1
|
Murillo-Léon M, Bastidas-Quintero AM, Steinfeldt T. Decoding Toxoplasma gondii virulence: the mechanisms of IRG protein inactivation. Trends Parasitol 2024; 40:805-819. [PMID: 39168720 DOI: 10.1016/j.pt.2024.07.009] [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: 05/31/2024] [Revised: 07/17/2024] [Accepted: 07/18/2024] [Indexed: 08/23/2024]
Abstract
Toxoplasmosis is a common parasitic zoonosis that can be life-threatening in immunocompromised patients. About one-third of the human population is infected with Toxoplasma gondii. Primary infection triggers an innate immune response wherein IFN-γ-induced host cell GTPases, namely IRG and GBP proteins, serve as a vital component for host cell resistance. In the past decades, interest in elucidating the function of these GTPase families in controlling various intracellular pathogens has emerged. Numerous T. gondii effectors were identified to inactivate particular IRG proteins. T. gondii is re-optimizing its effectors to combat IRG function and in this way secures transmission. We discuss the IRG-specific effectors employed by the parasite in murine infections, contributing to a better understanding of T. gondii virulence.
Collapse
Affiliation(s)
- Mateo Murillo-Léon
- Institute of Medical Microbiology and Hygiene, Medical Center University of Freiburg, 79104 Freiburg, Germany; CIBSS, Centre for Integrative Biological Signalling Studies, University of Freiburg, 79104 Freiburg, Germany; Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Aura María Bastidas-Quintero
- Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany; Institute of Virology, Medical Center University of Freiburg, 79104 Freiburg, Germany; Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Tobias Steinfeldt
- Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany; Institute of Virology, Medical Center University of Freiburg, 79104 Freiburg, Germany.
| |
Collapse
|
2
|
Lu YN, Shen XY, Lu JM, Jin GN, Lan HW, Xu X, Piao LX. Resveratrol inhibits Toxoplasma gondii-induced lung injury, inflammatory cascade and evidences of its mechanism of action. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 108:154522. [PMID: 36332392 DOI: 10.1016/j.phymed.2022.154522] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 10/03/2022] [Accepted: 10/21/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND Toxoplasma gondii is an opportunistic protozoan that can infect host to cause toxoplasmosis. We have previously reported that resveratrol (RSV) has protective effects against liver damage in T. gondii infected mice. However, the effect of RSV on lung injury caused by T. gondii infection and its mechanism of action remain unclear. PURPOSE In this work, we studied the protective effects of RSV on lung injury caused by T. gondii infection and explored the underlying mechanism. METHODS Molecular docking and localized surface plasmon resonance assay were used to detect the molecular interactions between RSV and target proteins. In vitro, the anti-T. gondii effects and potential anti-inflammatory mechanisms of RSV were investigated by quantitative competitive-PCR, RT-PCR, ELISA, Western blotting and immunofluorescence using RAW 264.7 cells infected with tachyzoites of T. gondii RH strain. In vivo, the effects of RSV on lung injury caused by T. gondii infection were assessed by observing pathological changes and the expression of inflammatory factors of lung. RESULTS RSV inhibited T. gondii loads and T. gondii-derived heat shock protein 70 (T.g.HSP70) expression in RAW 264.7 cells and lung tissues. Moreover, RSV interacts with T.g.HSP70 and toll-like receptor 4 (TLR4), respectively, and interferes with the interaction between T.g.HSP70 and TLR4. It also inhibited the overproduction of inducible nitric oxide synthase, TNF-α and high mobility group protein 1 (HMGB1) by down-regulating TLR4/nuclear factor kappa B (NF-κB) signaling pathway, which is consistent with the effect of TLR4 inhibitor CLI-095. In vivo, RSV improved the pathological lung damage produced by T. gondii infection, as well as decreased the number of inflammatory cells in bronchoalveolar lavage fluid and the release of HMGB1 and TNF-α. CONCLUSION These findings indicate that RSV can inhibit the proliferation of T. gondii and T.g.HSP70 expression both in vitro and in vivo. RSV can inhibit excessive inflammatory response by intervening T.g.HSP70 and HMGB1 mediated TLR4/NF-κB signaling pathway activation, thereby ameliorating lung injury caused by T. gondii infection. The present study provides new data that may be useful for the development of RSV as a new agent for the treatment of lung damage caused by T. gondii infection.
Collapse
Affiliation(s)
- Yu Nan Lu
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji 133002, Jilin Province, PR. China
| | - Xin Yu Shen
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji 133002, Jilin Province, PR. China
| | - Jing Mei Lu
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji 133002, Jilin Province, PR. China
| | - Guang Nan Jin
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji 133002, Jilin Province, PR. China
| | - Hui Wen Lan
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji 133002, Jilin Province, PR. China
| | - Xiang Xu
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji 133002, Jilin Province, PR. China.
| | - Lian Xun Piao
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji 133002, Jilin Province, PR. China.
| |
Collapse
|
3
|
Disruption of Toxoplasma gondii-Induced Host Cell DNA Replication Is Dependent on Contact Inhibition and Host Cell Type. mSphere 2022; 7:e0016022. [PMID: 35587658 PMCID: PMC9241542 DOI: 10.1128/msphere.00160-22] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The protozoan Toxoplasma gondii is a highly successful obligate intracellular parasite that, upon invasion of its host cell, releases an array of host-modulating protein effectors to counter host defenses and further its own replication and dissemination. Early studies investigating the impact of T. gondii infection on host cell function revealed that this parasite can force normally quiescent cells to activate their cell cycle program. Prior reports by two independent groups identified the dense granule protein effector HCE1/TEEGR as being solely responsible for driving host cell transcriptional changes through its direct interaction with the cyclin E regulatory complex DP1 and associated transcription factors. Our group independently identified HCE1/TEEGR through the presence of distinct repeated regions found in a number of host nuclear targeted parasite effectors and verified its central role in initiating host cell cycle changes. Additionally, we report here the time-resolved kinetics of host cell cycle transition in response to HCE1/TEEGR, using the fluorescence ubiquitination cell cycle indicator reporter line (FUCCI), and reveal the existence of a block in S-phase progression and host DNA synthesis in several cell lines commonly used in the study of T. gondii. Importantly, we have observed that this S-phase block is not due to additional dense granule effectors but rather is dependent on the host cell line background and contact inhibition status of the host monolayer in vitro. This work highlights intriguing differences in the host response to reprogramming by the parasite and raises interesting questions regarding how parasite effectors differentially manipulate the host cell depending on the in vitro or in vivo context. IMPORTANCEToxoplasma gondii chronically infects approximately one-third of the global population and can produce severe pathology in immunologically immature or compromised individuals. During infection, this parasite releases numerous host-targeted effector proteins that can dramatically alter the expression of a variety of host genes. A better understanding of parasite effectors and their host targets has the potential to not only provide ways to control infection but also inform us about our own basic biology. One host pathway that has been known to be altered by T. gondii infection is the cell cycle, and prior reports have identified a parasite effector, known as HCE1/TEEGR, as being responsible. In this report, we further our understanding of the kinetics of cell cycle transition induced by this effector and show that the capacity of HCE1/TEEGR to induce host cell DNA synthesis is dependent on both the cell type and the status of contact inhibition.
Collapse
|
4
|
Zhang L, Kang S, Chen H, Liao J, Sun M, Wu S, Xu Z, Xu L, Zhang X, Qin Q, Wei J. The roles of grouper TAK1 in regulating the infection of Singapore grouper iridovirus. FISH & SHELLFISH IMMUNOLOGY 2022; 124:164-173. [PMID: 35398221 DOI: 10.1016/j.fsi.2022.04.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 04/02/2022] [Accepted: 04/03/2022] [Indexed: 06/14/2023]
Abstract
Transforming growth factor-β activated kinase 1 (TAK1) is a member of the mitogen-activated protein kinase family. It is an upstream factor of the IκB kinase, which activates IKKα and IKKβ. TAK1 is a key factor in the induction of nuclear factor κB (NF-κB) and plays a crucial role in the activation of inflammatory responses. However, the roles of TAK1 during viral infection in teleost fish are largely unknown. In this study, we cloned a TAK1 homolog (HgTAK1) from the hybrid grouper (Epinephelus fuscoguttatus♂ × Epinephelus lanceolatus♀). The open reading frame of HgTAK1 consists of 1728 nucleotides encoding 575 amino acids, and the predicted molecular weight is 64.32 kDa HgTAK1 has an S_TKc domain, which consists of a serine/threonine protein kinase and a catalytic domain. Expression pattern analysis showed that HgTAK1 was distributed in all tested tissues, with abundant contents in the heart, head kidney, and blood. Additionally, HgTAK1 was distributed in the cytoplasm of grouper spleen (GS) cells. After Singapore grouper iridovirus (SGIV) infection, the expression of HgTAK1 increased in GS cells. Overexpression of HgTAK1 could promote the replication of SGIV in GS cells and inhibit the activation of NF-κB and IFN stimulated response elements (ISRE) in reporter assay. When co-expressed with IRF3 or HgIRF7 in GS cells, HgTAK1 obviously down-regulated IRF3- or IRF7-mediated the NF-κB and ISRE promoter induction. The interaction between HgTAK1 and IRF3 or IRF7 has been identified by co-immunoprecipitation assay. These findings provide a basis for understanding the innate immune mechanism of the grouper response to viral infection.
Collapse
Affiliation(s)
- Luhao Zhang
- University Joint Laboratory of Guangdong Province, Hong Kong and Macao Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, PR China
| | - Shaozhu Kang
- University Joint Laboratory of Guangdong Province, Hong Kong and Macao Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, PR China
| | - Hong Chen
- University Joint Laboratory of Guangdong Province, Hong Kong and Macao Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, PR China
| | - Jiaming Liao
- University Joint Laboratory of Guangdong Province, Hong Kong and Macao Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, PR China
| | - Mengshi Sun
- University Joint Laboratory of Guangdong Province, Hong Kong and Macao Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, PR China
| | - Siting Wu
- University Joint Laboratory of Guangdong Province, Hong Kong and Macao Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, PR China
| | - Zhuqing Xu
- University Joint Laboratory of Guangdong Province, Hong Kong and Macao Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, PR China
| | - Linting Xu
- University Joint Laboratory of Guangdong Province, Hong Kong and Macao Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, PR China
| | - Xin Zhang
- University Joint Laboratory of Guangdong Province, Hong Kong and Macao Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, PR China
| | - Qiwei Qin
- University Joint Laboratory of Guangdong Province, Hong Kong and Macao Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, PR China; Guangdong Laboratory for Lingnan Modern Agriculture, PR China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266000, PR China; Southern Marine Science and Engineering Guangdong Laboratory, Zhuhai, 528478, China.
| | - Jingguang Wei
- University Joint Laboratory of Guangdong Province, Hong Kong and Macao Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, PR China.
| |
Collapse
|
5
|
Sang R, Sun F, Zhou H, Wang M, Li H, Li C, Sun X, Zhao X, Zhang X. Immunomodulatory effects of Inonotus obliquus polysaccharide on splenic lymphocytes infected with Toxoplasma gondii via NF-κB and MAPKs pathways. Immunopharmacol Immunotoxicol 2021; 44:129-138. [PMID: 34918603 DOI: 10.1080/08923973.2021.2017453] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
CONTEXT As a medicinal and edible fungus, Inonotus obliquus has been traditionally used to prevent and treat various ailments. Inonotus obliquus polysaccharide (IOP) isolated from I. obliquus processes many biological activities, our series of in vivo studies have shown that IOP protects against Toxoplasma gondii infection. OBJECTIVE This study aimed to investigate the in vitro immunomodulatory effects and its mechanisms of IOP on mouse splenic lymphocytes infected with T. gondii. MATERIALS AND METHODS Mouse splenic lymphocytes were infected with T. gondii tachyzoites, and treated with different concentrations of IOP. The levels of cytokines and chemokines were measured by enzyme-linked immunosorbent assay (ELISA) and reverse transcription-polymerase chain reaction (RT-PCR). The expression of toll-like receptor 2 (TLR2) and TLR4, and the modulation of nuclear factor-kappa B (NF-κB) and mitogen-activated protein kinases (MAPKs) signaling pathways were determined by Western blot. RESULTS IOP significantly decreased the over-release of cytokine interleukin-1 beta (IL-1β), IL-4, IL-6, interferon-gamma (IFN-γ), and tumor necrosis factor-alpha (TNF-α) in supernatant from T. gondii-infected splenic lymphocytes. IOP also effectively inhibited the overexpression of cytokines and chemokine macrophage inflammatory protein-1 (MIP-1) and monocyte chemoattractant protein-1 (MCP-1) mRNA. Furthermore, IOP down-regulated TLR2 and TLR4 expressions and inhibited the over-phosphorylation of NF-κB p65 and inhibitor κBα (IκBα) in NF-κB signaling pathway and p38, c-Jun N-terminal kinase (JNK) in MAPKs signaling pathway. By observing the effect of IOP on TNF-α secretion after pretreatment with specific inhibitors, it was further confirmed that IOP was involved in the regulation of NF-κB, p38, and JNK signaling pathways. CONCLUSIONS These data indicate that IOP can inhibit the excessive inflammatory response caused by T. gondii infection through modulating NF-κB, p38, and JNK signaling pathways, and thus plays the in vitro anti-T. gondii role.
Collapse
Affiliation(s)
- Rui Sang
- Agricultural College, Yanbian University, Yanji, China
| | - Fuliang Sun
- Agricultural College, Yanbian University, Yanji, China
| | - Hongyuan Zhou
- Agricultural College, Yanbian University, Yanji, China
| | - Meng Wang
- Agricultural College, Yanbian University, Yanji, China
| | - Haitao Li
- Agricultural College, Yanbian University, Yanji, China.,Institute of Special Wild Economic Animals and Plants, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Chunting Li
- Agricultural College, Yanbian University, Yanji, China
| | - Xinhui Sun
- Agricultural College, Yanbian University, Yanji, China
| | - Xin Zhao
- Agricultural College, Yanbian University, Yanji, China
| | - Xuemei Zhang
- Agricultural College, Yanbian University, Yanji, China.,Engineering Research Center of North-East Cold Region Beef Cattle Science & Technology Innovation, Ministry of Education, Yanbian University, Yanji, China
| |
Collapse
|
6
|
Ihara F, Fereig RM, Himori Y, Kameyama K, Umeda K, Tanaka S, Ikeda R, Yamamoto M, Nishikawa Y. Toxoplasma gondii Dense Granule Proteins 7, 14, and 15 Are Involved in Modification and Control of the Immune Response Mediated via NF-κB Pathway. Front Immunol 2020; 11:1709. [PMID: 32849602 PMCID: PMC7412995 DOI: 10.3389/fimmu.2020.01709] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 06/26/2020] [Indexed: 11/13/2022] Open
Abstract
Toxoplasma gondii infects almost all warm-blooded animals, including humans, leading to both cellular and humoral immune responses in the host. The virulence of T. gondii is strain specific and is defined by secreted effector proteins that disturb host immunity. Here, we focus on nuclear factor-kappa B (NFκB) signaling, which regulates the induction of T-helper type 1 immunity. A luciferase assay for screening effector proteins, including ROPs and GRAs that have biological activity against an NFκB-dependent reporter plasmid, found that overexpression of GRA7, 14, and 15 of a type II strain resulted in a strong activity. Thus, our study was aimed at understanding the involvement of NFκB in the pathogenesis of toxoplasmosis through a comparative analysis of these three molecules. We found that GRA7 and GRA14 were partially involved in the activation of NFκB, whereas GRA15 was essential for NFκB activation. The deletion of GRA7, GRA14, and GRA15 in the type II Prugniaud (Pru) strain resulted in a defect in the nuclear translocation of RelA. Cells infected with the PruΔgra15 parasite showed reduced phosphorylation of inhibitor-κBα. GRA7, GRA14, and GRA15 deficiency decreased the levels of interleukin-6 in RAW246.7 cells, and RNA-seq analysis revealed that GRA7, GRA14, and GRA15 deficiency predominantly resulted in downregulation of gene expression mediated by NFκB. The virulence of all mutant strains increased, but PruΔgra14 only showed a slight increase in virulence. However, the intra-footpad injection of the highly-virulent type I RHΔgra14 parasites in mice resulted in increased virulence. This study shows that GRA7, 14, and 15-induced host immunity via NFκB limits parasite expansion.
Collapse
Affiliation(s)
- Fumiaki Ihara
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Japan
| | - Ragab M Fereig
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Japan.,Department of Animal Medicine, Faculty of Veterinary Medicine, South Valley University, Qena City, Egypt
| | - Yuu Himori
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Japan
| | - Kyohko Kameyama
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Japan
| | - Kosuke Umeda
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Japan
| | - Sachi Tanaka
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Japan.,Division of Animal Science, Department of Agricultural and Life Sciences, Faculty of Agriculture, Shinshu University, Nagano, Japan
| | - Rina Ikeda
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Japan
| | - Masahiro Yamamoto
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan.,Laboratory of Immunoparasitology, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Yoshifumi Nishikawa
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Japan
| |
Collapse
|
7
|
Guo Y, Xu Y, Xiong D, Zhou Y, Kang X, Meng C, Gu D, Jiao X, Pan Z. Molecular characterisation, expression and functional feature of TRAF6 in the King pigeon ( Columba livia). Innate Immun 2020; 26:490-504. [PMID: 32393097 PMCID: PMC7491236 DOI: 10.1177/1753425920920930] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
TNF receptor-associated factor 6 (TRAF6) is a signal transducer, which plays a pivotal role in triggering a variety of signalling cascades. Here, we cloned and identified the TRAF6 gene from the King pigeon. The open reading frame sequence of pigeon TRAF6 (piTRAF6) is 1638 bp long and encodes a 545 aa protein, including a low-complexity domain, RING finger, Zinc finger, coiled coil domain, and meprin and TRAF homology domain. The aa sequence of piTRAF6 shared a strong identity with that of other birds. PiTRAF6 transcripts were broadly expressed in all the tested tissues; piTRAF6 levels were the highest and lowest in the heart and stomach, respectively. Overexpression of piTRAF6 activated NF-κB in a dose-dependent manner and induced IFN-β expression. Upon piTRAF6 knockdown by small interfering RNAs, NF-κB activation was markedly inhibited in HEK293T cells. The expression of piTRAF6, as well as pro-inflammatory cytokines and antiviral molecules, were obviously increased after TLR ligand stimulation and Newcastle disease virus or Salmonella Pullorum inoculation. These results suggest that piTRAF6 may play a key immunoregulatory role in the innate immune response against viral and bacterial infections.
Collapse
Affiliation(s)
- Yaxin Guo
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, PR China.,Jiangsu Key Laboratory of Zoonosis, Yangzhou University, PR China.,Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou University, PR China.,Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou University, PR China
| | - Ying Xu
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, PR China.,Jiangsu Key Laboratory of Zoonosis, Yangzhou University, PR China.,Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou University, PR China.,Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou University, PR China
| | - Dan Xiong
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, PR China.,Jiangsu Key Laboratory of Zoonosis, Yangzhou University, PR China.,Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou University, PR China.,Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou University, PR China
| | - Yingying Zhou
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, PR China.,Jiangsu Key Laboratory of Zoonosis, Yangzhou University, PR China.,Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou University, PR China.,Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou University, PR China
| | - Xilong Kang
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, PR China.,Jiangsu Key Laboratory of Zoonosis, Yangzhou University, PR China.,Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou University, PR China.,Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou University, PR China
| | - Chuang Meng
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, PR China.,Jiangsu Key Laboratory of Zoonosis, Yangzhou University, PR China.,Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou University, PR China.,Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou University, PR China
| | - Dan Gu
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, PR China.,Jiangsu Key Laboratory of Zoonosis, Yangzhou University, PR China.,Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou University, PR China.,Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou University, PR China
| | - Xinan Jiao
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, PR China.,Jiangsu Key Laboratory of Zoonosis, Yangzhou University, PR China.,Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou University, PR China.,Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou University, PR China
| | - Zhiming Pan
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, PR China.,Jiangsu Key Laboratory of Zoonosis, Yangzhou University, PR China.,Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou University, PR China.,Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou University, PR China
| |
Collapse
|
8
|
Guo Y, Xu Y, Kang X, Meng C, Gu D, Zhou Y, Xiong D, Geng S, Jiao X, Pan Z. Molecular cloning and functional analysis of TRAF6 from Yangzhou great white goose Anser anser. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2019; 101:103435. [PMID: 31288047 DOI: 10.1016/j.dci.2019.103435] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 07/03/2019] [Accepted: 07/03/2019] [Indexed: 06/09/2023]
Abstract
TNF receptor-associated factor 6 (TRAF6) is an adaptor protein and an E3 ubiquitin ligase mediating multiple cell signaling pathway activation in a context-dependent manner. TRAF6 plays critical roles in innate immune response and regulates function of antigen-presenting cells. Here, we cloned the goose TRAF6 (goTRAF6) gene from a healthy Yangzhou great white goose (Anser anser), which had a typical TRAF structure and shared a high-sequence identity with TRAF6 of other birds. Quantitative real-time PCR revealed that goTRAF6 mRNA was broadly expressed in all the studied tissues, with highest expression in the heart and pectoral muscle. Overexpression of goTRAF6 caused NF-κB activation in a dose-dependent manner and substantially upregulated IFN-β expression in HEK293T cells. Following Toll-like receptor (TLR) ligand stimulation of goose peripheral blood mononuclear cells, goTRAF6 and downstream inflammatory cytokine mRNA levels considerably up-regulated, especially at early stages. Salmonella Enteritidis challenge caused overexpression of goTRAF6 and cytokine mRNA in all the examined organs. These findings demonstrated that goTRAF6 played a substantial role in TLR-TRAF6 signaling cascade, and further contributed to the antibacterial-responses in host.
Collapse
Affiliation(s)
- Yaxin Guo
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Jiangsu, 225009, China; Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, Jiangsu, 225009, China; Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou University, Yangzhou, Jiangsu, China; Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou University, Jiangsu, China
| | - Ying Xu
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Jiangsu, 225009, China; Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, Jiangsu, 225009, China; Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou University, Yangzhou, Jiangsu, China; Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou University, Jiangsu, China
| | - Xilong Kang
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Jiangsu, 225009, China; Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, Jiangsu, 225009, China; Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou University, Yangzhou, Jiangsu, China; Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou University, Jiangsu, China
| | - Chuang Meng
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Jiangsu, 225009, China; Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, Jiangsu, 225009, China; Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou University, Yangzhou, Jiangsu, China; Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou University, Jiangsu, China
| | - Dan Gu
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Jiangsu, 225009, China; Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, Jiangsu, 225009, China; Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou University, Yangzhou, Jiangsu, China; Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou University, Jiangsu, China
| | - Yingying Zhou
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Jiangsu, 225009, China; Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, Jiangsu, 225009, China; Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou University, Yangzhou, Jiangsu, China; Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou University, Jiangsu, China
| | - Dan Xiong
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Jiangsu, 225009, China; Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, Jiangsu, 225009, China; Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou University, Yangzhou, Jiangsu, China; Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou University, Jiangsu, China
| | - Shizhong Geng
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Jiangsu, 225009, China; Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, Jiangsu, 225009, China; Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou University, Yangzhou, Jiangsu, China; Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou University, Jiangsu, China
| | - Xinan Jiao
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Jiangsu, 225009, China; Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, Jiangsu, 225009, China; Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou University, Yangzhou, Jiangsu, China; Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou University, Jiangsu, China.
| | - Zhiming Pan
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Jiangsu, 225009, China; Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, Jiangsu, 225009, China; Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou University, Yangzhou, Jiangsu, China; Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou University, Jiangsu, China.
| |
Collapse
|
9
|
Nelson RH, Nelson DE. Signal Distortion: How Intracellular Pathogens Alter Host Cell Fate by Modulating NF-κB Dynamics. Front Immunol 2018; 9:2962. [PMID: 30619320 PMCID: PMC6302744 DOI: 10.3389/fimmu.2018.02962] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2018] [Accepted: 12/03/2018] [Indexed: 01/17/2023] Open
Abstract
By uncovering complex dynamics in the expression or localization of transcriptional regulators in single cells that were otherwise hidden at the population level, live cell imaging has transformed our understanding of how cells sense and orchestrate appropriate responses to changes in their internal state or extracellular environment. This has proved particularly true for the nuclear factor-kappaB (NF-κB) family of transcription factors, key regulators of the inflammatory response and innate immune function, which are capable of encoding information about the mode and intensity of stimuli in the dynamics of NF-κB nuclear accumulation and loss. While live cell imaging continues to serve as a useful tool in ongoing efforts to characterize the feedbacks that shape these dynamics and to connect dynamics to downstream gene expression, it is also proving invaluable for recent studies that seek to determine how intracellular pathogens subvert NF-κB signaling to survive and replicate within host cells by providing quantitative information about the pathogen and changes in NF-κB activity during different stages of an infection. Here, we provide a brief overview of NF-κB signaling in innate immune cells and review recent literature that uses live imaging to investigate the mechanisms by which bacterial and yeast pathogens modulate NF-κB in a variety of different host cell types to evade destruction or maintain the viability of an intracellular growth niche.
Collapse
Affiliation(s)
- Rachel H Nelson
- Cellular Generation and Phenotyping Core Facility, Wellcome Sanger Institute, Cambridge, United Kingdom
| | - David E Nelson
- Department of Biology, Middle Tennessee State University, Murfreesboro, TN, United States
| |
Collapse
|
10
|
Dzitko K, Grzybowski MM, Pawełczyk J, Dziadek B, Gatkowska J, Stączek P, Długońska H. Phytoecdysteroids as modulators of the Toxoplasma gondii growth rate in human and mouse cells. Parasit Vectors 2015; 8:422. [PMID: 26272689 PMCID: PMC4536731 DOI: 10.1186/s13071-015-1019-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2014] [Accepted: 07/27/2015] [Indexed: 02/01/2023] Open
Abstract
Background Searching for new effective drugs against human and animal toxoplasmosis we decided to test the anti-Toxoplasma potential of phytoecdysteroids (α-ecdysone and 20-hydroxyecdysone) characterized by the pleiotropic activity on mammalian organisms including the enhancement of host’s anti-parasitic defence. This objective was accomplished by the in vitro evaluation of T. gondii growth in phytoecdysteroid-treated immunocompetent cells of selected hosts: humans and two strains of inbred mice with genetically determined different susceptibility to toxoplasmosis. Methods Peripheral mononuclear blood cells were isolated from Toxoplasma-positive and Toxoplasma-negative women (N = 43) and men (N = 21). Non-infected mice (C57BL/6, N = 10 and BALB/c, N = 14) and mice (BALB/c, N = 10) challenged intraperitoneally with 5 tissue cysts of the T. gondii DX strain were also used in this study as a source of splenocytes. The effects of phytoecdysteroids on the viability of human PBMC and mouse splenocytes were evaluated using the MTT assay. The influence of phytoecdysteroids on PBMCs, splenocytes and T. gondii proliferation was measured using radioactivity tests (the level of 3[H] uracil incorporation by toxoplasms or 3[H] thymidine by PBMCs and splenocytes), which was confirmed by quantitative Real-Time PCR. Statistical analysis was performed using SigmaStat 3.5 (Systat Software GmbH). The best-fit IC50 curves were plotted using GraphPad Prism 6.0 (GraphPad Software, Inc.). Results Our results showed that phytoecdysteroids promote the multiplication of Toxoplasma in cultures of human or murine immune cells, in contrast to another apicomplexan parasite, Babesia gibsoni. Additionally, the tested phytoecdysteroids did not stimulate the in vitro secretion of the essential protective cytokines (IFN-γ, IL-2 and IL-10), neither by human nor by murine immune cells involved in an effective intracellular killing of the parasite. Conclusions Judging by the effect of phytoecdysteroids on the T. gondii proliferation, demonstrated for the first time in this study, it seems that these compounds should not be taken into consideration as potential medications to treat toxoplasmosis. Phytoecdysteroids included in the food are most likely not harmful for human or animal health but certain nutrients containing ecdysteroids at high concentrations could promote T. gondii proliferation in chronically infected and immunocompromised individuals. In order to assess the real impact of ecdysteroids on the course of natural T. gondii invasion, in vivo research should be undertaken because it cannot be ruled out that the in vivo effect will be different than the in vitro one. However, taking into account the possible stimulating effect of ecdysteroids on some opportunistic parasites (such as Toxoplasma or Strongyloides) further studies are necessary and should focus on the mechanisms of their action, which directly or indirectly enhance the parasite growth. Since ecdysteroids are considered as potential drugs, it is essential to determine their effect on various parasitic pathogens, which may infect the host at the same time, especially in immunocompromised individuals.
Collapse
Affiliation(s)
- Katarzyna Dzitko
- Department of Immunoparasitology, Faculty of Biology and Environmental Protection, University of Łódź, Banacha 12/16, 90-237, Łódź, Poland.
| | - Marcin Mikołaj Grzybowski
- Department of Immunoparasitology, Faculty of Biology and Environmental Protection, University of Łódź, Banacha 12/16, 90-237, Łódź, Poland.
| | - Jakub Pawełczyk
- Institute of Medical Biology of the Polish Academy of Sciences, Lodowa 106, 93-232, Łódź, Poland.
| | - Bożena Dziadek
- Department of Immunoparasitology, Faculty of Biology and Environmental Protection, University of Łódź, Banacha 12/16, 90-237, Łódź, Poland.
| | - Justyna Gatkowska
- Department of Immunoparasitology, Faculty of Biology and Environmental Protection, University of Łódź, Banacha 12/16, 90-237, Łódź, Poland.
| | - Paweł Stączek
- Department of Microbial Genetics, Faculty of Biology and Environmental Protection, University of Łódź, Banacha 12/16, 90-237, Łódź, Poland.
| | - Henryka Długońska
- Department of Immunoparasitology, Faculty of Biology and Environmental Protection, University of Łódź, Banacha 12/16, 90-237, Łódź, Poland.
| |
Collapse
|
11
|
Li YW, Li X, Wang Z, Mo ZQ, Dan XM, Luo XC, Li AX. Orange-spotted grouper Epinephelus coioides Tak1: molecular identification, expression analysis and functional study. JOURNAL OF FISH BIOLOGY 2015; 86:417-430. [PMID: 25677752 DOI: 10.1111/jfb.12550] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2014] [Accepted: 09/18/2014] [Indexed: 06/04/2023]
Abstract
In this study, the complementary (c)DNA sequence encoding orange-spotted grouper Epinephelus coioides Tak1 (ectak1) was cloned, which has an open reading frame of 1728 bp that encodes 575 amino acids (aa). Sequence analysis indicated that Ectak1 contains two characteristic conserved domains, i.e. an N-terminal serine-threonine protein kinase catalytic domain (27-275 aa) and a C-terminal coiled-coil region (499-562 aa). Ectak1 shares high sequence identity with Tak1 from other fish species, especially those of Nile tilapia Oreochromis niloticus (96%) and zebra mbuna Maylandia zebra (96%). ectak1 transcripts were expressed broadly in all of the tissues tested, but ectak1 expression was reduced mainly in the local infection sites (skin and gill) after infection with Cryptocaryon irritans. Intracellular localization analysis showed that Ectak1 was distributed mainly in the cytoplasm. A luciferase reporter assay showed that Ectak1 significantly impaired the NF-κB activity induced by E. coioides Myd88 and Traf6. Overall, these results suggest that Ectak1 functions to reduce the activity of NF-κB induced by toll-like receptor (TLR) signal molecules in HEK-293T cells, and it might have an important role in host defences against parasitic infections.
Collapse
Affiliation(s)
- Y W Li
- Key Laboratory of Aquatic Product Safety (Sun Yat-Sen University), Ministry of Education/State Key Laboratory of Biocontrol, The School of Life Sciences, Sun Yat-sen University, 135 Xingang West Street, Haizhu District, Guangzhou 510275, Guangdong Province, The People's Republic of China
| | - X Li
- Key Laboratory of Aquatic Product Safety (Sun Yat-Sen University), Ministry of Education/State Key Laboratory of Biocontrol, The School of Life Sciences, Sun Yat-sen University, 135 Xingang West Street, Haizhu District, Guangzhou 510275, Guangdong Province, The People's Republic of China
| | - Z Wang
- Key Laboratory of Aquatic Product Safety (Sun Yat-Sen University), Ministry of Education/State Key Laboratory of Biocontrol, The School of Life Sciences, Sun Yat-sen University, 135 Xingang West Street, Haizhu District, Guangzhou 510275, Guangdong Province, The People's Republic of China
| | - Z Q Mo
- College of Animal Science, South China Agricultural University, Guangzhou 510642, Guangdong Province, PR China
| | - X M Dan
- College of Animal Science, South China Agricultural University, Guangzhou 510642, Guangdong Province, PR China
| | - X C Luo
- School of Bioscience and Biotechnology, South China University of Technology, Guangzhou 510006, Guangdong Province, PR China
| | - A X Li
- Key Laboratory of Aquatic Product Safety (Sun Yat-Sen University), Ministry of Education/State Key Laboratory of Biocontrol, The School of Life Sciences, Sun Yat-sen University, 135 Xingang West Street, Haizhu District, Guangzhou 510275, Guangdong Province, The People's Republic of China
| |
Collapse
|
12
|
Zhao F, Li YW, Pan HJ, Shi CB, Luo XC, Li AX, Wu SQ. TAK1-binding proteins (TAB1 and TAB2) in grass carp (Ctenopharyngodon idella): identification, characterization, and expression analysis after infection with Ichthyophthirius multifiliis. FISH & SHELLFISH IMMUNOLOGY 2014; 38:389-399. [PMID: 24747054 DOI: 10.1016/j.fsi.2014.04.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2014] [Revised: 04/04/2014] [Accepted: 04/08/2014] [Indexed: 06/03/2023]
Abstract
Transforming growth factor-β activated kinase-1 (TAK1) is a key regulatory molecule in toll-like receptor (TLR), interleukin-1 (IL-1), and tumor necrosis factor (TNF) signaling pathways. The activation of TAK1 is specifically regulated by two TAK1-binding proteins, TAB1 and TAB2. However, the roles of TAB1 and TAB2 in fish have not been reported to date. In the present study, TAB1 (CiTAB1) and TAB2 (CiTAB2) in grass carp (Ctenopharyngodon idella) were identified and characterized, and their expression profiles were analyzed after fish were infected with the pathogenic ciliate Ichthyophthirius multifiliis. The full-length CiTAB1 cDNA is 1949 bp long with an open reading frame (ORF) of 1497 bp that encodes a putative protein of 498 amino acids containing a typical PP2Cc domain. The full-length CiTAB2 cDNA is 2967 bp long and contains an ORF of 2178 bp encoding a putative protein of 725 amino acids. Protein structure analysis revealed that CiTAB2 consists of three main structural domains: an N-terminal CUE domain, a coiled-coil domain, and a C-terminal ZnF domain. Multiple sequence alignment showed that CiTAB1 and CiTAB2 share high sequence identity with other known TAB1 and TAB2 proteins, and several conserved phosphorylation sites and an O-GlcNAc site were deduced in CiTAB1. Phylogenetic tree analysis demonstrated that CiTAB1 and CiTAB2 have the closest evolutionary relationship with TAB1 and TAB2 of Danio rerio, respectively. CiTAB1 and CiTAB2 were both widely expressed in all examined tissues with the highest levels in the heart and liver, respectively. After infection with I. multifiliis, the expressions of CiTAB1 and CiTAB2 were both significantly up-regulated in all tested tissues at most time points, which indicates that these proteins may be involved in the host immune response against I. multifiliis infection.
Collapse
Affiliation(s)
- Fei Zhao
- Key Laboratory of Fishery Drug Development, Ministry of Agriculture, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou 510380, PR China; State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, PR China
| | - Yan-Wei Li
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, PR China.
| | - Hou-Jun Pan
- Key Laboratory of Fishery Drug Development, Ministry of Agriculture, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou 510380, PR China
| | - Cun-Bin Shi
- Key Laboratory of Fishery Drug Development, Ministry of Agriculture, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou 510380, PR China
| | - Xiao-Chun Luo
- School of Bioscience and Bioengineering, South China University of Technology, Guangzhou 510006, PR China
| | - An-Xing Li
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, PR China
| | - Shu-Qin Wu
- Key Laboratory of Fishery Drug Development, Ministry of Agriculture, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou 510380, PR China.
| |
Collapse
|
13
|
Du J, An R, Chen L, Shen Y, Chen Y, Cheng L, Jiang Z, Zhang A, Yu L, Chu D, Shen Y, Luo Q, Chen H, Wan L, Li M, Xu X, Shen J. WITHDRAWN: Toxoplasma gondii virulence factor ROP18 inhibits the host NF-κB pathway by promoting p65 degradation. J Biol Chem 2014; 289:12578-92. [PMID: 24648522 PMCID: PMC4007449 DOI: 10.1074/jbc.m113.544718] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Revised: 03/10/2014] [Indexed: 01/09/2023] Open
Abstract
The obligate intracellular parasite Toxoplasma gondii secretes effector molecules into the host cell to modulate host immunity. Previous studies have shown that T. gondii could interfere with host NF-κB signaling to promote their survival, but the effectors of type I strains remain unclear. The polymorphic rhoptry protein ROP18 is a key serine/threonine kinase that phosphorylates host proteins to modulate acute virulence. Our data demonstrated that the N-terminal portion of ROP18 is associated with the dimerization domain of p65. ROP18 phosphorylates p65 at Ser-468 and targets this protein to the ubiquitin-dependent degradation pathway. The kinase activity of ROP18 is required for p65 degradation and suppresses NF-κB activation. Consistently, compared with wild-type ROP18 strain, ROP18 kinase-deficient type I parasites displayed a severe inability to inhibit NF-κB, culminating in the enhanced production of IL-6, IL-12, and TNF-α in infected macrophages. In addition, studies have shown that transgenic parasites carrying kinase-deficient ROP18 induce M1-biased activation. These results demonstrate for the first time that the virulence factor ROP18 in T. gondii type I strains is responsible for inhibiting the host NF-κB pathway and for suppressing proinflammatory cytokine expression, thus providing a survival advantage to the infectious agent.
Collapse
Affiliation(s)
- Jian Du
- From the Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Ran An
- From the Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Lijian Chen
- Department of Anesthesiology, the First Affiliated Hospital of Anhui Medical University, Hefei 230032, China
| | - Yuxian Shen
- Institute of Biopharmaceuticals, Anhui Medical University, Hefei 230032, China
| | - Ying Chen
- Institute of Biopharmaceuticals, Anhui Medical University, Hefei 230032, China
| | - Li Cheng
- From the Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Zhongru Jiang
- From the Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Aimei Zhang
- Central Laboratory of Affiliated Provincial Hospital of Anhui Medical University, Hefei 230001, China, and
| | - Li Yu
- Anhui Provincial Laboratory of Microbiology and Parasitology, Anhui Medical University, Hefei 230032, China
| | - Deyong Chu
- Anhui Provincial Laboratory of Microbiology and Parasitology, Anhui Medical University, Hefei 230032, China
| | - Yujun Shen
- Institute of Biopharmaceuticals, Anhui Medical University, Hefei 230032, China
| | - Qingli Luo
- Anhui Provincial Laboratory of Microbiology and Parasitology, Anhui Medical University, Hefei 230032, China
| | - He Chen
- Anhui Provincial Laboratory of Microbiology and Parasitology, Anhui Medical University, Hefei 230032, China
| | - Lijuan Wan
- From the Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Min Li
- Anhui Provincial Laboratory of Microbiology and Parasitology, Anhui Medical University, Hefei 230032, China
| | - Xiucai Xu
- Central Laboratory of Affiliated Provincial Hospital of Anhui Medical University, Hefei 230001, China, and
| | - Jilong Shen
- Anhui Provincial Laboratory of Microbiology and Parasitology, Anhui Medical University, Hefei 230032, China
| |
Collapse
|
14
|
Senderskiy IV, Timofeev SA, Seliverstova EV, Pavlova OA, Dolgikh VV. Secretion of Antonospora (Paranosema) locustae proteins into infected cells suggests an active role of microsporidia in the control of host programs and metabolic processes. PLoS One 2014; 9:e93585. [PMID: 24705470 PMCID: PMC3976299 DOI: 10.1371/journal.pone.0093585] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Accepted: 03/05/2014] [Indexed: 01/02/2023] Open
Abstract
Molecular tools of the intracellular protozoan pathogens Apicomplexa and Kinetoplastida for manipulation of host cell machinery have been the focus of investigation for approximately two decades. Microsporidia, fungi-related microorganisms forming another large group of obligate intracellular parasites, are characterized by development in direct contact with host cytoplasm (the majority of species), strong minimization of cell machinery, and acquisition of unique transporters to exploit host metabolic system. All the aforementioned features are suggestive of the ability of microsporidia to modify host metabolic and regulatory pathways. Seven proteins of the microsporidium Antonospora (Paranosema) locustae with predicted signal peptides but without transmembrane domains were overexpressed in Escherichia coli. Western-blot analysis with antibodies against recombinant products showed secretion of parasite proteins from different functional categories into the infected host cell. Secretion of parasite hexokinase and α/β-hydrolase was confirmed by immunofluorescence microscopy. In addition, this method showed specific accumulation of A. locustae hexokinase in host nuclei. Expression of hexokinase, trehalase, and two leucine-rich repeat proteins without any exogenous signal peptide led to their secretion in the yeast Pichia pastoris. In contrast, α/β-hydrolase was not found in the culture medium, though a significant amount of this enzyme accumulated in the yeast membrane fraction. These results suggest that microsporidia possess a broad set of enzymes and regulatory proteins secreted into infected cells to control host metabolic processes and molecular programs.
Collapse
Affiliation(s)
- Igor V. Senderskiy
- Laboratory of Microbiological Control, All-Russian Institute for Plant Protection, St. Petersburg, Pushkin, Russia
| | - Sergey A. Timofeev
- Laboratory of Microbiological Control, All-Russian Institute for Plant Protection, St. Petersburg, Pushkin, Russia
| | - Elena V. Seliverstova
- Laboratory of Renal Physiology, Sechenov Institute of Evolutionary Physiology and Biochemistry, St. Petersburg, Russia
| | - Olga A. Pavlova
- Laboratory of Microbiological Control, All-Russian Institute for Plant Protection, St. Petersburg, Pushkin, Russia
| | - Viacheslav V. Dolgikh
- Laboratory of Microbiological Control, All-Russian Institute for Plant Protection, St. Petersburg, Pushkin, Russia
- * E-mail:
| |
Collapse
|
15
|
Silmon de Monerri NC, Kim K. Pathogens hijack the epigenome: a new twist on host-pathogen interactions. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 184:897-911. [PMID: 24525150 DOI: 10.1016/j.ajpath.2013.12.022] [Citation(s) in RCA: 100] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Revised: 12/01/2013] [Accepted: 12/05/2013] [Indexed: 02/07/2023]
Abstract
Pathogens have evolved strategies to promote their survival by dramatically modifying the transcriptional profile and protein content of the host cells they infect. Modifications of the host transcriptome and proteome are mediated by pathogen-encoded effector molecules that modulate host cells through a variety of different mechanisms. Recent studies highlight the importance of the host chromatin and other epigenetic regulators as targets of pathogens. Host gene regulatory mechanisms may be targeted through cytoplasmic signaling, directly by pathogen effector proteins, and possibly by pathogen RNA. Although many of these changes are short-lived and persist only during the course of infection, several studies indicate that pathogens are able to induce long-term, heritable changes that are essential to pathogenesis of infectious diseases and persistence of pathogens within their hosts. In this review, we discuss how pathogens modulate the epigenome of host cells, a new and flourishing avenue of host-pathogen interaction studies.
Collapse
Affiliation(s)
- Natalie C Silmon de Monerri
- Departments of Medicine, Pathology, and Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York
| | - Kami Kim
- Departments of Medicine, Pathology, and Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York.
| |
Collapse
|
16
|
Yang N, Farrell A, Niedelman W, Melo M, Lu D, Julien L, Marth GT, Gubbels MJ, Saeij JPJ. Genetic basis for phenotypic differences between different Toxoplasma gondii type I strains. BMC Genomics 2013; 14:467. [PMID: 23837824 PMCID: PMC3710486 DOI: 10.1186/1471-2164-14-467] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Accepted: 07/03/2013] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Toxoplasma gondii has a largely clonal population in North America and Europe, with types I, II and III clonal lineages accounting for the majority of strains isolated from patients. RH, a particular type I strain, is most frequently used to characterize Toxoplasma biology. However, compared to other type I strains, RH has unique characteristics such as faster growth, increased extracellular survival rate and inability to form orally infectious cysts. Thus, to identify candidate genes that could account for these parasite phenotypic differences, we determined genetic differences and differential parasite gene expression between RH and another type I strain, GT1. Moreover, as differences in host cell modulation could affect Toxoplasma replication in the host, we determined differentially modulated host processes among the type I strains through host transcriptional profiling. RESULTS Through whole genome sequencing, we identified 1,394 single nucleotide polymorphisms (SNPs) and insertions/deletions (indels) between RH and GT1. These SNPs/indels together with parasite gene expression differences between RH and GT1 were used to identify candidate genes that could account for type I phenotypic differences. A polymorphism in dense granule protein, GRA2, determined RH and GT1 differences in the evasion of the interferon gamma response. In addition, host transcriptional profiling identified that genes regulated by NF-ĸB, such as interleukin (IL)-12p40, were differentially modulated by the different type I strains. We subsequently showed that this difference in NF-ĸB activation was due to polymorphisms in GRA15. Furthermore, we observed that RH, but not other type I strains, recruited phosphorylated IĸBα (a component of the NF-ĸB complex) to the parasitophorous vacuole membrane and this recruitment of p- IĸBα was partially dependent on GRA2. CONCLUSIONS We identified candidate parasite genes that could be responsible for phenotypic variation among the type I strains through comparative genomics and transcriptomics. We also identified differentially modulated host pathways among the type I strains, and these can serve as a guideline for future studies in examining the phenotypic differences among type I strains.
Collapse
Affiliation(s)
- Ninghan Yang
- Biology Department, Massachusetts Institute of Technology, 77 Massachusetts Ave, building 68-270, Cambridge, MA 02139, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Zhao F, Li YW, Pan HJ, Wu SQ, Shi CB, Luo XC, Li AX. Grass carp (Ctenopharyngodon idella) TRAF6 and TAK1: molecular cloning and expression analysis after Ichthyophthirius multifiliis infection. FISH & SHELLFISH IMMUNOLOGY 2013; 34:1514-1523. [PMID: 23542602 DOI: 10.1016/j.fsi.2013.03.003] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2013] [Revised: 03/01/2013] [Accepted: 03/11/2013] [Indexed: 06/02/2023]
Abstract
Ichthyophthirius multifiliis, a pathogenic ciliate parasite, infects almost all freshwater fish species and causes significant economic losses. Tumor necrosis factor receptor-associated factor 6 (TRAF6) and transforming growth factor-β-activated kinase 1 (TAK1) are two important signaling molecules involved in toll-like receptor (TLR) signal transduction. To date, the roles of TRAF6 and TAK1 in host defense against fish parasites are still poorly understood. In the present study, TRAF6 (CiTRAF6) and TAK1 (CiTAK1) were identified from grass carp (Ctenopharyngodon idella). The full-length cDNA sequence of CiTRAF6 (2250 bp) includes an open reading frame (ORF) of 1629 bp, which shows a high similarity to that of Cyprinus carpio TRAF6 and encodes a putative protein of 542 amino acids containing one RING domain, two zinc fingers, one coiled-coil region, and one MATH domain. The full-length CiTAK1 cDNA sequence is 2768 bp and includes an ORF of 1626 bp that encodes a putative protein of 541 amino acids containing a conserved serine/threonine protein kinase catalytic domain and a coiled-coil region. Phylogenetic analysis showed that CiTRAF6 and CiTAK1 were clustered with TRAF6 and TAK1 of other teleosts, respectively. CiTRAF6 and CiTAK1 were both constitutively expressed in all examined tissues but with varied expression levels. The highest expressions of CiTRAF6 and CiTAK1 were in the head kidney and spleen, respectively. The expression profiles of CiTRAF6 and CiTAK1 were detected in grass carp after I. multifiliis infection. Expressions of both genes were significantly up-regulated in the skin, gill, head kidney, and spleen at most time points after infection, indicating that CiTRAF6 and CiTAK1 may play essential roles in grass carp defense against I. multifiliis.
Collapse
Affiliation(s)
- Fei Zhao
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Haizhu District, Guangzhou, Guangdong Province 510275, PR China
| | | | | | | | | | | | | |
Collapse
|
18
|
A genome-wide RNA interference screen identifies caspase 4 as a factor required for tumor necrosis factor alpha signaling. Mol Cell Biol 2012; 32:3372-81. [PMID: 22733992 DOI: 10.1128/mcb.06739-11] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Tumor necrosis factor alpha (TNF-α) is a potent inflammatory cytokine secreted upon cellular stress as well as immunological stimuli and is implicated in the pathology of inflammatory diseases and cancer. The therapeutic potential of modifying TNF-α pathway activity has been realized in several diseases, and antagonists of TNF-α have reached clinical applications. While much progress in the understanding of signaling downstream of the TNF-α receptor complex has been made, the compendium of factors required for signal transduction is still not complete. In order to find novel regulators of proinflammatory signaling induced by TNF-α, we conducted a genome-wide small interfering RNA screen in human cells. We identified several new candidate modulators of TNF-α signaling, which were confirmed in independent experiments. Specifically, we show that caspase 4 is required for the induction of NF-κB activity, while it appears to be dispensable for the activation of the Jun N-terminal protein kinase signaling branch. Taken together, our experiments identify caspase 4 as a novel regulator of TNF-α-induced NF-κB signaling that is required for the activation of IκB kinase. We further provide the genome-wide RNA interference data set as a compendium in a format compliant with minimum information about an interfering RNA experiment (MAIRE).
Collapse
|
19
|
Rodrigues V, Cordeiro-da-Silva A, Laforge M, Ouaissi A, Silvestre R, Estaquier J. Modulation of mammalian apoptotic pathways by intracellular protozoan parasites. Cell Microbiol 2012; 14:325-33. [PMID: 22168464 DOI: 10.1111/j.1462-5822.2011.01737.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
During intracellular parasitic infections, pathogens and host cells take part in a complex web of events that are crucial for the outcome of the infection. Modulation of host cell apoptosis by pathogens attracted the attention of scientists during the last decade. Apoptosis is an efficient mechanism used by the host to control infection and limit pathogen multiplication and dissemination. In order to ensure completion of their complex life cycles and to guarantee transmission between different hosts, intracellular parasites have developed mechanisms to block apoptosis and sustain the viability of their host cells. Here, we review how some of the most prominent intracellular protozoan parasites modulate the main mammalian apoptotic pathways by emphasizing the advances from the last decade, which have begun to dissect this dynamic and complex interaction.
Collapse
|
20
|
Effects of Toxoplasma gondii genotype and absence of host MAL/Myd88 on the temporal regulation of gene expression in infected microglial cells. Exp Parasitol 2011; 129:409-13. [PMID: 21924265 DOI: 10.1016/j.exppara.2011.08.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2011] [Revised: 07/27/2011] [Accepted: 08/25/2011] [Indexed: 11/20/2022]
Abstract
The majority of strains of Toxoplasma gondii belong to three distinct clonal lines known as types I, II, and III. The outcome of the immune response to infection is influenced by the parasite strain type. The goal of this study was to examine differences in the kinetics of gene expression in microglial cells infected with types I, II, or III of T. gondii. In addition, a requirement for the integrity of host Toll-like receptor (TLR) signaling in parasite-mediated changes in gene expression was evaluated. Wild type murine microglial cells infected with T. gondii displayed different kinetic patterns of pro-inflammatory cytokine expression that were dependent on the parasite strain type. In general, types II and III elicited higher sustained responses compared to type I which induced fluctuating patterns of cytokine gene expression. Contrary to this, differences in the induction of anti-apoptotic gene expression were minimal among the different type strains throughout infection. Experiments with cells lacking the TLR adaptor molecules MAL and Myd88 showed a dependency on these factors for the pro-inflammatory response but not the anti-apoptotic response. The results show that the outcome of gene expression in T. gondii-infected microglial cells is dependent on the parasite strain type in a time-dependent manner and is selective to particular subsets of genes. The induction of an anti-apoptotic response by T. gondii infection in the absence of TLR signaling reflects a complex level of modulation of host functions by the parasite.
Collapse
|
21
|
Abstract
Intracellular parasitism has arisen only a few times during the long ancestry of protozoan parasites including in diverse groups such as microsporidians, kinetoplastids, and apicomplexans. Strategies used to gain entry differ widely from injection (e.g. microsporidians), active penetration of the host cell (e.g. Toxoplasma), recruitment of lysosomes to a plasma membrane wound (e.g. Trypanosoma cruzi), to host cell-mediated phagocytosis (e.g. Leishmania). The resulting range of intracellular niches is equally diverse ranging from cytosolic (e.g. T. cruzi) to residing within a non-fusigenic vacuole (e.g. Toxoplasma, Encephalitozoon) or a modified phagolysosome (e.g. Leishmania). These lifestyle choices influence access to nutrients, interaction with host cell signaling pathways, and detection by pathogen recognition systems. As such, intracellular life requires a repertoire of adaptations to assure entry-exit from the cell, as well as to thwart innate immune mechanisms and prevent clearance. Elucidating these pathways at the cellular and molecular level may identify key steps that can be targeted to reduce parasite survival or augment immunologic responses and thereby prevent disease.
Collapse
Affiliation(s)
- L David Sibley
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63100, USA.
| |
Collapse
|
22
|
Marugán-Hernández V, Alvarez-García G, Tomley F, Hemphill A, Regidor-Cerrillo J, Ortega-Mora LM. Identification of novel rhoptry proteins in Neospora caninum by LC/MS-MS analysis of subcellular fractions. J Proteomics 2011; 74:629-42. [PMID: 21315855 DOI: 10.1016/j.jprot.2011.02.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2010] [Revised: 02/01/2011] [Accepted: 02/02/2011] [Indexed: 11/30/2022]
Abstract
Apicomplexan parasites possess an apical complex that is composed of two secretory organelles recognized as micronemes and rhoptries. Rhoptry contents are secreted into the parasitophorous vacuole during the host cell invasion process. Several rhoptry proteins have been identified in Toxoplasma gondii and seem to be involved in host-pathogen interactions and some of them are considered to be important virulence factors. Only one rhoptry protein, NcROP2, has been identified and extensively characterized in the closely related parasite Neospora caninum, and this has showed immunoprotective properties. Thus, with the aim of increasing knowledge of the rhoptry protein repertoire in N. caninum, a subcellular fractionation of tachyzoites was performed to obtain fractions enriched for this secretory organelle. 2-D SDS-PAGE followed by MS and LC/MS-MS were applied for fraction analysis and 8 potential novel rhoptry components (NcROP1, 5, 8, 30 and NcRON2, 3, 4, 8) and several kinases, proteases and phosphatases proteins were identified with a high homology to those previously found in T. gondii. Their existence in N. caninum tachyzoites suggests their involvement in similar events or pathways that occur in T. gondii. These novel proteins may be considered as targets that could be useful in the future development of immunoprophylactic measures.
Collapse
Affiliation(s)
- V Marugán-Hernández
- SALUVET, Animal Health Department, Faculty of Veterinary Sciences, Complutense University of Madrid, Ciudad Universitaria s/n, Madrid, Spain
| | | | | | | | | | | |
Collapse
|
23
|
Abstract
The nuclear factor-κB (NF-κB) family of transcription factors plays a central part in the host response to infection by microbial pathogens, by orchestrating the innate and acquired host immune responses. The NF-κB proteins are activated by diverse signalling pathways that originate from many different cellular receptors and sensors. Many successful pathogens have acquired sophisticated mechanisms to regulate the NF-κB signalling pathways by deploying subversive proteins or hijacking the host signalling molecules. Here, we describe the mechanisms by which viruses and bacteria micromanage the host NF-κB signalling circuitry to favour the continued survival of the pathogen.
Collapse
Affiliation(s)
- Masmudur M Rahman
- Department of Molecular Genetics and Microbiology, College of Medicine, University of Florida, 1600 SW Archer Road, PO Box 100266, Gainesville, Florida, USA
| | | |
Collapse
|
24
|
Carmen JC, Sinai AP. The Differential Effect of Toxoplasma Gondii Infection on the Stability of BCL2-Family Members Involves Multiple Activities. Front Microbiol 2011; 2:1. [PMID: 21716958 PMCID: PMC3109420 DOI: 10.3389/fmicb.2011.00001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2010] [Accepted: 01/02/2011] [Indexed: 11/13/2022] Open
Abstract
The regulation of mitochondrial permeability, a key event in the initiation of apoptosis is governed by the opposing actions of the pro- and anti-apoptotic members of the BCL2-family of proteins. The BCL2-family can be classified further based on the number of BCL-homology (BH) domains they encode. Pathogen mediated modulation of BCL2-family members play a significant role in their ability to affect the apoptotic pathways in the infected host cell. The protozoan parasite Toxoplasma gondii establishes a profound blockade of apoptosis noted by a requirement for host NFκB activity and correlating with the selective degradation of pro-apoptotic BCL2-family members. In this study, we explore the potential activities associated with the inherent stability of the anti-apoptotic BCL2 as well as the selective degradation of the pro-apoptotic proteins BAX, BAD, and BID. We find that multiple activities govern the relative stability of BCL2-family members suggesting a complex and balanced network of stability-enhancing and–destabilizing activities are perturbed by parasite infection. The data leave open the possibility for both parasite induced host activities as well as the direct consequence of parasite effectors in governing the relative levels of BCL2-proteins in the course of infection.
Collapse
Affiliation(s)
- John Cherrington Carmen
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky College of Medicine Lexington, KY, USA
| | | |
Collapse
|
25
|
Rosowski EE, Lu D, Julien L, Rodda L, Gaiser RA, Jensen KDC, Saeij JPJ. Strain-specific activation of the NF-kappaB pathway by GRA15, a novel Toxoplasma gondii dense granule protein. ACTA ACUST UNITED AC 2011; 208:195-212. [PMID: 21199955 PMCID: PMC3023140 DOI: 10.1084/jem.20100717] [Citation(s) in RCA: 310] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
NF-κB is an integral component of the immune response to Toxoplasma gondii. Although evidence exists that T. gondii can directly modulate the NF-κB pathway, the parasite-derived effectors involved are unknown. We determined that type II strains of T. gondii activate more NF-κB than type I or type III strains, and using forward genetics we found that this difference is a result of the polymorphic protein GRA15, a novel dense granule protein which T. gondii secretes into the host cell upon invasion. A GRA15-deficient type II strain has a severe defect in both NF-κB nuclear translocation and NF-κB-mediated transcription. Furthermore, human cells expressing type II GRA15 also activate NF-κB, demonstrating that GRA15 alone is sufficient for NF-κB activation. Along with the rhoptry protein ROP16, GRA15 is responsible for a large part of the strain differences in the induction of IL-12 secretion by infected mouse macrophages. In vivo bioluminescent imaging showed that a GRA15-deficient type II strain grows faster compared with wild-type, most likely through its reduced induction of IFN-γ. These results show for the first time that a dense granule protein can modulate host signaling pathways, and dense granule proteins can therefore join rhoptry proteins in T. gondii's host cell-modifying arsenal.
Collapse
Affiliation(s)
- Emily E Rosowski
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | | | | | | | | | | | |
Collapse
|
26
|
A history of studies that examine the interactions of Toxoplasma with its host cell: Emphasis on in vitro models. Int J Parasitol 2010; 39:903-14. [PMID: 19630139 DOI: 10.1016/j.ijpara.2009.01.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
This review is a historical look at work carried out over the past 50 years examining interactions of Toxoplasma with the host cell and attempts to focus on some of the seminal experiments in the field. This early work formed the foundation for more recent studies aimed at identifying the host and parasite factors mediating key Toxoplasma-host cell interactions. We focus especially on those studies that were performed in vitro and provide discussions of the following general areas: (i) establishment of the parasitophorous vacuole, (ii) the requirement of specific host cell molecules for parasite replication, (iii) the scenarios under which the host cell can resist parasite replication and/or persistence, (iv) host species-specific and host strain-specific responses to Toxoplasma infection, and (v) Toxoplasma-induced immune modulation.
Collapse
|
27
|
Emmott E, Rodgers MA, Macdonald A, McCrory S, Ajuh P, Hiscox JA. Quantitative proteomics using stable isotope labeling with amino acids in cell culture reveals changes in the cytoplasmic, nuclear, and nucleolar proteomes in Vero cells infected with the coronavirus infectious bronchitis virus. Mol Cell Proteomics 2010; 9:1920-36. [PMID: 20467043 PMCID: PMC2938107 DOI: 10.1074/mcp.m900345-mcp200] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2009] [Revised: 04/09/2010] [Indexed: 01/19/2023] Open
Abstract
Virus-host interactions involve complex interplay between viral and host factors, rendering them an ideal target for proteomic analysis. Here we detail a high throughput quantitative proteomics analysis of Vero cells infected with the coronavirus infectious bronchitis virus (IBV), a positive strand RNA virus that replicates in the cytoplasm. Stable isotope labeling with amino acids in cell culture (SILAC) was used in conjunction with LC-MS/MS to identify and quantify 1830 cellular and two viral proteins from IBV-infected cells. Fractionation of cells into cytoplasmic, nuclear, and nucleolar extracts was used to reduce sample complexity and provide information on the trafficking of proteins between the different compartments. Each fraction showed a proportion of proteins exhibiting >or=2-fold changes in abundance. Ingenuity Pathway Analysis revealed that proteins that changed in response to infection could be grouped into different functional categories. These included proteins regulated by NF-kappaB- and AP-1-dependent pathways and proteins involved in the cytoskeleton and molecular motors. A luciferase-based reporter gene assay was used to validate the up-regulation of AP-1- and NF-kappaB-dependent transcription in IBV-infected cells and confirmed using immunofluorescence. Immunofluorescence was used to validate changes in the subcellular localization of vimentin and myosin VI in IBV-infected cells. The proteomics analysis also confirmed the presence of the viral nucleocapsid protein as localizing in the cytoplasm, nucleus, and nucleolus and the viral membrane protein in the cytoplasmic fraction. This research is the first application of SILAC to study total host cell proteome changes in response to positive sense RNA virus infection and illustrates the versatility of this technique as applied to infectious disease research.
Collapse
Affiliation(s)
- Edward Emmott
- From the ‡Institute of Molecular and Cellular Biology, Faculty of Biological Sciences and
- the §Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds LS2 9JT, United Kingdom and
| | - Mark A. Rodgers
- From the ‡Institute of Molecular and Cellular Biology, Faculty of Biological Sciences and
| | - Andrew Macdonald
- From the ‡Institute of Molecular and Cellular Biology, Faculty of Biological Sciences and
| | - Sarah McCrory
- From the ‡Institute of Molecular and Cellular Biology, Faculty of Biological Sciences and
| | - Paul Ajuh
- **Dundee Cell Products Ltd., James Lindsay Place, Dundee Technopole, Dundee DD1 5JJ, United Kingdom
| | - Julian A. Hiscox
- From the ‡Institute of Molecular and Cellular Biology, Faculty of Biological Sciences and
- the §Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds LS2 9JT, United Kingdom and
| |
Collapse
|
28
|
Leng J, Butcher BA, Denkers EY. Dysregulation of macrophage signal transduction by Toxoplasma gondii: past progress and recent advances. Parasite Immunol 2010; 31:717-28. [PMID: 19891610 DOI: 10.1111/j.1365-3024.2009.01122.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The opportunistic protozoan parasite Toxoplasma gondii is well known as a strong inducer of cell-mediated immunity, largely as a result of proinflammatory cytokine induction during in vivo infection. Yet, during intracellular infection the parasite suppresses signal transduction pathways leading to these proinflammatory responses. The opposing responses are likely to reflect the parasite's need to stimulate immunity allowing host survival and parasite persistence, and at the same time avoiding excessive responses that could result in parasite elimination and host immunopathology. This Review summarizes past and present investigations into the effects of Toxoplasma on host cell signal transduction. These studies reveal insight into the profound suppression of proinflammatory cytokine responses that occurs when the parasite infects macrophages and other cells of innate immunity.
Collapse
Affiliation(s)
- J Leng
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853-6401, USA
| | | | | |
Collapse
|
29
|
Brown KM, Blader IJ. The role of DNA microarrays in Toxoplasma gondii research, the causative agent of ocular toxoplasmosis. J Ocul Biol Dis Infor 2009; 2:214-222. [PMID: 20157353 PMCID: PMC2816810 DOI: 10.1007/s12177-009-9040-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2009] [Accepted: 11/06/2009] [Indexed: 12/11/2022] Open
Abstract
Ocular toxoplasmosis, which is caused by the protozoan parasite Toxoplasma gondii, is the leading cause of retinochoroiditis. Toxoplasma is an obligate intracellular pathogen that replicates within a parasitophorous vacuole. Infections are initiated by digestion of parasites deposited in cat feces or in undercooked meat. Parasites then disseminate to target tissues that include the retina where they then develop into long-lived asymptomatic tissue cysts. Occasionally, cysts reactivate and growth of newly emerged parasites must be controlled by the host's immune system or disease will occur. The mechanisms by which Toxoplasma grows within its host cell, encysts, and interacts with the host's immune system are important questions. Here, we will discuss how the use of DNA microarrays in transcriptional profiling, genotyping, and epigenetic experiments has impacted our understanding of these processes. Finally, we will discuss how these advances relate to ocular toxoplasmosis and how future research on ocular toxoplasmosis can benefit from DNA microarrays.
Collapse
Affiliation(s)
- Kevin M. Brown
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73034 USA
| | - Ira J. Blader
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73034 USA
| |
Collapse
|
30
|
Albuquerque SS, Carret C, Grosso AR, Tarun AS, Peng X, Kappe SHI, Prudêncio M, Mota MM. Host cell transcriptional profiling during malaria liver stage infection reveals a coordinated and sequential set of biological events. BMC Genomics 2009; 10:270. [PMID: 19534804 PMCID: PMC2706893 DOI: 10.1186/1471-2164-10-270] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2009] [Accepted: 06/17/2009] [Indexed: 12/29/2022] Open
Abstract
Background Plasmodium sporozoites migrate to the liver where they traverse several hepatocytes before invading the one inside which they will develop and multiply into thousands of merozoites. Although this constitutes an essential step of malaria infection, the requirements of Plasmodium parasites in liver cells and how they use the host cell for their own survival and development are poorly understood. Results To gain new insights into the molecular host-parasite interactions that take place during malaria liver infection, we have used high-throughput microarray technology to determine the transcriptional profile of P. berghei-infected hepatoma cells. The data analysis shows differential expression patterns for 1064 host genes starting at 6 h and up to 24 h post infection, with the largest proportion correlating specifically with the early stages of the infection process. A considerable proportion of those genes were also found to be modulated in liver cells collected from P. yoelii-infected mice 24 and 40 h after infection, strengthening the data obtained with the in vitro model and highlighting genes and pathways involved in the host response to rodent Plasmodium parasites. Conclusion Our data reveal that host cell infection by Plasmodium sporozoites leads to a coordinated and sequential set of biological events, ranging from the initial stage of stress response up to the engagement of host metabolic processes and the maintenance of cell viability throughout infection.
Collapse
Affiliation(s)
- Sónia S Albuquerque
- Unidade de Malária, Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Lisboa, Portugal.
| | | | | | | | | | | | | | | |
Collapse
|
31
|
BLADER IRAJ, SAEIJ JEROENP. Communication between Toxoplasma gondii and its host: impact on parasite growth, development, immune evasion, and virulence. APMIS 2009; 117:458-76. [PMID: 19400868 PMCID: PMC2810527 DOI: 10.1111/j.1600-0463.2009.02453.x] [Citation(s) in RCA: 135] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Toxoplasma gondii is an obligate intracellular protozoan parasite that can infect most warm-blooded animals and cause severe and life-threatening disease in developing fetuses and in immune-compromised patients. Although Toxoplasma was discovered over 100 years ago, we are only now beginning to appreciate the importance of the role that parasite modulation of its host has on parasite growth, bradyzoite development, immune evasion, and virulence. The goal of this review is to highlight these findings, to develop an integrated model for communication between Toxoplasma and its host, and to discuss new questions that arise out of these studies.
Collapse
Affiliation(s)
- IRA J. BLADER
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, BMSB 1034, 940 Stanton L. Young Blvd. Oklahoma City, OK 73104, USA.
| | - JEROEN P. SAEIJ
- Department of Biology, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Building 68-270, Cambridge, MA 02139, USA.
| |
Collapse
|
32
|
Jamieson SE, Cordell H, Petersen E, McLeod R, Gilbert RE, Blackwell JM. Host genetic and epigenetic factors in toxoplasmosis. Mem Inst Oswaldo Cruz 2009; 104:162-9. [PMID: 19430638 PMCID: PMC2735098 DOI: 10.1590/s0074-02762009000200006] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2008] [Accepted: 02/17/2009] [Indexed: 12/29/2022] Open
Abstract
Analysing human genetic variation provides a powerful tool in understanding risk factors for disease. Toxoplasma gondii acquired by the mother can be transmitted to the fetus. Infants with the most severe clinical signs in brain and eye are those infected early in pregnancy when fetal immunity is least well developed. Genetic analysis could provide unique insight into events in utero that are otherwise difficult to determine. We tested the hypothesis that propensity for T. gondii to cause eye disease is associated with genes previously implicated in congenital or juvenile onset ocular disease. Using mother-child pairs from Europe (EMSCOT) and child/parent trios from North America (NCCCTS), we demonstrated that ocular and brain disease in congenital toxoplasmosis associate with polymorphisms in ABCA4 encoding ATP-binding cassette transporter, subfamily A, member 4 previously associated with juvenile onset retinal dystrophies including Stargardt's disease. Polymorphisms at COL2A1 encoding type II collagen, previously associated with Stickler syndrome, associated only with ocular disease in congenital toxoplasmosis. Experimental studies showed that both ABCA4 and COL2A1 show isoform-specific epigenetic modifications consistent with imprinting, which provided an explanation for the patterns of inheritance observed. These genetic and epigenetic risk factors provide unique insight into molecular pathways in the pathogenesis of disease.
Collapse
Affiliation(s)
- Sarra E Jamieson
- Telethon Institute for Child Health Research, Centre for Child Health Research, The University of Western Australia, PO Box 855, West Perth, Western Australia 6872, Australia
- Cambridge Institute for Medical Research and Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Heather Cordell
- Cambridge Institute for Medical Research and Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, UK
- Institute of Human Genetics, Newcastle University, International Centre for Life, Central Parkway, Newcastle upon Tyne, UK
| | - Eskild Petersen
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| | - Rima McLeod
- Departments of Ophthalmology, Medicine, Pediatrics, Committees on Immunology, Molecular Medicine and Genetics, University of Chicago, Illinois, USA
- Michael Reese Hospital and Medical Center, Chicago, Illinois, USA
| | - Ruth E Gilbert
- Centre for Paediatric Epidemiology and Biostatistics, Institute of Child Health, University College London, London, UK
| | - Jenefer M Blackwell
- Telethon Institute for Child Health Research, Centre for Child Health Research, The University of Western Australia, PO Box 855, West Perth, Western Australia 6872, Australia
- Cambridge Institute for Medical Research and Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, UK
| |
Collapse
|
33
|
Miller CM, Boulter NR, Ikin RJ, Smith NC. The immunobiology of the innate response to Toxoplasma gondii. Int J Parasitol 2008; 39:23-39. [PMID: 18775432 DOI: 10.1016/j.ijpara.2008.08.002] [Citation(s) in RCA: 143] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2008] [Revised: 08/07/2008] [Accepted: 08/11/2008] [Indexed: 01/17/2023]
Abstract
Toxoplasma gondii is a unique intracellular parasite. It can infect a variety of cells in virtually all warm-blooded animals. It has a worldwide distribution and, overall, around one-third of people are seropositive for the parasite, with essentially the entire human population being at risk of infection. For most people, T. gondii causes asymptomatic infection but the parasite can cause serious disease in the immunocompromised and, if contracted for the first time during pregnancy, can cause spontaneous abortion or congenital defects, which have a substantial emotional, social and economic impact. Toxoplasma gondii provokes one of the most potent innate, pro-inflammatory responses of all infectious disease agents. It is also a supreme manipulator of the immune response so that innate immunity to T. gondii is a delicate balance between the parasite and its host involving a coordinated series of cellular interactions involving enterocytes, neutrophils, dendritic cells, macrophages and natural killer cells. Underpinning these interactions is the regulation of complex molecular reactions involving Toll-like receptors, activation of signalling pathways, cytokine production and activation of anti-microbial effector mechanisms including generation of reactive nitrogen and oxygen intermediates.
Collapse
Affiliation(s)
- Catherine M Miller
- Institute for the Biotechnology of Infectious Diseases, University of Technology, Sydney, PO Box 123, Broadway, NSW 2007, Australia
| | | | | | | |
Collapse
|
34
|
Jamieson SE, de Roubaix LA, Cortina-Borja M, Tan HK, Mui EJ, Cordell HJ, Kirisits MJ, Miller EN, Peacock CS, Hargrave AC, Coyne JJ, Boyer K, Bessieres MH, Buffolano W, Ferret N, Franck J, Kieffer F, Meier P, Nowakowska DE, Paul M, Peyron F, Stray-Pedersen B, Prusa AR, Thulliez P, Wallon M, Petersen E, McLeod R, Gilbert RE, Blackwell JM. Genetic and epigenetic factors at COL2A1 and ABCA4 influence clinical outcome in congenital toxoplasmosis. PLoS One 2008; 3:e2285. [PMID: 18523590 PMCID: PMC2390765 DOI: 10.1371/journal.pone.0002285] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2008] [Accepted: 04/11/2008] [Indexed: 01/26/2023] Open
Abstract
Background Primary Toxoplasma gondii infection during pregnancy can be transmitted to the fetus. At birth, infected infants may have intracranial calcification, hydrocephalus, and retinochoroiditis, and new ocular lesions can occur at any age after birth. Not all children who acquire infection in utero develop these clinical signs of disease. Whilst severity of disease is influenced by trimester in which infection is acquired by the mother, other factors including genetic predisposition may contribute. Methods and Findings In 457 mother-child pairs from Europe, and 149 child/parent trios from North America, we show that ocular and brain disease in congenital toxoplasmosis associate with polymorphisms in ABCA4 encoding ATP-binding cassette transporter, subfamily A, member 4. Polymorphisms at COL2A1 encoding type II collagen associate only with ocular disease. Both loci showed unusual inheritance patterns for the disease allele when comparing outcomes in heterozygous affected children with outcomes in affected children of heterozygous mothers. Modeling suggested either an effect of mother's genotype, or parent-of-origin effects. Experimental studies showed that both ABCA4 and COL2A1 show isoform-specific epigenetic modifications consistent with imprinting. Conclusions These associations between clinical outcomes of congenital toxoplasmosis and polymorphisms at ABCA4 and COL2A1 provide novel insight into the molecular pathways that can be affected by congenital infection with this parasite.
Collapse
Affiliation(s)
- Sarra E. Jamieson
- Cambridge Institute for Medical Research and Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrookes Hospital, Cambridge, United Kingdom
| | - Lee-Anne de Roubaix
- Cambridge Institute for Medical Research and Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrookes Hospital, Cambridge, United Kingdom
| | - Mario Cortina-Borja
- Centre for Paediatric Epidemiology and Biostatistics, Institute of Child Health, University College London, London, United Kingdom
| | - Hooi Kuan Tan
- Centre for Paediatric Epidemiology and Biostatistics, Institute of Child Health, University College London, London, United Kingdom
| | - Ernest J. Mui
- Departments of Ophthalmology, Medicine, Pediatrics, Committees on Immunology, Molecular Medicine, and Genetics, University of Chicago, and Michael Reese Hospital and Medical Center, Chicago, Illinois, United States of America
| | - Heather J. Cordell
- Cambridge Institute for Medical Research and Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrookes Hospital, Cambridge, United Kingdom
- Institute of Human Genetics, Newcastle University, International Centre for Life, Newcastle upon Tyne, United Kingdom
| | - Michael J. Kirisits
- Departments of Ophthalmology, Medicine, Pediatrics, Committees on Immunology, Molecular Medicine, and Genetics, University of Chicago, and Michael Reese Hospital and Medical Center, Chicago, Illinois, United States of America
| | - E. Nancy Miller
- Cambridge Institute for Medical Research and Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrookes Hospital, Cambridge, United Kingdom
| | - Christopher S. Peacock
- Cambridge Institute for Medical Research and Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrookes Hospital, Cambridge, United Kingdom
| | - Aubrey C. Hargrave
- Departments of Ophthalmology, Medicine, Pediatrics, Committees on Immunology, Molecular Medicine, and Genetics, University of Chicago, and Michael Reese Hospital and Medical Center, Chicago, Illinois, United States of America
| | - Jessica J. Coyne
- Departments of Ophthalmology, Medicine, Pediatrics, Committees on Immunology, Molecular Medicine, and Genetics, University of Chicago, and Michael Reese Hospital and Medical Center, Chicago, Illinois, United States of America
| | - Kenneth Boyer
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Rush University Medical Center, Chicago, Illinois, United States of America
| | | | - Wilma Buffolano
- Department of Paediatrics, University of Naples "Frederico II", Naples, Italy
| | - Nicole Ferret
- Service de Parasitologie et Mycologie, Hopital Archet II, Nice, France
| | | | - François Kieffer
- Department of Paediatrics, Institut de Puériculture, Paris, France
| | - Paul Meier
- Department of Biostatistics, Columbia University, New York, New York, United States of America
| | - Dorota E. Nowakowska
- Department of Fetal-Maternal Medicine and Gynecology, Medical University, Lodz, Rzgowska, Poland
| | - Malgorzata Paul
- Department and Clinic of Tropical and Parasitic Diseases, University of Medical Sciences, Poznań, Poland
| | - François Peyron
- Hospices Civils de Lyon, Service de Parasitologie, Hôpital de la Croix-Rousse, Lyon, France
| | - Babill Stray-Pedersen
- Department of Obstetrics and Gynaecology, University of Oslo, Rikshospitalet-Radiumhospitalet, Sognsvannsvn, Oslo, Norway
| | - Andrea-Romana Prusa
- Department of Pediatrics, Division of Neonatology, Congenital Disorders and Intensive Care, Medical University of Vienna, Vienna, Austria
| | | | - Martine Wallon
- Hospices Civils de Lyon, Service de Parasitologie, Hôpital de la Croix-Rousse, Lyon, France
| | - Eskild Petersen
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| | - Rima McLeod
- Departments of Ophthalmology, Medicine, Pediatrics, Committees on Immunology, Molecular Medicine, and Genetics, University of Chicago, and Michael Reese Hospital and Medical Center, Chicago, Illinois, United States of America
| | - Ruth E. Gilbert
- Centre for Paediatric Epidemiology and Biostatistics, Institute of Child Health, University College London, London, United Kingdom
| | - Jenefer M. Blackwell
- Cambridge Institute for Medical Research and Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrookes Hospital, Cambridge, United Kingdom
- * E-mail:
| |
Collapse
|
35
|
Abstract
Toxoplasma gondii is an obligate intracellular parasite that can infect virtually any nucleated cell. During invasion Toxoplasma creates the parasitophorous vacuole, a subcellular compartment that acts as an interface between the parasite and host, and serves as a platform for modulation of host cell functions that support parasite replication and infection. Spatial reorganization of host organelles and cytoskeleton around the parasitophorous vacuole are observed following entry, and recent evidence suggests this interior redecorating promotes parasite nutrient acquisition. New findings also reveal that Toxoplasma manipulates host signaling pathways by deploying parasite kinases and a phosphatase, including at least two that infiltrate the host nucleus. Toxoplasma infection additionally controls several cellular pathways to establish an anti-apoptotic environment, and subverts immune cells as a conduit for dissemination. In this review we discuss these recent developments in understanding how Toxoplasma achieves widespread success as a human and animal parasite by manipulating its host.
Collapse
Affiliation(s)
- J Laliberté
- Department of Microbiology and Immunology, University of Michigan Medical School, 1150 West Medical Center Drive, 5751 Medical Science Building II, Ann Arbor, Michigan 48109-0620, USA.
| | | |
Collapse
|
36
|
Abstract
Mammalian cells infected with Toxoplasma gondii are characterized by a profound reprogramming of gene expression. We examined whether such transcriptional responses were linked to changes in the cell cycle of the host. Human foreskin fibroblasts (HFFs) in the G(0)/G(1) phase of the cell cycle were infected with T. gondii and FACS analysis of DNA content was performed. Cell cycle profiles revealed a promotion into the S phase followed by an arrest towards the G(2)/M boundary with infection. This response was markedly different from that of growth factor stimulation which caused cell cycle entry and completion. Transcriptional profiles of T. gondii-infected HFF showed sustained increases in transcripts associated with a G(1)/S transition and DNA synthesis coupled to an abrogation of cell cycle regulators critical in G(2)/M transition relative to growth factor stimulation. These divergent responses correlated with a distinct temporal modulation of the critical cell cycle regulator kinase ERK by infection. While the kinetics of ERK phosphorylation by EGF showed rapid and sustained activation, infected cells displayed an oscillatory pattern of activation. Our results suggest that T. gondii infection induces and maintains a 'proliferation response' in the infected cell which may fulfill critical growth requirements of the parasite during intracellular residence.
Collapse
Affiliation(s)
- Robert E. Molestina
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky College of Medicine, Lexington, Kentucky, 40536 USA
| | - Nadia El-Guendy
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky College of Medicine, Lexington, Kentucky, 40536 USA
| | - Anthony P. Sinai
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky College of Medicine, Lexington, Kentucky, 40536 USA
| |
Collapse
|
37
|
El-Guendy NM, Sinai AP. Potential problems inherent in cell-based stable NF-kappaB-GFP reporter systems. Mol Cell Biochem 2008; 312:147-55. [PMID: 18327667 PMCID: PMC2647807 DOI: 10.1007/s11010-008-9730-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2007] [Accepted: 02/25/2008] [Indexed: 01/14/2023]
Abstract
The nuclear factor-kappaB (NF-kappaB) family of transcription factors plays a central role in numerous physiological processes including development, cell survival, immunity, and inflammation. We generated a series of stable clonal lines in mouse embryonic fibroblasts carrying NF-kappaB-GFP plasmid as a reporter. These cell lines were selected by flow cytometry for their high responsiveness to tumor necrosis factor (TNFalpha) or lipopolysaccharide (LPS), two classic NF-kappaB-inducing stimuli. Although all clones were generated from the same parental cell line, they each had a distinctive pattern of response to NF-kappaB stimuli. While exhibiting distinct profiles with regard to the GFP reporter, analysis of endogenous NF-kappaB downstream targets did not always show the same variability. This suggests that in the absence of confirmation of the signaling outcomes using endogenous outputs, considerable caution must be exercised in the interpretation of data using stable reporter systems.
Collapse
Affiliation(s)
- Nadia M. El-Guendy
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky College of Medicine, Lexington, Kentucky, 40536 USA
| | - Anthony P. Sinai
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky College of Medicine, Lexington, Kentucky, 40536 USA
| |
Collapse
|
38
|
Nelson MM, Jones AR, Carmen JC, Sinai AP, Burchmore R, Wastling JM. Modulation of the host cell proteome by the intracellular apicomplexan parasite Toxoplasma gondii. Infect Immun 2008; 76:828-44. [PMID: 17967855 PMCID: PMC2223483 DOI: 10.1128/iai.01115-07] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2007] [Revised: 09/19/2007] [Accepted: 10/21/2007] [Indexed: 01/09/2023] Open
Abstract
To investigate how intracellular parasites manipulate their host cell environment at the molecular level, we undertook a quantitative proteomic study of cells following infection with the apicomplexan parasite Toxoplasma gondii. Using conventional two-dimensional electrophoresis, difference gel electrophoresis (DIGE), and mass spectrometry, we identified host proteins that were consistently modulated in expression following infection. We detected modification of protein expression in key metabolic pathways, including glycolysis, lipid and sterol metabolism, mitosis, apoptosis, and structural-protein expression, suggestive of global reprogramming of cell metabolism by the parasite. Many of the differentially expressed proteins had not been previously implicated in the response to the parasite, while others provide important corroborative protein evidence for previously proposed hypotheses of pathogen-cell interactions. Significantly, over one-third of all modulated proteins were mitochondrial, and this was further investigated by DIGE analysis of a mitochondrion-enriched preparation from infected cells. Comparison of our proteomic data with previous transcriptional studies suggested that a complex relationship exits between transcription and protein expression that may be partly explained by posttranslational modifications of proteins and revealed the importance of investigating protein changes when interpreting transcriptional data. To investigate this further, we used phosphatase treatment and DIGE to demonstrate changes in the phosphorylation states of several key proteins following infection. Overall, our findings indicate that the host cell proteome responds in a dramatic way to T. gondii invasion, in terms of both protein expression changes and protein modifications, and reveal a complex and intimate molecular relationship between host and parasite.
Collapse
Affiliation(s)
- M M Nelson
- Department of Pre-Clinical Veterinary Science and Veterinary Pathology, Faculty of Veterinary Science, University of Liverpool, Liverpool L69 7ZJ, United Kingdom
| | | | | | | | | | | |
Collapse
|
39
|
Abstract
Apicomplexan parasites like Toxoplasma gondii are distinctive in their utilization of para site encoded motor systems to invade cells. Invasion results in the establishment of the parasitophorous vacuole (PV) within the infected cell. Most apicomplexans complete their intracellular tenure within the infected cell in the PV that is demarcated from the host cytoplasm by the parasitophorous vacuole membrane (PVM). In this chapter I focus on the events surrounding the formation of the PVM and selected activities attributed to it. Its central role as the interface between the parasite and its immediate environment, the host cytoplasm, is validated by the diversity of functions attributed to it. While functions in structural organization, nutrient acquisitions and signaling have been defined their molecular bases remain largely unknown. Several recent studies and the decoding of the Toxoplasma genome have set the stage for a rapid expansion in our understanding of the role of the PVM in parasite biology. Toxoplasma gondii, like all apicomplexan parasites are obligate intracellular pathogens. This family of parasites utilize their own actin-myosin based motor systems to gain entry into susceptible cells establishing themselves, in some cases transiently (e.g., Theileria spp) in specialized vacuolar compartment, the parasitophorous vacuole (PV). The T. gondii PV is highly dynamic compartment defining the replication permissive niche for the parasite. The delimiting membrane defining the parasitophorous vacuole, the parasitophorous vacuole membrane or PVM is increasingly being recognized as a specialized "organelle" that in the context of the infected cell is extracorporeal to the parent organism, the parasite. A systematic study of this enigmatic organelle has been severely limited by several issues. Primary among these is the fact that it is formed only in the context of the infected cell thereby limiting the amount of material. Secondly, unlike other cellular organelles that can often be purified by conventional approaches, the PVM, cannot be purified away from host cell organelles (see below). In spite of these significant obstacles considerable progress has been made in recent years toward understanding the biogenesis of the PVM, identification of its protein complement and the characterization of activities within it. These studies demonstrate that the PVM, on its own and by virtue of its interactions with cellular components, plays critical functions in the structural integrity of the vacuole, nutrient acquisition and the manipulation of cellular functions. In addition it appears that the repertoire of activities at the PVM is likely to be plastic reflecting temporal changes associated with the replicative phase of parasite growth. Finally, the PVM likely forms the foundation for the cyst wall as the parasite differentiates in the establishment of latent infection. As the critical border crossing between the parasite and invaded cell the study of the PVM provides a fertile area for new investigation aided by the recent decoding of the Toxoplasma genome (available at wwww.ToxoDB.org) and the application of proteomic analyses to basic questions in parasite biology.
Collapse
Affiliation(s)
- Anthony P Sinai
- Department of Microbiology Immunology and Molecular Genetics, University of Kentucky College of Medicine, 800 Rose St., Lexington, Kentucky 40536, USA.
| |
Collapse
|
40
|
Herman RK, Molestina RE, Sinai AP, Howe DK. The apicomplexan pathogen Neospora caninum inhibits host cell apoptosis in the absence of discernible NF-kappa B activation. Infect Immun 2007; 75:4255-62. [PMID: 17576757 PMCID: PMC1951197 DOI: 10.1128/iai.00418-07] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2007] [Revised: 04/26/2007] [Accepted: 06/07/2007] [Indexed: 12/14/2022] Open
Abstract
Neospora caninum, a causative agent of bovine abortions, is an apicomplexan parasite that is closely related to the human pathogen Toxoplasma gondii. Since a number of intracellular parasites, including T. gondii, have been shown to modulate host cell apoptosis, the present study was conducted to establish whether N. caninum is similarly capable of subverting apoptotic pathways in its host cells. Our results indicated that death receptor-mediated apoptosis is repressed during N. caninum infection, and the data further showed that the executioner caspase, caspase 3, does not become activated in the infected cells. Surprisingly, nuclear translocation of the NF-kappaB subunit p65 was not detected in N. caninum-infected cells, although this host transcription factor has been shown to upregulate prosurvival genes in cells infected with T. gondii. Consistent with these findings, the distinct accumulation of phosphorylated IkappaB that is seen at the parasitophorous vacuole membrane (PVM) of T. gondii was not apparent on the N. caninum PVM. Although a putative IkappaB kinase activity was detected in N. caninum extracts, thereby implying that this parasite is capable of modulating NF-kappaB translocation into the host cell nucleus, the data collectively suggest that a profound and sustained activation of the NF-kappaB pathway is not central to the ability of N. caninum to prevent apoptosis of their host cells.
Collapse
Affiliation(s)
- Rebecca K Herman
- Department of Veterinary Science, 108 Gluck Equine Research Center, University of Kentucky, Lexington, KY 40546-0099, USA
| | | | | | | |
Collapse
|
41
|
Bruchhaus I, Roeder T, Rennenberg A, Heussler VT. Protozoan parasites: programmed cell death as a mechanism of parasitism. Trends Parasitol 2007; 23:376-83. [PMID: 17588817 DOI: 10.1016/j.pt.2007.06.004] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2006] [Revised: 04/27/2007] [Accepted: 06/06/2007] [Indexed: 12/25/2022]
Abstract
Programmed cell death (PCD) is a potent mechanism to remove parasitized cells, but it has also been shown that protozoan parasites can induce or inhibit apoptosis in host cells. In recent years, it has become clear that unicellular parasites can also undergo PCD, meaning that they commit suicide in response to various stimuli. This review focuses on the role of protozoan PCD and on the interaction between protozoan parasites and the host cell death machinery from the perspective of parasite survival strategies.
Collapse
Affiliation(s)
- Iris Bruchhaus
- Bernhard Nocht Institute for Tropical Medicine, Bernhard-Nocht-Str. 74, 20359 Hamburg, Germany.
| | | | | | | |
Collapse
|
42
|
Abstract
The modulation of apoptosis has emerged as an important weapon in the pathogenic arsenal of multiple intracellular protozoan parasites. Cryptosporidium parvum, Leishmania spp., Trypanosoma cruzi, Theileria spp., Toxoplasma gondii and Plasmodium spp. have all been shown to inhibit the apoptotic response of their host cell. While the pathogen mediators responsible for this modulation are unknown, the parasites are interacting with multiple apoptotic regulatory systems to render their host cell refractory to apoptosis during critical phases of intracellular infection, including parasite invasion, establishment and replication. Additionally, emerging evidence suggests that the parasite life cycle stage impacts the modulation of apoptosis and possibly parasite differentiation. Dissection of the host-pathogen interactions involved in modulating apoptosis reveals a dynamic and complex interaction that recent studies are beginning to unravel.
Collapse
Affiliation(s)
- John C Carmen
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky College of Medicine, Lexington, KY 40536, USA
| | | |
Collapse
|
43
|
Abstract
Toxoplasma gondii, an intracellular protozoan parasite, can infect humans in 3 different ways: ingestion of tissue cysts, ingestion of oocysts, or congenital infection with tachyzoites. After proliferation of tachyzoites in various organs during the acute stage, the parasite forms cysts preferentially in the brain and establishes a chronic infection, which is a balance between host immunity and the parasite's evasion of the immune response. A variety of brain cells, including astrocytes and neurons, can be infected. In vitro studies using non-brain cells have demonstrated profound effects of the infection on gene expression of host cells, including molecules that promote the immune response and those involved in signal transduction pathways, suggesting that similar effects could occur in infected brain cells. Interferon-gamma is the essential mediator of the immune response to control T. gondii in the brain and to maintain the latency of chronic infection. Infection also induces the production of a variety of cytokines by microglia, astrocytes, and neurons, which promote or suppress inflammatory responses. The strain (genotype) of T. gondii, genetic factors of the host, and probably the route of infection and the stage (tachyzoite, cyst, or oocyst) of the parasite initiating infection all contribute to the establishment of a balance between the host and the parasite and affect the outcome of the infection.
Collapse
Affiliation(s)
- Vern B. Carruthers
- Department of Microbiology and Immunology, University of Michigan School of Medicine, Ann Arbor, MI 48109
| | - Yasuhiro Suzuki
- Center for Molecular Medicine and Infectious Diseases, Department of Biomedical Sciences and Pathobiology, Virginia-Maryland Regional College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061
- To whom correspondence should be addressed; tel: 540-231-2095, fax: 540-231-3426, e-mail:
| |
Collapse
|
44
|
Affiliation(s)
- Anthony P Sinai
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky College of Medicine, Lexington, Kentucky, United States of America.
| |
Collapse
|
45
|
Martin AM, Liu T, Lynn BC, Sinai AP. The Toxoplasma gondii Parasitophorous Vacuole Membrane: Transactions Across the Border. J Eukaryot Microbiol 2007; 54:25-8. [PMID: 17300514 DOI: 10.1111/j.1550-7408.2006.00230.x] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The obligate intracellular protozoan Toxoplasma gondii establishes its replication permissive niche within the infected host cell. This niche, the parasitophorous vacuole (PV), is delimited from the host cell cytoplasm by the PV membrane (PVM). In this chapter we highlight the roles of the PVM in the remodeling of host cell architecture, nutrient acquisition, the manipulation of signaling, and touch upon the potential roles in the parasite developmental cycle. We further present the PVM as a unique and dynamic "organelle" found only within the infected cell where it is established outside the parent organism. Despite its importance little is known about the biology of the PVM. There has, however, been a recent renewal of interest in the PVM, the study of which has become more tractable with the application of both classical approaches as well as genomic and proteomic analyses. In this review we discuss the diverse activities associated with the PVM and present pressing questions that remain to be elucidated regarding this enigmatic organelle.
Collapse
Affiliation(s)
- Angela M Martin
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, Lexington, Kentucky 40536, USA
| | | | | | | |
Collapse
|
46
|
Fouts AE, Boothroyd JC. Infection with Toxoplasma gondii bradyzoites has a diminished impact on host transcript levels relative to tachyzoite infection. Infect Immun 2006; 75:634-42. [PMID: 17088349 PMCID: PMC1828502 DOI: 10.1128/iai.01228-06] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Toxoplasma gondii, an intracellular pathogen, has the potential to infect nearly every warm-blooded animal but rarely causes morbidity. The ability for the parasite to convert to the bradyzoite stage and live inside slow-growing cysts that can go unnoticed by the host immune system allows for parasite persistence for the life of the infected host. This intracellular survival likely necessitates host cell modulation, and tachyzoites are known to modify a number of signaling cascades within the host to promote parasite survival. Little is known, however, about how bradyzoites manipulate their host cell. Microarrays were used to profile the host transcriptional changes caused by bradyzoite infection and compared to those of tachyzoite-infected and uninfected hosts cells 2 days postinfection in vitro. Infection resulted in chemokine, cytokine, extracellular matrix, and growth factor transcript level changes. A small group of genes were specifically induced by tachyzoite infection, including granulocyte-macrophage colony-stimulating factor, BCL2-related protein A1, and interleukin-24. Bradyzoite infection yielded only about half the changes seen with tachyzoite infection, and those changes that did occur were almost all of lower magnitude than those induced by tachyzoites. These results suggest that bradyzoites lead a more stealthy existence within the infected host cell.
Collapse
Affiliation(s)
- A E Fouts
- Department of Microbiology and Immunology, Fairchild Building D305, 300 Pasteur Dr., Stanford University School of Medicine, Stanford, CA 94305-5124, USA
| | | |
Collapse
|
47
|
Carmen JC, Hardi L, Sinai AP. Toxoplasma gondii inhibits ultraviolet light-induced apoptosis through multiple interactions with the mitochondrion-dependent programmed cell death pathway. Cell Microbiol 2006; 8:301-15. [PMID: 16441440 DOI: 10.1111/j.1462-5822.2005.00622.x] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Cells infected with the protozoan parasite Toxoplasma gondii are resistant to diverse apoptotic stimuli. In this study, we perform a detailed analysis of the manipulation of the mitochondrial arm of the apoptotic cascade by the parasite. Apoptosis was induced using irradiation with ultraviolet light (UV), and the kinetics of caspase activation, cytochrome c release and activation of the upstream signalling pathways were examined. The evidence clearly points to T. gondii targeting multiple steps in the transmission [inhibition of c-Jun N-terminal kinase (JNK) activation in response to UV], triggering (inhibition of cytochrome c release by affecting the balance of pro- and anti-apoptotic BCL-2 family members) and execution (inhibition of caspase 9 and caspase 3) phases of the apoptotic cascade. Interestingly, the multilevel pattern of inhibition that emerges suggests that the global inhibition of the mitochondrial arm of apoptosis is not likely to be contributed to by the small subset of mitochondria recruited to the T. gondii parasitophorous vacuole membrane.
Collapse
Affiliation(s)
- John C Carmen
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky College of Medicine, Lexington, KY 40536, USA
| | | | | |
Collapse
|