1
|
Goyal N, Barai A, Sen S, Kondabagil K. Amoebal Tubulin Cleavage Late during Infection Is a Characteristic Feature of Mimivirus but Not of Marseillevirus. Microbiol Spectr 2022; 10:e0275322. [PMID: 36453900 PMCID: PMC9769910 DOI: 10.1128/spectrum.02753-22] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 11/04/2022] [Indexed: 12/03/2022] Open
Abstract
Mimivirus and Marseillevirus infections of Acanthamoeba castellanii, like most other viral infections, induce cytopathic effects (CPE). The details of how they bring about CPE and to what extent and how they modify the host cytoskeletal network are unclear. In this study, we compared the rearrangement of the host cytoskeletal network induced by Mimivirus and Marseillevirus upon infection. We show that while both Mimivirus and Marseillevirus infections of A. castellanii cells cause retraction of acanthopodia and depolymerization of the host actin filament network, the Mimivirus infection also results in characteristic cleavage of the host tubulin, a phenomenon not previously reported with any intracellular pathogens. Furthermore, we show that the amoebal tubulin cleavage during Mimivirus infection is a post-replicative event. Because time-lapse microscopy showed that Mimivirus infection leads to the bursting of cells, releasing the virus, we hypothesize that tubulin cleavage together with actin depolymerization during the later stages of Mimivirus assembly is essential for cell lysis due to apoptotic/necrotic cell death. We also characterize the Mimivirus-encoded gp560, a Zn metalloprotease, however, the purified gp560 protein was unable to cleave the commercially available porcine brain tubulin. While protein synthesis is essential for causing the morphological changes in the case of Mimivirus, the proteins which are packaged in the viral capsid along with the genome are sufficient to induce CPE in the case of Marseillevirus. IMPORTANCE In general, intracellular pathogens target the cytoskeletal network to enable their life cycle inside the host. Pathogen-induced changes in the host cell morphology usually accompany global changes in the cytoskeleton resulting in cytopathic effects. While viruses have been shown to use the host actin cytoskeleton for entry and transport during early infection, the role of microtubules in the viral life cycle is only beginning to emerge. Here, we show that the giant viruses Mimivirus and Marseillevirus both induce depolymerization of the actin filament, Mimivirus also causes a characteristic cleavage of tubulin not previously reported for any intracellular pathogen. Because tubulin cleavage occurs late during infection, we hypothesize that tubulin cleavage aids in cell death and lysis rather than establishing infection. The different strategies used by viruses with similar host niches may help them survive in competition.
Collapse
Affiliation(s)
- Nisha Goyal
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai, India
| | - Amlan Barai
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai, India
| | - Shamik Sen
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai, India
| | - Kiran Kondabagil
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai, India
| |
Collapse
|
2
|
Silver LW, Cheng Y, Quigley BL, Robbins A, Timms P, Hogg CJ, Belov K. A targeted approach to investigating immune genes of an iconic Australian marsupial. Mol Ecol 2022; 31:3286-3303. [PMID: 35510793 PMCID: PMC9325493 DOI: 10.1111/mec.16493] [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: 08/29/2021] [Revised: 03/02/2022] [Accepted: 04/05/2022] [Indexed: 11/30/2022]
Abstract
Disease is a contributing factor to the decline of wildlife populations across the globe. Koalas, iconic yet declining Australian marsupials, are predominantly impacted by two pathogens, Chlamydia and koala retrovirus. Chlamydia is an obligate intracellular bacterium and one of the most widespread sexually transmitted infections in humans worldwide. In koalas, Chlamydia infections can present as asymptomatic or can cause a range of ocular and urogenital disease signs, such as conjunctivitis, cystitis and infertility. In this study, we looked at differences in response to Chlamydia in two northern populations of koalas using a targeted gene sequencing of 1209 immune genes in addition to genome‐wide reduced representation data. We identified two MHC Class I genes associated with Chlamydia disease progression as well as 25 single nucleotide polymorphisms across 17 genes that were associated with resolution of Chlamydia infection. These genes are involved in the innate immune response (TLR5) and defence (TLR5, IFNγ, SERPINE1, STAT2 and STX4). This study deepens our understanding of the role that genetics plays in disease progression in koalas and leads into future work that will use whole genome resequencing of a larger sample set to investigate in greater detail regions identified in this study. Elucidation of the role of host genetics in disease progression and resolution in koalas will directly contribute to better design of Chlamydia vaccines and management of koala populations which have recently been listed as “endangered.”
Collapse
Affiliation(s)
- Luke W Silver
- School of Life and Environmental Sciences, The University of Sydney, New South Wales, 2006, Australia
| | - Yuanyuan Cheng
- School of Life and Environmental Sciences, The University of Sydney, New South Wales, 2006, Australia
| | - Bonnie L Quigley
- Genecology Research Centre, University of the Sunshine Coast, 90 Sippy Downs Drive, Sippy Downs, Queensland, 4556, Australia.,Provectus Algae Pty Ltd, 5 Bartlett Road, Noosaville, Queensland, 4566, Australia
| | - Amy Robbins
- Genecology Research Centre, University of the Sunshine Coast, 90 Sippy Downs Drive, Sippy Downs, Queensland, 4556, Australia.,Endeavour Veterinary Ecology Pty Ltd, 1695 Pumicestone Road, Toorbul, Queensland, 4510, Australia
| | - Peter Timms
- Genecology Research Centre, University of the Sunshine Coast, 90 Sippy Downs Drive, Sippy Downs, Queensland, 4556, Australia
| | - Carolyn J Hogg
- School of Life and Environmental Sciences, The University of Sydney, New South Wales, 2006, Australia
| | - Katherine Belov
- School of Life and Environmental Sciences, The University of Sydney, New South Wales, 2006, Australia
| |
Collapse
|
3
|
Wen Y, Luo F, Zhao L, Su S, Lei W, Liu Y, Shi K, Li Z. Long Non-Coding RNA FGD5-AS1 Induced by Chlamydia trachomatis Infection Inhibits Apoptosis via Wnt/β-Catenin Signaling Pathway. Front Cell Infect Microbiol 2021; 11:701352. [PMID: 34568091 PMCID: PMC8460124 DOI: 10.3389/fcimb.2021.701352] [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: 05/22/2021] [Accepted: 07/15/2021] [Indexed: 12/12/2022] Open
Abstract
Background Chlamydia trachomatis (Ct) is one of the most common bacterial sexually transmitted infection (STI) pathogens in the world, but the exact pathogenic mechanism still needs to be further elucidated. Long non-coding RNAs (lncRNAs) have become vital regulators in many biological processes. Their role in the interaction between Ct and host cells has not been reported. Methods Microarrays were used to study the expression profiles of lncRNAs and mRNAs in HeLa cells at 12, 24, and 40 h post-infection (hpi). Differentially expressed lncRNAs and mRNAs were verified by RT-qPCR. Coding-non-coding (CNC) network analysis showed co-expression molecules of selected lncRNA. Western blot, flow cytometry, and indirect immunofluorescence were used to detect the effect of lncRNA FGD5-AS1 on apoptosis during Ct infection. Results Compared with the uninfected group, the number of differential lncRNAs were 2,130, 1,081, and 1,101 at 12, 24, and 40 hpi, and the number of differential mRNAs was 1,998, 1,129, and 1,330, respectively. Ct induced differential expression of large amounts of lncRNAs and mRNAs in HeLa cells, indicating that lncRNAs may play roles in the pathogenesis of Ct. RT-qPCR verified six differential lncRNAs and six differential mRNAs, confirming the reliability of the microarray. Among these molecules, lncRNA FGD5-AS1 was found to be upregulated at 12 and 24 hpi. Coding-non-coding (CNC) network analysis showed that co-expressed differential molecules of FGD5-AS1 at 12 and 24 hpi were enriched in the DNA replication and Wnt signaling pathway. The downregulation of FGD5-AS1 decreased the expression of β-catenin and inhibited the translocation of β-catenin and the DNA replication, while it promoted apoptosis of the host cells. Conclusions DNA replication and apoptosis of host cells were affected by upregulating FGD5-AS1 via Wnt/β-catenin pathway during Ct infection. This study provides evidence that lncRNAs are involved in the coaction between Ct and hosts, and provides new insights into the study of lncRNAs that regulate chlamydial infection.
Collapse
Affiliation(s)
- Yating Wen
- Institute of Pathogenic Biology, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| | - Fangzhen Luo
- Institute of Pathogenic Biology, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| | - Lanhua Zhao
- Institute of Pathogenic Biology, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| | - Shengmei Su
- Institute of Pathogenic Biology, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| | - Wenbo Lei
- Institute of Pathogenic Biology, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| | - Yi Liu
- Institute of Pathogenic Biology, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| | - Keliang Shi
- Institute of Pathogenic Biology, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| | - Zhongyu Li
- Institute of Pathogenic Biology, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| |
Collapse
|
4
|
He Z, Xiao J, Wang J, Lu S, Zheng K, Yu M, Liu J, Wang C, Ding N, Liang M, Wu Y. The Chlamydia psittaci Inclusion Membrane Protein 0556 Inhibits Human Neutrophils Apoptosis Through PI3K/AKT and NF-κB Signaling Pathways. Front Immunol 2021; 12:694573. [PMID: 34484191 PMCID: PMC8414580 DOI: 10.3389/fimmu.2021.694573] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 07/23/2021] [Indexed: 01/09/2023] Open
Abstract
Inclusion membrane proteins (Incs) play an important role in the structure and stability of chlamydial inclusion and the interaction between Chlamydia spp. and their hosts. Following Chlamydia infection through the respiratory tract, human polymorphonuclear neutrophils (hPMN) not only act as the primary immune cells reaching the lungs, but also serve as reservoir for Chlamydia. We have previously identified a Chlamydia psittaci hypothetical protein, CPSIT_0556, as a medium expressed inclusion membrane protein. However, the role of inclusion membrane protein, CPSIT_0556 in regulating hPMN functions remains unknown. In the present study, we found that CPSIT_0556 could not only inhibit hPMN apoptosis through the PI3K/Akt and NF-κB signaling pathways by releasing IL-8, but also delays procaspase-3 processing and inhibits caspase-3 activity in hPMN. Up-regulating the expression of anti-apoptotic protein Mcl-1 and down-regulating the expression of pro-apoptotic protein Bax could also inhibit the translocalization of Bax in the cytoplasm into the mitochondria, as well as induce the transfer of p65 NF-κB from the cytoplasm to the nucleus. Overall, our findings demonstrate that CPSIT_0556 could inhibit hPMN apoptosis through PI3K/Akt and NF-κB pathways and provide new insights towards understanding a better understanding of the molecular pathogenesis and immune escape mechanisms of C. psittaci.
Collapse
Affiliation(s)
- Zhangping He
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hengyang, China.,Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China.,Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, China
| | - Jian Xiao
- Department of Clinical Laboratory, The Affiliated Nanhua Hospital of University of South China, Hengyang, China
| | - Jianye Wang
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hengyang, China.,Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China.,Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, China
| | - Simin Lu
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hengyang, China.,Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China.,Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, China
| | - Kang Zheng
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hengyang, China.,Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China.,Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, China
| | - Maoying Yu
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hengyang, China.,Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China.,Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, China
| | - Jie Liu
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hengyang, China.,Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China.,Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, China
| | - Chuan Wang
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hengyang, China.,Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China.,Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, China
| | - Nan Ding
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hengyang, China.,Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China.,Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, China
| | - Mingxing Liang
- Department of Clinical Laboratory, The Affiliated Huaihua Hospital of University of South China, Huaihua, China
| | - Yimou Wu
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hengyang, China.,Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China.,Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, China
| |
Collapse
|
5
|
Sixt BS. Host cell death during infection with Chlamydia: a double-edged sword. FEMS Microbiol Rev 2021; 45:5902849. [PMID: 32897321 PMCID: PMC7794043 DOI: 10.1093/femsre/fuaa043] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 09/02/2020] [Indexed: 12/12/2022] Open
Abstract
The phylum Chlamydiae constitutes a group of obligate intracellular bacteria that infect a remarkably diverse range of host species. Some representatives are significant pathogens of clinical or veterinary importance. For instance, Chlamydia trachomatis is the leading infectious cause of blindness and the most common bacterial agent of sexually transmitted diseases. Chlamydiae are exceptionally dependent on their eukaryotic host cells as a consequence of their developmental biology. At the same time, host cell death is an integral part of the chlamydial infection cycle. It is therefore not surprising that the bacteria have evolved exquisite and versatile strategies to modulate host cell survival and death programs to their advantage. The recent introduction of tools for genetic modification of Chlamydia spp., in combination with our increasing awareness of the complexity of regulated cell death in eukaryotic cells, and in particular of its connections to cell-intrinsic immunity, has revived the interest in this virulence trait. However, recent advances also challenged long-standing assumptions and highlighted major knowledge gaps. This review summarizes current knowledge in the field and discusses possible directions for future research, which could lead us to a deeper understanding of Chlamydia's virulence strategies and may even inspire novel therapeutic approaches.
Collapse
Affiliation(s)
- Barbara S Sixt
- The Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå Centre for Microbial Research (UCMR), Department of Molecular Biology, Umeå University, SE-901 87 Umeå, Sweden
| |
Collapse
|
6
|
Negara KS, Suwiyoga K, Sudewi R, Astawa NM, Arijana GNK, Tunas K, Pemayun TGA. The role of caspase-dependent and caspase-independent pathways of apoptosis in the premature rupture of the membranes: A case-control study. Int J Reprod Biomed 2020; 18:439-448. [PMID: 32754679 PMCID: PMC7340985 DOI: 10.18502/ijrm.v13i6.7285] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 01/01/2019] [Accepted: 12/30/2019] [Indexed: 11/30/2022] Open
Abstract
Background Premature rupture of membrane (PROM) remains a problem in obstetrics, the mechanisms of PROM have not been clearly defined. Apoptosis is thought to play a key role in the mechanism, via caspase-dependent and caspase-independent pathways. Caspase-3, Apoptosis-inducing factor (AIF), and anti-apoptosis B-cell lymphoma 2 (Bcl-2) are hypothesized to be involved in PROM. Objective To determine the role of caspase-dependent and caspase-independent pathways in the mechanism of PROM. Materials and Methods This was a case-control study involving 42 pregnant women with gestational age between 20-42 wk. Participants were divided into the case group (with PROM) and control group (without PROM). Amniotic membranes were collected immediately after the delivery, and samples were taken from the site of membrane rupture. Immunohistochemical examination was done to determine the expression of Caspase-3, AIF, and Bcl-2. Results The expressions of Caspase-3 (OR = 9.75; 95% CI = 2.16-43.95; p = 0.001) and AIF (OR = 6.60; 95% CI = 1.48-29.36; p = 0.009) were significantly increased, whereas, Bcl-2 expressions (OR = 8.00; 95% CI = 1.79-35.74; p = 0.004) were significantly decreased in the case group. Conclusion High Caspase-3, AIF, and low Bcl-2 expression were the risk factors for PROM. Thus, it is evident that caspase-dependent and caspase-independent pathways are involved in the mechanism of PROM.
Collapse
Affiliation(s)
- Ketut Surya Negara
- Department of Obstetrics and Gynecology, Medical Faculty of Udayana University, Sanglah Hospital, Bali, Indonesia
| | - Ketut Suwiyoga
- Department of Obstetrics and Gynecology, Medical Faculty of Udayana University, Sanglah Hospital, Bali, Indonesia
| | - Raka Sudewi
- Department of Neurology, Medical Faculty of Udayana University, Sanglah Hospital Bali, Indonesia
| | | | | | - Ketut Tunas
- Department of Public Health, Dhyana Pura University Bali, Indonesia
| | - Tjokorda Gede Astawa Pemayun
- Department of Obstetrics and Gynecology, Medical Faculty of Udayana University, Sanglah Hospital, Bali, Indonesia
| |
Collapse
|
7
|
Leger T, Jouve C, Hininger-Favier I, Rigaudiere JP, Capel F, Sapin V, Moreau C, Charrier A, Demaison L. EPA is Cardioprotective in Male Rats Subjected to Sepsis, but ALA Is Not Beneficial. Antioxidants (Basel) 2020; 9:antiox9050371. [PMID: 32365668 PMCID: PMC7278601 DOI: 10.3390/antiox9050371] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 04/18/2020] [Accepted: 04/28/2020] [Indexed: 01/06/2023] Open
Abstract
It has been proven that dietary eicosapentaenoic acid (C20:5 n-3 or EPA) protects the heart against the deleterious effects of sepsis in female rats. We do not know if this is the case for male rodents. In this case, the efficiency of other n-3 polyunsaturated fatty acids (PUFAs) remains to be determined in both female and male rats. This study aimed at (i) determining whether dietary EPA is cardioprotective in septic male rats; (ii) evaluating the influence of dietary α-linolenic (C18:3 n-3 or ALA) on cardiac function during this pathology; and (iii) finding out the physiological and molecular mechanisms responsible for the observed effects. Sixty male rats were divided into three dietary groups. The animals were fed a diet deficient in n-3 PUFAs (DEF group), a diet enriched with ALA (ALA group) or a diet fortified with EPA (EPA group) for 6 weeks. Thereafter, each group was subdivided into 2 subgroups, one being subjected to cecal ligation and puncture (CLP) and the other undergoing a fictive surgery. Cardiac function was determined in vivo and ex vivo. Several parameters related to the inflammation process and oxidative stress were determined. Finally, the fatty acid compositions of circulating lipids and cardiac phospholipids were evaluated. The results of the ex vivo situation indicated that sepsis triggered cardiac damage in the DEF group. Conversely, the ex vivo data indicated that dietary ALA and EPA were cardioprotective by resolving the inflammation process and decreasing the oxidative stress. However, the measurements of the cardiac function in the in vivo situation modulated these conclusions. Indeed, in the in vivo situation, sepsis deteriorated cardiac mechanical activity in the ALA group. This was suspected to be due to a restricted coronary flow which was related to a lack of cyclooxygenase substrates in membrane phospholipids. Finally, only EPA proved to be beneficial in sepsis. Its action necessitates both resolution of inflammation and increased coronary perfusion. In that sense, dietary ALA, which does not allow the accumulation of vasodilator precursors in membrane lipids, cannot be protective during the pathology.
Collapse
Affiliation(s)
- Thibault Leger
- UNH, Unité de Nutrition Humaine, UMR 1019, Université Clermont Auvergne, INRAE, CRNH Auvergne, 63000 Clermont-Ferrand, France; (T.L.); (C.J.); (J.-P.R.); (F.C.); (C.M.); (A.C.)
| | - Chrystèle Jouve
- UNH, Unité de Nutrition Humaine, UMR 1019, Université Clermont Auvergne, INRAE, CRNH Auvergne, 63000 Clermont-Ferrand, France; (T.L.); (C.J.); (J.-P.R.); (F.C.); (C.M.); (A.C.)
| | | | - Jean-Paul Rigaudiere
- UNH, Unité de Nutrition Humaine, UMR 1019, Université Clermont Auvergne, INRAE, CRNH Auvergne, 63000 Clermont-Ferrand, France; (T.L.); (C.J.); (J.-P.R.); (F.C.); (C.M.); (A.C.)
| | - Frédéric Capel
- UNH, Unité de Nutrition Humaine, UMR 1019, Université Clermont Auvergne, INRAE, CRNH Auvergne, 63000 Clermont-Ferrand, France; (T.L.); (C.J.); (J.-P.R.); (F.C.); (C.M.); (A.C.)
| | - Vincent Sapin
- Department of Medical Biochemistry and Molecular Biology, CHU Clermont-Ferrand, 63000 Clermont-Ferrand, France;
| | - Clarisse Moreau
- UNH, Unité de Nutrition Humaine, UMR 1019, Université Clermont Auvergne, INRAE, CRNH Auvergne, 63000 Clermont-Ferrand, France; (T.L.); (C.J.); (J.-P.R.); (F.C.); (C.M.); (A.C.)
| | - Alice Charrier
- UNH, Unité de Nutrition Humaine, UMR 1019, Université Clermont Auvergne, INRAE, CRNH Auvergne, 63000 Clermont-Ferrand, France; (T.L.); (C.J.); (J.-P.R.); (F.C.); (C.M.); (A.C.)
| | - Luc Demaison
- UNH, Unité de Nutrition Humaine, UMR 1019, Université Clermont Auvergne, INRAE, CRNH Auvergne, 63000 Clermont-Ferrand, France; (T.L.); (C.J.); (J.-P.R.); (F.C.); (C.M.); (A.C.)
- Correspondence:
| |
Collapse
|
8
|
Chlamydia psittaci-Infected Dendritic Cells Communicate with NK Cells via Exosomes To Activate Antibacterial Immunity. Infect Immun 2019; 88:IAI.00541-19. [PMID: 31658957 DOI: 10.1128/iai.00541-19] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Accepted: 10/21/2019] [Indexed: 12/31/2022] Open
Abstract
Dendritic cells (DCs) and natural killer (NK) cells are critically involved in the early response against various bacterial microbes. Functional activation of infected DCs and NK cell-mediated gamma interferon (IFN-γ) secretion essentially contribute to the protective immunity against Chlamydia How DCs and NK cells cooperate during the antichlamydial response is not fully understood. Therefore, in the present study, we investigated the functional interplay between Chlamydia-infected DCs and NK cells. Our biochemical and cell biological experiments show that Chlamydia psittaci-infected DCs display enhanced exosome release. We find that such extracellular vesicles (referred to as dexosomes) do not contain infectious bacterial material but strongly induce IFN-γ production by NK cells. This directly affects C. psittaci growth in infected target cells. Furthermore, NK cell-released IFN-γ in cooperation with tumor necrosis factor alpha (TNF-α) and/or dexosomes augments apoptosis of both noninfected and infected epithelial cells. Thus, the combined effect of dexosomes and proinflammatory cytokines restricts C. psittaci growth and attenuates bacterial subversion of apoptotic host cell death. In conclusion, this provides new insights into the functional cooperation between DCs, dexosomes, and NK cells in the early steps of antichlamydial defense.
Collapse
|
9
|
Chen H, Wen Y, Li Z. Clear Victory for Chlamydia: The Subversion of Host Innate Immunity. Front Microbiol 2019; 10:1412. [PMID: 31333596 PMCID: PMC6619438 DOI: 10.3389/fmicb.2019.01412] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 06/05/2019] [Indexed: 12/14/2022] Open
Abstract
As obligate intracellular bacterial pathogens, members of the Chlamydia genera are the pivotal triggers for a wide range of infections, which can lead to blinding trachoma, pelvic inflammation, and respiratory diseases. Because of their restricted parasitism inside eukaryotic cells, the pathogens have to develop multiple strategies for adaptation with the hostile intracellular environment—intrinsically present in all host cells—to survive. The strategies that are brought into play at different stages of chlamydial development mainly involve interfering with diverse innate immune responses, such as innate immune recognition, inflammation, apoptosis, autophagy, as well as the manipulation of innate immune cells to serve as potential niches for chlamydial replication. This review will focus on the innate immune responses against chlamydial infection, highlighting the underlying molecular mechanisms used by the Chlamydia spp. to counteract host innate immune defenses. Insights into these subtle pathogenic mechanisms not only provide a rationale for the augmentation of immune responses against chlamydial infection but also open avenues for further investigation of the molecular mechanisms driving the survival of these clinically important pathogens in host innate immunity.
Collapse
Affiliation(s)
- Hongliang Chen
- Institute of Pathogenic Biology, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China.,Department of Clinical Microbiology Laboratory, Chenzhou No. 1 People's Hospital, Chenzhou, China
| | - Yating Wen
- Institute of Pathogenic Biology, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| | - Zhongyu Li
- Institute of Pathogenic Biology, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| |
Collapse
|
10
|
Visser JG, Van Staden ADP, Smith C. Harnessing Macrophages for Controlled-Release Drug Delivery: Lessons From Microbes. Front Pharmacol 2019; 10:22. [PMID: 30740053 PMCID: PMC6355695 DOI: 10.3389/fphar.2019.00022] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 01/09/2019] [Indexed: 01/15/2023] Open
Abstract
With the effectiveness of therapeutic agents ever decreasing and the increased incidence of multi-drug resistant pathogens, there is a clear need for administration of more potent, potentially more toxic, drugs. Alternatively, biopharmaceuticals may hold potential but require specialized protection from premature in vivo degradation. Thus, a paralleled need for specialized drug delivery systems has arisen. Although cell-mediated drug delivery is not a completely novel concept, the few applications described to date are not yet ready for in vivo application, for various reasons such as drug-induced carrier cell death, limited control over the site and timing of drug release and/or drug degradation by the host immune system. Here, we present our hypothesis for a new drug delivery system, which aims to negate these limitations. We propose transport of nanoparticle-encapsulated drugs inside autologous macrophages polarized to M1 phenotype for high mobility and treated to induce transient phagosome maturation arrest. In addition, we propose a significant shift of existing paradigms in the study of host-microbe interactions, in order to study microbial host immune evasion and dissemination patterns for their therapeutic utilization in the context of drug delivery. We describe a system in which microbial strategies may be adopted to facilitate absolute control over drug delivery, and without sacrificing the host carrier cells. We provide a comprehensive summary of the lessons we can learn from microbes in the context of drug delivery and discuss their feasibility for in vivo therapeutic application. We then describe our proposed "synthetic microbe drug delivery system" in detail. In our opinion, this multidisciplinary approach may hold the solution to effective, controlled drug delivery.
Collapse
Affiliation(s)
- Johan Georg Visser
- Department of Physiological Sciences, Stellenbosch University, Matieland, South Africa
| | | | - Carine Smith
- Department of Physiological Sciences, Stellenbosch University, Matieland, South Africa
| |
Collapse
|
11
|
Kerr MC, Gomez GA, Ferguson C, Tanzer MC, Murphy JM, Yap AS, Parton RG, Huston WM, Teasdale RD. Laser-mediated rupture of chlamydial inclusions triggers pathogen egress and host cell necrosis. Nat Commun 2017; 8:14729. [PMID: 28281536 PMCID: PMC5353685 DOI: 10.1038/ncomms14729] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 01/25/2017] [Indexed: 12/21/2022] Open
Abstract
Remarkably little is known about how intracellular pathogens exit the host cell in order to infect new hosts. Pathogenic chlamydiae egress by first rupturing their replicative niche (the inclusion) before rapidly lysing the host cell. Here we apply a laser ablation strategy to specifically disrupt the chlamydial inclusion, thereby uncoupling inclusion rupture from the subsequent cell lysis and allowing us to dissect the molecular events involved in each step. Pharmacological inhibition of host cell calpains inhibits inclusion rupture, but not subsequent cell lysis. Further, we demonstrate that inclusion rupture triggers a rapid necrotic cell death pathway independent of BAK, BAX, RIP1 and caspases. Both processes work sequentially to efficiently liberate the pathogen from the host cytoplasm, promoting secondary infection. These results reconcile the pathogen's known capacity to promote host cell survival and induce cell death.
Collapse
Affiliation(s)
- Markus C. Kerr
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Guillermo A. Gomez
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Charles Ferguson
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Maria C. Tanzer
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria 3052, Australia
| | - James M. Murphy
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Alpha S. Yap
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Robert G. Parton
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Wilhelmina M. Huston
- School of Life Sciences, University of Technology Sydney, Ultimo, New South Wales 2007, Australia
| | - Rohan D Teasdale
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| |
Collapse
|
12
|
Nogueira AT, Pedrosa AT, Carabeo RA. Manipulation of the Host Cell Cytoskeleton by Chlamydia. Curr Top Microbiol Immunol 2016; 412:59-80. [PMID: 27197645 DOI: 10.1007/82_2016_10] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
Chlamydiae are obligate intracellular pathogens. They undergo a biphasic developmental cycle differentiating between the infectious but metabolically quiescent elementary body and the vegetative, but non-infectious reticulate body. Chlamydia spends a significant portion of its development in the non-infectious stage, demanding an effective strategy of manipulating the host cells to ensure its intracellular survival and replication. A common target of all Chlamydia species studied so far is the host cell cytoskeleton, with past and recent findings revealing crucial roles in invasion, inclusion maintenance, nutrient acquisition, and egress. The molecular details of how Chlamydia co-opts the cytoskeleton is becoming clearer, with bacterial factors and their corresponding host cell targets identified.
Collapse
Affiliation(s)
- Ana T Nogueira
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| | - Antonio T Pedrosa
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| | - Rey A Carabeo
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA, USA.
| |
Collapse
|
13
|
Assis PA, Espíndola MS, Paula-Silva FWG, Rios WM, Pereira PAT, Leão SC, Silva CL, Faccioli LH. Mycobacterium tuberculosis expressing phospholipase C subverts PGE2 synthesis and induces necrosis in alveolar macrophages. BMC Microbiol 2014; 14:128. [PMID: 24886263 PMCID: PMC4057917 DOI: 10.1186/1471-2180-14-128] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Accepted: 05/06/2014] [Indexed: 01/15/2023] Open
Abstract
Background Phospholipases C (PLCs) are virulence factors found in several bacteria. In Mycobacterium tuberculosis (Mtb) they exhibit cytotoxic effects on macrophages, but the mechanisms involved in PLC-induced cell death are not fully understood. It has been reported that induction of cell necrosis by virulent Mtb is coordinated by subversion of PGE2, an essential factor in cell membrane protection. Results Using two Mtb clinical isolates carrying genetic variations in PLC genes, we show that the isolate 97-1505, which bears plcA and plcB genes, is more resistant to alveolar macrophage microbicidal activity than the isolate 97-1200, which has all PLC genes deleted. The isolate 97-1505 also induced higher rates of alveolar macrophage necrosis, and likewise inhibited COX-2 expression and PGE2 production. To address the direct effect of mycobacterial PLC on cell necrosis and PGE2 inhibition, both isolates were treated with PLC inhibitors prior to macrophage infection. Interestingly, inhibition of PLCs affected the ability of the isolate 97-1505 to induce necrosis, leading to cell death rates similar to those induced by the isolate 97-1200. Finally, PGE2 production by Mtb 97-1505-infected macrophages was restored to levels similar to those produced by 97-1200-infected cells. Conclusions Mycobacterium tuberculosis bearing PLCs genes induces alveolar macrophage necrosis, which is associated to subversion of PGE2 production.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Lúcia H Faccioli
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Av Cafe, s/n, Ribeirão Preto, SP 14040-903, Brazil.
| |
Collapse
|
14
|
Briken V. Mycobacterium tuberculosis genes involved in regulation of host cell death. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 783:93-102. [PMID: 23468105 DOI: 10.1007/978-1-4614-6111-1_5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The topic of host cell death response upon Mycobacterium tuberculosis (Mtb) infection has been a controversial one [1]. Recent findings demonstrate that one of the important confounding factors was most likely the fact that while Mtb inhibits host cell apoptosis induction early during the infection it clearly induces a necrotic form of cell death during later infection stages [2, 3]. This bi-phasic intracellular lifestyle in regard to host cell death manipulation is emerging as a common theme shared with other facultative and obligate intracellular bacterial pathogens such as Chlamydia and Legionella [4-6]. Accordingly, the list of discovered bacterial proteins involved in host cell apoptosis inhibition is growing [7, 8]. At the same time it is clearly beneficial for the resistance of the host to overcome the bacterial apoptosis block during the early stage of the infection [9-11]. Hence, host cell components have evolved to recognize intracellular pathogens and mediate host cell apoptosis induction if necessary [12]. There have been several reviews on the various aspects of the host cell death response upon Mtb infection [1, 3, 13-15]. Thus in this chapter I will focus on the pathogen side of the equation and describe the tremendous progress that has been made in the identification and characterization of Mtb genes involved in manipulation of host cell death pathways.
Collapse
Affiliation(s)
- Volker Briken
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742, USA.
| |
Collapse
|
15
|
Abstract
Pathogenicity of Chlamydia and Chlamydia-related bacteria could be partially mediated by an enhanced activation of the innate immune response. The study of this host pathogen interaction has proved challenging due to the restricted in vitro growth of these strict intracellular bacteria and the lack of genetic tools to manipulate their genomes. Despite these difficulties, the interactions of Chlamydiales with the innate immune cells and their effectors have been studied thoroughly. This review aims to point out the role of pattern recognition receptors and signal molecules (cytokines, reactive oxygen species) of the innate immune response in the pathogenesis of chlamydial infection. Besides inducing clearance of the bacteria, some of these effectors may be used by the Chlamydia to establish chronic infections or to spread. Thus, the induced innate immune response seems to be variable depending on the species and/or the serovar, making the pattern more complex. It remains crucial to determine the common players of the innate immune response in order to help define new treatment strategies and to develop effective vaccines. The excellent growth in phagocytic cells of some Chlamydia-related organisms such as Waddlia chondrophila supports their use as model organisms to study conserved features important for interactions between the innate immunity and Chlamydia.
Collapse
Affiliation(s)
- Brigida Rusconi
- Institute of Microbiology, University of Lausanne and University Hospital Center, Lausanne, Switzerland
| | | |
Collapse
|
16
|
El-Salem M, Raghunath PN, Marzec M, Liu X, Kasprzycka M, Robertson E, Wasik MA. Activation of mTORC1 signaling pathway in AIDS-related lymphomas. THE AMERICAN JOURNAL OF PATHOLOGY 2009; 175:817-24. [PMID: 19608873 DOI: 10.2353/ajpath.2009.080451] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Using immunohistochemistry with antibodies against the phosphoserine residues in both S6rp and 4E binding protein 1, we identified the activation of the mammalian target of rapamycin (mTORC)1 pathway in 29 cases of AIDS-related lymphoma. These cases represented a diverse spectrum of histological types of non-Hodgkin lymphoma (24 cases) and classic Hodgkin lymphoma (five cases). mTORC1 was also activated in the hyperplastic but not involuted follicles of HIV-associated lymphadenopathy in eight cases, supporting the notion that mTORC1 activation is a common feature of transformed lymphocytes irrespective of either their reactive or malignant phenotype. We also found that in B-cell lines that represent diffuse large B-cell lymphoma, Burkitt lymphoma, Epstein-Barr virus-infected lymphocytes, and human herpesvirus 8-positive primary effusion lymphoma, inhibitors of Syk, MEK, and, seemingly, phosphoinositide 3 kinases suppressed mTORC1 activation, in particular when these inhibitors were used in combination. These findings indicate that AIDS-related lymphoma and other histologically similar types of lymphomas that are derived from transformed B lymphocytes may display clinical responses to inhibitors that directly target mTORC1 or, possibly, upstream activators of the mTORC1 pathway.
Collapse
Affiliation(s)
- Mouna El-Salem
- University of Pennsylvania Medical Center, Department of Pathology and Laboratory Medicine, 3400 Spruce Street, 7.106 Founders Pavilion, Philadelphia, PA 19104, USA
| | | | | | | | | | | | | |
Collapse
|
17
|
Abstract
The exit of intracellular pathogens from host cells is an important step in the infectious cycle, but is poorly understood. It has recently emerged that microbial exit is a process that can be directed by organisms from within the cell, and is not simply a consequence of the physical or metabolic burden that is imposed on the host cell. This Review summarizes our current knowledge on the diverse mechanisms that are used by intracellular pathogens to exit cells. An integrated understanding of the diversity that exists for microbial exit pathways represents a new horizon in the study of host-pathogen interactions.
Collapse
|
18
|
Kohlhoff SA, Kutlin A, Riska P, Roblin PM, Roman C, Hammerschlag MR. In vitro models of acute and long-term continuous infection of human respiratory epithelial cells with Chlamydophila pneumoniae have opposing effects on host cell apoptosis. Microb Pathog 2007; 44:34-42. [PMID: 17888618 DOI: 10.1016/j.micpath.2007.08.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2007] [Accepted: 08/01/2007] [Indexed: 02/01/2023]
Abstract
Persistent infection with the obligate intracellular pathogen Chlamydophila pneumoniae has been implicated in the pathogenesis of many chronic diseases, but its mechanism remains unclear. Many pathogens have been found to modulate cellular apoptosis in order to survive and multiply. Chlamydial species were shown to both induce and inhibit host cell apoptosis depending on the experimental conditions. We utilized in vitro models of acute and long-term continuous (LTC) infection with the same cell line (HEp-2) and chlamydial isolate (TW-183) used in both models. Host cell apoptosis in infected and uninfected cells was assessed by fluorescence microscopy and flow cytometry. While acute infection induced apoptosis 72 h post-infection, LTC-infected cells had low rates of apoptosis and showed resistance to different exogenous inducers of apoptosis (sorbitol, serum withdrawal, hydrogen peroxide) when compared to uninfected cells. Chronicity of infection appears to be a critical factor in the modulation of host cell apoptosis by C. pneumoniae. Induction of apoptosis may help to propagate the infection, while inhibition of apoptosis could help protect the organism in chronic infection.
Collapse
Affiliation(s)
- Stephan A Kohlhoff
- Department of Pediatrics, SUNY Downstate Medical Center, 450 Clarkson Avenue, Brooklyn, NY 11203, USA.
| | | | | | | | | | | |
Collapse
|
19
|
Hybiske K, Stephens RS. Mechanisms of host cell exit by the intracellular bacterium Chlamydia. Proc Natl Acad Sci U S A 2007; 104:11430-5. [PMID: 17592133 PMCID: PMC2040915 DOI: 10.1073/pnas.0703218104] [Citation(s) in RCA: 350] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The mechanisms that mediate the release of intracellular bacteria from cells are poorly understood, particularly for those that live within a cellular vacuole. The release pathway of the obligate intracellular bacterium Chlamydia from cells is unknown. Using a GFP-based approach to visualize chlamydial inclusions within cells by live fluorescence videomicroscopy, we identified that Chlamydia release occurred by two mutually exclusive pathways. The first, lysis, consisted of an ordered sequence of membrane permeabilizations: inclusion, nucleus and plasma membrane rupture. Treatment with protease inhibitors abolished inclusion lysis. Intracellular calcium signaling was shown to be important for plasma membrane breakdown. The second release pathway was a packaged release mechanism, called extrusion. This slow process resulted in a pinching of the inclusion, protrusion out of the cell within a cell membrane compartment, and ultimately detachment from the cell. Treatment of Chlamydia-infected cells with specific pharmacological inhibitors of cellular factors demonstrated that extrusion required actin polymerization, neuronal Wiskott-Aldrich syndrome protein, myosin II and Rho GTPase. The participation of Rho was unique in that it functioned late in extrusion. The dual nature of release characterized for Chlamydia has not been observed as a strategy for intracellular bacteria.
Collapse
Affiliation(s)
- Kevin Hybiske
- Division of Infectious Diseases, School of Public Health, University of California, Berkeley, CA 94720
| | - Richard S. Stephens
- Division of Infectious Diseases, School of Public Health, University of California, Berkeley, CA 94720
- *To whom correspondence should be addressed. E-mail:
| |
Collapse
|
20
|
Yaraei K, Campbell LA, Zhu X, Liles WC, Kuo CC, Rosenfeld ME. Chlamydia pneumoniae augments the oxidized low-density lipoprotein-induced death of mouse macrophages by a caspase-independent pathway. Infect Immun 2005; 73:4315-22. [PMID: 15972525 PMCID: PMC1168605 DOI: 10.1128/iai.73.7.4315-4322.2005] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2004] [Revised: 02/03/2005] [Accepted: 02/28/2005] [Indexed: 11/20/2022] Open
Abstract
Chlamydia pneumoniae is a common respiratory pathogen that is associated with an increased risk of cardiovascular disease. However, the mechanisms by which C. pneumoniae contributes to cardiovascular disease have not been determined yet. C. pneumoniae infection may accelerate the death of cells within atherosclerotic lesions and contribute to the formation of unstable lesions. To test this hypothesis, the impact of C. pneumoniae infection on the death of lipid-loaded mouse macrophages was investigated. It was observed that RAW 264.7 cells are highly susceptible to the toxic effects of oxidized low-density lipoprotein (LDL) and exhibit markers of cell death within 24 h of treatment with as little as 5 microg/ml oxidized LDL. Subsequent infection with either live C. pneumoniae or heat-killed or UV-inactivated C. pneumoniae at a low multiplicity of infection for 24 to 72 h stimulated both additional binding of annexin V and the uptake of propidium iodide. Thus, C. pneumoniae augments the effects of oxidized LDL on cell death independent of a sustained infection. However, unlike oxidized LDL, C. pneumoniae infection does not activate caspase 3 or induce formation of the mitochondrial transition pore or the fragmentation of DNA, all of which are classical markers of apoptosis. Furthermore, primary bone marrow macrophages isolated from mice deficient in Toll-like receptor 2 (TLR-2) but not TLR-4 are resistant to C. pneumoniae-induced death. These data suggest that C. pneumoniae kills cells by a caspase-independent pathway and that the process is potentially mediated by activation of TLR-2.
Collapse
Affiliation(s)
- Kambiz Yaraei
- Department of Pathobiology, Box 353410, University of Washington, Seattle, Washington 98195, USA
| | | | | | | | | | | |
Collapse
|
21
|
Knodler LA, Finlay BB, Steele-Mortimer O. The Salmonella effector protein SopB protects epithelial cells from apoptosis by sustained activation of Akt. J Biol Chem 2005; 280:9058-64. [PMID: 15642738 DOI: 10.1074/jbc.m412588200] [Citation(s) in RCA: 181] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Invasion of epithelial cells by Salmonella enterica is mediated by bacterial "effector" proteins that are delivered into the host cell by a type III secretion system. Although primarily known for their roles in actin rearrangements and membrane ruffling, translocated effectors also affect host cell processes that are not directly associated with invasion. Here, we show that SopB/SigD, an effector with phosphoinositide phosphatase activity, has anti-apoptotic activity in Salmonella-infected epithelial cells. Salmonella induced the sustained activation of Akt/protein kinase B, a pro-survival kinase, in a SopB-dependent manner. Failure to activate Akt resulted in increased levels of apoptosis after infection with a sopB deletion mutant (DeltasopB). Furthermore, cells infected with wild type bacteria, but not the DeltasopB strain, were protected from camptothecin-induced cleavage of caspase-3 and subsequent apoptosis. The anti-apoptotic activity of SopB was dependent on its phosphatase activity, because a catalytically inactive mutant was unable to protect cells from the effects of camptothecin. Finally, small interfering RNA was used to demonstrate the essential role of Akt in SopB-mediated protection against apoptosis. These results provide new insights into the mechanisms of apoptosis and highlight how bacterial effectors can intercept signaling pathways to manipulate host responses.
Collapse
Affiliation(s)
- Leigh A Knodler
- Laboratory of Intracellular Parasites, NIAID, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, Montana 59840, USA
| | | | | |
Collapse
|
22
|
Byrne GI, Ojcius DM. Chlamydia and apoptosis: life and death decisions of an intracellular pathogen. Nat Rev Microbiol 2004; 2:802-8. [PMID: 15378044 DOI: 10.1038/nrmicro1007] [Citation(s) in RCA: 153] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The chlamydiae are important obligate intracellular prokaryotic pathogens that, each year, are responsible for millions of human infections involving the eye, genital tract, respiratory tract, vasculature and joints. The chlamydiae grow in cytoplasmic vesicles in susceptible host cells, which include the mucosal epithelium, vascular endothelium, smooth muscle cells, circulating monocytes and recruited or tissue-specific macrophages. One important pathogenic strategy that chlamydiae have evolved to promote their survival is the modulation of programmed cell death pathways in infected host cells. The chlamydiae can elicit the induction of host cell death, or apoptosis, under some circumstances and actively inhibit apoptosis under others. This subtle pathogenic mechanism highlights the manner in which these highly successful pathogens take control of infected cells to promote their own survival - even under the most adverse circumstances.
Collapse
Affiliation(s)
- Gerald I Byrne
- Department of Molecular Sciences, University of Tennessee Health Science Center, 858 Madison Avenue, Memphis, Tennessee 38163, USA.
| | | |
Collapse
|
23
|
Yilmaz O, Jungas T, Verbeke P, Ojcius DM. Activation of the phosphatidylinositol 3-kinase/Akt pathway contributes to survival of primary epithelial cells infected with the periodontal pathogen Porphyromonas gingivalis. Infect Immun 2004; 72:3743-51. [PMID: 15213114 PMCID: PMC427421 DOI: 10.1128/iai.72.7.3743-3751.2004] [Citation(s) in RCA: 184] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Porphyromonas gingivalis, an important periodontal pathogen, infects primary gingival epithelial cells (GECs). Despite the large number of bacteria that replicate inside the GECs, the host cell remains viable. We demonstrate that P. gingivalis triggers rapid and reversible surface phosphatidylserine exposure through a mechanism requiring caspase activation. However, after 1 day of infection, the bacteria no longer induce phosphatidylserine externalization and instead protect infected cells against apoptosis. Infection exerts its effect at the level of mitochondria, as P. gingivalis also blocks depolarization of the mitochondrial transmembrane potential and cytochrome c release. Interestingly, protein kinase B/Akt is phosphorylated during infection, which can be blocked with the phosphatidylinositol 3-kinase (PI3K) inhibitor LY294002. Suppression of the PI3K/Akt pathway following staurosporine treatment results in mitochondrial-membrane depolarization, cytochrome c release, DNA fragmentation, and increased apoptosis of infected GECs. Thus, P. gingivalis stimulates early surface exposure of phosphatidylserine, which could downmodulate the inflammatory response, while also promoting host cell survival through the PI3K/Akt pathway.
Collapse
Affiliation(s)
- Ozlem Yilmaz
- Department of Pathobiology, University of Washington, School of Public Health, HSC Box 357238, Seattle, WA 98195, USA.
| | | | | | | |
Collapse
|
24
|
Schwarzenbacher R, Stenner-Liewen F, Liewen H, Robinson H, Yuan H, Bossy-Wetzel E, Reed JC, Liddington RC. Structure of the Chlamydia protein CADD reveals a redox enzyme that modulates host cell apoptosis. J Biol Chem 2004; 279:29320-4. [PMID: 15087448 DOI: 10.1074/jbc.m401268200] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The Chlamydia protein CADD (Chlamydia protein associating with death domains) has been implicated in the modulation of host cell apoptosis via binding to the death domains of tumor necrosis factor family receptors. Transfection of CADD into mammalian cells induces apoptosis. Here we present the CADD crystal structure, which reveals a dimer of seven-helix bundles. Each bundle contains a di-iron center adjacent to an internal cavity, forming an active site similar to that of methane mono-oxygenase hydrolase. We further show that CADD mutants lacking critical metal-coordinating residues are substantially less effective in inducing apoptosis but retain their ability to bind to death domains. We conclude that CADD is a novel redox protein toxin unique to Chlamydia species and propose that both its redox activity and death domain binding ability are required for its biological activity.
Collapse
|