1
|
Duan N, Lin M, Zhang W, Yan Q, Chien RC, Budachetri K, Denton S, Kawahara J, Lakritz J, Zhong Y, Dong Y, Rikihisa Y. Development of Etf-3-specific nanobodies to prevent Ehrlichia infection and LNP-mRNA delivery in cellular and murine models. Microbiol Res 2025; 292:128027. [PMID: 39705831 DOI: 10.1016/j.micres.2024.128027] [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: 08/07/2024] [Revised: 12/02/2024] [Accepted: 12/13/2024] [Indexed: 12/23/2024]
Abstract
Ehrlichia chaffeensis is an obligatory intracellular bacterium that infects monocytes and macrophages and causes human monocytic ehrlichiosis. Ehrlichia translocated factor-3 (Etf-3) is a type IV secretion system effector that binds host-cell ferritin light chain and induces ferritinophagy, thus increasing cellular labile iron pool for Ehrlichia proliferation. To further characterize roles of Etf-3 in Ehrlichia infection, we produced immune libraries of Etf-3-specific nanobodies (Nbs). Based on distinct complementarity-determining region 3 sequences, we identified 16 and 15 families of anti-Etf-3 Nbs that could specifically bind the N- and C-terminal halves of Etf-3, respectively. Transfection with plasmids encoding the anti-Etf-3 Nbs N48 and N51, but not N59, significantly inhibited E. chaffeensis infection in HEK293 cells. All three Nbs colocalized with Etf-3-GFP in co-transfected RF/6A cells, but N48 and N51 had significantly higher binding affinities for recombinant Etf-3. Etf-3-GFP transfection-induced ferritinophagy and endogenous ferritin degradation was abrogated in HEK293 cells co-transfected with N48 or N51, but not with N59. To efficiently express Nbs in the infected host-cell cytoplasm, lipid nanoparticles-encapsulated mRNAs (LNP-mRNAs) encoding N48, N51, or N59 were created for delivery into cells or mice. Incubation of HEK293 cells or inoculation of mice with LNP-mRNA-N48 or LNP-mRNA-N51 significantly inhibited E. chaffeensis infection compared to those with LNP-mRNA-N59 or without LNP-mRNA. Our results demonstrate that Etf-3-specific Nbs delivered via LNP-mRNAs can inhibit Etf-3 functions and Ehrlichia infection.
Collapse
Affiliation(s)
- Nan Duan
- Department of Veterinary Biosciences, The Ohio State University, 1925 Coffey Road, Columbus, OH 43210, United States
| | - Mingqun Lin
- Department of Veterinary Biosciences, The Ohio State University, 1925 Coffey Road, Columbus, OH 43210, United States
| | - Wenqing Zhang
- Department of Veterinary Biosciences, The Ohio State University, 1925 Coffey Road, Columbus, OH 43210, United States
| | - Qi Yan
- Department of Veterinary Biosciences, The Ohio State University, 1925 Coffey Road, Columbus, OH 43210, United States
| | - Rory C Chien
- Department of Veterinary Biosciences, The Ohio State University, 1925 Coffey Road, Columbus, OH 43210, United States
| | - Khemraj Budachetri
- Department of Veterinary Biosciences, The Ohio State University, 1925 Coffey Road, Columbus, OH 43210, United States
| | - Stephen Denton
- Department of Veterinary Biosciences, The Ohio State University, 1925 Coffey Road, Columbus, OH 43210, United States
| | - Jeffrey Kawahara
- Department of Veterinary Biosciences, The Ohio State University, 1925 Coffey Road, Columbus, OH 43210, United States
| | - Jeffrey Lakritz
- Department of Veterinary Preventive Medicine, The Ohio State University, 1925 Coffey Road, Columbus, OH 43210, United States
| | - Yichen Zhong
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| | - Yizhou Dong
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| | - Yasuko Rikihisa
- Department of Veterinary Biosciences, The Ohio State University, 1925 Coffey Road, Columbus, OH 43210, United States.
| |
Collapse
|
2
|
Croci C, Erriquez L, Bisaglia B, Bellinzona G, Olivieri E, Sassera D, Castelli M. Genome sequence of Ehrlichia muris from Ixodes ricinus collected in Italy on a migratory bird provides epidemiological and evolutionary insights. Ticks Tick Borne Dis 2024; 15:102409. [PMID: 39488869 DOI: 10.1016/j.ttbdis.2024.102409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 10/04/2024] [Accepted: 10/06/2024] [Indexed: 11/05/2024]
Abstract
Ticks are prominent vectors of several zoonotic diseases. Tick-borne pathogens include the members of the genus Ehrlichia, which are obligate intracellular bacteria infecting immune and hematopoietic cells. Ehrlichia muris predominantly affects rodents, but was also reported to be a human pathogen. The known geographical distribution of this bacterium ranges from Asia, to the USA and eastern Europe. In the present work, we report the finding of E. muris in an Ixodes ricinus tick collected from a migratory bird (Turdus iliacus) in Italy, southern Europe. We sequenced the total DNA from this tick sample, and, thanks to a dedicated bioinformatic pipeline, selectively assembled the genome of the bacterium, which represents the first one for E. muris from Europe. Phylogenetic and comparative genomic analyses were then performed. Accounting for tick species distribution, bird migratory routes, and molecular phylogeny of the bacterium, it is likely that this bird transported the tick to Italy from an endemic area of E. muris, such as eastern Europe. In addition, comparative genomic analyses highlighted that E. muris and other Ehrlichia spp. display copy number variations in two families of membrane proteins, likely due to recent gene duplication, deletion and recombination events. These differences are probably a source of variability for surface antigens to evade host immunity, with a potential role in host adaptation and specificity. The present results underline the impact of migratory birds on the spread of tick-borne pathogens towards non-endemic areas, highlighting the need for further epidemiological surveillance at bird ringing stations in Italy, and advocating further investigations on possible local transmission of E. muris in competent mammalian hosts.
Collapse
Affiliation(s)
- Carlo Croci
- Department of Biology and Biotechnology, University of Pavia, Italy
| | - Luca Erriquez
- Department of Biology and Biotechnology, University of Pavia, Italy
| | | | - Greta Bellinzona
- Department of Biology and Biotechnology, University of Pavia, Italy
| | - Emanuela Olivieri
- Istituto Zooprofilattico Sperimentale della Lombardia e dell'Emilia Romagna, Pavia, Italy
| | - Davide Sassera
- Department of Biology and Biotechnology, University of Pavia, Italy; Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Michele Castelli
- Department of Biology and Biotechnology, University of Pavia, Italy.
| |
Collapse
|
3
|
Chien RC, Lin M, Duan N, Denton S, Kawahara J, Rikihisa Y. RipE expression correlates with high ATP levels in Ehrlichia, which confers resistance during the extracellular stage to facilitate a new cycle of infection. Front Cell Infect Microbiol 2024; 14:1416577. [PMID: 39411319 PMCID: PMC11473500 DOI: 10.3389/fcimb.2024.1416577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 08/28/2024] [Indexed: 10/19/2024] Open
Abstract
Ehrlichiosis is a potentially life-threatening disease caused by infection with the obligatory intracellular bacteria Ehrlichia species. Ehrlichia japonica infection of mice provides an animal model of ehrlichiosis as it recapitulates full-spectrum and lethal ehrlichiosis in humans. The E. japonica transposon mutant of EHF0962, which encodes a previously uncharacterized hypothetical protein, is attenuated in both infection and virulence in mice. EHF0962 was hence named here as resistance-inducing protein of Ehrlichia (RipE). Using this ΔripE mutant, we studied how RipE protein contributes to Ehrlichia pathogenesis. Ehrlichia species have an intracellular developmental cycle and a brief extracellular stage to initiate a new cycle of infection. Majority of RipE proteins were expressed on the surface of the smaller infectious dense-core stage of bacteria. Extracellular ΔripE E. japonica contained significantly less adenosine triphosphate (ATP) and lost infectivity more rapidly in culture compared with wild-type (WT) E. japonica. Genetic complementation in the ΔripE mutant or overexpression of ripE in WT E. japonica significantly increased bacterial ATP levels, and RipE-overexpressing E. japonica was more virulent in mice than WT E. japonica. RipE is conserved among Ehrlichia species. Immunization of mice with recombinant RipE induced an in vitro infection-neutralizing antibody, significantly prolonged survival time after a lethal dose of E. japonica challenge, and cross-protected mice from infection by Ehrlichia chaffeensis, the agent of human monocytic ehrlichiosis. Our findings shed light on the extracellular stage of Ehrlichia, highlighting the importance of RipE and ATP levels in Ehrlichia for extracellular resistance and the next cycle of infection. Thus, RipE is a critical Ehrlichia protein for infection as such can be a potential vaccine target for ehrlichiosis.
Collapse
Affiliation(s)
| | | | | | | | | | - Yasuko Rikihisa
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
4
|
Bonin JL, Torres SR, Marcinkiewicz AL, Duhamel GE, Yang X, Pal U, DiSpirito JM, Nowak TA, Lin YP, MacNamara KC. Impact of E. muris infection on B. burgdorferi-induced joint pathology in mice. Front Immunol 2024; 15:1430419. [PMID: 39229265 PMCID: PMC11368855 DOI: 10.3389/fimmu.2024.1430419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 07/30/2024] [Indexed: 09/05/2024] Open
Abstract
Tick-borne infections are increasing in the United States and around the world. The most common tick-borne disease in the United States is Lyme disease caused by infection with the spirochete Borrelia burgdorferi (Bb), and pathogenesis varies from subclinical to severe. Bb infection is transmitted by Ixodes ticks, which can carry multiple other microbial pathogens, including Ehrlichia species. To address how the simultaneous inoculation of a distinct pathogen impacted the course of Bb-induced disease, we used C57BL/6 (B6) mice which are susceptible to Bb infection but develop only mild joint pathology. While infection of B6 mice with Bb alone resulted in minimal inflammatory responses, mice co-infected with both Bb and the obligate intracellular pathogen Ehrlichia muris (Em) displayed hematologic changes, inflammatory cytokine production, and emergency myelopoiesis similar to what was observed in mice infected only with Em. Moreover, infection of B6 mice with Bb alone resulted in no detectable joint inflammation, whereas mice co-infected with both Em and Bb exhibited significant inflammation of the ankle joint. Our findings support the concept that co-infection with Ehrlichia can exacerbate inflammation, resulting in more severe Bb-induced disease.
Collapse
Affiliation(s)
- Jesse L. Bonin
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY, United States
| | - Steven R. Torres
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY, United States
| | - Ashley L. Marcinkiewicz
- Division of Infectious Disease, Wadsworth Center, New York State Department of Health, Albany, NY, United States
| | - Gerald E. Duhamel
- New York State Animal Health Diagnostic Center and Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
| | - Xiuli Yang
- Department of Veterinary Medicine, Virginia-Maryland Regional College of Veterinary Medicine, University of Maryland, College Park, MD, United States
| | - Utpal Pal
- Department of Veterinary Medicine, Virginia-Maryland Regional College of Veterinary Medicine, University of Maryland, College Park, MD, United States
| | - Julia M. DiSpirito
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY, United States
| | - Tristan A. Nowak
- Division of Infectious Disease, Wadsworth Center, New York State Department of Health, Albany, NY, United States
- Department of Biomedical Sciences, State University of New York at Albany, Albany, NY, United States
| | - Yi-Pin Lin
- Division of Infectious Disease, Wadsworth Center, New York State Department of Health, Albany, NY, United States
- Department of Biomedical Sciences, State University of New York at Albany, Albany, NY, United States
| | - Katherine C. MacNamara
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY, United States
| |
Collapse
|
5
|
Li M, Yang N, Li X, Duan N, Qin S, Wang M, Zhou Y, Jin Y, Wu W, Jin S, Cheng Z. Host Cells Upregulate Phosphate Transporter PIT1 to Inhibit Ehrlichia chaffeensis Intracellular Growth. Int J Mol Sci 2024; 25:7895. [PMID: 39063137 PMCID: PMC11276888 DOI: 10.3390/ijms25147895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 07/12/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024] Open
Abstract
Ehrlichia chaffeensis infects and proliferates inside monocytes or macrophages and causes human monocytic ehrlichiosis (HME), an emerging life-threatening tick-borne zoonosis. After internalization, E. chaffeensis resides in specialized membrane-bound inclusions, E. chaffeensis-containing vesicles (ECVs), to evade from host cell innate immune responses and obtain nutrients. However, mechanisms exploited by host cells to inhibit E. chaffeensis growth in ECVs are still largely unknown. Here we demonstrate that host cells recognize E. chaffeensis Ech_1067, a penicillin-binding protein, and then upregulate the expression of PIT1, which is a phosphate transporter and transports phosphate from ECVs to the cytosol to inhibit bacterial growth. We found that host cells upregulate the PIT1 expression upon E. chaffeensis infection using transcriptome sequencing, qRT-PCR and Western blotting, and PIT1 is localized on the ECV membrane in infected THP-1 cells using confocal microscopy. Silence of PIT1 using shRNA enhances E. chaffeensis intracellular growth. Finally, we found that E. chaffeensis Ech_1067 induces the upregulation of PIT1 expression through the MyD88-NF-κB pathway using recombinant protein for stimulation and siRNA for silence. Our findings deepen the understanding of the innate immune responses of host cells to inhibit bacterial intracellular growth and facilitate the development of new therapeutics for HME.
Collapse
Affiliation(s)
- Meifang Li
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin 300071, China; (M.L.); (N.Y.); (X.L.); (N.D.); (S.Q.); (M.W.); (Y.Z.); (Y.J.); (W.W.); (S.J.)
- Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Nan Yang
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin 300071, China; (M.L.); (N.Y.); (X.L.); (N.D.); (S.Q.); (M.W.); (Y.Z.); (Y.J.); (W.W.); (S.J.)
- Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Xiaoxiao Li
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin 300071, China; (M.L.); (N.Y.); (X.L.); (N.D.); (S.Q.); (M.W.); (Y.Z.); (Y.J.); (W.W.); (S.J.)
- Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Nan Duan
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin 300071, China; (M.L.); (N.Y.); (X.L.); (N.D.); (S.Q.); (M.W.); (Y.Z.); (Y.J.); (W.W.); (S.J.)
- Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Shanhua Qin
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin 300071, China; (M.L.); (N.Y.); (X.L.); (N.D.); (S.Q.); (M.W.); (Y.Z.); (Y.J.); (W.W.); (S.J.)
- Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Mengyao Wang
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin 300071, China; (M.L.); (N.Y.); (X.L.); (N.D.); (S.Q.); (M.W.); (Y.Z.); (Y.J.); (W.W.); (S.J.)
- Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Yuhong Zhou
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin 300071, China; (M.L.); (N.Y.); (X.L.); (N.D.); (S.Q.); (M.W.); (Y.Z.); (Y.J.); (W.W.); (S.J.)
- Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Yongxin Jin
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin 300071, China; (M.L.); (N.Y.); (X.L.); (N.D.); (S.Q.); (M.W.); (Y.Z.); (Y.J.); (W.W.); (S.J.)
- Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Weihui Wu
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin 300071, China; (M.L.); (N.Y.); (X.L.); (N.D.); (S.Q.); (M.W.); (Y.Z.); (Y.J.); (W.W.); (S.J.)
- Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Shouguang Jin
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin 300071, China; (M.L.); (N.Y.); (X.L.); (N.D.); (S.Q.); (M.W.); (Y.Z.); (Y.J.); (W.W.); (S.J.)
- Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Zhihui Cheng
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin 300071, China; (M.L.); (N.Y.); (X.L.); (N.D.); (S.Q.); (M.W.); (Y.Z.); (Y.J.); (W.W.); (S.J.)
- Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| |
Collapse
|
6
|
Yang N, Li M, Qin S, Duan N, Li X, Zhou Y, Wang M, Jin Y, Wu W, Cheng Z. Ehrlichia chaffeensis Etf-3 Induces Host RAB15 Upregulation for Bacterial Intracellular Growth. Int J Mol Sci 2024; 25:2551. [PMID: 38473798 DOI: 10.3390/ijms25052551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 02/18/2024] [Accepted: 02/20/2024] [Indexed: 03/14/2024] Open
Abstract
Ehrlichia chaffeensis infects human monocytes or macrophages and causes human monocytic ehrlichiosis (HME), an emerging life-threatening zoonosis. After internalization, E. chaffeensis resides in membrane-bound inclusions, E. chaffeensis-containing vesicles (ECVs), which have early endosome-like characteristics and fuse with early autophagosomes but not lysosomes, to evade host innate immune microbicidal mechanisms and obtain nutrients for bacterial intracellular growth. The mechanisms exploited by E. chaffeensis to modulate intracellular vesicle trafficking in host cells have not been comprehensively studied. Here, we demonstrate that E. chaffeensis type IV secretion system (T4SS) effector Etf-3 induces RAB15 upregulation in host cells and that RAB15, which is localized on ECVs, inhibits ECV fusion with lysosomes and induces autophagy. We found that E. chaffeensis infection upregulated RAB15 expression using qRT-PCR, and RAB15 was colocalized with E. chaffeensis using confocal microscopy. Silence of RAB15 using siRNA enhanced ECV maturation to late endosomes and fusion with lysosomes, as well as inhibited host cell autophagy. Overexpression of Etf-3 in host cells specifically induced RAB15 upregulation and autophagy. Our findings deepen the understanding of E. chaffeensis pathogenesis and adaptation in hosts as well as the function of RAB15 and facilitate the development of new therapeutics for HME.
Collapse
Affiliation(s)
- Nan Yang
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin 300071, China
- Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Meifang Li
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin 300071, China
- Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Shanhua Qin
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin 300071, China
- Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Nan Duan
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin 300071, China
- Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Xiaoxiao Li
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin 300071, China
- Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Yuhong Zhou
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin 300071, China
- Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Mengyao Wang
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin 300071, China
- Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Yongxin Jin
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin 300071, China
- Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Weihui Wu
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin 300071, China
- Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Zhihui Cheng
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin 300071, China
- Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| |
Collapse
|
7
|
Sharma AK, Ismail N. Non-Canonical Inflammasome Pathway: The Role of Cell Death and Inflammation in Ehrlichiosis. Cells 2023; 12:2597. [PMID: 37998332 PMCID: PMC10670716 DOI: 10.3390/cells12222597] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 11/01/2023] [Accepted: 11/07/2023] [Indexed: 11/25/2023] Open
Abstract
Activating inflammatory caspases and releasing pro-inflammatory mediators are two essential functions of inflammasomes which are triggered in response to pathogen-associated molecular patterns (PAMPs) or danger-associated molecular patterns (DAMPs). The canonical inflammasome pathway involves the activation of inflammasome and its downstream pathway via the adaptor ASC protein, which causes caspase 1 activation and, eventually, the cleavage of pro-IL-1b and pro-IL-18. The non-canonical inflammasome pathway is induced upon detecting cytosolic lipopolysaccharide (LPS) by NLRP3 inflammasome in Gram-negative bacteria. The activation of NLRP3 triggers the cleavage of murine caspase 11 (human caspase 4 or caspase 5), which results in the formation of pores (via gasdermin) to cause pyroptosis. Ehrlichia is an obligately intracellular bacterium which is responsible for causing human monocytic ehrlichiosis (HME), a potentially lethal disease similar to toxic shock syndrome and septic shock syndrome. Several studies have indicated that canonical and non-canonical inflammasome activation is a crucial pathogenic mechanism that induces dysregulated inflammation and host cellular death in the pathophysiology of HME. Mechanistically, the activation of canonical and non-canonical inflammasome pathways affected by virulent Ehrlichia infection is due to a block in autophagy. This review aims to explore the significance of non-canonical inflammasomes in ehrlichiosis, and how the pathways involving caspases (with the exception of caspase 1) contribute to the pathophysiology of severe and fatal ehrlichiosis. Improving our understanding of the non-canonical inflammatory pathway that cause cell death and inflammation in ehrlichiosis will help the advancement of innovative therapeutic, preventative, and diagnostic approaches to the treatment of ehrlichiosis.
Collapse
Affiliation(s)
| | - Nahed Ismail
- Department of Pathology, College of Medicine, University of Illinois at Chicago, Chicago, IL 60607, USA;
| |
Collapse
|
8
|
Clemente TM, Angara RK, Gilk SD. Establishing the intracellular niche of obligate intracellular vacuolar pathogens. Front Cell Infect Microbiol 2023; 13:1206037. [PMID: 37645379 PMCID: PMC10461009 DOI: 10.3389/fcimb.2023.1206037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 07/21/2023] [Indexed: 08/31/2023] Open
Abstract
Obligate intracellular pathogens occupy one of two niches - free in the host cell cytoplasm or confined in a membrane-bound vacuole. Pathogens occupying membrane-bound vacuoles are sequestered from the innate immune system and have an extra layer of protection from antimicrobial drugs. However, this lifestyle presents several challenges. First, the bacteria must obtain membrane or membrane components to support vacuole expansion and provide space for the increasing bacteria numbers during the log phase of replication. Second, the vacuole microenvironment must be suitable for the unique metabolic needs of the pathogen. Third, as most obligate intracellular bacterial pathogens have undergone genomic reduction and are not capable of full metabolic independence, the bacteria must have mechanisms to obtain essential nutrients and resources from the host cell. Finally, because they are separated from the host cell by the vacuole membrane, the bacteria must possess mechanisms to manipulate the host cell, typically through a specialized secretion system which crosses the vacuole membrane. While there are common themes, each bacterial pathogen utilizes unique approach to establishing and maintaining their intracellular niches. In this review, we focus on the vacuole-bound intracellular niches of Anaplasma phagocytophilum, Ehrlichia chaffeensis, Chlamydia trachomatis, and Coxiella burnetii.
Collapse
Affiliation(s)
| | | | - Stacey D. Gilk
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, United States
| |
Collapse
|
9
|
Fisher DJ, Beare PA. Recent advances in genetic systems in obligate intracellular human-pathogenic bacteria. Front Cell Infect Microbiol 2023; 13:1202245. [PMID: 37404720 PMCID: PMC10315504 DOI: 10.3389/fcimb.2023.1202245] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 05/22/2023] [Indexed: 07/06/2023] Open
Abstract
The ability to genetically manipulate a pathogen is fundamental to discovering factors governing host-pathogen interactions at the molecular level and is critical for devising treatment and prevention strategies. While the genetic "toolbox" for many important bacterial pathogens is extensive, approaches for modifying obligate intracellular bacterial pathogens were classically limited due in part to the uniqueness of their obligatory lifestyles. Many researchers have confronted these challenges over the past two and a half decades leading to the development of multiple approaches to construct plasmid-bearing recombinant strains and chromosomal gene inactivation and deletion mutants, along with gene-silencing methods enabling the study of essential genes. This review will highlight seminal genetic achievements and recent developments (past 5 years) for Anaplasma spp., Rickettsia spp., Chlamydia spp., and Coxiella burnetii including progress being made for the still intractable Orientia tsutsugamushi. Alongside commentary of the strengths and weaknesses of the various approaches, future research directions will be discussed to include methods for C. burnetii that should have utility in the other obligate intracellular bacteria. Collectively, the future appears bright for unraveling the molecular pathogenic mechanisms of these significant pathogens.
Collapse
Affiliation(s)
- Derek J. Fisher
- School of Biological Sciences, Southern Illinois University, Carbondale, IL, United States
| | - Paul A. Beare
- Rocky Mountain Laboratory, National Institute of Health, Hamilton, MT, United States
| |
Collapse
|
10
|
Gordon JL, Oliva Chavez AS, Martinez D, Vachiery N, Meyer DF. Possible biased virulence attenuation in the Senegal strain of Ehrlichia ruminantium by ntrX gene conversion from an inverted segmental duplication. PLoS One 2023; 18:e0266234. [PMID: 36800354 PMCID: PMC9937504 DOI: 10.1371/journal.pone.0266234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 03/16/2022] [Indexed: 02/18/2023] Open
Abstract
Ehrlichia ruminantium is a tick-borne intracellular pathogen of ruminants that causes heartwater, a disease present in Sub-saharan Africa, islands in the Indian Ocean and the Caribbean, inducing significant economic losses. At present, three avirulent strains of E. ruminantium (Gardel, Welgevonden and Senegal isolates) have been produced by a process of serial passaging in mammalian cells in vitro, but unfortunately their use as vaccines do not offer a large range of protection against other strains, possibly due to the genetic diversity present within the species. So far no genetic basis for virulence attenuation has been identified in any E. ruminantium strain that could offer targets to facilitate vaccine production. Virulence attenuated Senegal strains have been produced twice independently, and require many fewer passages to attenuate than the other strains. We compared the genomes of a virulent and attenuated Senegal strain and identified a likely attenuator gene, ntrX, a global transcription regulator and member of a two-component system that is linked to environmental sensing. This gene has an inverted partial duplicate close to the parental gene that shows evidence of gene conversion in different E. ruminantium strains. The pseudogenisation of the gene in the avirulent Senegal strain occurred by gene conversion from the duplicate to the parent, transferring a 4 bp deletion which is unique to the Senegal strain partial duplicate amongst the wild isolates. We confirmed that the ntrX gene is not expressed in the avirulent Senegal strain by RT-PCR. The inverted duplicate structure combined with the 4 bp deletion in the Senegal strain can explain both the attenuation and the faster speed of attenuation in the Senegal strain relative to other strains of E. ruminantium. Our results identify nrtX as a promising target for the generation of attenuated strains of E. ruminantium by random or directed mutagenesis that could be used for vaccine production.
Collapse
Affiliation(s)
- Jonathan L. Gordon
- CIRAD, UMR ASTRE, Petit-Bourg, Guadeloupe, France
- ASTRE, CIRAD, INRAe, Univ Montpellier, Montpellier, France
| | - Adela S. Oliva Chavez
- CIRAD, UMR ASTRE, Petit-Bourg, Guadeloupe, France
- ASTRE, CIRAD, INRAe, Univ Montpellier, Montpellier, France
| | | | | | - Damien F. Meyer
- CIRAD, UMR ASTRE, Petit-Bourg, Guadeloupe, France
- ASTRE, CIRAD, INRAe, Univ Montpellier, Montpellier, France
- * E-mail:
| |
Collapse
|
11
|
Liang Q, Yan J, Zhang S, Yang N, Li M, Jin Y, Bai F, Wu W, Cheng Z. CtrA activates the expression of glutathione S-transferase conferring oxidative stress resistance to Ehrlichia chaffeensis. Front Cell Infect Microbiol 2022; 12:1081614. [PMID: 36579340 PMCID: PMC9791040 DOI: 10.3389/fcimb.2022.1081614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 11/24/2022] [Indexed: 12/14/2022] Open
Abstract
Ehrlichia chaffeensis, the causative agent of human monocytic ehrlichiosis (HME), is a Gram-negative obligatory intracellular bacterium, which infects and multiplies in human monocytes and macrophages. Host immune cells produce reactive oxygen species (ROS) to eliminate E. chaffeensis upon infection. E. chaffeensis global transcriptional regulator CtrA activates the expression of GshA and GshB to synthesize glutathione (GSH), the most potent natural antioxidant, upon oxidative stress to combat ROS damage. However, the mechanisms exploited by E. chaffeensis to utilize GSH are still unknown. Here, we found that in E. chaffeensis CtrA activated the expression of glutathione S-transferase (GST) upon oxidative stress, and E. chaffeensis GST utilizes GSH to eliminate ROS and confers the oxidative stress resistance to E. chaffeensis. We found that CtrA bound to the promoter regions of 211 genes, including gst, in E. chaffeensis using chromatin immunoprecipitation coupled to deep sequencing (ChIP-seq). Recombinant E. chaffeensis CtrA directly bound to the gst promoter region determined with electrophoretic mobility shift assay (EMSA), and activated the gst expression determined with reporter assay. Recombinant GST showed GSH conjugation activity towards its typical substrate 2,4-dinitrochlorobenzene (CDNB) in vitro and peptide nucleic acid (PNA) transfection of E. chaffeensis, which can knock down the gst transcription level, reduced bacterial survival upon oxidative stress. Our results demonstrate that E. chaffeensis CtrA regulates GSH utilization, which plays a critical role in resistance to oxidative stress, and aid in the development of new therapeutics for HME.
Collapse
|
12
|
Liu H, Knox CA, Jakkula LUMR, Wang Y, Peddireddi L, Ganta RR. Evaluating EcxR for Its Possible Role in Ehrlichia chaffeensis Gene Regulation. Int J Mol Sci 2022; 23:12719. [PMID: 36361509 PMCID: PMC9657007 DOI: 10.3390/ijms232112719] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 10/17/2022] [Accepted: 10/19/2022] [Indexed: 04/14/2024] Open
Abstract
Ehrlichia chaffeensis, a tick-transmitted intraphagosomal bacterium, is the causative agent of human monocytic ehrlichiosis. The pathogen also infects several other vertebrate hosts. E. chaffeensis has a biphasic developmental cycle during its growth in vertebrate monocytes/macrophages and invertebrate tick cells. Host- and vector-specific differences in the gene expression from many genes of E. chaffeensis are well documented. It is unclear how the organism regulates gene expression during its developmental cycle and for its adaptation to vertebrate and tick host cell environments. We previously mapped promoters of several E. chaffeensis genes which are recognized by its only two sigma factors: σ32 and σ70. In the current study, we investigated in assessing five predicted E. chaffeensis transcription regulators; EcxR, CtrA, MerR, HU and Tr1 for their possible roles in regulating the pathogen gene expression. Promoter segments of three genes each transcribed with the RNA polymerase containing σ70 (HU, P28-Omp14 and P28-Omp19) and σ32 (ClpB, DnaK and GroES/L) were evaluated by employing multiple independent molecular methods. We report that EcxR binds to all six promoters tested. Promoter-specific binding of EcxR to several gene promoters results in varying levels of gene expression enhancement. This is the first detailed molecular characterization of transcription regulators where we identified EcxR as a gene regulator having multiple promoter-specific interactions.
Collapse
Affiliation(s)
| | | | | | | | | | - Roman R. Ganta
- Center of Excellence for Vector-Borne Diseases (CEVBD), Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA
| |
Collapse
|
13
|
Yan J, Liang Q, Chai Z, Duan N, Li X, Liu Y, Yang N, Li M, Jin Y, Bai F, Wu W, Cheng Z. Glutathione Synthesis Regulated by CtrA Protects Ehrlichia chaffeensis From Host Cell Oxidative Stress. Front Microbiol 2022; 13:846488. [PMID: 35432225 PMCID: PMC9005958 DOI: 10.3389/fmicb.2022.846488] [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: 12/31/2021] [Accepted: 02/08/2022] [Indexed: 11/13/2022] Open
Abstract
Ehrlichia chaffeensis, a small Gram-negative obligatory intracellular bacterium, infects human monocytes or macrophages, and causes human monocytic ehrlichiosis, one of the most prevalent, life-threatening emerging zoonoses. Reactive oxygen species are produced by the host immune cells in response to bacterial infections. The mechanisms exploited by E. chaffeensis to resist oxidative stress have not been comprehensively demonstrated. Here, we found that E. chaffeensis encodes two functional enzymes, GshA and GshB, to synthesize glutathione that confers E. chaffeensis the oxidative stress resistance, and that the expression of gshA and gshB is upregulated by CtrA, a global transcriptional regulator, upon oxidative stress. We found that in E. chaffeensis, the expression of gshA and gshB was upregulated upon oxidative stress using quantitative RT-PCR. Ehrlichia chaffeensis GshA or GshB restored the ability of Pseudomonas aeruginosa GshA or GshB mutant to cope with oxidative stress, respectively. Recombinant E. chaffeensis CtrA directly bound to the promoters of gshA and gshB, determined with electrophoretic mobility shift assay, and activated the expression of gshA and gshB determined with reporter assay. Peptide nucleic acid transfection of E. chaffeensis, which reduced the CtrA protein level, inhibited the oxidative stress-induced upregulation of gshA and gshB. Our findings provide insights into the function and regulation of the two enzymes critical for E. chaffeensis resistance to oxidative stress and may deepen our understanding of E. chaffeensis pathogenesis and adaptation in hosts.
Collapse
Affiliation(s)
- Jiaqi Yan
- Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, China
| | - Qi'an Liang
- Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, China
| | - Zhouyi Chai
- Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, China
| | - Nan Duan
- Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, China
| | - Xiaoxiao Li
- Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, China
| | - Yajing Liu
- Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, China
| | - Nan Yang
- Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, China
| | - Meifang Li
- Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, China
| | - Yongxin Jin
- Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, China
| | - Fang Bai
- Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, China
| | - Weihui Wu
- Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, China
| | - Zhihui Cheng
- Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, China
| |
Collapse
|
14
|
Rikihisa Y. The "Biological Weapons" of Ehrlichia chaffeensis: Novel Molecules and Mechanisms to Subjugate Host Cells. Front Cell Infect Microbiol 2022; 11:830180. [PMID: 35155275 PMCID: PMC8834651 DOI: 10.3389/fcimb.2021.830180] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 12/20/2021] [Indexed: 12/22/2022] Open
Abstract
Ehrlichia chaffeensis is an obligatory intracellular bacterium that causes human monocytic ehrlichiosis, an emerging, potentially fatal tick-borne infectious disease. The bacterium enters human cells via the binding of its unique outer-membrane invasin EtpE to the cognate receptor DNase X on the host-cell plasma membrane; this triggers actin polymerization and filopodia formation at the site of E. chaffeensis binding, and blocks activation of phagocyte NADPH oxidase that catalyzes the generation of microbicidal reactive oxygen species. Subsequently, the bacterium replicates by hijacking/dysregulating host-cell functions using Type IV secretion effectors. For example, the Ehrlichia translocated factor (Etf)-1 enters mitochondria and inhibits mitochondria-mediated apoptosis of host cells. Etf-1 also induces autophagy mediated by the small GTPase RAB5, the result being the liberation of catabolites for proliferation inside host cells. Moreover, Etf-2 competes with the RAB5 GTPase-activating protein, for binding to RAB5-GTP on the surface of E. chaffeensis inclusions, which blocks GTP hydrolysis and consequently prevents the fusion of inclusions with host-cell lysosomes. Etf-3 binds ferritin light chain to induce ferritinophagy to obtain intracellular iron. To enable E. chaffeensis to rapidly adapt to the host environment and proliferate, the bacterium must acquire host membrane cholesterol and glycerophospholipids for the purpose of producing large amounts of its own membrane. Future studies on the arsenal of unique Ehrlichia molecules and their interplay with host-cell components will undoubtedly advance our understanding of the molecular mechanisms of obligatory intracellular infection and may identify hitherto unrecognized signaling pathways of human hosts. Such data could be exploited for development of treatment and control measures for ehrlichiosis as well as other ailments that potentially could involve the same host-cell signaling pathways that are appropriated by E. chaffeensis.
Collapse
Affiliation(s)
- Yasuko Rikihisa
- Laboratory of Molecular, Cellular, and Environmental Rickettsiology, Department of Veterinary Biosciences, College of Veterinary Medicine, Infectious Diseases Institute, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
15
|
Ismail N, Sharma A, Soong L, Walker DH. Review: Protective Immunity and Immunopathology of Ehrlichiosis. ZOONOSES (BURLINGTON, MASS.) 2022; 2:10.15212/zoonoses-2022-0009. [PMID: 35876763 PMCID: PMC9300479 DOI: 10.15212/zoonoses-2022-0009] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Human monocytic ehrlichiosis, a tick transmitted infection, ranges in severity from apparently subclinical to a fatal toxic shock-like fatal disease. Models in immunocompetent mice range from an abortive infection to uniformly lethal depending on the infecting Ehrlichia species, dose of inoculum, and route of inoculation. Effective immunity is mediated by CD4+ T lymphocytes and gamma interferon. Lethal infection occurs with early overproduction of proinflammatory cytokines and overproduction of TNF alpha and IL-10 by CD8+ T lymphocytes. Furthermore, fatal ehrlichiosis is associated with signaling via TLR 9/MyD88 with upregulation of several inflammasome complexes and secretion of IL-1 beta, IL-1 alpha, and IL-18 by hepatic mononuclear cells, suggesting activation of canonical and noncanonical inflammasome pathways, a deleterious role for IL-18, and the protective role for caspase 1. Autophagy promotes ehrlichial infection, and MyD88 signaling hinders ehrlichial infection by inhibiting autophagy induction and flux. Activation of caspase 11 during infection of hepatocytes by the lethal ehrlichial species after interferon alpha receptor signaling results in the production of inflammasome-dependent IL-1 beta, extracellular secretion of HMGB1, and pyroptosis. The high level of HMGB1 in lethal ehrlichiosis suggests a role in toxic shock. Studies of primary bone marrow-derived macrophages infected by highly avirulent or mildly avirulent ehrlichiae reveal divergent M1 and M2 macrophage polarization that links with generation of pathogenic CD8 T cells, neutrophils, and excessive inflammation or with strong expansion of protective Th1 and NKT cells, resolution of inflammation and clearance of infection, respectively.
Collapse
Affiliation(s)
- Nahed Ismail
- Clinical Microbiology, Laboratory Medicine, University of Illinois at Chicago-College of Medicine, University of Illinois Hospitals & Health Science System, Chicago, IL
| | - Aditya Sharma
- Clinical Microbiology, Laboratory Medicine, University of Illinois at Chicago-College of Medicine, University of Illinois Hospitals & Health Science System, Chicago, IL
| | - Lynn Soong
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX
- Department of Pathology, Center for Biodefense & Emerging Infectious Diseases, University of Texas Medical Branch, Galveston, TX
| | - David H. Walker
- Department of Pathology, Center for Biodefense & Emerging Infectious Diseases, University of Texas Medical Branch, Galveston, TX
| |
Collapse
|
16
|
Duan N, Ma X, Cui H, Wang Z, Chai Z, Yan J, Li X, Feng Y, Cao Y, Jin Y, Bai F, Wu W, Rikihisa Y, Cheng Z. Insights into the mechanism regulating the differential expression of the P28-OMP outer membrane proteins in obligatory intracellular pathogen Ehrlichia chaffeensis. Emerg Microbes Infect 2021; 10:461-471. [PMID: 33660592 PMCID: PMC7971322 DOI: 10.1080/22221751.2021.1899054] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Ehrlichia chaffeensis causes human monocytic ehrlichiosis (HME), which is one of the most prevalent, life-threatening emerging infectious zoonoses. The life cycle of E. chaffeensis includes ticks and mammals, in which E. chaffeensis proteins are expressed differentially contributing to bacterial survival and infection. Among the E. chaffeensis P28-OMP outer membrane proteins, OMP-1B and P28 are predominantly expressed in tick cells and mammalian macrophages, respectively. The mechanisms regulating this differential expression have not been comprehensively studied. Here, we demonstrate that the transcriptional regulators EcxR and Tr1 regulate the differential expression of omp-1B and p28 in E. chaffeensis. Recombinant E. chaffeensis Tr1 bound to the promoters of omp-1B and p28, and transactivated omp-1B and p28 promoter-EGFP fusion constructs in Escherichia coli. The consensus sequence of Tr1 binding motifs was AC/TTATA as determined with DNase I footprint assay. Tr1 showed a higher affinity towards the p28 promoter than the omp-1B promoter as determined with surface plasmon resonance. EcxR activated the tr1 expression in response to a temperature decrease. At 37°C low level of Tr1 activated the p28 expression. At 25°C high level of Tr1 activated the omp-1B expression, while repressing the p28 expression by binding to an additional site upstream of the p28 gene. Our data provide insights into a novel mechanism mediated by Tr1 regulating E. chaffeensis differential gene expression, which may aid in the development of new therapeutics for HME.
Collapse
Affiliation(s)
- Nan Duan
- Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, People's Republic of China
| | - Xiaohui Ma
- Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, People's Republic of China
| | - Heting Cui
- Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, People's Republic of China
| | - Zhexuan Wang
- Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, People's Republic of China
| | - Zhouyi Chai
- Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, People's Republic of China
| | - Jiaqi Yan
- Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, People's Republic of China
| | - Xiaoxiao Li
- Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, People's Republic of China
| | - Yingxing Feng
- Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, People's Republic of China
| | - Yu Cao
- Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, People's Republic of China
| | - Yongxin Jin
- Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, People's Republic of China
| | - Fang Bai
- Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, People's Republic of China
| | - Weihui Wu
- Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, People's Republic of China
| | - Yasuko Rikihisa
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
| | - Zhihui Cheng
- Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, People's Republic of China
| |
Collapse
|
17
|
Immune Response and Apoptosis-Related Pathways Induced by Aeromonas schubertii Infection of Hybrid Snakehead ( Channa maculata♀ × Channa argus♂). Pathogens 2021; 10:pathogens10080997. [PMID: 34451461 PMCID: PMC8401259 DOI: 10.3390/pathogens10080997] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 07/27/2021] [Accepted: 08/04/2021] [Indexed: 11/17/2022] Open
Abstract
Aeromonas schubertii is the etiological pathogen of internal organ nodules in snakehead fish. Infections with A. schubertii produce a significant economic loss in aquaculture. Therefore, it is important to examine the immune mechanisms by which snakeheads defend against A. schubertii infection. In this study, we established a hybrid snakehead infection model by intraperitoneal injection of A. schubertii that produced internal organ nodules. The splenic immune response of infected fish was examined at the transcriptome level by Illumina-seq analysis. Results showed 14,796 differentially expressed genes (DEGs) following A. schubertii infection, including 4441 up-regulated unigenes and 10,355 down-regulated unigenes. KEGG analysis showed 2084 DEGs to be involved in 192 pathways, 14 of which were immune-related. Twelve DEGs were used to validate quantitative real-time PCR results with RNA-seq data. Time-course expression analysis of six genes demonstrated modulation of the snakehead immune response by A. schubertii. Furthermore, transcriptome analysis identified a substantial number of DEGs that were involved in the apoptosis signaling pathway. TUNEL analysis of infected spleens confirmed the presence of apoptotic cells. This study provided new information for a further understanding of the pathogenesis of A. schubertii in snakeheads, which can be used to prevent and possibly treat A. schubertii infections.
Collapse
|
18
|
Functional Characterization of Multiple Ehrlichia chaffeensis Sodium (Cation)/Proton Antiporter Genes Involved in the Bacterial pH Homeostasis. Int J Mol Sci 2021; 22:ijms22168420. [PMID: 34445146 PMCID: PMC8395091 DOI: 10.3390/ijms22168420] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 07/30/2021] [Accepted: 08/02/2021] [Indexed: 11/16/2022] Open
Abstract
Ehrlichia chaffeensis causes human monocytic ehrlichiosis. Little is known about how this and other related tick-borne rickettsia pathogens maintain pH homeostasis in acidified phagosomes and the extracellular milieu. The membrane-bound sodium (cation)/proton antiporters are found in a wide range of organisms aiding pH homeostasis. We recently reported a mutation in an antiporter gene of E. chaffeensis (ECH_0379) which causes bacterial in vivo attenuation. The E. chaffeensis genome contains 10 protein coding sequences encoding for predicted antiporters. We report here that nine of these genes are transcribed during the bacterial growth in macrophages and tick cells. All E. chaffeensis antiporter genes functionally complemented antiporter deficient Escherichia coli. Antiporter activity for all predicted E. chaffeensis genes was observed at pH 5.5, while gene products of ECH_0179 and ECH_0379 were also active at pH 8.0, and ECH_0179 protein was complemented at pH 7.0. The antiporter activity was independently verified for the ECH_0379 protein by proteoliposome diffusion analysis. This is the first description of antiporters in E. chaffeensis and demonstrates that the pathogen contains multiple antiporters with varying biological functions, which are likely important for the pH homeostasis of the pathogen’s replicating and infectious forms.
Collapse
|
19
|
Mutations in Ehrlichia chaffeensis Genes ECH_0660 and ECH_0665 Cause Transcriptional Changes in Response to Zinc or Iron Limitation. J Bacteriol 2021; 203:e0002721. [PMID: 33875547 PMCID: PMC8316085 DOI: 10.1128/jb.00027-21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Ehrlichia chaffeensis causes human monocytic ehrlichiosis by replicating within phagosomes of monocytes/macrophages. A function disruption mutation within the pathogen's ECH_0660 gene, which encodes a phage head-to-tail connector protein, resulted in the rapid clearance of the pathogen in vivo, while aiding in induction of sufficient immunity in a host to protect against wild-type infection challenge. In this study, we describe the characterization of a cluster of seven genes spanning from ECH_0659 to ECH_0665, which contained four genes encoding bacterial phage proteins, including the ECH_0660 gene. Assessment of the promoter region upstream of the first gene of the seven genes (ECH_0659) in Escherichia coli demonstrated transcriptional enhancement under zinc and iron starvation conditions. Furthermore, transcription of the seven genes was significantly higher under zinc and iron starvation conditions for E. chaffeensis carrying a mutation in the ECH_0660 gene compared to the wild-type pathogen. In contrast, for the ECH_0665 gene mutant with the function disruption, transcription from the genes was mostly similar to that of the wild type or was moderately downregulated. Recently, we reported that this mutation caused a minimal impact on the pathogen's in vivo growth, as it persisted similarly to the wild type. The current study is the first to describe how zinc and iron contribute to E. chaffeensis biology. Specifically, we demonstrated that the functional disruption in the gene encoding the phage head-to-tail connector protein in E. chaffeensis results in the enhanced transcription of seven genes, including those encoding phage proteins, under zinc and iron limitation. IMPORTANCE Ehrlichia chaffeensis, a tick-transmitted bacterium, causes human monocytic ehrlichiosis by replicating within phagosomes of monocytes/macrophages. A function disruption mutation within the pathogen's gene encoding a phage head-to-tail connector protein resulted in the rapid clearance of the pathogen in vivo, while aiding in induction of sufficient immunity in a host to protect against wild-type infection challenge. In the current study, we investigated if the functional disruption in the phage head-to-tail connector protein gene caused transcriptional changes resulting from metal ion limitations. This is the first study describing how zinc and iron may contribute to E. chaffeensis replication.
Collapse
|
20
|
Revisiting Ehrlichia ruminantium Replication Cycle Using Proteomics: The Host and the Bacterium Perspectives. Microorganisms 2021; 9:microorganisms9061144. [PMID: 34073568 PMCID: PMC8229282 DOI: 10.3390/microorganisms9061144] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 04/28/2021] [Accepted: 05/07/2021] [Indexed: 12/16/2022] Open
Abstract
The Rickettsiales Ehrlichia ruminantium, the causal agent of the fatal tick-borne disease Heartwater, induces severe damage to the vascular endothelium in ruminants. Nevertheless, E. ruminantium-induced pathobiology remains largely unknown. Our work paves the way for understanding this phenomenon by using quantitative proteomic analyses (2D-DIGE-MS/MS, 1DE-nanoLC-MS/MS and biotin-nanoUPLC-MS/MS) of host bovine aorta endothelial cells (BAE) during the in vitro bacterium intracellular replication cycle. We detect 265 bacterial proteins (including virulence factors), at all time-points of the E. ruminantium replication cycle, highlighting a dynamic bacterium–host interaction. We show that E. ruminantium infection modulates the expression of 433 host proteins: 98 being over-expressed, 161 under-expressed, 140 detected only in infected BAE cells and 34 exclusively detected in non-infected cells. Cystoscape integrated data analysis shows that these proteins lead to major changes in host cell immune responses, host cell metabolism and vesicle trafficking, with a clear involvement of inflammation-related proteins in this process. Our findings led to the first model of E. ruminantium infection in host cells in vitro, and we highlight potential biomarkers of E. ruminantium infection in endothelial cells (such as ROCK1, TMEM16K, Albumin and PTPN1), which may be important to further combat Heartwater, namely by developing non-antibiotic-based strategies.
Collapse
|
21
|
Patterson LL, Byerly CD, McBride JW. Anaplasmataceae: Dichotomous Autophagic Interplay for Infection. Front Immunol 2021; 12:642771. [PMID: 33912170 PMCID: PMC8075259 DOI: 10.3389/fimmu.2021.642771] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 03/15/2021] [Indexed: 12/19/2022] Open
Abstract
Autophagy is a vital conserved degradative process that maintains cellular homeostasis by recycling or eliminating dysfunctional cellular organelles and proteins. More recently, autophagy has become a well-recognized host defense mechanism against intracellular pathogens through a process known as xenophagy. On the host-microbe battlefield many intracellular bacterial pathogens have developed the ability to subvert xenophagy to establish infection. Obligately intracellular bacterial pathogens of the Anaplasmataceae family, including Ehrlichia chaffeensis, Anaplasma phaogocytophilium and Orientia tsutsugamushi have developed a dichotomous strategy to exploit the host autophagic pathway to obtain nutrients while escaping lysosomal destruction for intracellular survival within the host cell. In this review, the recent findings regarding how these master manipulators engage and inhibit autophagy for infection are explored. Future investigation to understand mechanisms used by Anaplasmataceae to exploit autophagy may advance novel antimicrobial therapies and provide new insights into how intracellular microbes exploit autophagy to survive.
Collapse
Affiliation(s)
- LaNisha L Patterson
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| | - Caitlan D Byerly
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| | - Jere W McBride
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States.,Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, United States.,Center for Biodefense and Emerging Infectious Diseases, University of Texas Medical Branch, Galveston, TX, United States.,Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX, United States.,Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, United States
| |
Collapse
|
22
|
Lin M, Xiong Q, Chung M, Daugherty SC, Nagaraj S, Sengamalay N, Ott S, Godinez A, Tallon LJ, Sadzewicz L, Fraser C, Dunning Hotopp JC, Rikihisa Y. Comparative Analysis of Genome of Ehrlichia sp. HF, a Model Bacterium to Study Fatal Human Ehrlichiosis. BMC Genomics 2021; 22:11. [PMID: 33407096 PMCID: PMC7789307 DOI: 10.1186/s12864-020-07309-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 12/07/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND The genus Ehrlichia consists of tick-borne obligatory intracellular bacteria that can cause deadly diseases of medical and agricultural importance. Ehrlichia sp. HF, isolated from Ixodes ovatus ticks in Japan [also referred to as I. ovatus Ehrlichia (IOE) agent], causes acute fatal infection in laboratory mice that resembles acute fatal human monocytic ehrlichiosis caused by Ehrlichia chaffeensis. As there is no small laboratory animal model to study fatal human ehrlichiosis, Ehrlichia sp. HF provides a needed disease model. However, the inability to culture Ehrlichia sp. HF and the lack of genomic information have been a barrier to advance this animal model. In addition, Ehrlichia sp. HF has several designations in the literature as it lacks a taxonomically recognized name. RESULTS We stably cultured Ehrlichia sp. HF in canine histiocytic leukemia DH82 cells from the HF strain-infected mice, and determined its complete genome sequence. Ehrlichia sp. HF has a single double-stranded circular chromosome of 1,148,904 bp, which encodes 866 proteins with a similar metabolic potential as E. chaffeensis. Ehrlichia sp. HF encodes homologs of all virulence factors identified in E. chaffeensis, including 23 paralogs of P28/OMP-1 family outer membrane proteins, type IV secretion system apparatus and effector proteins, two-component systems, ankyrin-repeat proteins, and tandem repeat proteins. Ehrlichia sp. HF is a novel species in the genus Ehrlichia, as demonstrated through whole genome comparisons with six representative Ehrlichia species, subspecies, and strains, using average nucleotide identity, digital DNA-DNA hybridization, and core genome alignment sequence identity. CONCLUSIONS The genome of Ehrlichia sp. HF encodes all known virulence factors found in E. chaffeensis, substantiating it as a model Ehrlichia species to study fatal human ehrlichiosis. Comparisons between Ehrlichia sp. HF and E. chaffeensis will enable identification of in vivo virulence factors that are related to host specificity, disease severity, and host inflammatory responses. We propose to name Ehrlichia sp. HF as Ehrlichia japonica sp. nov. (type strain HF), to denote the geographic region where this bacterium was initially isolated.
Collapse
Affiliation(s)
- Mingqun Lin
- Department of Veterinary Biosciences, The Ohio State University, 1925 Coffey Road, Columbus, OH, 43210, USA.
| | - Qingming Xiong
- Department of Veterinary Biosciences, The Ohio State University, 1925 Coffey Road, Columbus, OH, 43210, USA
| | - Matthew Chung
- Institute for Genome Sciences, University of Maryland School of Medicine, 801 W. Baltimore St, Baltimore, MD, 21201, USA
| | - Sean C Daugherty
- Institute for Genome Sciences, University of Maryland School of Medicine, 801 W. Baltimore St, Baltimore, MD, 21201, USA
| | - Sushma Nagaraj
- Institute for Genome Sciences, University of Maryland School of Medicine, 801 W. Baltimore St, Baltimore, MD, 21201, USA
| | - Naomi Sengamalay
- Institute for Genome Sciences, University of Maryland School of Medicine, 801 W. Baltimore St, Baltimore, MD, 21201, USA
| | - Sandra Ott
- Institute for Genome Sciences, University of Maryland School of Medicine, 801 W. Baltimore St, Baltimore, MD, 21201, USA
| | - Al Godinez
- Institute for Genome Sciences, University of Maryland School of Medicine, 801 W. Baltimore St, Baltimore, MD, 21201, USA
| | - Luke J Tallon
- Institute for Genome Sciences, University of Maryland School of Medicine, 801 W. Baltimore St, Baltimore, MD, 21201, USA
| | - Lisa Sadzewicz
- Institute for Genome Sciences, University of Maryland School of Medicine, 801 W. Baltimore St, Baltimore, MD, 21201, USA
| | - Claire Fraser
- Institute for Genome Sciences, University of Maryland School of Medicine, 801 W. Baltimore St, Baltimore, MD, 21201, USA
- Department of Medicine, University of Maryland School of Medicine, 801 W. Baltimore St, Baltimore, MD, 21201, USA
| | - Julie C Dunning Hotopp
- Institute for Genome Sciences, University of Maryland School of Medicine, 801 W. Baltimore St, Baltimore, MD, 21201, USA
- Department of Microbiology and Immunology, University of Maryland School of Medicine, 801 W. Baltimore St, Baltimore, MD, 21201, USA
- Greenebaum Cancer Center, University of Maryland School of Medicine, 801 W. Baltimore St, Baltimore, MD, 21201, USA
| | - Yasuko Rikihisa
- Department of Veterinary Biosciences, The Ohio State University, 1925 Coffey Road, Columbus, OH, 43210, USA.
| |
Collapse
|
23
|
Wang Y, Nair ADS, Alhassan A, Jaworski DC, Liu H, Trinkl K, Hove P, Ganta CK, Burkhardt N, Munderloh UG, Ganta RR. Multiple Ehrlichia chaffeensis Genes Critical for Its Persistent Infection in a Vertebrate Host Are Identified by Random Mutagenesis Coupled with In Vivo Infection Assessment. Infect Immun 2020; 88:e00316-20. [PMID: 32747600 PMCID: PMC7504954 DOI: 10.1128/iai.00316-20] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 07/23/2020] [Indexed: 12/14/2022] Open
Abstract
Ehrlichia chaffeensis, a tick-transmitted obligate intracellular rickettsial agent, causes human monocytic ehrlichiosis. In recent reports, we described substantial advances in developing random and targeted gene disruption methods to investigate the functions of E. chaffeensis genes. We reported earlier that the Himar1 transposon-based random mutagenesis is a valuable tool in defining E. chaffeensis genes critical for its persistent growth in vivo in reservoir and incidental hosts. The method also aided in extending studies focused on vaccine development and immunity. Here, we describe the generation and mapping of 55 new mutations. To define the critical nature of the bacterial genes, infection experiments were carried out in the canine host with pools of mutant organisms. Infection evaluation in the physiologically relevant host by molecular assays and by xenodiagnoses allowed the identification of many proteins critical for the pathogen's persistent in vivo growth. Genes encoding proteins involved in biotin biosynthesis, protein synthesis and fatty acid biosynthesis, DNA repair, electron transfer, and a component of a multidrug resistance (MDR) efflux pump were concluded to be essential for the pathogen's in vivo growth. Three known immunodominant membrane proteins, i.e., two 28-kDa outer membrane proteins (P28/OMP) and a 120-kDa surface protein, were also recognized as necessary for the pathogen's obligate intracellular life cycle. The discovery of many E. chaffeensis proteins crucial for its continuous in vivo growth will serve as a major resource for investigations aimed at defining pathogenesis and developing novel therapeutics for this and related pathogens of the rickettsial family Anaplasmataceae.
Collapse
Affiliation(s)
- Ying Wang
- Center of Excellence for Vector-Borne Diseases (CEVBD), Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas, USA
| | - Arathy D S Nair
- Center of Excellence for Vector-Borne Diseases (CEVBD), Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas, USA
| | - Andy Alhassan
- Center of Excellence for Vector-Borne Diseases (CEVBD), Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas, USA
- Department of Pathobiology, School of Veterinary Medicine, St. George's University, West Indies, Grenada
| | - Deborah C Jaworski
- Center of Excellence for Vector-Borne Diseases (CEVBD), Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas, USA
| | - Huitao Liu
- Center of Excellence for Vector-Borne Diseases (CEVBD), Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas, USA
| | - Kathleen Trinkl
- Center of Excellence for Vector-Borne Diseases (CEVBD), Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas, USA
| | - Paidashe Hove
- Center of Excellence for Vector-Borne Diseases (CEVBD), Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas, USA
- Department of Pathobiology, School of Veterinary Medicine, St. George's University, West Indies, Grenada
| | - Charan K Ganta
- Center of Excellence for Vector-Borne Diseases (CEVBD), Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas, USA
| | - Nicole Burkhardt
- Department of Entomology, University of Minnesota, St. Paul, Minnesota, USA
| | - Ulrike G Munderloh
- Department of Entomology, University of Minnesota, St. Paul, Minnesota, USA
| | - Roman R Ganta
- Center of Excellence for Vector-Borne Diseases (CEVBD), Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas, USA
| |
Collapse
|
24
|
An Entry-Triggering Protein of Ehrlichia Is a New Vaccine Candidate against Tick-Borne Human Monocytic Ehrlichiosis. mBio 2020; 11:mBio.00895-20. [PMID: 32723916 PMCID: PMC7387794 DOI: 10.1128/mbio.00895-20] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The incidence of tick-borne diseases has risen dramatically in the past two decades and continues to rise. Discovered in 1986 and designated a nationally notifiable disease in 1998 by the Centers for Disease Control and Prevention, human monocytic ehrlichiosis, which is caused by the bacterium Ehrlichia chaffeensis, is one of the most prevalent, life-threatening, emerging tick-borne zoonoses in the United States. We investigated the role of the E. chaffeensis protein EtpE in transmission of the bacterium from tick to human cells and in vaccinated dogs with EtpE to assess the efficacy of vaccination against E. chaffeensis-infected tick challenge. Our results help fill gaps in our understanding of E. chaffeensis-derived protective antigens that could be used in a candidate vaccine for immunization of humans to counter tick-transmitted ehrlichiosis. Ehrlichia chaffeensis is an obligatory intracellular bacterium that causes human monocytic ehrlichiosis, an emerging disease transmitted by the Lone Star tick, Amblyomma americanum. E. chaffeensis outer membrane protein entry triggering protein of Ehrlichia (EtpE) is necessary for bacterial entry into human cells. We investigated the role of EtpE in transmission of the bacteria from tick to human cells and whether or not vaccination with EtpE can prevent transmission of ehrlichiae from ticks to mammals. An antiserum against the recombinant C terminus of EtpE (rEtpE-C), which binds a mammalian cell-surface receptor and triggers bacterial entry, significantly inhibited E. chaffeensis transmission from infected tick cells to human monocytes in culture. Each of five specific-pathogen-free dogs were vaccinated with rEtpE-C along with an immunostimulating complex or were sham vaccinated with the complex alone. Dogs vaccinated with rEtpE-C developed high antibody titers against rEtpE-C and produced interferon-γ-secreting cells, as assessed with the ELISpot assay. All 10 dogs were challenged with A. americanum adult ticks infected as nymphs by syringe inoculation with E. chaffeensis. Upon challenge, both the vaccinated and control dogs became infected by day 1 post-tick attachment, but the majority of rEtpE-C-vaccinated dogs rapidly cleared the infection from the bloodstream as soon as day 7, whereas most of sham-vaccinated dogs remained infected at day 35. Peripheral blood leukocytes from vaccinated dogs had significantly elevated interferon-γ mRNA levels and secreted significantly elevated interferon-γ soon after tick attachment. Thus, the EtpE-C vaccine represents the first ehrlichial protein vaccine demonstrated to reduce bacterial infection in mammals upon challenge with infected ticks.
Collapse
|
25
|
Mogg M, Wang HH, Baker A, Derouen Z, Borski J, Grant WE. Increased Incidence of Ehrlichia chaffeensis Infections in the United States, 2012 Through 2016. Vector Borne Zoonotic Dis 2020; 20:547-550. [PMID: 32077809 DOI: 10.1089/vbz.2019.2595] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Human ehrlichioses are tick-borne diseases that have been increasing in incidence in the United States during recent years. Ehrlichia chaffeensis is one of the primary bacteria that cause ehrlichiosis in humans, which typically results in fever-like symptoms, but may also be fatal if left untreated. E. chaffeensis infections are reported to the Centers for Disease Control and Prevention (CDC) through the National Notifiable Diseases Surveillance System (NNDSS). This study analyzed the cases of E. chaffeensis infections reported by the NNDSS from 2012 through 2016. There were 6786 cases and the incidence rate was 4.46 cases per million persons per year. The demographic group most commonly infected was white males between the ages of 40 and 64. Infections were most abundant in the southeast and midwest, particularly in Arkansas, Missouri, Tennessee, and Oklahoma, as well as much of the east coast. The number of cases reported each year from 2012 through 2016 was higher than the number reported in any of the previous 4 years. Ongoing surveillance and reporting of tick-borne diseases are critical to inform public health practice and guide disease treatment and prevention efforts.
Collapse
Affiliation(s)
- Michael Mogg
- Department of Management, Texas A&M University, College Station, Texas, USA
| | - Hsiao-Hsuan Wang
- Department of Wildlife and Fisheries Sciences, Texas A&M University, College Station, Texas, USA
| | - Adam Baker
- College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| | - Zakary Derouen
- Department of Ecosystem Science and Management; Texas A&M University, College Station, Texas, USA
| | - Jennifer Borski
- Department of Wildlife and Fisheries Sciences, Texas A&M University, College Station, Texas, USA
| | - William E Grant
- Department of Wildlife and Fisheries Sciences, Texas A&M University, College Station, Texas, USA
| |
Collapse
|
26
|
Delaby M, Panis G, Viollier PH. Bacterial cell cycle and growth phase switch by the essential transcriptional regulator CtrA. Nucleic Acids Res 2020; 47:10628-10644. [PMID: 31598724 PMCID: PMC6847485 DOI: 10.1093/nar/gkz846] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Revised: 09/13/2019] [Accepted: 10/05/2019] [Indexed: 11/13/2022] Open
Abstract
Many bacteria acquire dissemination and virulence traits in G1-phase. CtrA, an essential and conserved cell cycle transcriptional regulator identified in the dimorphic alpha-proteobacterium Caulobacter crescentus, first activates promoters in late S-phase and then mysteriously switches to different target promoters in G1-phase. We uncovered a highly conserved determinant in the DNA-binding domain (DBD) of CtrA uncoupling this promoter switch. We also show that it reprograms CtrA occupancy in stationary cells inducing a (p)ppGpp alarmone signal perceived by the RNA polymerase beta subunit. A simple side chain modification in a critical residue within the core DBD imposes opposing developmental phenotypes and transcriptional activities of CtrA and a proximal residue can direct CtrA towards activation of the dispersal (G1-phase) program. Hence, we propose that this conserved determinant in the CtrA primary structure dictates promoter reprogramming during the growth transition in other alpha-proteobacteria that differentiate from replicative cells into dispersal cells.
Collapse
Affiliation(s)
- Marie Delaby
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Gaël Panis
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Patrick H Viollier
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
27
|
Lin M, Grandinetti G, Hartnell LM, Bliss D, Subramaniam S, Rikihisa Y. Host membrane lipids are trafficked to membranes of intravacuolar bacterium Ehrlichia chaffeensis. Proc Natl Acad Sci U S A 2020; 117:8032-8043. [PMID: 32193339 PMCID: PMC7149431 DOI: 10.1073/pnas.1921619117] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Ehrlichia chaffeensis, a cholesterol-rich and cholesterol-dependent obligate intracellular bacterium, partially lacks genes for glycerophospholipid biosynthesis. We found here that E. chaffeensis is dependent on host glycerolipid biosynthesis, as an inhibitor of host long-chain acyl CoA synthetases, key enzymes for glycerolipid biosynthesis, significantly reduced bacterial proliferation. E. chaffeensis cannot synthesize phosphatidylcholine or cholesterol but encodes enzymes for phosphatidylethanolamine (PE) biosynthesis; however, exogenous NBD-phosphatidylcholine, Bodipy-PE, and TopFluor-cholesterol were rapidly trafficked to ehrlichiae in infected cells. DiI (3,3'-dioctadecylindocarbocyanine)-prelabeled host-cell membranes were unidirectionally trafficked to Ehrlichia inclusion and bacterial membranes, but DiI-prelabeled Ehrlichia membranes were not trafficked to host-cell membranes. The trafficking of host-cell membranes to Ehrlichia inclusions was dependent on both host endocytic and autophagic pathways, and bacterial protein synthesis, as the respective inhibitors blocked both infection and trafficking of DiI-labeled host membranes to Ehrlichia In addition, DiI-labeled host-cell membranes were trafficked to autophagosomes induced by the E. chaffeensis type IV secretion system effector Etf-1, which traffic to and fuse with Ehrlichia inclusions. Cryosections of infected cells revealed numerous membranous vesicles inside inclusions, as well as multivesicular bodies docked on the inclusion surface, both of which were immunogold-labeled by a GFP-tagged 2×FYVE protein that binds to phosphatidylinositol 3-phosphate. Focused ion-beam scanning electron microscopy of infected cells validated numerous membranous structures inside bacteria-containing inclusions. Our results support the notion that Ehrlichia inclusions are amphisomes formed through fusion of early endosomes, multivesicular bodies, and early autophagosomes induced by Etf-1, and they provide host-cell glycerophospholipids and cholesterol that are necessary for bacterial proliferation.
Collapse
Affiliation(s)
- Mingqun Lin
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210
| | - Giovanna Grandinetti
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Lisa M Hartnell
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Donald Bliss
- National Library of Medicine, National Institutes of Health, Bethesda, MD 20894
| | - Sriram Subramaniam
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Yasuko Rikihisa
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210;
| |
Collapse
|
28
|
Bekebrede H, Lin M, Teymournejad O, Rikihisa Y. Discovery of in vivo Virulence Genes of Obligatory Intracellular Bacteria by Random Mutagenesis. Front Cell Infect Microbiol 2020; 10:2. [PMID: 32117791 PMCID: PMC7010607 DOI: 10.3389/fcimb.2020.00002] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 01/06/2020] [Indexed: 11/13/2022] Open
Abstract
Ehrlichia spp. are emerging tick-borne obligatory intracellular bacteria that cause febrile and sometimes fatal diseases with abnormal blood cell counts and signs of hepatitis. Ehrlichia HF strain provides an excellent mouse disease model of fatal human ehrlichiosis. We recently obtained and established stable culture of Ehrlichia HF strain in DH82 canine macrophage cell line, and obtained its whole genome sequence and annotation. To identify genes required for in vivo virulence of Ehrlichia, we constructed random insertional HF strain mutants by using Himar1 transposon-based mutagenesis procedure. Of total 158 insertional mutants isolated via antibiotic selection in DH82 cells, 74 insertions were in the coding regions of 55 distinct protein-coding genes, including TRP120 and multi-copy genes, such as p28/omp-1, virB2, and virB6. Among 84 insertions mapped within the non-coding regions, seven are located in the putative promoter region since they were within 50 bp upstream of the seven distinct genes. Using limited dilution methods, nine stable clonal mutants that had no apparent defect for multiplication in DH82 cells, were obtained. Mouse virulence of seven mutant clones was similar to that of wild-type HF strain, whereas two mutant clones showed significantly retarded growth in blood, livers, and spleens, and the mice inoculated with them lived longer than mice inoculated with wild-type. The two clones contained mutations in genes encoding a conserved hypothetical protein and a staphylococcal superantigen-like domain protein, respectively, and both genes are conserved among Ehrlichia spp., but lack homology to other bacterial genes. Inflammatory cytokine mRNA levels in the liver of mice infected with the two mutants were significantly diminished than those infected with HF strain wild-type, except IL-1β and IL-12 p40 in one clone. Thus, we identified two Ehrlichia virulence genes responsible for in vivo infection, but not for infection and growth in macrophages.
Collapse
Affiliation(s)
| | | | | | - Yasuko Rikihisa
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
29
|
|
30
|
Ehrlichia chaffeensis Outer Membrane Protein 1-Specific Human Antibody-Mediated Immunity Is Defined by Intracellular TRIM21-Dependent Innate Immune Activation and Extracellular Neutralization. Infect Immun 2019; 87:IAI.00383-19. [PMID: 31548319 PMCID: PMC6867850 DOI: 10.1128/iai.00383-19] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 09/18/2019] [Indexed: 01/05/2023] Open
Abstract
Antibodies are essential for immunity against Ehrlichia chaffeensis, and protective mechanisms involve blocking of ehrlichial attachment or complement and Fcγ-receptor-dependent destruction. In this study, we determined that major outer membrane protein 1 (OMP-19) hypervariable region 1 (HVR1)-specific human monoclonal antibodies (huMAbs) are protective through conventional extracellular neutralization and, more significantly, through a novel intracellular TRIM21-mediated mechanism. Antibodies are essential for immunity against Ehrlichia chaffeensis, and protective mechanisms involve blocking of ehrlichial attachment or complement and Fcγ-receptor-dependent destruction. In this study, we determined that major outer membrane protein 1 (OMP-19) hypervariable region 1 (HVR1)-specific human monoclonal antibodies (huMAbs) are protective through conventional extracellular neutralization and, more significantly, through a novel intracellular TRIM21-mediated mechanism. Addition of OMP-1-specific huMAb EHRL-15 (IgG1) prevented infection by blocking attachment/entry, a mechanism previously reported; conversely, OMP-1-specific huMAb EHRL-4 (IgG3) engaged intracellular TRIM21 and initiated an immediate innate immune response and rapid intracellular degradation of ehrlichiae. EHRL-4-TRIM21-mediated inhibition was significantly impaired in TRIM21 knockout THP-1 cells. EHRL-4 interacted with cytosolic Fc receptor TRIM21, observed by confocal microscopy and confirmed by co-immunoprecipitation. E. chaffeensis-EHRL-4-TRIM21 complexes caused significant upregulation of proinflammatory cytokine/chemokine transcripts and resulted in rapid (<30 min) nuclear accumulation of NF-κB and TRIM21 and ehrlichial destruction. We investigated the role of TRIM21 in the autophagic clearance of ehrlichiae in the presence of EHRL-4. Colocalization between EHRL-4-opsonized ehrlichiae, polyubiquitinated TRIM21, autophagy regulators (ULK1 and beclin 1) and effectors (LC3 and p62), and lysosome-associated membrane protein 2 (LAMP2) was observed. Moreover, autophagic flux defined by conversion of LC3I to LC3II and accumulation and degradation of p62 was detected, and EHRL-4-mediated degradation of E. chaffeensis was abrogated by the autophagy inhibitor 3-methyladenine. Our results demonstrate that huMAbs are capable of inhibiting E. chaffeensis infection by distinct effector mechanisms: extracellularly by neutralization and intracellularly by engaging TRIM21, which mediates a rapid innate immune response that mobilizes the core autophagy components, triggering localized selective autophagic degradation of ehrlichiae.
Collapse
|
31
|
Rogan MR, Patterson LL, Wang JY, McBride JW. Bacterial Manipulation of Wnt Signaling: A Host-Pathogen Tug-of-Wnt. Front Immunol 2019; 10:2390. [PMID: 31681283 PMCID: PMC6811524 DOI: 10.3389/fimmu.2019.02390] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 09/23/2019] [Indexed: 12/27/2022] Open
Abstract
The host-pathogen interface is a crucial battleground during bacterial infection in which host defenses are met with an array of bacterial counter-mechanisms whereby the invader aims to make the host environment more favorable to survival and dissemination. Interestingly, the eukaryotic Wnt signaling pathway has emerged as a key player in the host and pathogen tug-of-war. Although studied for decades as a regulator of embryogenesis, stem cell maintenance, bone formation, and organogenesis, Wnt signaling has recently been shown to control processes related to bacterial infection in the human host. Wnt signaling pathways contribute to cell cycle control, cytoskeleton reorganization during phagocytosis and cell migration, autophagy, apoptosis, and a number of inflammation-related events. Unsurprisingly, bacterial pathogens have evolved strategies to manipulate these Wnt-associated processes in order to enhance infection and survival within the human host. In this review, we examine the different ways human bacterial pathogens with distinct host cell tropisms and lifestyles exploit Wnt signaling for infection and address the potential of harnessing Wnt-related mechanisms to combat infectious disease.
Collapse
Affiliation(s)
- Madison R. Rogan
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| | - LaNisha L. Patterson
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| | - Jennifer Y. Wang
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| | - Jere W. McBride
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, United States
- Center for Biodefense and Emerging Infectious Diseases, University of Texas Medical Branch, Galveston, TX, United States
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, United States
| |
Collapse
|
32
|
Sanchez-Vicente S, Tagliafierro T, Coleman JL, Benach JL, Tokarz R. Polymicrobial Nature of Tick-Borne Diseases. mBio 2019; 10:e02055-19. [PMID: 31506314 PMCID: PMC6737246 DOI: 10.1128/mbio.02055-19] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 08/07/2019] [Indexed: 01/08/2023] Open
Abstract
Tick-borne diseases have doubled in the last 12 years, and their geographic distribution has spread as well. The clinical spectrum of tick-borne diseases can range from asymptomatic to fatal infections, with a disproportionate incidence in children and the elderly. In the last few years, new agents have been discovered, and genetic changes have helped in the spread of pathogens and ticks. Polymicrobial infections, mostly in Ixodes scapularis, can complicate diagnostics and augment disease severity. Amblyomma americanum ticks have expanded their range, resulting in a dynamic and complex situation, possibly fueled by climate change. To document these changes, using molecular biology strategies for pathogen detection, an assessment of 12 microbes (9 pathogens and 3 symbionts) in three species of ticks was done in Suffolk County, New York. At least one agent was detected in 63% of I. scapularis ticksBorrelia burgdorferi was the most prevalent pathogen (57% in adults; 27% in nymphs), followed by Babesia microti (14% in adults; 15% in nymphs), Anaplasma phagocytophilum (14% in adults; 2% in nymphs), Borrelia miyamotoi (3% in adults), and Powassan virus (2% in adults). Polymicrobial infections were detected in 22% of I. scapularis ticks, with coinfections of B. burgdorferi and B. microti (9%) and of B. burgdorferi and A. phagocytophilum (7%). Three Ehrlichia species were detected in 4% of A. americanum ticks. The rickettsiae constituted the largest prokaryotic biomass of all the ticks tested and included Rickettsia amblyommatis, Rickettsia buchneri, and Rickettsia montanensis The high rates of polymicrobial infection in ticks present an opportunity to study the biological interrelationships of pathogens and their vectors.IMPORTANCE Tick-borne diseases have increased in prevalence in the United States and abroad. The reasons for these increases are multifactorial, but climate change is likely to be a major factor. One of the main features of the increase is the geographic expansion of tick vectors, notably Amblyomma americanum, which has brought new pathogens to new areas. The clinical spectrum of tick-borne diseases can range from asymptomatic to fatal infections, with a disproportionate incidence in children and the elderly. In addition, new pathogens that are cotransmitted by Ixodes scapularis have been discovered and have led to difficult diagnoses and to disease severity. Of these, Borrelia burgdorferi, the agent of Lyme disease, continues to be the most frequently transmitted pathogen. However, Babesia microti, Borrelia miyamotoi (another spirochete), Anaplasma phagocytophilum, and Powassan virus are frequent cotransmitted agents. Polymicrobial infection has important consequences for the diagnosis and management of tick-borne diseases.
Collapse
Affiliation(s)
- Santiago Sanchez-Vicente
- Department of Molecular Genetics and Microbiology and Center for Infectious Diseases, Stony Brook University, Stony Brook, New York, USA
| | - Teresa Tagliafierro
- Center for Infection and Immunity, Mailman School of Public Health, Columbia University, New York, New York, USA
| | - James L Coleman
- Department of Molecular Genetics and Microbiology and Center for Infectious Diseases, Stony Brook University, Stony Brook, New York, USA
| | - Jorge L Benach
- Department of Molecular Genetics and Microbiology and Center for Infectious Diseases, Stony Brook University, Stony Brook, New York, USA
| | - Rafal Tokarz
- Center for Infection and Immunity, Mailman School of Public Health, Columbia University, New York, New York, USA
| |
Collapse
|
33
|
Hu W, Chan H, Lu L, Wong KT, Wong SH, Li MX, Xiao ZG, Cho CH, Gin T, Chan MTV, Wu WKK, Zhang L. Autophagy in intracellular bacterial infection. Semin Cell Dev Biol 2019; 101:41-50. [PMID: 31408699 DOI: 10.1016/j.semcdb.2019.07.014] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 06/06/2019] [Accepted: 07/30/2019] [Indexed: 12/11/2022]
Abstract
Autophagy is a conserved intracellular degradation process enclosing the bulk of cytosolic components for lysosomal degradation to maintain cellular homeostasis. Accumulating evidences showed that a specialized form of autophagy, known as xenophagy, could serve as an innate immune response to defend against pathogens invading inside the host cells. Correspondingly, infectious pathogens have developed a variety of strategies to disarm xenophagy, leading to a prolonged and persistent intracellular colonization. In this review, we first summarize the current knowledge about the general mechanisms of intracellular bacterial infections and xenophagy. We then focus on the ongoing battle between these two processes.
Collapse
Affiliation(s)
- Wei Hu
- Department of Gastroenterology, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong, PR China; Department of Anesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong, China
| | - Hung Chan
- Department of Anesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong, China
| | - Lan Lu
- Sichuan Industrial Institute of Antibiotics, Chengdu University, Chengdu, Sichuan, PR China
| | - Kam Tak Wong
- Department of Microbiology, The Chinese University of Hong Kong, Hong Kong, China
| | - Sunny H Wong
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China; State Key Laboratory of Digestive Diseases, Li Ka Shing Institute of Health Sciences, and Centre for Gut Microbiota Research, The Chinese University of Hong Kong, Hong Kong, China
| | - Ming X Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, PR China
| | - Zhan G Xiao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, PR China
| | - Chi H Cho
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, PR China
| | - Tony Gin
- Department of Anesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong, China
| | - Matthew T V Chan
- Department of Anesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong, China.
| | - William K K Wu
- Department of Anesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong, China; State Key Laboratory of Digestive Diseases, Li Ka Shing Institute of Health Sciences, and Centre for Gut Microbiota Research, The Chinese University of Hong Kong, Hong Kong, China.
| | - Lin Zhang
- Department of Anesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong, China; State Key Laboratory of Digestive Diseases, Li Ka Shing Institute of Health Sciences, and Centre for Gut Microbiota Research, The Chinese University of Hong Kong, Hong Kong, China.
| |
Collapse
|
34
|
Behar SM, Briken V. Apoptosis inhibition by intracellular bacteria and its consequence on host immunity. Curr Opin Immunol 2019; 60:103-110. [PMID: 31228759 DOI: 10.1016/j.coi.2019.05.007] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 05/07/2019] [Accepted: 05/11/2019] [Indexed: 02/08/2023]
Abstract
Regulated cell death via apoptosis not only is important for organismal homeostasis but also serves as an innate defense mechanism. The engulfment of apoptotic infected cells, a process known as efferocytosis, is a common pathway for the destruction of many intracellular bacteria. Some pathogens take advantage of efferocytosis to prevent activation of macrophages and thereby facilitate their dissemination. Conversely, many obligate intracellular bacterial pathogens and some facultative-intracellular bacteria inhibit apoptosis, preventing efferocytosis, and evading innate host defenses. The molecular mechanism of bacterial effectors includes secreted proteins that bind to and inhibit apoptosis cell signaling pathways. We provide an overview of the known bacterial effectors, their host cell targets and their importance for the virulence of human pathogens.
Collapse
Affiliation(s)
- Samuel M Behar
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, USA.
| | - Volker Briken
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD, USA.
| |
Collapse
|
35
|
Tominello TR, Oliveira ERA, Hussain SS, Elfert A, Wells J, Golden B, Ismail N. Emerging Roles of Autophagy and Inflammasome in Ehrlichiosis. Front Immunol 2019; 10:1011. [PMID: 31134081 PMCID: PMC6517498 DOI: 10.3389/fimmu.2019.01011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 04/23/2019] [Indexed: 12/16/2022] Open
Abstract
Human monocytic ehrlichiosis (HME) is a potentially life-threatening tick-borne rickettsial disease (TBRD) caused by the obligate intracellular Gram-negative bacteria, Ehrlichia. Fatal HME presents with acute ailments of sepsis and toxic shock-like symptoms that can evolve to multi-organ failure and death. Early clinical and laboratory diagnosis of HME are problematic due to non-specific flu-like symptoms and limitations in the current diagnostic testing. Several studies in murine models showed that cell-mediated immunity acts as a “double-edged sword” in fatal ehrlichiosis. Protective components are mainly formed by CD4 Th1 and NKT cells, in contrast to deleterious effects originated from neutrophils and TNF-α-producing CD8 T cells. Recent research has highlighted the central role of the inflammasome and autophagy as part of innate immune responses also leading to protective or pathogenic scenarios. Recognition of pathogen-associated molecular patterns (PAMPS) or damage-associated molecular patterns (DAMPS) triggers the assembly of the inflammasome complex that leads to multiple outcomes. Recognition of PAMPs or DAMPs by such complexes can result in activation of caspase-1 and -11, secretion of the pro-inflammatory cytokines IL-1β and IL-18 culminating into dysregulated inflammation, and inflammatory cell death known as pyroptosis. The precise functions of inflammasomes and autophagy remain unexplored in infections with obligate intracellular rickettsial pathogens, such as Ehrlichia. In this review, we discuss the intracellular innate immune surveillance in ehrlichiosis involving the regulation of inflammasome and autophagy, and how this response influences the innate and adaptive immune responses against Ehrlichia. Understanding such mechanisms would pave the way in research for novel diagnostic, preventative and therapeutic approaches against Ehrlichia and other rickettsial diseases.
Collapse
Affiliation(s)
- Tyler R Tominello
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Edson R A Oliveira
- Department of Pathology, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Shah S Hussain
- Department of Pathology, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Amr Elfert
- Department of Pathology, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Jakob Wells
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Brandon Golden
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Nahed Ismail
- Department of Pathology, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| |
Collapse
|
36
|
Role and Function of the Type IV Secretion System in Anaplasma and Ehrlichia Species. Curr Top Microbiol Immunol 2019; 413:297-321. [PMID: 29536364 DOI: 10.1007/978-3-319-75241-9_12] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The obligatory intracellular pathogens Anaplasma phagocytophilum and Ehrlichia chaffeensis proliferate within membrane-bound vacuoles of human leukocytes and cause potentially fatal emerging infectious diseases. Despite the reductive genome evolution in this group of bacteria, genes encoding the type IV secretion system (T4SS), which is homologous to the VirB/VirD4 system of the plant pathogen Agrobacterium tumefaciens, have been expanded and are highly expressed in A. phagocytophilum and E. chaffeensis in human cells. Of six T4SS effector proteins identified in them, roles and functions have been described so far only for ankyrin repeat domain-containing protein A (AnkA), Anaplasma translocated substrate 1 (Ats-1), and Ehrlichia translocated factor 1 (Etf-1, ECH0825). These effectors are abundantly produced and secreted into the host cytoplasm during infection, but not toxic to host cells. They contain eukaryotic protein motifs or organelle localization signals and have distinct subcellular localization, target to specific host cell molecules to promote infection. Ats-1 and Etf-1 are orthologous proteins, subvert two important innate immune mechanisms against intracellular infection, cellular apoptosis and autophagy, and manipulate autophagy to gain nutrients from host cells. Although Ats-1 and Etf-1 have similar functions and roles in obligatory intracellular infection, they are specifically adapted to the distinct membrane-bound intracellular niche of A. phagocytophilum and E. chaffeensis, respectively. Ectopic expression of these effectors enhances respective bacterial infection, whereas intracellular delivery of antibodies against these effectors or targeted knockdown of the effector with antisense peptide nucleic acid significantly impairs bacterial infection. Thus, both T4SSs have evolved as important survival and nutritional virulence mechanism in these obligatory intracellular bacteria. Future studies on the functions of Anaplasma and Ehrlichia T4SS effector molecules and signaling pathways will undoubtedly advance our understanding of the complex interplay between obligatory intracellular pathogens and their hosts. Such data can be applied toward the treatment and control of anaplasmosis and ehrlichiosis.
Collapse
|
37
|
Functional Replacement of the BioC and BioH Proteins of Escherichia coli Biotin Precursor Biosynthesis by Ehrlichia chaffeensis Novel Proteins. Curr Microbiol 2019; 76:626-636. [PMID: 30915508 DOI: 10.1007/s00284-019-01669-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 03/05/2019] [Indexed: 01/22/2023]
Abstract
The biosynthesis of the pimelate moiety of biotin in Escherichia coli requires two specialized proteins, BioC and BioH. However, the enzymes that have BioC- or BioH-like activities show remarkable sequence diversity among biotin-producing bacteria. Here, we report that the intracellular rickettsial pathogen Ehrlichia chaffeensis encodes two novel proteins, BioT and BioU, which functionally replace the E. coli BioC and BioH proteins, respectively. The desthiobiotin assays demonstrated that these two proteins make pimeloyl-acyl carrier protein (ACP) from the substrate malonyl-ACP with the aid of the FAS II pathway, through the expected pimeloyl-ACP methyl ester intermediate. BioT and BioU homologues seem restricted to the species of Ehrlichia and its close relative, Anaplasma. Taken together, the synthesis of the biotin precursor in E. chaffeensis appears to be catalyzed by two novel BioC- and BioH-like proteins.
Collapse
|
38
|
Spier A, Stavru F, Cossart P. Interaction between Intracellular Bacterial Pathogens and Host Cell Mitochondria. Microbiol Spectr 2019; 7:10.1128/microbiolspec.bai-0016-2019. [PMID: 30848238 PMCID: PMC11590420 DOI: 10.1128/microbiolspec.bai-0016-2019] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Indexed: 12/31/2022] Open
Abstract
Mitochondria are essential and highly dynamic organelles whose morphology is determined by a steady-state balance between fusion and fission. Mitochondrial morphology and function are tightly connected. Because they are involved in many important cellular processes, including energy production, cell-autonomous immunity, and apoptosis, mitochondria present an attractive target for pathogens. Here, we explore the relationship between host cell mitochondria and intracellular bacteria, with a focus on mitochondrial morphology and function, as well as apoptosis. Modulation of apoptosis can allow bacteria to establish their replicative niche or support bacterial dissemination. Furthermore, bacteria can manipulate mitochondrial morphology and function through secreted effector proteins and can also contribute to the establishment of a successful infection, e.g., by favoring access to nutrients and/or evasion of the immune system.
Collapse
Affiliation(s)
- Anna Spier
- Institut Pasteur, Unité des Interactions Bactéries-Cellules, Paris, France
- Institut National de la Recherche Agronomique, USC2020, Paris, France
- Bio Sorbonne Paris Cité, Université Paris Diderot, Paris, France
- Institut National de la Santé et de la Recherche Médicale, U604, Paris, France
| | - Fabrizia Stavru
- Institut Pasteur, Unité des Interactions Bactéries-Cellules, Paris, France
- Institut National de la Recherche Agronomique, USC2020, Paris, France
- Centre National de la Recherche Scientifique, SNC 5101, France
- Institut National de la Santé et de la Recherche Médicale, U604, Paris, France
| | - Pascale Cossart
- Institut Pasteur, Unité des Interactions Bactéries-Cellules, Paris, France
- Institut National de la Santé et de la Recherche Médicale, U604, Paris, France
- Institut National de la Recherche Agronomique, USC2020, Paris, France
| |
Collapse
|
39
|
Devitt KA, Gardner JA. Intravascular large B-cell lymphoma: The Great Imitator. Autops Case Rep 2019; 8:e2018055. [PMID: 30775328 PMCID: PMC6360824 DOI: 10.4322/acr.2018.055] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 10/08/2018] [Indexed: 11/23/2022]
Abstract
Intravascular large B-cell lymphoma (IVLBCL) is a rare subtype of diffuse large B-cell lymphoma with an estimated incidence of less than one per million. Unlike other hematopoietic malignancies, lymphadenopathy and hepatosplenomegaly are uncommon, and patients typically present with nonspecific symptoms. IVLBCL presents a diagnostic challenge and patients are usually diagnosed late in the disease course, if at all, and the prognosis is poor. The differential diagnosis is broad, and physicians often pursue multiple diagnostic possibilities during patient workup. We present a case of IVLBCL discovered at autopsy in an 80-year-old male who presented with history and symptoms pointing to the tick-borne illness ehrlichiosis.
Collapse
Affiliation(s)
- Katherine Ann Devitt
- University of Vermont Medical Center, Department of Pathology and Laboratory Medicine. Burlington, Vermont, USA
| | - Juli-Anne Gardner
- University of Vermont Medical Center, Department of Pathology and Laboratory Medicine. Burlington, Vermont, USA
| |
Collapse
|
40
|
Ge Y, Guo G, Ge B, Yin H, Yin H. The spleen microbiota of small wild mammals reveals distinct patterns with tick-borne bacteria. PLoS Negl Trop Dis 2018; 12:e0006499. [PMID: 29975692 PMCID: PMC6033388 DOI: 10.1371/journal.pntd.0006499] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 05/04/2018] [Indexed: 12/15/2022] Open
Abstract
Background Wild mammals serve as reservoirs for a variety of microbes and play an important role in the enzootic cycles of these microbes. Some of them are vector-borne bacteria in the genera Anaplasma, Ehrlichia and Rickettsia of the order Rickettsiales, which can cause febrile illnesses in human beings as well as animals. Anaplasma spp., Ehrlichia spp. and many spotted fever group (SFG) Rickettsia spp. are transmitted to mammalian hosts by tick vectors during blood meals. As a powerful sequencing method, the next generation sequencing can reveal the complexity of bacterial communities in humans and animals. Compared with limited studies on blood microbiota, however, much fewer studies have been carried out on spleen microbiota, which is very scarce in wild mammals. Chongming Island is the third biggest island in China. It was unclear whether there were any vector-borne bacteria in Chongming Island. In the present study, we explored the bacterial microbiota in the spleens of wild mice and shrews from the rural areas of Chongming Island and investigated the prevalence of vector-borne bacteria. Methodology/Principal findings Genomic DNAs were extracted from the spleen samples of 35 mice and shrews. The 16S rDNA V3-V4 regions of the DNA extracts were amplified by PCR and subjected to the 16S rDNA-targeted metagenomic sequencing on an Illumina MiSeq platform. All the 35 spleen samples obtained data with sufficient coverage (99.7–99.9%) for analysis. More than 1,300,000 sequences were obtained after quality control and classified into a total of 1,967 operational taxonomic units (OTUs) clustered at 97% similarity. The two most abundant bacterial phyla were Firmicutes and Proteobacteria according to the analysis of rarefied sequences. Among the bacterial communities detected in this study, Anaplasma, Rickettsia and Coxiella were adjacently clustered by hierarchical analysis. Significant differences in many bacterial features between Anaplasma-positive and Anaplasma-negative samples were identified by LEfSe analysis and Wilcoxon rank-sum test, suggesting that the Anaplasma-infection of small wild mammals was associated with a specific pattern of spleen microbiota. Conclusions/Significance Our study has comprehensively characterized the complex bacterial profiles in the spleens of wild mice and shrews from Chongming Island, Shanghai city. This work has revealed distinct spleen bacterial communities associated with tick-borne bacteria in wild animals. The detection of tick-borne bacteria highlights the risk of contracting pathogens with public health importance upon tick-exposure in the studied areas. In this study, the 16S rDNA-targeted metagenomic sequencing was used to determine the bacterial community and diversity in the spleens of small wild mammals from China. The 16S rDNAs were amplified from the spleen genomic DNAs of 35 small wild mice and shrews and sequenced by Illumina MiSeq technology. More than 1,300,000 sequences were obtained after quality control and classified into a total of 1,967 operational taxonomic units (OTUs) clustered at 97% similarity. The two most abundant bacterial phyla were Firmicutes and Proteobacteria according to the analysis of rarefied sequences. Within the bacterial communities detected in this study, vector-borne bacteria, Anaplasma, Rickettsia and Coxiella, were adjacently clustered by hierarchical analysis. Significant differences in many bacterial features between Anaplasma-positive and Anaplasma-negative samples were observed, suggesting that the infection of small wild mammals with Anaplasma is associated with a distinct pattern of spleen microbiota. This study has revealed the complex bacterial profiles in the spleens of wild mice and shrews. The detection of vector-borne bacteria highlights the role of wild mice and shrews as animal reservoirs with potential public health importance in the studied areas.
Collapse
Affiliation(s)
- Yan Ge
- Department of Immunology and Pathogen Biology, Tongji University School of Medicine, Shanghai, China
- * E-mail:
| | - Guangpu Guo
- Departement of Biotechnology, College of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Baoxue Ge
- Department of Immunology and Pathogen Biology, Tongji University School of Medicine, Shanghai, China
| | - Hongmei Yin
- Department of Immunology and Pathogen Biology, Tongji University School of Medicine, Shanghai, China
| | - Hong Yin
- The State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Gansu, China
| |
Collapse
|
41
|
Ehrlichia chaffeensis TRP75 Interacts with Host Cell Targets Involved in Homeostasis, Cytoskeleton Organization, and Apoptosis Regulation To Promote Infection. mSphere 2018; 3:3/2/e00147-18. [PMID: 29643078 PMCID: PMC5909120 DOI: 10.1128/msphere.00147-18] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 03/24/2018] [Indexed: 01/06/2023] Open
Abstract
Human monocytic ehrlichiosis (HME) is caused by an obligatory intracellular bacterium, E. chaffeensis, and is one of the most prevalent, life-threatening emerging infectious zoonoses in the United States. The mechanisms through which E. chaffeensis invades and establishes an intracellular niche are not well understood but are dependent on secreted ehrlichial effector proteins. The significance of this study is in addressing how intracellular pathogens, particularly those with small genomes such as Ehrlichia, exploit a limited number of secreted effector proteins such as tandem repeat proteins (TRPs) to manipulate complex eukaryotes and to regulate host cell processes through molecular pathogen-host interplay. The results of our studies highlight the broader role of ehrlichial TRPs in promoting infection and help define the mechanisms through which obligately intracellular bacteria modulate host cell function for survival. Ehrlichia chaffeensis is an obligately intracellular bacterium that exhibits tropism for mononuclear phagocytes. The mechanisms involved in E. chaffeensis infection of the host cell and evasion of host defenses are not fully defined, but a subset of type 1 secreted tandem repeat protein (TRP) effectors play important roles. Recently, we determined molecular interactions of TRP120, TRP47, and TRP32 with the eukaryotic host cell. In this investigation, we used yeast two-hybrid analysis to reveal that another E. chaffeensis tandem repeat protein, TRP75, interacts with a diverse group of human proteins associated with organismal and tissue homeostasis, multiple metabolic processes and regulation, response to reactive oxygen species, signal transduction, and protein modifications. Thirteen identified host target proteins associated with actin cytoskeleton reorganization or apoptosis were examined in detail and confirmed to interact with TRP75 at different levels as determined by coimmunoprecipitation assays. These protein interactions were visualized by immunofluorescence confocal microscopy during infection and colocalized with Ehrlichia morulae with different intensities. Moreover, small interfering RNAs (siRNAs) (n = 86) were used to knock down identified TRP75-interacting host proteins separately, and their influence on ehrlichial infection was investigated by real-time quantitative PCR (qPCR). Knockdown of 74/86 (86%) TRP75 target proteins had a significant negative effect on ehrlichial infection. The results of this study further support the idea of a role of Ehrlichia TRPs as effectors that interact with a complex array of host proteins to promote ehrlichial infection. IMPORTANCE Human monocytic ehrlichiosis (HME) is caused by an obligatory intracellular bacterium, E. chaffeensis, and is one of the most prevalent, life-threatening emerging infectious zoonoses in the United States. The mechanisms through which E. chaffeensis invades and establishes an intracellular niche are not well understood but are dependent on secreted ehrlichial effector proteins. The significance of this study is in addressing how intracellular pathogens, particularly those with small genomes such as Ehrlichia, exploit a limited number of secreted effector proteins such as tandem repeat proteins (TRPs) to manipulate complex eukaryotes and to regulate host cell processes through molecular pathogen-host interplay. The results of our studies highlight the broader role of ehrlichial TRPs in promoting infection and help define the mechanisms through which obligately intracellular bacteria modulate host cell function for survival.
Collapse
|
42
|
Grohmann E, Christie PJ, Waksman G, Backert S. Type IV secretion in Gram-negative and Gram-positive bacteria. Mol Microbiol 2018; 107:455-471. [PMID: 29235173 PMCID: PMC5796862 DOI: 10.1111/mmi.13896] [Citation(s) in RCA: 231] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 12/07/2017] [Accepted: 12/09/2017] [Indexed: 02/06/2023]
Abstract
Type IV secretion systems (T4SSs) are versatile multiprotein nanomachines spanning the entire cell envelope in Gram-negative and Gram-positive bacteria. They play important roles through the contact-dependent secretion of effector molecules into eukaryotic hosts and conjugative transfer of mobile DNA elements as well as contact-independent exchange of DNA with the extracellular milieu. In the last few years, many details on the molecular mechanisms of T4SSs have been elucidated. Exciting structures of T4SS complexes from Escherichia coli plasmids R388 and pKM101, Helicobacter pylori and Legionella pneumophila have been solved. The structure of the F-pilus was also reported and surprisingly revealed a filament composed of pilin subunits in 1:1 stoichiometry with phospholipid molecules. Many new T4SSs have been identified and characterized, underscoring the structural and functional diversity of this secretion superfamily. Complex regulatory circuits also have been shown to control T4SS machine production in response to host cell physiological status or a quorum of bacterial recipient cells in the vicinity. Here, we summarize recent advances in our knowledge of 'paradigmatic' and emerging systems, and further explore how new basic insights are aiding in the design of strategies aimed at suppressing T4SS functions in bacterial infections and spread of antimicrobial resistances.
Collapse
Affiliation(s)
- Elisabeth Grohmann
- Beuth University of Applied Sciences Berlin, Life Sciences and Technology, D-13347 Berlin, Germany
| | - Peter J. Christie
- Department of Microbiology and Molecular Genetics, The University of Texas Medical School at Houston, 6431 Fannin St, Houston, Texas 77030, USA
| | - Gabriel Waksman
- Institute of Structural and Molecular Biology, University College London and Birkbeck College, London WC1E 7HX, United Kingdom
| | - Steffen Backert
- Friedrich Alexander University Erlangen-Nuremberg, Department of Biology, Division of Microbiology, Staudtstrasse 5, D-91058 Erlangen, Germany
| |
Collapse
|
43
|
Ehrlichia chaffeensis and Its Invasin EtpE Block Reactive Oxygen Species Generation by Macrophages in a DNase X-Dependent Manner. mBio 2017; 8:mBio.01551-17. [PMID: 29162709 PMCID: PMC5698551 DOI: 10.1128/mbio.01551-17] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The obligatory intracellular pathogen Ehrlichia chaffeensis lacks most genes that confer resistance to oxidative stress but can block reactive oxygen species (ROS) generation by host monocytes-macrophages. Bacterial and host molecules responsible for this inhibition have not been identified. To infect host cells, Ehrlichia uses the C terminus of its surface invasin, entry-triggering protein of Ehrlichia (EtpE; EtpE-C), which directly binds the mammalian cell surface receptor glycosylphosphatidylinositol-anchored protein DNase X. We investigated whether EtpE-C binding to DNase X blocks ROS production by mouse bone marrow-derived macrophages (BMDMs). On the basis of a luminol-dependent chemiluminescence assay, E. chaffeensis inhibited phorbol myristate acetate (PMA)-induced ROS generation by BMDMs from wild-type, but not DNase X−/−, mice. EtpE-C is critical for inhibition, as recombinant EtpE-C (rEtpE-C)-coated latex beads, but not recombinant N-terminal EtpE-coated or uncoated beads, inhibited PMA-induced ROS generation by BMDMs from wild-type mice. DNase X is required for this inhibition, as none of these beads inhibited PMA-induced ROS generation by BMDMs from DNase X−/− mice. Previous studies showed that E. chaffeensis does not block ROS generation in neutrophils, a cell type that is a potent ROS generator but is not infected by E. chaffeensis. Human and mouse peripheral blood neutrophils did not express DNase X. Our findings point to a unique survival mechanism of ROS-sensitive obligate intramonocytic bacteria that involves invasin EtpE binding to DNase X on the host cell surface. This is the first report of bacterial invasin having such a subversive activity on ROS generation. Ehrlichia chaffeensis preferentially infects monocytes-macrophages and causes a life-threatening emerging tick-transmitted infectious disease called human monocytic ehrlichiosis. Ehrlichial infection, and hence the disease, depends on the ability of this bacterium to avoid or overcome powerful microbicidal mechanisms of host monocytes-macrophages, one of which is the generation of ROS. Our findings reveal that an ehrlichial surface invasin, EtpE, not only triggers bacterial entry but also blocks ROS generation by host macrophages through its host cell receptor, DNase X. As ROS sensitivity is an Achilles’ heel of this group of pathogens, understanding the mechanism by which E. chaffeensis rapidly blocks ROS generation suggests a new approach for developing effective anti-infective measures. The discovery of a ROS-blocking pathway is also important, as modulation of ROS generation is important in a variety of ailments and biological processes.
Collapse
|
44
|
Kader M, Alaoui-EL-Azher M, Vorhauer J, Kode BB, Wells JZ, Stolz D, Michalopoulos G, Wells A, Scott M, Ismail N. MyD88-dependent inflammasome activation and autophagy inhibition contributes to Ehrlichia-induced liver injury and toxic shock. PLoS Pathog 2017; 13:e1006644. [PMID: 29049365 PMCID: PMC5663626 DOI: 10.1371/journal.ppat.1006644] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 10/31/2017] [Accepted: 09/11/2017] [Indexed: 01/19/2023] Open
Abstract
Severe hepatic inflammation is a common cause of acute liver injury following systemic infection with Ehrlichia, obligate Gram-negative intracellular bacteria that lack lipopolysaccharide (LPS). We have previously shown that type I IFN (IFN-I) and inflammasome activation are key host-pathogenic mediators that promote excessive inflammation and liver damage following fatal Ehrlichia infection. However, the underlying signals and mechanisms that regulate protective immunity and immunopathology during Ehrlichia infection are not well understood. To address this issue, we compared susceptibility to lethal Ixodes ovatus Ehrlichia (IOE) infection between wild type (WT) and MyD88-deficient (MyD88-/-) mice. We show here that MyD88-/- mice exhibited decreased inflammasome activation, attenuated liver injury, and were more resistant to lethal infection than WT mice, despite suppressed protective immunity and increased bacterial burden in the liver. MyD88-dependent inflammasome activation was also dependent on activation of the metabolic checkpoint kinase mammalian target of rapamycin complex 1 (mTORC1), inhibition of autophagic flux, and defective mitophagy in macrophages. Blocking mTORC1 signaling in infected WT mice and primary macrophages enhanced bacterial replication and attenuated inflammasome activation, suggesting autophagy promotes bacterial replication while inhibiting inflammasome activation. Finally, our data suggest TLR9 and IFN-I are upstream signaling mechanisms triggering MyD88-mediated mTORC1 and inflammasome activation in macrophages following Ehrlichia infection. This study reveals that Ehrlichia-induced liver injury and toxic shock are mediated by MyD88-dependent inflammasome activation and autophagy inhibition.
Collapse
MESH Headings
- Animals
- Autophagy/immunology
- Blotting, Western
- Disease Models, Animal
- Ehrlichia/immunology
- Ehrlichiosis/immunology
- Ehrlichiosis/metabolism
- Enzyme-Linked Immunosorbent Assay
- Female
- Flow Cytometry
- Fluorescent Antibody Technique
- Image Processing, Computer-Assisted
- In Situ Nick-End Labeling
- Inflammasomes/immunology
- Inflammasomes/metabolism
- Liver Failure, Acute/immunology
- Liver Failure, Acute/metabolism
- Liver Failure, Acute/microbiology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Microscopy, Confocal
- Microscopy, Electron, Transmission
- Myeloid Differentiation Factor 88/immunology
- Myeloid Differentiation Factor 88/metabolism
- Real-Time Polymerase Chain Reaction
- Shock, Septic/immunology
- Shock, Septic/metabolism
Collapse
Affiliation(s)
- Muhamuda Kader
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Mounia Alaoui-EL-Azher
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Jennie Vorhauer
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Bhushan B Kode
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Jakob Z. Wells
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Donna Stolz
- Center for Biologic Imaging, University of Pittsburgh Medical School, Pittsburgh, Pennsylvania, United States of America
| | - George Michalopoulos
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Alan Wells
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Melanie Scott
- Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Nahed Ismail
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| |
Collapse
|
45
|
Abstract
Intracellular pathogens often exploit RAB functions to establish a safe haven in which to survive and proliferate. Ehrlichia chaffeensis, an obligatory intracellular bacterium, resides in specialized membrane-bound inclusions that have early endosome-like characteristics, e.g., resident RAB5 GTPase and RAB5 effectors, including VPS34 (the catalytic subunit of class III phosphatidylinositol 3-kinase), but the inclusions lack late endosomal or lysosomal markers. Within inclusions, Ehrlichia obtains host-derived nutrients by inducing RAB5-regulated autophagy using Ehrlichia translocated factor-1 deployed by its type IV secretion system. This manipulation of RAB5 by a bacterial molecule offers a simple strategy for Ehrlichia to avoid destruction in lysosomes and obtain nutrients, membrane components, and a homeostatic intra-host-cell environment in which to grow.
Collapse
Affiliation(s)
- Yasuko Rikihisa
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, USA,CONTACT Yasuko Rikihisa Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, 1925 Coffey Road, Columbus, OH 43210-1093, USA
| |
Collapse
|
46
|
Abstract
A vast number of novel tick-related microorganisms and tick-borne disease agents have been identified in the past 20 years, and more are being described due to several factors, from the curiosity of clinicians faced with unusual clinical syndromes to new tools used by microbiologists and entomologists. Borrelioses, ehrlichioses, anaplasmosis, and tick-borne rickettsial diseases are some of the emerging diseases that have been described throughout the world in recent years. In this article, we focus on the bacterial agents and diseases that have been recognized in the past 3 years and refer to major recent reviews of other recognized infections.
Collapse
|
47
|
Sharma P, Teymournejad O, Rikihisa Y. Peptide Nucleic Acid Knockdown and Intra-host Cell Complementation of Ehrlichia Type IV Secretion System Effector. Front Cell Infect Microbiol 2017. [PMID: 28638803 PMCID: PMC5461285 DOI: 10.3389/fcimb.2017.00228] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Survival of Ehrlichia chaffeensis depends on obligatory intracellular infection. One of the barriers to E. chaffeensis research progress has been the inability, using conventional techniques, to generate knock-out mutants for genes essential for intracellular infection. This study examined the use of Peptide Nucleic Acids (PNAs) technology to interrupt type IV secretion system (T4SS) effector protein expression in E. chaffeensis followed by intracellular complementation of the effector to determine its requirement for infection. Successful E. chaffeensis infection depends on the E. chaffeensis-specific T4SS protein effector, ehrlichial translocated factor-1 (Etf-1), which induces Rab5-regulated autophagy to provide host cytosolic nutrients required for E. chaffeensis proliferation. Etf-1 is also imported by host cell mitochondria where it inhibits host cell apoptosis to prolong its infection. We designed a PNA specific to Etf-1 and showed that the PNA bound to the target region of single-stranded Etf-1 RNA using a competitive binding assay. Electroporation of E. chaffeensis with this PNA significantly reduced Etf-1 mRNA and protein, and the bacteria's ability to induce host cell autophagy and infect host cells. Etf-1 PNA-mediated inhibition of ehrlichial Etf-1 expression and E. chaffeensis infection could be intracellularly trans-complemented by ectopic expression of Etf-1-GFP in host cells. These data affirmed the critical role of bacterial T4SS effector in host cell autophagy and E. chaffeensis infection, and demonstrated the use of PNA to analyze the gene functions of obligate intracellular bacteria.
Collapse
Affiliation(s)
- Pratibha Sharma
- Department of Veterinary Biosciences, Ohio State UniversityColumbus, OH, United States
| | - Omid Teymournejad
- Department of Veterinary Biosciences, Ohio State UniversityColumbus, OH, United States
| | - Yasuko Rikihisa
- Department of Veterinary Biosciences, Ohio State UniversityColumbus, OH, United States
| |
Collapse
|
48
|
Abstract
Human ehrlichiosis and anaplasmosis are acute febrile tick-borne infectious diseases caused by various members from the genera Ehrlichia and Anaplasma. Ehrlichia chaffeensis is the major etiologic agent of human monocytotropic ehrlichiosis (HME), while Anaplasma phagocytophilum is the major cause of human granulocytic anaplasmosis (HGA). The clinical manifestations of HME and HGA ranges from subclinical to potentially life-threatening diseases associated with multi-organ failure. Macrophages and neutrophils are the major target cells for Ehrlichia and Anaplasma, respectively. The threat to public health is increasing with newly emerging ehrlichial and anaplasma agents, yet vaccines for human ehrlichioses and anaplasmosis are not available, and therapeutic options are limited. This article reviews recent advances in the understanding of HME and HGA.
Collapse
|
49
|
Hallez R, Delaby M, Sanselicio S, Viollier PH. Hit the right spots: cell cycle control by phosphorylated guanosines in alphaproteobacteria. Nat Rev Microbiol 2017; 15:137-148. [PMID: 28138140 DOI: 10.1038/nrmicro.2016.183] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The class Alphaproteobacteria includes Gram-negative free-living, symbiotic and obligate intracellular bacteria, as well as important plant, animal and human pathogens. Recent work has established the key antagonistic roles that phosphorylated guanosines, cyclic-di-GMP (c-di-GMP) and the alarmones guanosine tetraphosphate and guanosine pentaphosphate (collectively referred to as (p)ppGpp), have in the regulation of the cell cycle in these bacteria. In this Review, we discuss the insights that have been gained into the regulation of the initiation of DNA replication and cytokinesis by these second messengers, with a particular focus on the cell cycle of Caulobacter crescentus. We explore how the fluctuating levels of c-di-GMP and (p)ppGpp during the progression of the cell cycle and under conditions of stress control the synthesis and proteolysis of key regulators of the cell cycle. As these signals also promote bacterial interactions with host cells, the enzymes that control (p)ppGpp and c-di-GMP are attractive antibacterial targets.
Collapse
Affiliation(s)
- Régis Hallez
- Bacterial Cell cycle and Development (BCcD), Unité de recherche en biologie des micro-organismes (URBM), University of Namur, 61 Rue de Bruxelles, Namur 5000, Belgium
| | - Marie Delaby
- Department of Microbiology and Molecular Medicine, Institute of Genetics &Genomics in Geneva (iGE3), Faculty of Medicine, University of Geneva, CH-1211 Geneva, Switzerland
| | - Stefano Sanselicio
- Department of Microbiology and Molecular Medicine, Institute of Genetics &Genomics in Geneva (iGE3), Faculty of Medicine, University of Geneva, CH-1211 Geneva, Switzerland.,Present address: Molecular Genetics Group, Groningen Biomolecular Sciences and Biotechnology Institute, Centre for Synthetic Biology, University of Groningen, Nijenborgh 7, 9747 AG Groningen, The Netherlands
| | - Patrick H Viollier
- Department of Microbiology and Molecular Medicine, Institute of Genetics &Genomics in Geneva (iGE3), Faculty of Medicine, University of Geneva, CH-1211 Geneva, Switzerland
| |
Collapse
|
50
|
Tago D, Meyer DF. Economic Game Theory to Model the Attenuation of Virulence of an Obligate Intracellular Bacterium. Front Cell Infect Microbiol 2016; 6:86. [PMID: 27610355 PMCID: PMC4997789 DOI: 10.3389/fcimb.2016.00086] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 08/09/2016] [Indexed: 12/19/2022] Open
Abstract
Diseases induced by obligate intracellular pathogens have a large burden on global human and animal health. Understanding the factors involved in the virulence and fitness of these pathogens contributes to the development of control strategies against these diseases. Based on biological observations, a theoretical model using game theory is proposed to explain how obligate intracellular bacteria interact with their host. The equilibrium in such a game shows that the virulence and fitness of the bacterium is host-triggered and by changing the host's defense system to which the bacterium is confronted, an evolutionary process leads to an attenuated strain. Although, the attenuation procedure has already been conducted in practice in order to develop an attenuated vaccine (e.g., with Ehrlichia ruminantium), there was a lack of understanding of the theoretical basis behind this process. Our work provides a model to better comprehend the existence of different phenotypes and some underlying evolutionary mechanisms for the virulence of obligate intracellular bacteria.
Collapse
Affiliation(s)
- Damian Tago
- La Recherche Agronomique Pour le Développement (CIRAD), UMR Contrôle des Maladies Animales, Exotiques et Émergentes (CMAEE)Petit-Bourg, France; Institut National de la Recherche Agronomique, UMR1309 CMAEEMontpellier, France
| | - Damien F Meyer
- La Recherche Agronomique Pour le Développement (CIRAD), UMR Contrôle des Maladies Animales, Exotiques et Émergentes (CMAEE)Petit-Bourg, France; Institut National de la Recherche Agronomique, UMR1309 CMAEEMontpellier, France
| |
Collapse
|