation and kinematic description of long actin tracks induced by spherical beads. Biophys J 2010; 99:2793-802. [PMID: 21044576 PMCID: PMC2966028 DOI: 10.1016/j.bpj.2010.08.058] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2010] [Revised: 08/11/2010] [Accepted: 08/27/2010] [Indexed: 11/28/2022] Open
Abstract
We report an in vitro study comparing the growth of long actin tails induced by spherical beads coated with the verprolin central acidic domain of the polymerization enzyme N-WASP to that induced by Listeria monocytogenes in similar cellular extracts. The tracks behind the beads show characteristic differences in shape and curvature from those left by the bacteria, which have an elongated shape and a similar polymerization-inducing enzyme distributed only on the rear surface of the cell. The experimental tracks are simulated using a generalized kinematic model, which incorporates three modes of bead rotation with respect to the tail. The results show that the trajectories of spherical beads are mechanically deterministic rather than random, as suggested by stochastic models. Assessment of the bead rotation and its mechanistic basis offers insights into the biological function of actin-based motility.
Collapse
Affiliation(s)
- Hyeran Kang
- Department of Physics, Brown University, Providence, Rhode Island
| | | | - Vivek B. Shenoy
- School of Engineering, Brown University, Providence, Rhode Island
| | - Jay X. Tang
- Department of Physics, Brown University, Providence, Rhode Island
- School of Engineering, Brown University, Providence, Rhode Island
| |
Collapse
|
28
|
Thepparit C, Bourchookarn A, Petchampai N, Barker SA, Macaluso KR. Interaction of Rickettsia felis with histone H2B facilitates the infection of a tick cell line. MICROBIOLOGY-SGM 2010; 156:2855-2863. [PMID: 20558510 PMCID: PMC3068691 DOI: 10.1099/mic.0.041400-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Haematophagous arthropods are the primary vectors in the transmission of Rickettsia, yet the molecular mechanisms mediating the rickettsial infection of arthropods remain elusive. This study utilized a biotinylated protein pull-down assay together with LC-MS/MS to identify interaction between Ixodes scapularis histone H2B and Rickettsia felis. Co-immunoprecipitation of histone with rickettsial cell lysate demonstrated the association of H2B with R. felis proteins, including outer-membrane protein B (OmpB), a major rickettsial adhesin molecule. The rickettsial infection of tick ISE6 cells was reduced by approximately 25 % via RNA-mediated H2B-depletion or enzymic treatment of histones. The interaction of H2B with the rickettsial adhesin OmpB suggests a role for H2B in mediating R. felis internalization into ISE6 cells.
Collapse
Affiliation(s)
- Chutima Thepparit
- Department of Pathobiological Sciences, Louisiana State University, School of Veterinary Medicine, Baton Rouge, LA 70803, USA
| | - Apichai Bourchookarn
- Department of Technology and Industries, Faculty of Science and Technology, Prince of Songkla University, Pattani 94000, Thailand
| | - Natthida Petchampai
- Department of Pathobiological Sciences, Louisiana State University, School of Veterinary Medicine, Baton Rouge, LA 70803, USA
| | - Steven A Barker
- Department of Comparative Biomedical Sciences, Louisiana State University, School of Veterinary Medicine, Baton Rouge, LA 70803, USA
| | - Kevin R Macaluso
- Department of Pathobiological Sciences, Louisiana State University, School of Veterinary Medicine, Baton Rouge, LA 70803, USA
| |
Collapse
|
29
|
Abstract
The vascular endothelium is the main target of a limited number of infectious agents, Rickettsia, Ehrlichia ruminantium, and Orientia tsutsugamushi are among them. These arthropod-transmitted obligately-intracellular bacteria cause serious systemic diseases that are not infrequently lethal. In this review, we discuss the bacterial biology, vector biology, and clinical aspects of these conditions with particular emphasis on the interactions of these bacteria with the vascular endothelium and how it responds to intracellular infection. The study of these bacteria in relevant in vivo models is likely to offer new insights into the physiology of the endothelium that have not been revealed by other models.
Collapse
Affiliation(s)
- Gustavo Valbuena
- Department of Pathology and Center for Biodefense and Emerging Infectious Diseases, The University of Texas Medical Branch, 301 University Boulevard, Galveston, Texas 77555-0609, USA.
| | | |
Collapse
|
30
|
Abstract
One century after the first description of rickettsiae as human pathogens, the rickettsiosis remained poorly understood diseases. These microorganisms are indeed characterized by a strictly intracellular location which has, for long, prohibited their detailed study. Within the last ten years, the completion of the genome sequences of several strains allowed gaining a better knowledge about the molecular mechanisms involved in rickettsia pathogenicity. Here, we summarized available data concerning the critical steps of rickettsia-host cell interactions that should contribute to tissue injury and diseases, that is, adhesion, phagosomal escape, motility, and intracellular survival of the bacteria.
Collapse
Affiliation(s)
- Premanand Balraj
- Unité des Rickettsies, URMITE IRD-CNRS 6236, Faculté de Médecine, Marseille, France
| | | | | |
Collapse
|
31
|
Sabaneyeva EV, Derkacheva ME, Benken KA, Fokin SI, Vainio S, Skovorodkin IN. Actin-based mechanism of holospora obtusa trafficking in Paramecium caudatum. Protist 2009; 160:205-19. [PMID: 19231281 DOI: 10.1016/j.protis.2008.11.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2008] [Accepted: 11/08/2008] [Indexed: 12/25/2022]
Abstract
Holospora obtusa, an alpha-proteobacterium, is an obligate endonuclear pathogen of the ciliate Paramecium caudatum. It is engulfed by the host cell in the course of phagocytosis but soon escapes from the phagosome and is transported across the host cell cytoplasm to the paramecium macronucleus. Electron microscopy reveals a comet-like tail resembling that of Listeria trailing after H. obtusa in the host cytoplasm. In this study we investigated the role of the host cell actin and Arp3 in the process of infection with Holospora. Cytochalasin D treatment significantly reduced the rate of nuclear infection. Using immunocytochemistry and experimental infection of GFP-actin-transfected paramecia we demonstrated that the Paramecium actin1-1 took part in the bacterial escape from the phagosome, its trafficking in the cytoplasm and entry into the host macronucleus. Rapid assembly/disassembly of actin filaments in P. caudatum led to quick loss of actin1-1 from the trails left by H. obtusa. Immunocytochemistry using anti-bovine Arp3 antibodies demonstrated the presence of Arp3 in these trails. Our data indicate that details of H. obtusa infection are rather similar to those of Listeria and Rickettsia.
Collapse
Affiliation(s)
- Elena V Sabaneyeva
- Department of Cytology and Histology, St.-Petersburg State University, 199034 St.-Petersburg, Russia
| | | | | | | | | | | |
Collapse
|
32
|
Fujishima M. Infection and Maintenance of Holospora Species in Paramecium caudatum. ENDOSYMBIONTS IN PARAMECIUM 2009. [DOI: 10.1007/978-3-540-92677-1_8] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
33
|
Chen LF, Sexton DJ. What's New in Rocky Mountain Spotted Fever? Infect Dis Clin North Am 2008; 22:415-32, vii-viii. [DOI: 10.1016/j.idc.2008.03.008] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
34
|
Emerging and re-emerging rickettsioses: endothelial cell infection and early disease events. Nat Rev Microbiol 2008; 6:375-86. [PMID: 18414502 DOI: 10.1038/nrmicro1866] [Citation(s) in RCA: 196] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
35
|
Lane BJ, Mutchler C, Al Khodor S, Grieshaber SS, Carabeo RA. Chlamydial entry involves TARP binding of guanine nucleotide exchange factors. PLoS Pathog 2008; 4:e1000014. [PMID: 18383626 PMCID: PMC2279300 DOI: 10.1371/journal.ppat.1000014] [Citation(s) in RCA: 112] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Chlamydia trachomatis attachment to cells induces the secretion of the elementary body–associated protein TARP (Translocated Actin Recruiting Protein). TARP crosses the plasma membrane where it is immediately phosphorylated at tyrosine residues by unknown host kinases. The Rac GTPase is also activated, resulting in WAVE2 and Arp2/3-dependent recruitment of actin to the sites of chlamydia attachment. We show that TARP participates directly in chlamydial invasion activating the Rac-dependent signaling cascade to recruit actin. TARP functions by binding two distinct Rac guanine nucleotide exchange factors (GEFs), Sos1 and Vav2, in a phosphotyrosine-dependent manner. The tyrosine phosphorylation profile of the sequence YEPISTENIYESI within TARP, as well as the transient activation of the phosphatidylinositol 3-kinase (PI3-K), appears to determine which GEF is utilized to activate Rac. The first and second tyrosine residues, when phosphorylated, are utilized by the Sos1/Abi1/Eps8 and Vav2, respectively, with the latter requiring the lipid phosphatidylinositol 3,4,5-triphosphate. Depletion of these critical signaling molecules by siRNA resulted in inhibition of chlamydial invasion to varying degrees, owing to a possible functional redundancy of the two pathways. Collectively, these data implicate TARP in signaling to the actin cytoskeleton remodeling machinery, demonstrating a mechanism by which C. trachomatis invades non-phagocytic cells. The human pathogen Chlamydia trachomatis is the causative agent of the most prevalent bacterial sexually transmitted disease in industrialized nations, and of the preventable blinding condition trachoma in developing countries. Survival and replication of chlamydial species occur exclusively inside a host cell, and thus, gaining access to the protective intracellular niche is an absolute requirement. This report describes how the chlamydia protein TARP, which is secreted at the base of the bacteria and across the host membrane, acts as a scaffold to which host signaling proteins bind. This assembly of the complex of signaling proteins, which include Sos1, Abi1, Eps8, and Vav2 results in the remodeling of the host cytoskeleton to facilitate engulfment of the infecting chlamydia. We conclude that these proteins have a role in chlamydia based on a number of observations including their interaction with the TARP protein, their ability to switch on known signaling participants in chlamydia invasion, their localization at the site of chlamydia entry, and the inhibition of chlamydia invasion in their absence. Altogether, the data functionally link TARP with signaling pathways that function in chlamydial invasion, demonstrating the direct involvement of TARP in the invasion of host cells by C. trachomatis.
Collapse
Affiliation(s)
- B. Josh Lane
- Department of Microbiology and Immunology, University of Louisville Medical School, Louisville, Kentucky, United States of America
| | - Charla Mutchler
- Department of Microbiology and Immunology, University of Louisville Medical School, Louisville, Kentucky, United States of America
| | - Souhaila Al Khodor
- Department of Microbiology and Immunology, University of Louisville Medical School, Louisville, Kentucky, United States of America
| | - Scott S. Grieshaber
- Department of Oral Biology, University of Florida School of Dentistry, Gainesville, Florida, United States of America
| | - Rey A. Carabeo
- Department of Microbiology and Immunology, University of Louisville Medical School, Louisville, Kentucky, United States of America
- * E-mail:
| |
Collapse
|
36
|
Valbuena G, Walker DH. The endothelium as a target for infections. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2007; 1:171-98. [PMID: 18039112 DOI: 10.1146/annurev.pathol.1.110304.100031] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The endothelial cells lining vascular and lymphatic vessels are targets of several infectious agents, including viruses and bacteria, that lead to dramatic changes in their functions. Understanding the pathophysiological mechanisms that cause the clinical manifestations of those infections has been advanced through the use of animal models and in vitro systems; however, there are also abundant studies that explore the consequences of endothelial infection in vitro without supporting evidence that endothelial cells are actual in vivo targets of infection in human diseases. This article defines criteria for considering an infection as truly endothelium-targeted and reviews the literature that offers insights into the pathogenesis of human endothelial-target infections.
Collapse
|
37
|
Rydkina E, Sahni A, Silverman DJ, Sahni SK. Comparative analysis of host-cell signalling mechanisms activated in response to infection with Rickettsia conorii and Rickettsia typhi. J Med Microbiol 2007; 56:896-906. [PMID: 17577053 DOI: 10.1099/jmm.0.47050-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The Gram-negative intracellular bacteria Rickettsia conorii and Rickettsia typhi are the aetiological agents of Mediterranean spotted fever and endemic typhus, respectively, in humans. Infection of endothelial cells (ECs) lining vessel walls, and the resultant vascular inflammation and haemostatic alterations are salient pathogenetic features of both of these rickettsial diseases. An important consideration, however, is that dramatic differences in the intracellular motility and accumulation patterns for spotted fever versus typhus group rickettsiae have been documented, suggesting the possibility of unique and potentially different interactions with host cells. This study characterized and compared R. conorii- and R. typhi-mediated effects on cultured human ECs. The DNA-binding activity of nuclear transcription factor-kappaB (NF-kappaB) and the phosphorylation status of stress-activated p38 kinase were determined as indicators of NF-kappaB and p38 activation. R. conorii infection resulted in a biphasic activation of NF-kappaB, with an early increase in DNA-binding activity at 3 h, followed by a later peak at 24 h. The activated NF-kappaB species were composed mainly of RelA p65-p50 heterodimers and p50 homodimers. R. typhi infection of ECs resulted in only early activation of NF-kappaB at 3 h, composed primarily of p65-p50 heterodimers. Whilst R. conorii infection induced increased phosphorylation of p38 kinase (threefold mean induction) with the maximal response at 3 h, a considerably less-intense response peaking at about 6 h post-infection was found with R. typhi. Furthermore, mRNA expression of the chemokines interleukin (IL)-8 and monocyte chemoattractant protein-1 in ECs infected with either Rickettsia species was higher than the corresponding controls, but there were distinct differences in the secretion patterns for IL-8, suggesting the possibility of involvement of post-transcriptional control mechanisms or differences in the release from intracellular storage sites. Thus, the intensity and kinetics of host-cell responses triggered by spotted fever and typhus species exhibit distinct variations that could subsequently lead to differences in the extent of endothelial activation and inflammation and serve as important determinants of pathogenesis.
Collapse
Affiliation(s)
- Elena Rydkina
- Hematology-Oncology Unit, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Abha Sahni
- Hematology-Oncology Unit, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - David J Silverman
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Sanjeev K Sahni
- Hematology-Oncology Unit, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| |
Collapse
|
38
|
Carabeo RA, Dooley CA, Grieshaber SS, Hackstadt T. Rac interacts with Abi-1 and WAVE2 to promote an Arp2/3-dependent actin recruitment during chlamydial invasion. Cell Microbiol 2007; 9:2278-88. [PMID: 17501982 DOI: 10.1111/j.1462-5822.2007.00958.x] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Chlamydiae are Gram-negative obligate intracellular pathogens to which access to an intracellular environment is fundamental to their development. Chlamydial attachment to host cells induces the activation of the Rac GTPase, which is required for the localization of WAVE2 at the sites of chlamydial entry. Co-immunoprecipitation experiments demonstrated that Chlamydia trachomatis infection promoted the interaction of Rac with WAVE2 and Abi-1, but not with IRSp53. siRNA depletion of WAVE2 and Abi-1 abrogated chlamydia-induced actin recruitment and significantly reduced the uptake of the pathogen by the depleted cells. Chlamydia invasion also requires the Arp2/3 complex as demonstrated by its localization to the sites of chlamydial attachment and the reduced efficiency of chlamydial invasion in cells overexpressing the VCA domain of the neural Wiskott-Aldrich syndrome protein. Thus, C. trachomatis activates Rac and promotes its interaction with WAVE2 and Abi-1 to activate the Arp2/3 complex resulting in the induction of actin cytoskeletal rearrangements that are required for invasion.
Collapse
Affiliation(s)
- Rey A Carabeo
- Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, KY, USA
| | | | | | | |
Collapse
|
39
|
Carlsson F, Brown EJ. Actin-based motility of intracellular bacteria, and polarized surface distribution of the bacterial effector molecules. J Cell Physiol 2006; 209:288-96. [PMID: 16826602 DOI: 10.1002/jcp.20721] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Several intracellular bacterial pathogens, including species of Listeria, Rickettsia, Shigella, Mycobacteria, and Burkholderia, have evolved mechanisms to exploit the actin polymerization machinery of their hosts to induce actin-based motility, enabling these pathogens to spread between host cells without exposing themselves to the extracellular milieu. Efficient cell-to-cell spread requires directional motility, which the bacteria may achieve by concentrating the effector molecules at one pole of their cell body, thereby restricting polymerization of monomeric actin into actin tails to this pole. The study of the molecular processes involved in the initiation of actin tail formation at the bacterial surface, and subsequent actin-based motility, has provided much insight into the pathogenesis of infections caused by these bacteria and into the cell biology of actin dynamics. Concomitantly, this field of research has provided an opportunity to understand the mechanisms whereby bacteria can achieve a polarized distribution of surface proteins. This review will describe the process of actin-based motility of intracellular bacteria, and the mechanisms by which bacteria can obtain a polarized distribution of their surface proteins.
Collapse
Affiliation(s)
- Fredric Carlsson
- Program in Microbial Pathogenesis and Host Defense, University of California, San Francisco, California 94158-2517, USA.
| | | |
Collapse
|
40
|
Jewett TJ, Fischer ER, Mead DJ, Hackstadt T. Chlamydial TARP is a bacterial nucleator of actin. Proc Natl Acad Sci U S A 2006; 103:15599-604. [PMID: 17028176 PMCID: PMC1622868 DOI: 10.1073/pnas.0603044103] [Citation(s) in RCA: 148] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Chlamydia trachomatis entry into host cells results from a parasite-directed remodeling of the actin cytoskeleton. A type III secreted effector, TARP (translocated actin recruiting phosphoprotein), has been implicated in the recruitment of actin to the site of internalization. To elucidate the role of TARP in actin recruitment, we identified host cell proteins that associated with recombinant GST-TARP fusions. TARP directly associated with actin, and this interaction promoted actin nucleation as determined by in vitro polymerization assays. Domain analysis of TARP identified an actin-binding domain that bears structural and primary amino acid sequence similarity to WH2 domain family proteins. In addition, a proline-rich domain was found to promote TARP oligomerization and was required for TARP-dependent nucleation of new actin filaments. Our findings reveal a mechanism by which chlamydiae induce localized cytoskeletal changes by the translocated effector TARP during entry into host cells.
Collapse
Affiliation(s)
- Travis J. Jewett
- Host–Parasite Interactions Section, Laboratory of Intracellular Parasites, and
| | - Elizabeth R. Fischer
- RTS Microscopy Unit, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840
| | - David J. Mead
- Host–Parasite Interactions Section, Laboratory of Intracellular Parasites, and
| | - Ted Hackstadt
- Host–Parasite Interactions Section, Laboratory of Intracellular Parasites, and
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|
41
|
Abstract
Rickettsiae are well known as intracellular pathogens of animals, humans, and plants and facultative and unorganized symbionts of invertebrates. No close relative of mitochondria has yet been associated with nutritional or developmental dependency of its host cell or organism. We have found a mycetomic Rickettsia that is a strict obligatory symbiont of the parthenogenetic booklouse Liposcelis bostrychophila (Psocoptera). These rickettsiae show an evolutionary transition from a solitary to a primary mycetomic bacterium adapted to the development of its host. These intracellular and intranuclear bacteria reside in specialized cells in several tissues. Their distribution changes markedly with the development of their host. The most advanced phenotype is a paired mycetome in the abdomen, described for the first time for Rickettsia and this host order. The mycetomic rickettsiae of two parthenogenetic book lice species are in the spotted fever group and in the basal limoniae group. While mycetomic bacteria are well known for their metabolic or light-emitting functions, these rickettsiae have an essential role in the early development of the oocyte. Removal of the Rickettsia stops egg production and reproduction in the book louse. In two phylogenetically distant psocopteran species, Rickettsia are shown to be associated with four transitional stages from free bacteria, infected cells, through single mycetocytes to organ-forming mycetomes.
Collapse
Affiliation(s)
- M Alejandra Perotti
- School of Biological Sciences, University of Wales Bangor, Bangor, Gwynedd LL57 2UW, UK
| | | | | | | |
Collapse
|
42
|
Grieshaber SS, Grieshaber NA, Miller N, Hackstadt T. Chlamydia trachomatis Causes Centrosomal Defects Resulting in Chromosomal Segregation Abnormalities. Traffic 2006; 7:940-9. [PMID: 16882039 DOI: 10.1111/j.1600-0854.2006.00439.x] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Chlamydiae traffic along microtubules to the microtubule organizing center (MTOC) to establish an intracellular niche within the host cell. Trafficking to the MTOC is dynein dependent although the activating and cargo-linking function of the dynactin complex is supplanted by unknown chlamydial protein(s). We demonstrate that once localized to the MTOC, the chlamydial inclusion maintains a tight association with cellular centrosomes. This association is sustained through mitosis and leads to a significant increase in supernumerary centrosomes, abnormal spindle poles, and chromosomal segregation defects. Chlamydial infection thus can lead to chromosome instability in cells that recover from infection.
Collapse
Affiliation(s)
- Scott S Grieshaber
- Host-Parasite Interactions Section, Laboratory of Intracellular Parasites, NIAID, NIH, Rocky Mountain Laboratories, Hamilton, MT 59840, USA
| | | | | | | |
Collapse
|
43
|
Ogata H, La Scola B, Audic S, Renesto P, Blanc G, Robert C, Fournier PE, Claverie JM, Raoult D. Genome sequence of Rickettsia bellii illuminates the role of amoebae in gene exchanges between intracellular pathogens. PLoS Genet 2006; 2:e76. [PMID: 16703114 PMCID: PMC1458961 DOI: 10.1371/journal.pgen.0020076] [Citation(s) in RCA: 233] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2005] [Accepted: 04/04/2006] [Indexed: 11/18/2022] Open
Abstract
The recently sequenced Rickettsia felis genome revealed an unexpected plasmid carrying several genes usually associated with DNA transfer, suggesting that ancestral rickettsiae might have been endowed with a conjugation apparatus. Here we present the genome sequence of Rickettsia bellii, the earliest diverging species of known rickettsiae. The 1,552,076 base pair-long chromosome does not exhibit the colinearity observed between other rickettsia genomes, and encodes a complete set of putative conjugal DNA transfer genes most similar to homologues found in Protochlamydia amoebophila UWE25, an obligate symbiont of amoebae. The genome exhibits many other genes highly similar to homologues in intracellular bacteria of amoebae. We sought and observed sex pili-like cell surface appendages for R. bellii. We also found that R. bellii very efficiently multiplies in the nucleus of eukaryotic cells and survives in the phagocytic amoeba, Acanthamoeba polyphaga. These results suggest that amoeba-like ancestral protozoa could have served as a genetic "melting pot" where the ancestors of rickettsiae and other bacteria promiscuously exchanged genes, eventually leading to their adaptation to the intracellular lifestyle within eukaryotic cells.
Collapse
Affiliation(s)
- Hiroyuki Ogata
- Structural and Genomic Information Laboratory, Centre National de la Recherche Scientifique UPR-2589, Institut de Biologie Structurale et Microbiologie, Parc Scientifique de Luminy, Marseille, France
- * To whom correspondence should be addressed. E-mail: (HO); (DR)
| | - Bernard La Scola
- Unité des Rickettsies, Centre National de la Recherche Scientifique UMR-6020, IFR-48, Faculté de Médecine, Université de la Méditerranée, Marseille, France
| | - Stéphane Audic
- Structural and Genomic Information Laboratory, Centre National de la Recherche Scientifique UPR-2589, Institut de Biologie Structurale et Microbiologie, Parc Scientifique de Luminy, Marseille, France
| | - Patricia Renesto
- Unité des Rickettsies, Centre National de la Recherche Scientifique UMR-6020, IFR-48, Faculté de Médecine, Université de la Méditerranée, Marseille, France
| | - Guillaume Blanc
- Structural and Genomic Information Laboratory, Centre National de la Recherche Scientifique UPR-2589, Institut de Biologie Structurale et Microbiologie, Parc Scientifique de Luminy, Marseille, France
| | - Catherine Robert
- Unité des Rickettsies, Centre National de la Recherche Scientifique UMR-6020, IFR-48, Faculté de Médecine, Université de la Méditerranée, Marseille, France
| | - Pierre-Edouard Fournier
- Unité des Rickettsies, Centre National de la Recherche Scientifique UMR-6020, IFR-48, Faculté de Médecine, Université de la Méditerranée, Marseille, France
| | - Jean-Michel Claverie
- Structural and Genomic Information Laboratory, Centre National de la Recherche Scientifique UPR-2589, Institut de Biologie Structurale et Microbiologie, Parc Scientifique de Luminy, Marseille, France
| | - Didier Raoult
- Unité des Rickettsies, Centre National de la Recherche Scientifique UMR-6020, IFR-48, Faculté de Médecine, Université de la Méditerranée, Marseille, France
- * To whom correspondence should be addressed. E-mail: (HO); (DR)
| |
Collapse
|
44
|
Eto DS, Sundsbak JL, Mulvey MA. Actin-gated intracellular growth and resurgence of uropathogenic Escherichia coli. Cell Microbiol 2006; 8:704-17. [PMID: 16548895 DOI: 10.1111/j.1462-5822.2006.00691.x] [Citation(s) in RCA: 109] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Strains of uropathogenic Escherichia coli (UPEC) can invade terminally differentiated superficial bladder epithelial cells and subsequently multiply, forming large biofilm-like inclusions referred to as pods. In contrast, within immature bladder cells UPEC enter a more quiescent state and often fail to replicate appreciably. As immature bladder epithelial cells undergo terminal differentiation the actin cytoskeleton is radically diminished, a phenomenon that we reasoned could influence the intracellular fate of UPEC. Here we show that UPEC within undifferentiated bladder cells is trafficked into acidic compartments having key features of late endosomes and lysosomes. These UPEC-containing vacuoles are often enmeshed within a network of actin filaments, the disruption of which stimulates intravacuolar growth and efflux of UPEC in cell culture-based studies. In this in vitro model system, release of UPEC into the host cytosol further stimulates intracellular bacterial growth and the rapid development of pod-like inclusions. These inclusions, as well as those observed using an in vivo mouse model, develop in association with cytokeratin intermediate filaments that may act as scaffolding for intracellular biofilm formation. Our data suggest an aetiological basis for recurrent urinary tract infections, linking bladder cell differentiation and the accompanying redistribution of actin microfilaments with the resurgence of UPEC from quiescent intravacuolar reservoirs within the bladder epithelium.
Collapse
Affiliation(s)
- Danelle S Eto
- Pathology Department, Division of Cell Biology and Immunology, University of Utah, Salt Lake City, UT 84112-0565, USA
| | | | | |
Collapse
|
45
|
Abstract
Listeria, Rickettsia, Burkholderia, Shigella and Mycobacterium species subvert cellular actin dynamics to facilitate their movement within the host cytosol and to infect neighbouring cells while evading host immune surveillance and promoting their intracellular survival. 'Attaching and effacing' Escherichia coli do not enter host cells but attach intimately to the cell surface, inducing motile actin-rich pedestals, the function of which is currently unclear. The molecular basis of actin-based motility of these bacterial pathogens reveals novel insights about bacterial pathogenesis and fundamental host-cell pathways.
Collapse
Affiliation(s)
- Joanne M Stevens
- Division of Microbiology, Institute for Animal Health, Compton Laboratory, Berkshire, RG20 7NN, UK
| | | | | |
Collapse
|
46
|
Abstract
Rickettsiae are obligate intracellular alpha-proteobacteria that primarily target the microvascular endothelium. In the last two decades, new rickettsial pathogens have been associated with human illness around the world. Clinically, the common denominator in all rickettsioses is the development of increased microvascular permeability, leading to cerebral and non-cardiogenic pulmonary edema. With the development of powerful research tools, advances in the understanding of rickettsial pathogenesis have been dramatic. Entry into the host cell is followed by rapid escape into the cytoplasm to avoid phagolysosomal fusion. Spotted fever group rickettsiae induce actin polymerization via a group of proteins called RickA, which promote nucleation of actin monomers via the Arp2/3 complex at one rickettsial pole, propelling the bacteria across the cytoplasm and into neighboring cells. Damage to the host cell is most likely multifactorial. The most extensively studied mechanism is the generation of reactive oxygen species (ROS) and downregulation of enzymes involved in protection against oxidative injury. The significance of ROS-mediated cellular damage in vivo is beginning to be elucidated. The main pathogenic mechanism is increased microvascular permeability leading to profound metabolic disturbances in the extravascular compartment. The underlying factors responsible for those changes are beginning to be elucidated in vitro and include direct effects of intracellular rickettsiae, cytokines, and possibly activated coagulation factors--all of which most likely modify interendothelial junctions. Our knowledge on rickettsial pathogenesis will continue to expand in the near future as new research tools become available.
Collapse
Affiliation(s)
- Juan P Olano
- Center for Biodefense and Emerging Infectious Diseases, University of Texas Medical Branch, Galveston, TX 77555-0609, USA.
| |
Collapse
|
47
|
Kurtti TJ, Simser JA, Baldridge GD, Palmer AT, Munderloh UG. Factors influencing in vitro infectivity and growth of Rickettsia peacockii (Rickettsiales: Rickettsiaceae), an endosymbiont of the Rocky Mountain wood tick, Dermacentor andersoni (Acari, Ixodidae). J Invertebr Pathol 2005; 90:177-86. [PMID: 16288906 PMCID: PMC1625098 DOI: 10.1016/j.jip.2005.09.001] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2005] [Revised: 09/02/2005] [Accepted: 09/09/2005] [Indexed: 11/29/2022]
Abstract
Rickettsia peacockii, a spotted fever group rickettsia, is a transovarially transmitted endosymbiont of Rocky Mountain wood ticks, Dermacentor andersoni. This rickettsia, formerly known as the East Side Agent and restricted to female ticks, was detected in a chronically infected embryonic cell line, DAE100, from D. andersoni. We examined infectivity, ability to induce cytopathic effect (CPE) and host cell specificity of R. peacockii using cultured arthropod and mammalian cells. Aposymbiotic DAE100 cells were obtained using oxytetracycline or incubation at 37 degrees C. Uninfected DAE100 sublines grew faster than the parent line, indicating R. peacockii regulation of host cell growth. Nevertheless, DAE100 cellular defenses exerted partial control over R. peacockii growth. Rickettsiae existed free in the cytosol of DAE100 cells or within autophagolysosomes. Exocytosed rickettsiae accumulated in the medium and were occasionally contained within host membranes. R. peacockii multiplied in other cell lines from the hard ticks D. andersoni, Dermacentor albipictus, Ixodes scapularis, and Ixodes ricinus; the soft tick Carios capensis; and the lepidopteran Trichoplusia ni. Lines from the tick Amblyomma americanum, the mosquito Aedes albopictus, and two mammalian cell lines were non-permissive to R. peacockii. High cell densities facilitated rickettsial spread within permissive cell cultures, and an inoculum of one infected to nine uninfected cells resulted in the greatest yield of infected tick cells. Cell-free R. peacockii also were infectious for tick cells and centrifugation onto cell layers enhanced infectivity approximately 100-fold. The ability of R. peacockii to cause mild CPE suggests that its pathogenicity is not completely muted. An analysis of R. peacockii-cell interactions in comparison to pathogenic rickettsiae will provide insights into host cell colonization mechanisms.
Collapse
Affiliation(s)
- Timothy J Kurtti
- Department of Entomology, University of Minnesota, St. Paul, MN 55108, USA.
| | | | | | | | | |
Collapse
|
48
|
Iwatani K, Dohra H, Lang BF, Burger G, Hori M, Fujishima M. Translocation of an 89-kDa periplasmic protein is associated with Holospora infection. Biochem Biophys Res Commun 2005; 337:1198-205. [PMID: 16236256 DOI: 10.1016/j.bbrc.2005.09.175] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2005] [Accepted: 09/25/2005] [Indexed: 11/23/2022]
Abstract
The symbiotic bacterium Holospora obtusa infects the macronucleus of the ciliate Paramecium caudatum. After ingestion by its host, an infectious form of Holospora with an electron-translucent tip passes through the host digestive vacuole and penetrates the macronuclear envelope with this tip. To investigate the underlying molecular mechanism of this process, we raised a monoclonal antibody against the tip-specific 89-kDa protein, sequenced this partially, and identified the corresponding complete gene. The deduced protein sequence carries two actin-binding motifs. Indirect immunofluorescence microscopy shows that during escape from the host digestive vacuole, the 89-kDa proteins translocates from the inside to the outside of the tip. When the bacterium invades the macronucleus, the 89-kDa protein is left behind at the entry point of the nuclear envelope. Transmission electron microscopy shows the formation of fine fibrous structures that co-localize with the antibody-labeled regions of the bacterium. Our findings suggest that the 89-kDa protein plays a role in Holospora's escape from the host digestive vacuole, the migration through the host cytoplasm, and the invasion into the macronucleus.
Collapse
Affiliation(s)
- Koichi Iwatani
- Biological Institute, Faculty of Science, Yamaguchi University, Yoshida 1677-1, Yamaguchi 753-8512, Japan
| | | | | | | | | | | |
Collapse
|
49
|
Balter A, Tang JX. Hydrodynamic stability of helical growth at low Reynolds number. PHYSICAL REVIEW. E, STATISTICAL, NONLINEAR, AND SOFT MATTER PHYSICS 2005; 71:051912. [PMID: 16089576 DOI: 10.1103/physreve.71.051912] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2004] [Indexed: 05/03/2023]
Abstract
A cylindrical object growing at a low Reynolds number can spontaneously develop a helical shape. We have studied this phenomenon numerically, and our results may shed some light on the spontaneous formation of helical tails of a dense protein network observed in experiments on actin based motility. We also identify an unstable critical pitch angle which separates helices that straighten into rods from helices that flatten into planar curves as they grow. At the critical angle the pitch angle remains constant, whereas both helical diameter and pitch increase with the helical contour length.
Collapse
Affiliation(s)
- Ariel Balter
- Department of Physics, Indiana University, Bloomington, IN 47405, USA
| | | |
Collapse
|
50
|
Abstract
The actin cytoskeleton is harnessed by several pathogenic bacteria that are capable of entering into non-phagocytic cells, the so-called 'invasive bacteria'. Among them, a few also exploit the host actin cytoskeleton to move intra- and inter-cellularly. Our knowledge of the basic mechanisms underlying actin-based motility has dramatically increased and the list of bacteria that are able to move in this way is also increasing including not only Listeria, Shigella and Rickettsia species but also Mycobacterium marinum and Burkholderia pseudomallei. In all cases the central player is the Arp2/3 complex. Vaccinia virus moves intracellularly on microtubules and just after budding, triggers actin polymerization and the formation of protrusions similar to that of adherent enteropathogenic Escherichia coli, that involve the Arp2/3 complex and facilitate its inter-cellular spread.
Collapse
Affiliation(s)
- Edith Gouin
- Unité des Interactions Bactéries-cellules, Institut Pasteur, 28 Rue du Docteur Roux, Paris 75015, France
| | | | | |
Collapse
|