1
|
Kwon SY, Thi-Thu Ngo H, Son J, Hong Y, Min JJ. Exploiting bacteria for cancer immunotherapy. Nat Rev Clin Oncol 2024; 21:569-589. [PMID: 38840029 DOI: 10.1038/s41571-024-00908-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/17/2024] [Indexed: 06/07/2024]
Abstract
Immunotherapy has revolutionized the treatment of cancer but continues to be constrained by limited response rates, acquired resistance, toxicities and high costs, which necessitates the development of new, innovative strategies. The discovery of a connection between the human microbiota and cancer dates back 4,000 years, when local infection was observed to result in tumour eradication in some individuals. However, the true oncological relevance of the intratumoural microbiota was not recognized until the turn of the twentieth century. The intratumoural microbiota can have pivotal roles in both the pathogenesis and treatment of cancer. In particular, intratumoural bacteria can either promote or inhibit cancer growth via remodelling of the tumour microenvironment. Over the past two decades, remarkable progress has been made preclinically in engineering bacteria as agents for cancer immunotherapy; some of these bacterial products have successfully reached the clinical stages of development. In this Review, we discuss the characteristics of intratumoural bacteria and their intricate interactions with the tumour microenvironment. We also describe the many strategies used to engineer bacteria for use in the treatment of cancer, summarizing contemporary data from completed and ongoing clinical trials. The work described herein highlights the potential of bacteria to transform the landscape of cancer therapy, bridging ancient wisdom with modern scientific innovation.
Collapse
Affiliation(s)
- Seong-Young Kwon
- Institute for Molecular Imaging and Theranostics, Chonnam National University Medical School, Jeonnam, Republic of Korea
- Department of Nuclear Medicine, Chonnam National University Medical School and Hwasun Hospital, Jeonnam, Republic of Korea
| | - Hien Thi-Thu Ngo
- Institute for Molecular Imaging and Theranostics, Chonnam National University Medical School, Jeonnam, Republic of Korea
- Department of Biomedical Sciences, Chonnam National University Medical School, Jeonnam, Republic of Korea
- Department of Biochemistry, Hanoi Medical University, Hanoi, Vietnam
| | - Jinbae Son
- CNCure Biotech, Jeonnam, Republic of Korea
| | - Yeongjin Hong
- Institute for Molecular Imaging and Theranostics, Chonnam National University Medical School, Jeonnam, Republic of Korea
- CNCure Biotech, Jeonnam, Republic of Korea
- Department of Microbiology and Immunology, Chonnam National University Medical School, Jeonnam, Republic of Korea
- National Immunotherapy Innovation Center, Chonnam National University, Jeonnam, Republic of Korea
| | - Jung-Joon Min
- Institute for Molecular Imaging and Theranostics, Chonnam National University Medical School, Jeonnam, Republic of Korea.
- Department of Nuclear Medicine, Chonnam National University Medical School and Hwasun Hospital, Jeonnam, Republic of Korea.
- Department of Biomedical Sciences, Chonnam National University Medical School, Jeonnam, Republic of Korea.
- CNCure Biotech, Jeonnam, Republic of Korea.
- Department of Microbiology and Immunology, Chonnam National University Medical School, Jeonnam, Republic of Korea.
- National Immunotherapy Innovation Center, Chonnam National University, Jeonnam, Republic of Korea.
| |
Collapse
|
2
|
Shi W, Zhang Q, Li H, Du D, Ma X, Wang J, Jiang J, Liu C, Kou L, Ren J. Biofilm Formation, Motility, and Virulence of Listeria monocytogenes Are Reduced by Deletion of the Gene lmo0159, a Novel Listerial LPXTG Surface Protein. Microorganisms 2024; 12:1354. [PMID: 39065121 PMCID: PMC11278909 DOI: 10.3390/microorganisms12071354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 06/27/2024] [Accepted: 06/29/2024] [Indexed: 07/28/2024] Open
Abstract
Listeria monocytogenes (L. monocytogenes) is a foodborne pathogen that causes listeriosis in humans and other animals. Surface proteins with the LPXTG motif have important roles in the virulence of L. monocytogenes. Lmo0159 is one such protein, but little is known about its role in L. monocytogenes virulence, motility, and biofilm formation. Here, we constructed and characterized a deletion mutant of lmo0159 (∆lmo0159). We analyzed not only the capacity of biofilm formation, motility, attachment, and intracellular growth in different cell types but also LD50; bacterial load in mice's liver, spleen, and brain; expression of virulence genes; and survival time of mice after challenge. The results showed that the cross-linking density of the biofilm of ∆lmo0159 strain was lower than that of WT by microscopic examination. The expression of biofilm-formation and virulence genes also decreased in the biofilm state. Subsequently, the growth and motility of ∆lmo0159 in the culture medium were enhanced. Conversely, the growth and motility of L. monocytogenes were attenuated by ∆lmo0159 at both the cellular and mouse levels. At the cellular level, ∆lmo0159 reduced plaque size; accelerated scratch healing; and attenuated the efficiency of adhesion, invasion, and intracellular proliferation in swine intestinal epithelial cells (SIEC), RAW264.7, mouse-brain microvascular endothelial cells (mBMEC), and human-brain microvascular endothelial cells (hCMEC/D3). The expression of virulence genes was also inhibited. At the mouse level, the LD50 of the ∆lmo0159 strain was 100.97 times higher than that of the WT strain. The bacterial load of the ∆lmo0159 strain in the liver and spleen was lower than that of the WT strain. In a mouse model of intraperitoneal infection, the deletion of the lmo0159 gene significantly prolonged the survival time of the mice, suggesting that the lmo0159 deletion mutant also exhibited reduced virulence. Thus, our study identified lmo0159 as a novel virulence factor among L. monocytogenes LPXTG proteins.
Collapse
Affiliation(s)
- Weidi Shi
- College of Animal Science and Technology, Shihezi University, Shihezi 832000, China; (W.S.); (Q.Z.); (H.L.); (J.J.); (C.L.); (L.K.); (J.R.)
| | - Qiwen Zhang
- College of Animal Science and Technology, Shihezi University, Shihezi 832000, China; (W.S.); (Q.Z.); (H.L.); (J.J.); (C.L.); (L.K.); (J.R.)
| | - Honghuan Li
- College of Animal Science and Technology, Shihezi University, Shihezi 832000, China; (W.S.); (Q.Z.); (H.L.); (J.J.); (C.L.); (L.K.); (J.R.)
| | - Dongdong Du
- Analysis and Testing Center, Xinjiang Academy of Agriculture and Reclamation Science, Shihezi 832000, China;
| | - Xun Ma
- College of Animal Science and Technology, Shihezi University, Shihezi 832000, China; (W.S.); (Q.Z.); (H.L.); (J.J.); (C.L.); (L.K.); (J.R.)
| | - Jing Wang
- College of Animal Science and Technology, Shihezi University, Shihezi 832000, China; (W.S.); (Q.Z.); (H.L.); (J.J.); (C.L.); (L.K.); (J.R.)
- Key Laboratory of Control and Prevention of Animal Disease, Xinjiang Production & Construction, Shihezi 832000, China
| | - Jianjun Jiang
- College of Animal Science and Technology, Shihezi University, Shihezi 832000, China; (W.S.); (Q.Z.); (H.L.); (J.J.); (C.L.); (L.K.); (J.R.)
| | - Caixia Liu
- College of Animal Science and Technology, Shihezi University, Shihezi 832000, China; (W.S.); (Q.Z.); (H.L.); (J.J.); (C.L.); (L.K.); (J.R.)
| | - Lijun Kou
- College of Animal Science and Technology, Shihezi University, Shihezi 832000, China; (W.S.); (Q.Z.); (H.L.); (J.J.); (C.L.); (L.K.); (J.R.)
| | - Jingjing Ren
- College of Animal Science and Technology, Shihezi University, Shihezi 832000, China; (W.S.); (Q.Z.); (H.L.); (J.J.); (C.L.); (L.K.); (J.R.)
| |
Collapse
|
3
|
Wang J, Cui M, Liu Y, Chen M, Xu J, Xia J, Sun J, Jiang L, Fang W, Song H, Cheng C. The mitochondrial carboxylase PCCA interacts with Listeria monocytogenes phospholipase PlcB to modulate bacterial survival. Appl Environ Microbiol 2024; 90:e0213523. [PMID: 38727222 PMCID: PMC11218614 DOI: 10.1128/aem.02135-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 03/19/2024] [Indexed: 06/19/2024] Open
Abstract
Listeria monocytogenes, a prominent foodborne pathogen responsible for zoonotic infections, owes a significant portion of its virulence to the presence of the phospholipase PlcB. In this study, we performed an in-depth examination of the intricate relationship between L. monocytogenes PlcB and host cell mitochondria, unveiling a novel participant in bacterial survival: the mitochondrial carboxylase propionyl-coenzyme A carboxylase (PCCA). Our investigation uncovered previously unexplored levels of interaction and colocalization between PCCA and PlcB within host cells, with particular emphasis on the amino acids 504-508 of PCCA, which play a pivotal role in this partnership. To assess the effect of PCCA expression on L. monocytogenes proliferation, PCCA expression levels were manipulated by siRNA-si-PCCA or pCMV-N-HA-PCCA plasmid transfection. Our findings demonstrated a clear inverse correlation between PCCA expression levels and the proliferation of L. monocytogenes. Furthermore, the effect of L. monocytogenes infection on PCCA expression was investigated by assessing PCCA mRNA and protein expression in HeLa cells infected with L. monocytogenes. These results indicate that L. monocytogenes infection did not significantly alter PCCA expression. These findings led us to propose that PCCA represents a novel participant in L. monocytogenes survival, and its abundance has a detrimental impact on bacterial proliferation. This suggests that L. monocytogenes may employ PlcB-PCCA interactions to maintain stable PCCA expression, representing a unique pro-survival strategy distinct from that of other intracellular bacterial pathogens. IMPORTANCE Mitochondria represent attractive targets for pathogenic bacteria seeking to modulate host cellular processes to promote their survival and replication. Our current study has uncovered mitochondrial carboxylase propionyl-coenzyme A carboxylase (PCCA) as a novel host cell protein that interacts with L. monocytogenes PlcB. The results demonstrate that PCCA plays a negative regulatory role in L. monocytogenes infection, as heightened PCCA levels are associated with reduced bacterial survival and persistence. However, L. monocytogenes may exploit the PlcB-PCCA interaction to maintain stable PCCA expression and establish a favorable intracellular milieu for bacterial infection. Our findings shed new light on the intricate interplay between bacterial pathogens and host cell mitochondria, while also highlighting the potential of mitochondrial metabolic enzymes as antimicrobial agents.
Collapse
Affiliation(s)
- Jing Wang
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection and Internet Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology and College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, Zhejiang, China
| | - Mingzhu Cui
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection and Internet Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology and College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, Zhejiang, China
| | - Yucong Liu
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection and Internet Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology and College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, Zhejiang, China
| | - Mianmian Chen
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection and Internet Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology and College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, Zhejiang, China
| | - Jiali Xu
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection and Internet Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology and College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, Zhejiang, China
| | - Jing Xia
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection and Internet Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology and College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, Zhejiang, China
| | - Jing Sun
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection and Internet Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology and College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, Zhejiang, China
| | - Lingli Jiang
- Ningbo College of Health Sciences, Ningbo, Zhejiang, China
| | - Weihuan Fang
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection and Internet Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology and College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, Zhejiang, China
| | - Houhui Song
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection and Internet Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology and College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, Zhejiang, China
| | - Changyong Cheng
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection and Internet Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology and College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, Zhejiang, China
| |
Collapse
|
4
|
Fischer K, Bradlerova M, Decker T, Supper V. Vγ9+Vδ2+ T cell control of Listeria monocytogenes growth in infected epithelial cells requires butyrophilin 3A genes. Sci Rep 2023; 13:18651. [PMID: 37903831 PMCID: PMC10616279 DOI: 10.1038/s41598-023-45587-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 10/21/2023] [Indexed: 11/01/2023] Open
Abstract
Intracellular bacteria produce antigens, which serve as potent activators of γδ T cells. Phosphoantigens are presented via a complex of butyrophilins (BTN) to signal infection to human Vγ9+Vδ2+ T cells. Here, we established an in vitro system allowing for studies of Vγ9+Vδ2+ T cell activity in coculture with epithelial cells infected with the intracellular bacterial pathogen Listeria monocytogenes. We report that the Vγ9+Vδ2+ T cells efficiently control L. monocytogenes growth in such cultures. This effector function requires the expression of members of the BTN3A family on epithelial cells. Specifically, we observed a BTN3A1-independent BTN3A3 activity to present antigen to Vγ9+Vδ2+ T cells. Since BTN3A1 is the only BTN3A associated with phosphoantigen presentation, our study suggests that BTN3A3 may present different classes of antigens to mediate Vγ9+Vδ2+ T cell effector function against L. monocytogenes-infected epithelia.
Collapse
Affiliation(s)
- Katrin Fischer
- Max Perutz Labs, Vienna Biocenter Campus (VBC), Vienna, Austria
- Department of Microbiology, Immunobiology and Genetics, Center for Molecular Biology, University of Vienna, Vienna, Austria
| | - Michaela Bradlerova
- Max Perutz Labs, Vienna Biocenter Campus (VBC), Vienna, Austria
- Department of Microbiology, Immunobiology and Genetics, Center for Molecular Biology, University of Vienna, Vienna, Austria
| | - Thomas Decker
- Max Perutz Labs, Vienna Biocenter Campus (VBC), Vienna, Austria.
- Department of Microbiology, Immunobiology and Genetics, Center for Molecular Biology, University of Vienna, Vienna, Austria.
| | - Verena Supper
- Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria
| |
Collapse
|
5
|
Ding YD, Shu LZ, He RS, Chen KY, Deng YJ, Zhou ZB, Xiong Y, Deng H. Listeria monocytogenes: a promising vector for tumor immunotherapy. Front Immunol 2023; 14:1278011. [PMID: 37868979 PMCID: PMC10587691 DOI: 10.3389/fimmu.2023.1278011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 09/25/2023] [Indexed: 10/24/2023] Open
Abstract
Cancer receives enduring international attention due to its extremely high morbidity and mortality. Immunotherapy, which is generally expected to overcome the limits of traditional treatments, serves as a promising direction for patients with recurrent or metastatic malignancies. Bacteria-based vectors such as Listeria monocytogenes take advantage of their unique characteristics, including preferential infection of host antigen presenting cells, intracellular growth within immune cells, and intercellular dissemination, to further improve the efficacy and minimize off-target effects of tailed immune treatments. Listeria monocytogenes can reshape the tumor microenvironment to bolster the anti-tumor effects both through the enhancement of T cells activity and a decrease in the frequency and population of immunosuppressive cells. Modified Listeria monocytogenes has been employed as a tool to elicit immune responses against different tumor cells. Currently, Listeria monocytogenes vaccine alone is insufficient to treat all patients effectively, which can be addressed if combined with other treatments, such as immune checkpoint inhibitors, reactivated adoptive cell therapy, and radiotherapy. This review summarizes the recent advances in the molecular mechanisms underlying the involvement of Listeria monocytogenes vaccine in anti-tumor immunity, and discusses the most concerned issues for future research.
Collapse
Affiliation(s)
- Yi-Dan Ding
- Medical College, Nanchang University, Nanchang, China
| | - Lin-Zhen Shu
- Medical College, Nanchang University, Nanchang, China
| | - Rui-Shan He
- Medical College, Nanchang University, Nanchang, China
| | - Kai-Yun Chen
- Office of Clinical Trials Administration, The Fourth Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yan-Juan Deng
- Department of Pathology, The Fourth Affiliated Hospital of Nanchang University, Nanchang, China
- Tumor Immunology Institute, Nanchang University, Nanchang, China
| | - Zhi-Bin Zhou
- Department of Pathology, The Fourth Affiliated Hospital of Nanchang University, Nanchang, China
- Tumor Immunology Institute, Nanchang University, Nanchang, China
| | - Ying Xiong
- Department of General Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Huan Deng
- Department of Pathology, The Fourth Affiliated Hospital of Nanchang University, Nanchang, China
- Tumor Immunology Institute, Nanchang University, Nanchang, China
| |
Collapse
|
6
|
Abstract
Listeria monocytogenes is a Gram-positive facultative intracellular pathogen that can cause severe invasive infections upon ingestion with contaminated food. Clinically, listerial disease, or listeriosis, most often presents as bacteremia, meningitis or meningoencephalitis, and pregnancy-associated infections manifesting as miscarriage or neonatal sepsis. Invasive listeriosis is life-threatening and a main cause of foodborne illness leading to hospital admissions in Western countries. Sources of contamination can be identified through international surveillance systems for foodborne bacteria and strains' genetic data sharing. Large-scale whole genome studies have increased our knowledge on the diversity and evolution of L. monocytogenes, while recent pathophysiological investigations have improved our mechanistic understanding of listeriosis. In this article, we present an overview of human listeriosis with particular focus on relevant features of the causative bacterium, epidemiology, risk groups, pathogenesis, clinical manifestations, and treatment and prevention.
Collapse
Affiliation(s)
- Merel M Koopmans
- Amsterdam UMC, University of Amsterdam, Department of Neurology, Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - Matthijs C Brouwer
- Amsterdam UMC, University of Amsterdam, Department of Neurology, Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - José A Vázquez-Boland
- Infection Medicine, Edinburgh Medical School (Biomedical Sciences), University of Edinburgh, Edinburgh, United Kingdom
| | - Diederik van de Beek
- Amsterdam UMC, University of Amsterdam, Department of Neurology, Amsterdam Neuroscience, Amsterdam, the Netherlands
| |
Collapse
|
7
|
Role of mitochondria in regulating immune response during bacterial infection. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2023; 374:159-200. [PMID: 36858655 DOI: 10.1016/bs.ircmb.2022.10.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Mitochondria are dynamic organelles of eukaryotes involved in energy production and fatty acid oxidation. Besides maintaining ATP production, calcium signaling, cellular apoptosis, and fatty acid synthesis, mitochondria are also known as the central hub of the immune system as it regulates the innate immune pathway during infection. Mitochondria mediated immune functions mainly involve regulation of reactive oxygen species production, inflammasome activation, cytokine secretion, and apoptosis of infected cells. Recent findings indicate that cellular mitochondria undergo constant biogenesis, fission, fusion and degradation, and these dynamics regulate cellular immuno-metabolism. Several intracellular pathogens target and modulate these normal functions of mitochondria to facilitate their own survival and growth. De-regulation of mitochondrial functions and dynamics favors bacterial infection and pathogens are able to protect themselves from mitochondria mediated immune responses. Here, we will discuss how mitochondria mediated anti-bacterial immune pathways help the host to evade pathogenic insult. In addition, examples of bacterial pathogens modulating mitochondrial metabolism and dynamics will also be elaborated. Study of these interactions between the mitochondria and bacterial pathogens during infection will lead to a better understanding of the mitochondrial metabolism pathways and dynamics important for the establishment of bacterial diseases. In conclusion, detailed studies on how mitochondria regulate the immune response during bacterial infection can open up new avenues to develop mitochondria centric anti-bacterial therapeutics.
Collapse
|
8
|
Septicaemic Listeriosis in a White-Faced Saki (Pithecia pithecia). J Comp Pathol 2022; 194:7-13. [DOI: 10.1016/j.jcpa.2022.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 02/16/2022] [Accepted: 03/11/2022] [Indexed: 11/18/2022]
|
9
|
Ling Z, Zhao D, Xie X, Yao H, Wang Y, Kong S, Chen X, Pan Z, Jiao X, Yin Y. inlF Enhances Listeria monocytogenes Early-Stage Infection by Inhibiting the Inflammatory Response. Front Cell Infect Microbiol 2022; 11:748461. [PMID: 35223532 PMCID: PMC8866704 DOI: 10.3389/fcimb.2021.748461] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 11/09/2021] [Indexed: 12/16/2022] Open
Abstract
The internalin family proteins, which carry the leucine repeat region structural motif, play diverse roles in Listeria monocytogenes (Lm) infection and pathogenesis. Although Internalin F, encoded by inlF, was identified more than 20 years ago, its role in the Lm anti-inflammatory response remains unknown. Lm serotype 4b isolates are associated with the majority of listeriosis outbreaks, but the function of InlF in these strains is not fully understood. In this study, we aimed to elucidate the role of inlF in modulating the inflammatory response and pathogenesis of the 4b strain Lm NTSN. Strikingly, although inlF was highly expressed at the transcriptional level during infection of five non-phagocytic cell types, it was not involved in adherence or invasion. Conversely, inlF did contributed to Lm adhesion and invasion of macrophages, and dramatically suppressed the expression of pro-inflammatory cytokines interleukin (IL)-1β and tumor necrosis factor (TNF-α). Consistent with the in vitro results, during Lm infection mice, inlF significantly inhibited the expression of IL-1β and IL-6 in the spleen, as well as IL-1β, IL-6, and TNF-α in the liver. More importantly, inlF contributed to Lm colonization in the spleen, liver, and ileum during the early stage of mouse infection via intragastric administration, inducing severe inflammatory injury and histopathologic changes in the late stage. To our knowledge, this is the first report to demonstrate that inlF mediates the inhibition of the pro-inflammatory response and contributes to the colonization and survival of Lm during the early stage of infection in mice. Our research partly explains the high pathogenicity of serovar 4b strains and will lead to new insights into the pathogenesis and immune evasion of Lm.
Collapse
Affiliation(s)
- Zhiting Ling
- Jangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, China
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, The Ministry of Agriculture and Rural Affairs of the People’s Republic of China, Yangzhou University, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, China
- Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonosis, Yangzhou University, Yangzhou, China
| | - Dan Zhao
- Jangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, China
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, The Ministry of Agriculture and Rural Affairs of the People’s Republic of China, Yangzhou University, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, China
- Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonosis, Yangzhou University, Yangzhou, China
| | - Xinyu Xie
- Jangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, China
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, The Ministry of Agriculture and Rural Affairs of the People’s Republic of China, Yangzhou University, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, China
- Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonosis, Yangzhou University, Yangzhou, China
| | - Hao Yao
- Jangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, China
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, The Ministry of Agriculture and Rural Affairs of the People’s Republic of China, Yangzhou University, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, China
- Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonosis, Yangzhou University, Yangzhou, China
| | - Yuting Wang
- Jangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, China
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, The Ministry of Agriculture and Rural Affairs of the People’s Republic of China, Yangzhou University, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, China
- Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonosis, Yangzhou University, Yangzhou, China
| | - Suwei Kong
- Jangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, China
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, The Ministry of Agriculture and Rural Affairs of the People’s Republic of China, Yangzhou University, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, China
- Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonosis, Yangzhou University, Yangzhou, China
| | - Xiang Chen
- Jangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, China
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, The Ministry of Agriculture and Rural Affairs of the People’s Republic of China, Yangzhou University, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, China
- Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonosis, Yangzhou University, Yangzhou, China
| | - Zhiming Pan
- Jangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, China
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, The Ministry of Agriculture and Rural Affairs of the People’s Republic of China, Yangzhou University, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, China
- Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonosis, Yangzhou University, Yangzhou, China
| | - Xin’an Jiao
- Jangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, China
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, The Ministry of Agriculture and Rural Affairs of the People’s Republic of China, Yangzhou University, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, China
- Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonosis, Yangzhou University, Yangzhou, China
- *Correspondence: Xin’an Jiao, ; Yuelan Yin,
| | - Yuelan Yin
- Jangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, China
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, The Ministry of Agriculture and Rural Affairs of the People’s Republic of China, Yangzhou University, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, China
- Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonosis, Yangzhou University, Yangzhou, China
- *Correspondence: Xin’an Jiao, ; Yuelan Yin,
| |
Collapse
|
10
|
Li R, Liang Q, Tian S, Zhang Y, Liu S, Ou Q, Chen Z, Wang C. Hemolysin function of Listeria is related to biofilm formation: transcriptomics analysis. Vet Res 2022; 53:113. [PMID: 36587206 PMCID: PMC9805692 DOI: 10.1186/s13567-022-01124-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 11/07/2022] [Indexed: 01/02/2023] Open
Abstract
Listeriolysin O (LLO) is the main virulence protein of Listeria monocytogenes (LM), that helps LM escape lysosomes. We previously found that the cellular immune response elicited by L.ivanovii (LI) is weaker than that elicited by LM. We speculated that this may be related to the function of ivanolysin O (ILO). Here, we constructed hemolysin gene deletion strain, LIΔilo, and a modified strain, LIΔilo::hly, in which ilo was replaced by hly. Prokaryotic transcriptome sequencing was performed on LI, LIΔilo, and LIΔilo::hly. Transcriptome differences between the three strains were compared, and genes and pathways with significant differences between the three strains were analyzed. Prokaryotic transcriptome sequencing results revealed the relationship of ilo to the ribosome, quorum sensing, and phosphotransferase system (PTS) pathways, etc. LIΔilo exhibited attenuated biofilm formation ability compared to LI. Biofilm formation was significantly recovered or even increased after replenishing hly. After knocking out ilo, the relative expression levels of some virulence genes, including sigB, prfA, actA, smcL, and virR, were up-regulated compared to LI. After replenishing hly, these genes were down-regulated compared to LIΔilo. The trend and degree of such variation were not completely consistent when cultured in media containing only monosaccharides or disaccharides. The results confirmed that hemolysin is related to some important biological properties of Listeria, including biofilm formation and virulence gene expression levels. This is the first comprehensive study on ILO function at the transcriptomic level and the first evidence of a relationship between Listeria hemolysin and biofilm formation.
Collapse
Affiliation(s)
- Ruidan Li
- grid.13291.380000 0001 0807 1581Department of Public Health Laboratory Sciences, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610061 China ,Shen Zhen Biomed Alliance Biotech Group Co., Ltd, Shenzhen, 518057 China
| | - Qian Liang
- grid.13291.380000 0001 0807 1581Department of Public Health Laboratory Sciences, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610061 China ,Shen Zhen Biomed Alliance Biotech Group Co., Ltd, Shenzhen, 518057 China
| | - Sicheng Tian
- grid.13291.380000 0001 0807 1581Department of Public Health Laboratory Sciences, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610061 China
| | - Yunwen Zhang
- grid.13291.380000 0001 0807 1581Department of Public Health Laboratory Sciences, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610061 China
| | - Sijing Liu
- grid.13291.380000 0001 0807 1581Department of Public Health Laboratory Sciences, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610061 China
| | - Qian Ou
- grid.13291.380000 0001 0807 1581Department of Public Health Laboratory Sciences, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610061 China
| | - Zhaobin Chen
- Shen Zhen Biomed Alliance Biotech Group Co., Ltd, Shenzhen, 518057 China
| | - Chuan Wang
- grid.13291.380000 0001 0807 1581Department of Public Health Laboratory Sciences, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610061 China
| |
Collapse
|
11
|
Bagatella S, Tavares-Gomes L, Oevermann A. Listeria monocytogenes at the interface between ruminants and humans: A comparative pathology and pathogenesis review. Vet Pathol 2021; 59:186-210. [PMID: 34856818 DOI: 10.1177/03009858211052659] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The bacterium Listeria monocytogenes (Lm) is widely distributed in the environment as a saprophyte, but may turn into a lethal intracellular pathogen upon ingestion. Invasive infections occur in numerous species worldwide, but most commonly in humans and farmed ruminants, and manifest as distinct forms. Of those, neuroinfection is remarkably threatening due to its high mortality. Lm is widely studied not only as a pathogen but also as an essential model for intracellular infections and host-pathogen interactions. Many aspects of its ecology and pathogenesis, however, remain unclear and are rarely addressed in its natural hosts. This review highlights the heterogeneity and adaptability of Lm by summarizing its association with the environment, farm animals, and disease. It also provides current knowledge on key features of the pathology and (molecular) pathogenesis of various listeriosis forms in naturally susceptible species with a special focus on ruminants and on the neuroinvasive form of the disease. Moreover, knowledge gaps on pathomechanisms of listerial infections and relevant unexplored topics in Lm pathogenesis research are highlighted.
Collapse
Affiliation(s)
- Stefano Bagatella
- Division of Neurological Sciences, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Leticia Tavares-Gomes
- Division of Neurological Sciences, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Anna Oevermann
- Division of Neurological Sciences, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| |
Collapse
|
12
|
Quereda JJ, Morón-García A, Palacios-Gorba C, Dessaux C, García-del Portillo F, Pucciarelli MG, Ortega AD. Pathogenicity and virulence of Listeria monocytogenes: A trip from environmental to medical microbiology. Virulence 2021; 12:2509-2545. [PMID: 34612177 PMCID: PMC8496543 DOI: 10.1080/21505594.2021.1975526] [Citation(s) in RCA: 78] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 08/23/2021] [Accepted: 08/25/2021] [Indexed: 01/02/2023] Open
Abstract
Listeria monocytogenes is a saprophytic gram-positive bacterium, and an opportunistic foodborne pathogen that can produce listeriosis in humans and animals. It has evolved an exceptional ability to adapt to stress conditions encountered in different environments, resulting in a ubiquitous distribution. Because some food preservation methods and disinfection protocols in food-processing environments cannot efficiently prevent contaminations, L. monocytogenes constitutes a threat to human health and a challenge to food safety. In the host, Listeria colonizes the gastrointestinal tract, crosses the intestinal barrier, and disseminates through the blood to target organs. In immunocompromised individuals, the elderly, and pregnant women, the pathogen can cross the blood-brain and placental barriers, leading to neurolisteriosis and materno-fetal listeriosis. Molecular and cell biology studies of infection have proven L. monocytogenes to be a versatile pathogen that deploys unique strategies to invade different cell types, survive and move inside the eukaryotic host cell, and spread from cell to cell. Here, we present the multifaceted Listeria life cycle from a comprehensive perspective. We discuss genetic features of pathogenic Listeria species, analyze factors involved in food contamination, and review bacterial strategies to tolerate stresses encountered both during food processing and along the host's gastrointestinal tract. Then we dissect host-pathogen interactions underlying listerial pathogenesis in mammals from a cell biology and systemic point of view. Finally, we summarize the epidemiology, pathophysiology, and clinical features of listeriosis in humans and animals. This work aims to gather information from different fields crucial for a comprehensive understanding of the pathogenesis of L. monocytogenes.
Collapse
Affiliation(s)
- Juan J. Quereda
- Departamento de Producción y Sanidad Animal, Salud Pública Veterinaria y Ciencia y Tecnología de los Alimentos, Facultad de Veterinaria, Universidad Cardenal Herrera-CEU, CEU Universities. Valencia, Spain
| | - Alvaro Morón-García
- Departamento de Biología Celular. Facultad de Ciencias Biológicas, Universidad Complutense de Madrid. Madrid, Spain
| | - Carla Palacios-Gorba
- Departamento de Producción y Sanidad Animal, Salud Pública Veterinaria y Ciencia y Tecnología de los Alimentos, Facultad de Veterinaria, Universidad Cardenal Herrera-CEU, CEU Universities. Valencia, Spain
| | - Charlotte Dessaux
- Departamento de Biotecnología Microbiana, Centro Nacional de Biotecnología (CNB)- Consejo Superior De Investigaciones Científicas (CSIC), Madrid, Spain
| | - Francisco García-del Portillo
- Departamento de Biotecnología Microbiana, Centro Nacional de Biotecnología (CNB)- Consejo Superior De Investigaciones Científicas (CSIC), Madrid, Spain
| | - M. Graciela Pucciarelli
- Departamento de Biotecnología Microbiana, Centro Nacional de Biotecnología (CNB)- Consejo Superior De Investigaciones Científicas (CSIC), Madrid, Spain
- Centro de Biología Molecular ‘Severo Ochoa’. Departamento de Biología Molecular, Facultad de Ciencias, Universidad Autónoma de Madrid. Madrid, Spain
| | - Alvaro D. Ortega
- Departamento de Biología Celular. Facultad de Ciencias Biológicas, Universidad Complutense de Madrid. Madrid, Spain
- Departamento de Biotecnología Microbiana, Centro Nacional de Biotecnología (CNB)- Consejo Superior De Investigaciones Científicas (CSIC), Madrid, Spain
| |
Collapse
|
13
|
Howell LM, Forbes NS. Bacteria-based immune therapies for cancer treatment. Semin Cancer Biol 2021; 86:1163-1178. [PMID: 34547442 DOI: 10.1016/j.semcancer.2021.09.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 09/03/2021] [Accepted: 09/12/2021] [Indexed: 12/23/2022]
Abstract
Engineered bacterial therapies that target the tumor immune landscape offer a new class of cancer immunotherapy. Salmonella enterica and Listeria monocytogenes are two species of bacteria that have been engineered to specifically target tumors and serve as delivery vessels for immunotherapies. Therapeutic bacteria have been engineered to deliver cytokines, gene silencing shRNA, and tumor associated antigens that increase immune activation. Bacterial therapies stimulate both the innate and adaptive immune system, change the immune dynamics of the tumor microenvironment, and offer unique strategies for targeting tumors. Bacteria have innate adjuvant properties, which enable both the delivered molecules and the bacteria themselves to stimulate immune responses. Bacterial immunotherapies that deliver cytokines and tumor-associated antigens have demonstrated clinical efficacy. Harnessing the diverse set of mechanisms that Salmonella and Listeria use to alter the tumor-immune landscape has the potential to generate many new and effective immunotherapies.
Collapse
Affiliation(s)
- Lars M Howell
- Department of Chemical Engineering, University of Massachusetts, Amherst, United States
| | - Neil S Forbes
- Department of Chemical Engineering, University of Massachusetts, Amherst, United States.
| |
Collapse
|
14
|
Pombinho R, Pinheiro J, Resende M, Meireles D, Jalkanen S, Sousa S, Cabanes D. Stabilin-1 plays a protective role against Listeria monocytogenes infection through the regulation of cytokine and chemokine production and immune cell recruitment. Virulence 2021; 12:2088-2103. [PMID: 34374322 PMCID: PMC8366540 DOI: 10.1080/21505594.2021.1958606] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Scavenger receptors are part of a complex surveillance system expressed by host cells to efficiently orchestrate innate immune response against bacterial infections. Stabilin-1 (STAB-1) is a scavenger receptor involved in cell trafficking, inflammation, and cancer; however, its role in infection remains to be elucidated. Listeria monocytogenes (Lm) is a major intracellular human food-borne pathogen causing severe infections in susceptible hosts. Using a mouse model of infection, we demonstrate here that STAB-1 controls Lm-induced cytokine and chemokine production and immune cell accumulation in Lm-infected organs. We show that STAB-1 also regulates the recruitment of myeloid cells in response to Lm infection and contributes to clear circulating bacteria. In addition, whereas STAB-1 appears to promote bacterial uptake by macrophages, infection by pathogenic Listeria induces the down regulation of STAB-1 expression and its delocalization from the host cell membrane. We propose STAB-1 as a new SR involved in the control of Lm infection through the regulation of host defense mechanisms, a process that would be targeted by bacterial virulence factors to promote infection.
Collapse
Affiliation(s)
- Rita Pombinho
- Instituto De Investigação E Inovação Em Saúde - i3S, Universidade Do Porto, Porto, Portugal.,Group of Molecular Microbiology, Instituto De Biologia Molecular E Celular - IBMC, Porto, Portugal
| | - Jorge Pinheiro
- Instituto De Investigação E Inovação Em Saúde - i3S, Universidade Do Porto, Porto, Portugal.,Group of Molecular Microbiology, Instituto De Biologia Molecular E Celular - IBMC, Porto, Portugal
| | - Mariana Resende
- Instituto De Investigação E Inovação Em Saúde - i3S, Universidade Do Porto, Porto, Portugal.,Microbiology and Immunology of Infection, Instituto De Biologia Molecular E Celular - IBMC, Porto, Portugal
| | - Diana Meireles
- Instituto De Investigação E Inovação Em Saúde - i3S, Universidade Do Porto, Porto, Portugal.,Group of Molecular Microbiology, Instituto De Biologia Molecular E Celular - IBMC, Porto, Portugal
| | - Sirpa Jalkanen
- MediCity Research Laboratory and Department of Medical Microbiology and Immunology, University of Turku, Turku, Finland
| | - Sandra Sousa
- Instituto De Investigação E Inovação Em Saúde - i3S, Universidade Do Porto, Porto, Portugal.,Group of Molecular Microbiology, Instituto De Biologia Molecular E Celular - IBMC, Porto, Portugal
| | - Didier Cabanes
- Instituto De Investigação E Inovação Em Saúde - i3S, Universidade Do Porto, Porto, Portugal.,Group of Molecular Microbiology, Instituto De Biologia Molecular E Celular - IBMC, Porto, Portugal
| |
Collapse
|
15
|
Zhang X, Wang R, Luo J, Xia D, Zhou C. Detection of meningoencephalitis caused by Listeria monocytogenes with ischemic stroke-like onset using metagenomics next-generation sequencing: A case report. Medicine (Baltimore) 2021; 100:e26802. [PMID: 34397834 PMCID: PMC8341303 DOI: 10.1097/md.0000000000026802] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 07/12/2021] [Indexed: 01/04/2023] Open
Abstract
RATIONALE Listeria monocytogenes (L. monocytogenes) is a compatible intracellular bacterial pathogen that can invade different mammalian cells and reach the central nervous system (CNS), leading to meningoencephalitis and brain abscesses. In the diagnosis of L. monocytogenes meningoencephalitis (LMM), conventional tests are often reported as negative due to antibiotic therapy or low bacterial content in cerebrospinal fluid. To date, prompt diagnosis and accurate treatment remain a challenge for patients with Listeria infections. PATIENT CONCERNS Here, we report a case of a 64-year-old male diagnosed with LMM by using metagenomics next-generation sequencing (mNGS). DIAGNOSIS LMM was confirmed by mNGS analysis of cerebrospinal fluid. INTERVENTIONS The patient was treated with piperacillin and sensitive antibiotics. OUTCOMES The patient could walk independently about 1 month after admission and was discharged from the hospital. LESSONS This case highlights the value of mNGS in the diagnosis of LMM and emphasizes the inadequate sensitivity of conventional diagnostic methods for Listeria infection.
Collapse
Affiliation(s)
- Xiaobo Zhang
- Department of Neurology, The First People's Hospital of Changde City, Changde, Hunan, China
| | - Ruying Wang
- Department of Neurology, The First People's Hospital of Changde City, Changde, Hunan, China
| | - Jie Luo
- Department of Neurology, The First People's Hospital of Changde City, Changde, Hunan, China
| | - Danni Xia
- Department of Laboratory, The First People's Hospital of Changde City, Changde, Hunan, China
| | - Chaojun Zhou
- Department of Neurology, The First People's Hospital of Changde City, Changde, Hunan, China
| |
Collapse
|
16
|
Muri J, Feng Q, Wolleb H, Shamshiev A, Ebner C, Tortola L, Broz P, Carreira EM, Kopf M. Cyclopentenone Prostaglandins and Structurally Related Oxidized Lipid Species Instigate and Share Distinct Pro- and Anti-inflammatory Pathways. Cell Rep 2021; 30:4399-4417.e7. [PMID: 32234476 DOI: 10.1016/j.celrep.2020.03.019] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 12/11/2019] [Accepted: 03/05/2020] [Indexed: 01/06/2023] Open
Abstract
Oxidized lipids play a critical role in a variety of diseases with two faces: pro- and anti-inflammatory. The molecular mechanisms of this Janus-faced activity remain largely unknown. Here, we have identified that cyclopentenone-containing prostaglandins such as 15d-PGJ2 and structurally related oxidized phospholipid species possess a dual and opposing bioactivity in inflammation, depending on their concentration. Exposure of dendritic cells (DCs)/macrophages to low concentrations of such lipids before Toll-like receptor (TLR) stimulation instigates an anti-inflammatory response mediated by nuclear factor erythroid 2-related factor 2 (Nrf2)-dependent inhibition of nuclear factor κB (NF-κB) activation and downstream targets. By contrast, high concentrations of such lipids upon TLR activation of DCs/macrophages result in inflammatory apoptosis characterized by mitochondrial depolarization and caspase-8-mediated interleukin (IL)-1β maturation independently of Nrf2 and the classical inflammasome pathway. These results uncover unexpected pro- and anti-inflammatory activities of physiologically relevant lipid species generated by enzymatic and non-enzymatic oxidation dependent on their concentration, a phenomenon known as hormesis.
Collapse
Affiliation(s)
- Jonathan Muri
- Institute of Molecular Health Sciences, ETH Zürich, 8093 Zürich, Switzerland
| | - Qian Feng
- Institute of Molecular Health Sciences, ETH Zürich, 8093 Zürich, Switzerland
| | - Helene Wolleb
- Laboratory of Organic Chemistry, ETH Zürich, 8093 Zürich, Switzerland
| | - Abdijapar Shamshiev
- Institute of Molecular Health Sciences, ETH Zürich, 8093 Zürich, Switzerland
| | - Christian Ebner
- Laboratory of Organic Chemistry, ETH Zürich, 8093 Zürich, Switzerland
| | - Luigi Tortola
- Institute of Molecular Health Sciences, ETH Zürich, 8093 Zürich, Switzerland
| | - Petr Broz
- Department of Biochemistry, University of Lausanne, 1066 Epalinges, Switzerland
| | - Erick M Carreira
- Laboratory of Organic Chemistry, ETH Zürich, 8093 Zürich, Switzerland
| | - Manfred Kopf
- Institute of Molecular Health Sciences, ETH Zürich, 8093 Zürich, Switzerland.
| |
Collapse
|
17
|
Antilisterial Potential of Lactic Acid Bacteria in Eliminating Listeria monocytogenes in Host and Ready-to-Eat Food Application. MICROBIOLOGY RESEARCH 2021. [DOI: 10.3390/microbiolres12010017] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Listeriosis is a severe food borne disease with a mortality rate of up to 30% caused by pathogenic Listeria monocytogenes via the production of several virulence factors including listeriolysin O (LLO), transcriptional activator (PrfA), actin (Act), internalin (Int), etc. It is a foodborne disease predominantly causing infections through consumption of contaminated food and is often associated with ready-to-eat food (RTE) and dairy products. Common medication for listeriosis such as antibiotics might cause an eagle effect and antibiotic resistance if it is overused. Therefore, exploration of the use of lactic acid bacteria (LAB) with probiotic characteristics and multiple antimicrobial properties is increasingly getting attention for their capability to treat listeriosis, vaccine development, and hurdle technologies. The antilisterial gene, a gene coding to produce antimicrobial peptide (AMP), one of the inhibitory substances found in LAB, is one of the potential key factors in listeriosis treatment, coupled with the vast array of functions and strategies; this review summarizes the various strategies by LAB against L. monocytogenes and the prospect in development of a ‘generally regarded as safe’ LAB for treatment of listeriosis.
Collapse
|
18
|
Besic V, Habibolahi F, Noël B, Rupp S, Genovesio A, Lebreton A. Coordination of transcriptional and translational regulations in human epithelial cells infected by Listeria monocytogenes. RNA Biol 2020; 17:1492-1507. [PMID: 32584699 PMCID: PMC7549700 DOI: 10.1080/15476286.2020.1777380] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 05/04/2020] [Accepted: 05/28/2020] [Indexed: 12/12/2022] Open
Abstract
The invasion of mammalian cells by intracellular bacterial pathogens reshuffles their gene expression and functions; however, we lack dynamic insight into the distinct control levels that shape the host response. Here, we have addressed the respective contribution of transcriptional and translational regulations during a time-course of infection of human intestinal epithelial cells by an epidemic strain of Listeria monocytogenes, using transcriptome analysis paralleled with ribosome profiling. Upregulations were dominated by early transcriptional activation of pro-inflammatory genes, whereas translation inhibition appeared as the major driver of downregulations. Instead of a widespread but transient shutoff, translation inhibition affected specifically and durably transcripts encoding components of the translation machinery harbouring a 5'-terminal oligopyrimidine motif. Pre-silencing the most repressed target gene (PABPC1) slowed down the intracellular multiplication of Listeria monocytogenes, suggesting that the infected host cell can benefit from the repression of genes involved in protein synthesis and thereby better control infection.
Collapse
Affiliation(s)
- Vinko Besic
- Bacterial Infection & RNA Destiny Group, Institut de biologie de l’ENS (IBENS), École normale supérieure, CNRS, INSERM, Université PSL, Paris, France
| | - Fatemeh Habibolahi
- Bacterial Infection & RNA Destiny Group, Institut de biologie de l’ENS (IBENS), École normale supérieure, CNRS, INSERM, Université PSL, Paris, France
- Computational Biology and Bioinformatics Group, Institut de biologie de l’ENS (IBENS), École normale supérieure, CNRS, INSERM, Université PSL, Paris, France
| | - Benoît Noël
- Bacterial Infection & RNA Destiny Group, Institut de biologie de l’ENS (IBENS), École normale supérieure, CNRS, INSERM, Université PSL, Paris, France
- Computational Biology and Bioinformatics Group, Institut de biologie de l’ENS (IBENS), École normale supérieure, CNRS, INSERM, Université PSL, Paris, France
| | - Sebastian Rupp
- Bacterial Infection & RNA Destiny Group, Institut de biologie de l’ENS (IBENS), École normale supérieure, CNRS, INSERM, Université PSL, Paris, France
| | - Auguste Genovesio
- Computational Biology and Bioinformatics Group, Institut de biologie de l’ENS (IBENS), École normale supérieure, CNRS, INSERM, Université PSL, Paris, France
| | - Alice Lebreton
- Bacterial Infection & RNA Destiny Group, Institut de biologie de l’ENS (IBENS), École normale supérieure, CNRS, INSERM, Université PSL, Paris, France
- INRAE, IBENS, Paris, France
| |
Collapse
|
19
|
Pathogenic Biohacking: Induction, Modulation and Subversion of Host Transcriptional Responses by Listeria monocytogenes. Toxins (Basel) 2020; 12:toxins12050294. [PMID: 32380645 PMCID: PMC7290974 DOI: 10.3390/toxins12050294] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 05/01/2020] [Accepted: 05/03/2020] [Indexed: 12/23/2022] Open
Abstract
During infection, the foodborne bacterial pathogen Listeria monocytogenes dynamically influences the gene expression profile of host cells. Infection-induced transcriptional changes are a typical feature of the host-response to bacteria and contribute to the activation of protective genes such as inflammatory cytokines. However, by using specialized virulence factors, bacterial pathogens can target signaling pathways, transcription factors, and epigenetic mechanisms to alter host gene expression, thereby reprogramming the response to infection. Therefore, the transcriptional profile that is established in the host is delicately balanced between antibacterial responses and pathogenesis, where any change in host gene expression might significantly influence the outcome of infection. In this review, we discuss the known transcriptional and epigenetic processes that are engaged during Listeria monocytogenes infection, the virulence factors that can remodel them, and the impact these processes have on the outcome of infection.
Collapse
|
20
|
Roberts BN, Chakravarty D, Gardner J, Ricke SC, Donaldson JR. Listeria monocytogenes Response to Anaerobic Environments. Pathogens 2020; 9:pathogens9030210. [PMID: 32178387 PMCID: PMC7157565 DOI: 10.3390/pathogens9030210] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 03/09/2020] [Accepted: 03/10/2020] [Indexed: 12/19/2022] Open
Abstract
Listeria monocytogenes is a Gram-positive facultative anaerobic bacterium that is responsible for the disease, listeriosis. It is particularly lethal in pregnant women, the fetus, the elderly and the immunocompromised. The pathogen survives and replicates over a wide range of temperatures (4 to 42 °C), pH, salt and oxygen concentrations. Because it can withstand various environments, L. monocytogenes is a major concern in food processing industries, especially in dairy products and ready-to-eat fruits, vegetables and deli meats. The environment in which the pathogen is exposed can influence the expression of virulence genes. For instance, studies have shown that variations in oxygen availability can impact resistance to stressors. Further investigation is needed to understand the essential genes required for the growth of L. monocytogenes in anaerobic conditions. Therefore, the purpose of this review is to highlight the data on L. monocytogenes under known environmental stresses in anaerobic environments and to focus on gaps in knowledge that may be advantageous to study in order to better understand the pathogenicity of the bacterium.
Collapse
Affiliation(s)
- Brandy N. Roberts
- Department of Biological Sciences, Mississippi State University, Mississippi State, MS 39762, USA;
| | - Damayanti Chakravarty
- Cell and Molecular Biology, The University of Southern Mississippi, Hattiesburg, MS 39406, USA; (D.C.); (J.C.G.III)
| | - J.C. Gardner
- Cell and Molecular Biology, The University of Southern Mississippi, Hattiesburg, MS 39406, USA; (D.C.); (J.C.G.III)
| | - Steven C. Ricke
- Center for Food Safety, Department of Food Science, University of Arkansas, Fayetteville, AR 72704, USA;
| | - Janet R. Donaldson
- Cell and Molecular Biology, The University of Southern Mississippi, Hattiesburg, MS 39406, USA; (D.C.); (J.C.G.III)
- Correspondence: ; Tel.: +1-601-206-6795
| |
Collapse
|
21
|
Wu Z, Cheng Y, Yang Y, Gao Y, Sun X, Wang L, Sun Q, Zhang J, Xu X. In vitro and in vivo anti-Listeria effect of Succinoglycan Riclin through regulating MAPK/IL-6 axis and metabolic profiling. Int J Biol Macromol 2020; 150:802-813. [PMID: 32057883 DOI: 10.1016/j.ijbiomac.2020.02.088] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 01/30/2020] [Accepted: 02/10/2020] [Indexed: 12/31/2022]
Abstract
Infectious diseases such as Listeria monocytogenes infection pose a great threat to the health of human beings and the development of livestock and poultry farming. Currently the treatment of Listeria infection mainly relies on antibiotics, which may result in excessive antibiotic residues in livestock and poultry products, as well as causing an increase in the occurrence of zoonotic diseases. Here, we demonstrate that Succinoglycan Riclin promoted the clearance of Listeria in the in vitro and in vivo infection model. The expression and secretion of inflammatory cytokines including IL-6 and IL-1β were significantly increased after Riclin treatment upon infection. The protective effect of Riclin was mainly through activating MAPK/IL-6 axis. HO-1/IL-1β signaling pathway was less important in this process. Moreover, Riclin caused significant metabolic changes including pathways involved in glycolysis, protein synthesis and oxidative stress during Listeria infection. These results suggest a potential use of Succinoglycan Riclin as non-antibiotic preventive and therapeutic anti-microbial agent in livestock and poultry farming and human diseases.
Collapse
Affiliation(s)
- Zhuhui Wu
- Center for Molecular Metabolism, Nanjing University of Science & Technology, Nanjing 210094, PR China
| | - Yingying Cheng
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, PR China
| | - Yunxia Yang
- Center for Molecular Metabolism, Nanjing University of Science & Technology, Nanjing 210094, PR China
| | - Yan Gao
- Center for Molecular Metabolism, Nanjing University of Science & Technology, Nanjing 210094, PR China
| | - Xiaqing Sun
- Center for Molecular Metabolism, Nanjing University of Science & Technology, Nanjing 210094, PR China
| | - Lei Wang
- Center for Molecular Metabolism, Nanjing University of Science & Technology, Nanjing 210094, PR China
| | - Qi Sun
- Center for Molecular Metabolism, Nanjing University of Science & Technology, Nanjing 210094, PR China
| | - Jianfa Zhang
- Center for Molecular Metabolism, Nanjing University of Science & Technology, Nanjing 210094, PR China
| | - Xi Xu
- Center for Molecular Metabolism, Nanjing University of Science & Technology, Nanjing 210094, PR China.
| |
Collapse
|
22
|
Liang W, Chen K, Gong W, Yoshimura T, Le Y, Wang Y, Wang JM. The Contribution of Chemoattractant GPCRs, Formylpeptide Receptors, to Inflammation and Cancer. Front Endocrinol (Lausanne) 2020; 11:17. [PMID: 32038501 PMCID: PMC6993212 DOI: 10.3389/fendo.2020.00017] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 01/09/2020] [Indexed: 12/14/2022] Open
Abstract
A hallmark of inflammatory responses is leukocyte mobilization, which is mediated by pathogen and host released chemotactic factors that activate Gi-protein-coupled seven-transmembrane receptors (GPCRs) on host cell surface. Formylpeptide receptors (FPRs, Fprs in mice) are members of the chemoattractant GPCR family, shown to be critical in myeloid cell trafficking during infection, inflammation, immune responses, and cancer progression. Accumulating evidence demonstrates that both human FPRs and murine Fprs are involved in a number of patho-physiological processes because of their expression on a wide variety of cell types in addition to myeloid cells. The unique capacity of FPRs (Fprs) to interact with numerous structurally unrelated chemotactic ligands enables these receptors to participate in orchestrated disease initiation, progression, and resolution. One murine Fpr member, Fpr2, and its endogenous agonist peptide, Cathelicidin-related antimicrobial peptide (CRAMP), have been demonstrated as key mediators of colon mucosal homeostasis and protection from inflammation and associated tumorigenesis. Recent availability of genetically engineered mouse models greatly expanded the understanding of the role of FPRs (Fprs) in pathophysiology that places these molecules in the list of potential targets for therapeutic intervention of diseases.
Collapse
Affiliation(s)
- Weiwei Liang
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Peking University, Beijing, China
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, United States
| | - Keqiang Chen
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, United States
| | - Wanghua Gong
- Basic Research Program, Leidos Biomedical Research, Inc., Frederick, MD, United States
| | - Teizo Yoshimura
- Department of Pathology and Experimental Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Yingying Le
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
| | - Ying Wang
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Peking University, Beijing, China
| | - Ji Ming Wang
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, United States
| |
Collapse
|
23
|
Intranasal vaccination with Listeria ivanovii as vector of Mycobacterium tuberculosis antigens promotes specific lung-localized cellular and humoral immune responses. Sci Rep 2020; 10:302. [PMID: 31942003 PMCID: PMC6962167 DOI: 10.1038/s41598-019-57245-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 12/12/2019] [Indexed: 11/08/2022] Open
Abstract
We have previously demonstrated that a recombinant Listeria ivanovii (LI) strain expressing the ESAT-6 or Ag85C protein of Mycobacterium tuberculosis (Mtb) as a tuberculosis (TB) vaccine candidates induced antigen-specific cellular immune responses after intravenous immunization of mice. However, whether such recombinant strains could induce desired immune responses in the lung, where TB infection occurs, is not clear. In this paper, C57BL/6 J mice were intranasally vaccinated with attenuated LIΔactAplcB-Rv3875 (Δ refers to gene deletion in the bacterial genome) or LIΔactAplcB-Rv0129c, the two vaccine candidates that utilize LI as an antigen delivery vector. Bacterial load in the target organs, histological changes in the infected organs, the percentage of specific cytokine-secreting T cells in the lung and spleen, IgG levels in the serum and secretory IgA (SIgA) levles in bronchoalveolar lavage (BAL) fluid were determined at specific days post inoculation (dpi). The results showed that both strains were mainly confined to the lung and were eliminated at 10 dpi. The histological damage caused by the infection in the lung was slight and recovered by day 5. Intranasal vaccination of the mice twice at an interval of 4 weeks notably elicited TB antigen-specific CD4+ and CD8+ T cell responses in the lung and SIgA secretion in the pulmonary mucosa, and significantly enhanced the percentage of double-functional CD8+ T cells (IFN-γ+ TNF-α+ CD8+). To our knowledge, this is the first report regarding the used of LI vector vaccines to induce promising lung-localized cellular and humoral immune responses by intranasal vaccination. These data suggest that LI could be a novel and promising live vector to construct an intranasal vaccine against respiratory diseases.
Collapse
|
24
|
Inducible degradation of lncRNA Sros1 promotes IFN-γ-mediated activation of innate immune responses by stabilizing Stat1 mRNA. Nat Immunol 2019; 20:1621-1630. [PMID: 31740800 DOI: 10.1038/s41590-019-0542-7] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 10/08/2019] [Indexed: 02/06/2023]
Abstract
Interferon-γ (IFN-γ) is essential for the innate immune response to intracellular bacteria. Noncoding RNAs and RNA-binding proteins (RBPs) need to be further considered in studies of regulation of the IFN-γ-activated signaling pathway in macrophages. In the present study, we found that the microRNA miR-1 promoted IFN-γ-mediated clearance of Listeria monocytogenes in macrophages by indirectly stabilizing the Stat1 messenger RNA through the degradation of the cytoplasmic long noncoding RNA Sros1. Inducible degradation or genetic loss of Sros1 led to enhanced IFN-γ-dependent activation of the innate immune response. Mechanistically, Sros1 blocked the binding of Stat1 mRNA to the RBP CAPRIN1, which stabilized the Stat1 mRNA and, consequently, promoted IFN-γ-STAT1-mediated innate immunity. These observations shed light on the complex RNA-RNA regulatory networks involved in cytokine-initiated innate responses in host-pathogen interactions.
Collapse
|
25
|
Mathipa MG, Thantsha MS, Bhunia AK. Lactobacillus casei expressing Internalins A and B reduces Listeria monocytogenes interaction with Caco-2 cells in vitro. Microb Biotechnol 2019; 12:715-729. [PMID: 30989823 PMCID: PMC6559204 DOI: 10.1111/1751-7915.13407] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 03/20/2019] [Accepted: 03/22/2019] [Indexed: 12/28/2022] Open
Abstract
Listeria monocytogenes has been implicated in a number of outbreaks including the recent largest outbreak in South Africa. Current methods for prevention of foodborne L. monocytogenes infection are inadequate, thus raising a need for an alternative strategy. Probiotic bioengineering is considered a prevailing approach to enhance the efficacy of probiotics for targeted control of pathogens. Here, the ability of Lactobacillus casei expressing the L. monocytogenes invasion proteins Internalins A and B (inlAB) to prevent infection was investigated. The inlAB operon was cloned and surface‐expressed on L. casei resulting in a recombinant strain, LbcInlAB, and subsequently, its ability to inhibit adhesion, invasion and translocation of L. monocytogenes through enterocyte‐like Caco‐2 cells was examined. Cell surface expression of InlAB on the LbcInlAB was confirmed by Western blotting and immunofluorescence staining. The LbcInlAB strain showed significantly higher (P < 0.0001) adherence, invasion and translocation of Caco‐2 cells than the wild‐type L. casei strain (LbcWT), as well as reduced L. monocytogenes adhesion, invasion and transcellular passage through the cell monolayer than LbcWT. Furthermore, pre‐exposure of Caco‐2 cells to LbcInlAB significantly reduced L. monocytogenes‐induced cell cytotoxicity and epithelial barrier dysfunction. These results suggest that InlAB‐expressing L. casei could be a potential practical approach for prevention of listeriosis.
Collapse
Affiliation(s)
- Moloko G Mathipa
- Department of Biochemistry, Genetics, and Microbiology, University of Pretoria, Pretoria, South Africa.,Molecular Food Microbiology Laboratory, Department of Food Science, Purdue University, West Lafayette, IN, USA
| | - Mapitsi S Thantsha
- Department of Biochemistry, Genetics, and Microbiology, University of Pretoria, Pretoria, South Africa
| | - Arun K Bhunia
- Molecular Food Microbiology Laboratory, Department of Food Science, Purdue University, West Lafayette, IN, USA.,Department of Comparative Pathobiology, Purdue University, West Lafayette, IN, USA.,Purdue Institute of Inflammation, Immunology, and Infectious Disease, Purdue University, West Lafayette, IN, USA
| |
Collapse
|
26
|
Drolia R, Bhunia AK. Crossing the Intestinal Barrier via Listeria Adhesion Protein and Internalin A. Trends Microbiol 2019; 27:408-425. [PMID: 30661918 DOI: 10.1016/j.tim.2018.12.007] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 11/11/2018] [Accepted: 12/14/2018] [Indexed: 12/24/2022]
Abstract
The intestinal epithelial cell lining provides the first line of defense, yet foodborne pathogens such as Listeria monocytogenes can overcome this barrier; however, the underlying mechanism is not well understood. Though the host M cells in Peyer's patch and the bacterial invasion protein internalin A (InlA) are involved, L. monocytogenes can cross the gut barrier in their absence. The interaction of Listeria adhesion protein (LAP) with the host cell receptor (heat shock protein 60) disrupts the epithelial barrier, promoting bacterial translocation. InlA aids L. monocytogenes transcytosis via interaction with the E-cadherin receptor, which is facilitated by epithelial cell extrusion and goblet cell exocytosis; however, LAP-induced cell junction opening may be an alternative bacterial strategy for InlA access to E-cadherin and its translocation. Here, we summarize the strategies that L. monocytogenes employs to circumvent the intestinal epithelial barrier and compare and contrast these strategies with other enteric bacterial pathogens. Additionally, we provide implications of recent findings for food safety regulations.
Collapse
Affiliation(s)
- Rishi Drolia
- Molecular Food Microbiology Laboratory, Department of Food Science, Purdue University, West Lafayette, IN 47907, USA
| | - Arun K Bhunia
- Molecular Food Microbiology Laboratory, Department of Food Science, Purdue University, West Lafayette, IN 47907, USA; Department of Comparative Pathobiology, Purdue University, West Lafayette, IN 47907, USA; Purdue Institute of Inflammation, Immunology and Infectious Disease, Purdue University, West Lafayette, IN 47907, USA.
| |
Collapse
|
27
|
|
28
|
Yao H, Kang M, Wang Y, Feng Y, Kong S, Cai X, Ling Z, Chen S, Jiao X, Yin Y. An essential role for hfq involved in biofilm formation and virulence in serotype 4b Listeria monocytogenes. Microbiol Res 2018; 215:148-154. [DOI: 10.1016/j.micres.2018.07.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 06/22/2018] [Accepted: 07/07/2018] [Indexed: 11/28/2022]
|
29
|
Weiß E, Kretschmer D. Formyl-Peptide Receptors in Infection, Inflammation, and Cancer. Trends Immunol 2018; 39:815-829. [PMID: 30195466 DOI: 10.1016/j.it.2018.08.005] [Citation(s) in RCA: 121] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 08/02/2018] [Accepted: 08/12/2018] [Indexed: 12/15/2022]
Abstract
Formyl-peptide receptors (FPRs) recognize bacterial and mitochondrial formylated peptides as well as endogenous non-formylated peptides and even lipids. FPRs are expressed on various host cell types but most strongly on neutrophils and macrophages. After the discovery of FPRs on leukocytes, it was assumed that these receptors predominantly govern a proinflammatory response resulting in chemotaxis, degranulation, and oxidative burst during infection. However, it is clear that the activation of FPRs has more complex consequences and can also promote the resolution of inflammation. Recent studies have highlighted associations between FPR function and inflammatory conditions, including inflammatory disorders, cancer, and infection. In this review we discuss these recent findings.
Collapse
Affiliation(s)
- Elisabeth Weiß
- Infection Biology, Interfaculty Institute for Microbiology and Infection Medicine Tübingen (IMIT), University of Tübingen, Auf der Morgenstelle 28, 72076 Tübingen, Germany
| | - Dorothee Kretschmer
- Infection Biology, Interfaculty Institute for Microbiology and Infection Medicine Tübingen (IMIT), University of Tübingen, Auf der Morgenstelle 28, 72076 Tübingen, Germany.
| |
Collapse
|
30
|
Chen G, Wu M, Liu W, Xie M, Zhang W, Fan E, Liu Q. Reactive oxygen species inhibits Listeria monocytogenes invasion into HepG2 epithelial cells. Food Sci Nutr 2018; 6:1501-1507. [PMID: 30258592 PMCID: PMC6145247 DOI: 10.1002/fsn3.615] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 01/27/2018] [Accepted: 01/30/2018] [Indexed: 11/10/2022] Open
Abstract
Listeria monocytogenes (Lm) can colonize human gastrointestinal tract and subsequently cross the intestinal barrier. Reactive oxygen species (ROS) are produced by NADPH oxidase. However, the role of ROS in bacterial invasion remains to be less understood. Herein, we investigated the impact of ROS on Lm invasion to HepG2 using NADPH oxidase inhibitor, diphenyleneiodonium chloride (DPI), as well as the ROS scavenger, N-acetyl cysteine (NAC). Our results showed that inhibiting ROS increased the invasive capability of Lm. Moreover, after Lm infection, inflammatory cytokines such as tumor necrosis factor alpha (TNF-α) and interleukin 1beta (IL-1β) in HepG2 were significantly upregulated. However, after inhibiting ROS, the expression levels of TNF-α and IL-1β were downregulated, indicating a failure of host cells to activate the immune mechanism. Taken together, ROS in Lm might be as a signal for host cells to sense Lm invasion and then stimulate cells to activate the immune mechanism.
Collapse
Affiliation(s)
- Guo‐wei Chen
- School of Medical Instrument and Food EngineeringUniversity of Shanghai for Science and TechnologyShanghaiChina
| | - Man Wu
- School of Medical Instrument and Food EngineeringUniversity of Shanghai for Science and TechnologyShanghaiChina
| | - Wu‐kang Liu
- School of Medical Instrument and Food EngineeringUniversity of Shanghai for Science and TechnologyShanghaiChina
| | - Man‐man Xie
- School of Medical Instrument and Food EngineeringUniversity of Shanghai for Science and TechnologyShanghaiChina
| | - Wei‐sheng Zhang
- Anorectal Department of Gansu Provincial HospitalLanzhouChina
| | - En‐guo Fan
- School of Medical Instrument and Food EngineeringUniversity of Shanghai for Science and TechnologyShanghaiChina
- Institute of Biochemistry and Molecular BiologyZBMZUniversity of FreiburgFreiburgGermany
| | - Qing Liu
- School of Medical Instrument and Food EngineeringUniversity of Shanghai for Science and TechnologyShanghaiChina
| |
Collapse
|
31
|
Wang G, Lin A, Han Q, Zhao H, Tian Z, Zhang J. IFN-γ protects from apoptotic neutrophil-mediated tissue injury during acute Listeria monocytogenes infection. Eur J Immunol 2018; 48:1470-1480. [PMID: 29935120 DOI: 10.1002/eji.201847491] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 05/24/2018] [Accepted: 06/08/2018] [Indexed: 02/07/2023]
Abstract
Listeria monocytogenes (LM) is a foodborne Gram-positive intracellular pathogen that can cause listeriosis in humans and animals. Although phagocytes are known to be involved in the response to this infection, the role of neutrophils is not entirely clear. Here, we have demonstrated that soon after LM infection, a large number of IFN-γ-producing neutrophils quickly accumulated in the spleen, blood, and peritoneal cavity. Both in vivo and in vitro experiments demonstrated that neutrophils were an important source of IFN-γ. IFN-γ played a critical protective role against acute LM infection, as demonstrated by the poor survival of Ifng-/- mice. Moreover, IFN-γ promoted bacterial clearance by the neutrophils, thereby inhibiting LM-induced neutrophil apoptosis and spleen damage. In addition to this, IFN-γ could effectively drive macrophage-mediated phagocytosis of apoptotic neutrophils, which was accompanied with TGF-β secretion and was involved in protection against tissue injury. Importantly, by phagocytizing apoptotic neutrophils, macrophages obtained myeloperoxidase, an important bactericidal molecule only produced by neutrophils, which further promoted the antibacterial activity of macrophages. These findings demonstrate that neutrophils are an important source of IFN-γ at the early stage of LM infection, which is characterized by both LM elimination and tissue-protective effects.
Collapse
Affiliation(s)
- Guan Wang
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Ang Lin
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Qiuju Han
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Huajun Zhao
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Zhigang Tian
- School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Jian Zhang
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, Jinan, China
| |
Collapse
|
32
|
Xie M, Ding C, Guo L, Chen G, Zeng H, Liu Q. Evaluation of Caco-2 cells response to Listeria monocytogenes virulence factors by RT-PCR. Microb Pathog 2018; 120:79-84. [PMID: 29715536 DOI: 10.1016/j.micpath.2018.04.059] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 04/16/2018] [Accepted: 04/27/2018] [Indexed: 01/25/2023]
Abstract
Listeria monocytogenes expresses various virulence factors enabling the invasion and multiplying in host cells, and together induces cytokines transcription. In order to explore the relationship between virulence factors of L. monocytogenes wild-type EGD-e and cellular response in human colonic epithelial cell line(Caco-2), we constructed mutant strains with in-frame deletions of critical virulence genes of inlA, inlB, hly, actA and virulence regulatory factor prfA from EGD-e, respectively. Compared with EGD-e, mutant strains showed significantly decreased invasion and apoptosis in Caco-2 cells. However, mutant strains were capable to evoke cytokines transcription of interleukin-8 (IL-8), mononuclear chemoattractant protein-1 (MCP-1), tumor necrosis factor-a (TNF-a), interleukin-1β (IL-1β), interleukin-6 (IL-6) and CXCL-2 production in Caco-2 cells. Interestingly, EGD-e Δhly-infected Caco-2 cells showed a significant decrease of IL-6, IL-8 and MCP-1 transcription compared with EGD-e at 1 h post-infection. Simultaneously, EGD-e ΔinlB-infected cells showed a decrease in IL-6 transcription, while EGD-e ΔactA-infected cells reflected a decrease in MCP-1 transcription. Virulence genes play a role in inflammatory transcription, but the interaction between pathogenic bacteria and the host cells predominates in inflammatory transcription. Overall, the data showed cellular response of Caco-2 cells infected with EGD-e mutant strains.
Collapse
Affiliation(s)
- Manman Xie
- School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai 200093, PR China
| | - Chengchao Ding
- School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai 200093, PR China
| | - Liang Guo
- School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai 200093, PR China
| | - Guowei Chen
- School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai 200093, PR China
| | - Haijuan Zeng
- School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai 200093, PR China
| | - Qing Liu
- School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai 200093, PR China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Shandong 266071, PR China.
| |
Collapse
|
33
|
|
34
|
Chen K, Bao Z, Gong W, Tang P, Yoshimura T, Wang JM. Regulation of inflammation by members of the formyl-peptide receptor family. J Autoimmun 2017; 85:64-77. [PMID: 28689639 PMCID: PMC5705339 DOI: 10.1016/j.jaut.2017.06.012] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 06/29/2017] [Indexed: 12/14/2022]
Abstract
Inflammation is associated with a variety of diseases. The hallmark of inflammation is leukocyte infiltration at disease sites in response to pathogen- or damage-associated chemotactic molecular patterns (PAMPs and MAMPs), which are recognized by a superfamily of seven transmembrane, Gi-protein-coupled receptors (GPCRs) on cell surface. Chemotactic GPCRs are composed of two major subfamilies: the classical GPCRs and chemokine GPCRs. Formyl-peptide receptors (FPRs) belong to the classical chemotactic GPCR subfamily with unique properties that are increasingly appreciated for their expression on diverse host cell types and the capacity to interact with a plethora of chemotactic PAMPs and MAMPs. Three FPRs have been identified in human: FPR1-FPR3, with putative corresponding mouse counterparts. FPR expression was initially described in myeloid cells but subsequently in many non-hematopoietic cells including cancer cells. Accumulating evidence demonstrates that FPRs possess multiple functions in addition to controlling inflammation, and participate in the processes of many pathophysiologic conditions. They are not only critical mediators of myeloid cell trafficking, but are also implicated in tissue repair, angiogenesis and protection against inflammation-associated tumorigenesis. A series recent discoveries have greatly expanded the scope of FPRs in host defense which uncovered the essential participation of FPRs in step-wise trafficking of myeloid cells including neutrophils and dendritic cells (DCs) in host responses to bacterial infection, tissue injury and wound healing. Also of great interest is the FPRs are exploited by malignant cancer cells for their growth, invasion and metastasis. In this article, we review the current understanding of FPRs concerning their expression in a vast array of cell types, their involvement in guiding leukocyte trafficking in pathophysiological conditions, and their capacity to promote the differentiation of immune cells, their participation in tumor-associated inflammation and cancer progression. The close association of FPRs with human diseases and cancer indicates their potential as targets for the development of therapeutics.
Collapse
Affiliation(s)
- Keqiang Chen
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, 21702, USA
| | - Zhiyao Bao
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, 21702, USA; Department of Pulmonary & Critical Care Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China
| | - Wanghua Gong
- Basic Research Program, Leidos Biomedical Research, Inc., Frederick, MD, 21702, USA
| | - Peng Tang
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, 21702, USA; Breast and Thyroid Surgery, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Teizo Yoshimura
- Department of Pathology and Experimental Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, 700-8558, Japan
| | - Ji Ming Wang
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, 21702, USA.
| |
Collapse
|
35
|
Kortebi M, Milohanic E, Mitchell G, Péchoux C, Prevost MC, Cossart P, Bierne H. Listeria monocytogenes switches from dissemination to persistence by adopting a vacuolar lifestyle in epithelial cells. PLoS Pathog 2017; 13:e1006734. [PMID: 29190284 PMCID: PMC5708623 DOI: 10.1371/journal.ppat.1006734] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 11/04/2017] [Indexed: 12/26/2022] Open
Abstract
Listeria monocytogenes causes listeriosis, a foodborne disease that poses serious risks to fetuses, newborns and immunocompromised adults. This intracellular bacterial pathogen proliferates in the host cytosol and exploits the host actin polymerization machinery to spread from cell-to-cell and disseminate in the host. Here, we report that during several days of infection in human hepatocytes or trophoblast cells, L. monocytogenes switches from this active motile lifestyle to a stage of persistence in vacuoles. Upon intercellular spread, bacteria gradually stopped producing the actin-nucleating protein ActA and became trapped in lysosome-like vacuoles termed Listeria-Containing Vacuoles (LisCVs). Subpopulations of bacteria resisted degradation in LisCVs and entered a slow/non-replicative state. During the subculture of host cells harboring LisCVs, bacteria showed a capacity to cycle between the vacuolar and the actin-based motility stages. When ActA was absent, such as in ΔactA mutants, vacuolar bacteria parasitized host cells in the so-called “viable but non-culturable” state (VBNC), preventing their detection by conventional colony counting methods. The exposure of infected cells to high doses of gentamicin did not trigger the formation of LisCVs, but selected for vacuolar and VBNC bacteria. Together, these results reveal the ability of L. monocytogenes to enter a persistent state in a subset of epithelial cells, which may favor the asymptomatic carriage of this pathogen, lengthen the incubation period of listeriosis, and promote bacterial survival during antibiotic therapy. L. monocytogenes is a model intracellular pathogen that replicates in the cytoplasm of mammalian cells and disseminate in the host using actin-based motility. Here, we reveal that L. monocytogenes changes its lifestyle and persists in lysosomal vacuoles during long-term infection of human hepatocytes and trophoblast cells. When the virulence factor ActA is not expressed, subpopulations of vacuolar bacteria enter a dormant viable but non-culturable (VBNC) state. This novel facet of the L. monocytogenes intracellular life could contribute to the asymptomatic carriage of this pathogen in epithelial tissues and render it tolerant to antibiotic therapy and undetectable by routine culture techniques.
Collapse
Affiliation(s)
- Mounia Kortebi
- Micalis Institute, Inra, AgroParisTech, Université Paris-Saclay, Equipe Epigénétique et Microbiologie Cellulaire, Jouy-en-Josas, France
| | - Eliane Milohanic
- Micalis Institute, Inra, AgroParisTech, Université Paris-Saclay, Equipe Epigénétique et Microbiologie Cellulaire, Jouy-en-Josas, France
| | - Gabriel Mitchell
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, California, United States of America
| | - Christine Péchoux
- Unité GABI, Inra, AgroParisTech, Université Paris-Saclay, Plate-Forme MIMA2, Jouy-en-Josas, France
| | | | - Pascale Cossart
- Institut Pasteur, Unité des interactions Bactéries-Cellules, Paris, France
- Inserm, U604, Paris, France
- Inra, USC2020, Paris, France
| | - Hélène Bierne
- Micalis Institute, Inra, AgroParisTech, Université Paris-Saclay, Equipe Epigénétique et Microbiologie Cellulaire, Jouy-en-Josas, France
- * E-mail:
| |
Collapse
|
36
|
Alice AF, Kramer G, Bambina S, Baird JR, Bahjat KS, Gough MJ, Crittenden MR. Amplifying IFN-γ Signaling in Dendritic Cells by CD11c-Specific Loss of SOCS1 Increases Innate Immunity to Infection while Decreasing Adaptive Immunity. THE JOURNAL OF IMMUNOLOGY 2017; 200:177-185. [PMID: 29150567 DOI: 10.4049/jimmunol.1700909] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 10/23/2017] [Indexed: 01/23/2023]
Abstract
Although prophylactic vaccines provide protective humoral immunity against infectious agents, vaccines that elicit potent CD8 T cell responses are valuable tools to shape and drive cellular immunity against cancer and intracellular infection. In particular, IFN-γ-polarized cytotoxic CD8 T cell immunity is considered optimal for protective immunity against intracellular Ags. Suppressor of cytokine signaling (SOCS)1 is a cross-functional negative regulator of TLR and cytokine receptor signaling via degradation of the receptor-signaling complex. We hypothesized that loss of SOCS1 in dendritic cells (DCs) would improve T cell responses by accentuating IFN-γ-directed immune responses. We tested this hypothesis using a recombinant Listeria monocytogenes vaccine platform that targets CD11c+ DCs in mice in which SOCS1 is selectively deleted in all CD11c+ cells. Unexpectedly, in mice lacking SOCS1 expression in CD11c+ cells, we observed a decrease in CD8+ T cell response to the L. monocytogenes vaccine. NK cell responses were also decreased in mice lacking SOCS1 expression in CD11c+ cells but did not explain the defect in CD8+ T cell immunity. We found that DCs lacking SOCS1 expression were functional in driving Ag-specific CD8+ T cell expansion in vitro but that this process was defective following infection in vivo. Instead, monocyte-derived innate TNF-α and inducible NO synthase-producing DCs dominated the antibacterial response. Thus, loss of SOCS1 in CD11c+ cells skewed the balance of immune response to infection by increasing innate responses while decreasing Ag-specific adaptive responses to infectious Ags.
Collapse
Affiliation(s)
- Alejandro F Alice
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland, OR 97213; and
| | - Gwen Kramer
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland, OR 97213; and
| | - Shelly Bambina
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland, OR 97213; and
| | - Jason R Baird
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland, OR 97213; and
| | - Keith S Bahjat
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland, OR 97213; and
| | - Michael J Gough
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland, OR 97213; and
| | - Marka R Crittenden
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland, OR 97213; and .,The Oregon Clinic, Portland, OR 97213
| |
Collapse
|
37
|
|
38
|
Shi TY, Zhang YF, Shi XH, Wen XH, Dong X, Meng J, Li HY, Yuan XX, Zheng Y, Lu YW. A rare case of meningoencephalitis by Listeria monocytogenes in systemic lupus erythematosus: case report and review. Clin Rheumatol 2017; 37:271-275. [PMID: 28785856 DOI: 10.1007/s10067-017-3783-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 07/18/2017] [Accepted: 07/31/2017] [Indexed: 02/05/2023]
Abstract
Patients with systemic lupus erythematosus (SLE) have a high risk of infection. Central nervous system infection and neuropsychiatric SLE are both major causes of death. It is vital to distinguish between these two conditions to improve prognosis due to the treatment paradigms required for each condition. Here, we report one case of meningoencephalitis by Listeria monocytogenes (LM) in a patient with SLE who presented with fever and developed headache and altered in consciousness in the hospital. The cerebrospinal fluid culture was positive for LM, and magnetic resonance imaging (MRI) findings were suggestive of ependymitis and periventricular white matter lesions. Amoxicillin/sulbactam, trimethoprim-sulfamethoxazole, and rifampicin were administered for 8 weeks. The patient had a relative good recovery without serious neurological sequelae after a follow-up of nearly 2 years. MRI abnormalities also had obvious resolution.
Collapse
Affiliation(s)
- Tian-Yan Shi
- Department of Rheumatology and Clinical Immunology, Beijing Chao Yang Hospital, Capital Medical University, No. 8 GongTi South Road, ChaoYang District, Beijing, 100020, China
| | - Yong-Feng Zhang
- Department of Rheumatology and Clinical Immunology, Beijing Chao Yang Hospital, Capital Medical University, No. 8 GongTi South Road, ChaoYang District, Beijing, 100020, China
| | - Xu-Hua Shi
- Department of Rheumatology and Clinical Immunology, Beijing Chao Yang Hospital, Capital Medical University, No. 8 GongTi South Road, ChaoYang District, Beijing, 100020, China
| | - Xiao-Hong Wen
- Department of Rheumatology and Clinical Immunology, Beijing Chao Yang Hospital, Capital Medical University, No. 8 GongTi South Road, ChaoYang District, Beijing, 100020, China
| | - Xin Dong
- Department of Rheumatology and Clinical Immunology, Beijing Chao Yang Hospital, Capital Medical University, No. 8 GongTi South Road, ChaoYang District, Beijing, 100020, China
| | - Juan Meng
- Department of Rheumatology and Clinical Immunology, Beijing Chao Yang Hospital, Capital Medical University, No. 8 GongTi South Road, ChaoYang District, Beijing, 100020, China
| | - Hai-Yun Li
- Department of Rheumatology and Clinical Immunology, Beijing Chao Yang Hospital, Capital Medical University, No. 8 GongTi South Road, ChaoYang District, Beijing, 100020, China
| | - Xiao-Xu Yuan
- Department of Rheumatology and Clinical Immunology, Beijing Chao Yang Hospital, Capital Medical University, No. 8 GongTi South Road, ChaoYang District, Beijing, 100020, China
| | - Yi Zheng
- Department of Rheumatology and Clinical Immunology, Beijing Chao Yang Hospital, Capital Medical University, No. 8 GongTi South Road, ChaoYang District, Beijing, 100020, China.
| | - Yue-Wu Lu
- Department of Rheumatology and Clinical Immunology, Beijing Chao Yang Hospital, Capital Medical University, No. 8 GongTi South Road, ChaoYang District, Beijing, 100020, China.
| |
Collapse
|
39
|
Rolhion N, Cossart P. How the study of Listeria monocytogenes has led to new concepts in biology. Future Microbiol 2017; 12:621-638. [PMID: 28604108 DOI: 10.2217/fmb-2016-0221] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The opportunistic intracellular bacterial pathogen Listeria monocytogenes has in 30 years emerged as an exceptional bacterial model system in infection biology. Research on this bacterium has provided considerable insight into how pathogenic bacteria adapt to mammalian hosts, invade eukaryotic cells, move intracellularly, interfere with host cell functions and disseminate within tissues. It also contributed to unveil features of normal host cell pathways and unsuspected functions of previously known cellular proteins. This review provides an updated overview of our knowledge on this pathogen. In many examples, findings on L. monocytogenes provided the basis for new concepts in bacterial regulation, cell biology and infection processes.
Collapse
Affiliation(s)
- Nathalie Rolhion
- Département de Biologie Cellulaire et Infection, Unité des Interactions Bactéries-Cellules, Institut Pasteur, F-75015 Paris, France.,INSERM, U604, F-75015 Paris, France.,INRA, Unité sous-contrat 2020, F-75015 Paris, France
| | - Pascale Cossart
- Département de Biologie Cellulaire et Infection, Unité des Interactions Bactéries-Cellules, Institut Pasteur, F-75015 Paris, France.,INSERM, U604, F-75015 Paris, France.,INRA, Unité sous-contrat 2020, F-75015 Paris, France
| |
Collapse
|
40
|
Zhou X, Zhang B, Cui Y, Chen S, Teng Z, Lu G, Wang J, Deng X. Curcumin Promotes the Clearance of Listeria monocytogenes both In Vitro and In Vivo by Reducing Listeriolysin O Oligomers. Front Immunol 2017; 8:574. [PMID: 28567044 PMCID: PMC5434164 DOI: 10.3389/fimmu.2017.00574] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Accepted: 04/28/2017] [Indexed: 01/28/2023] Open
Abstract
The pore-forming toxin listeriolysin O (LLO), an essential virulence factor that is secreted by Listeria monocytogenes (L. monocytogenes), is responsible for bacterial breaching at the phagosomal membranes and subsequent release into the cytoplasm; it cannot be recognized by the host immune system. The vital role that LLO plays in bacterial pathogenicity and evading host immune clearance makes this virulence a promising target for addressing L. monocytogenes infection. In this study, we hypothesized that curcumin, a polyphenol derived from turmeric that could effectively inhibit LLO pore-forming activity, might be useful in the prevention or treatment of L. monocytogenes infection. Thus, the in vitro protective effects of curcumin against L. monocytogenes infection by targeting LLO were assessed via hemolytic activity assays, cytotoxicity tests, intracellular growth assays, and confocal microscopy. Our results revealed that treating infected macrophages with curcumin can lead to a decrease in LLO-mediated bacteria phagosomal escape and limit the intracellular growth of L. monocytogenes. Moreover, results from animal experiments show that this natural compound effectively increases protection against bacterial infection and helps the host to clear the invading pathogen completely from an animal model, establishing it as a potent antagonist of L. monocytogenes. The results from our molecular modeling and mutational analysis demonstrated that curcumin directly engages with domains 2 and 4 of LLO, thereby decreasing the hemolytic activity of LLO by influencing its oligomerization. Taken together, these results suggest that, as an antitoxin agent, curcumin can be further developed into a novel therapy against L. monocytogenes infections by targeting LLO.
Collapse
Affiliation(s)
- Xuan Zhou
- Center of Infection and Immunity, The First Hospital, Jilin University, Changchun, China.,Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Bing Zhang
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Yumei Cui
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Shuiye Chen
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Zihao Teng
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Gejin Lu
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Jianfeng Wang
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Xuming Deng
- Center of Infection and Immunity, The First Hospital, Jilin University, Changchun, China.,Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| |
Collapse
|
41
|
Valderrama C, Clark A, Urano F, Unanue ER, Carrero JA. Listeria monocytogenes induces an interferon-enhanced activation of the integrated stress response that is detrimental for resolution of infection in mice. Eur J Immunol 2017; 47:830-840. [PMID: 28267207 PMCID: PMC5450196 DOI: 10.1002/eji.201646856] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Revised: 01/31/2017] [Accepted: 02/28/2017] [Indexed: 01/26/2023]
Abstract
Type I interferons (IFNs) induce a detrimental response during Listeria monocytogenes (L. monocytogenes) infection. We were interested in identifying mechanisms linking IFN signaling to negative host responses against L. monocytogenes infection. Herein, we found that infection of myeloid cells with L. monocytogenes led to a coordinated induction of type I IFNs and activation of the integrated stress response (ISR). Infected cells did not induce Xbp1 splicing or BiP upregulation, indicating that the unfolded protein response was not triggered. CHOP (Ddit3) gene expression was upregulated during the ISR activation induced by L. monocytogenes. Myeloid cells deficient in either type I IFN signaling or PKR activation had less upregulation of CHOP following infection. CHOP‐deficient mice showed lower expression of innate immune cytokines and were more resistant than wild‐type counterparts following L. monocytogenes infection. These findings indicate that L. monocytogenes infection induces type I IFNs, which activate the ISR through PKR, which contributes to a detrimental outcome in the infected host.
Collapse
Affiliation(s)
- Carolina Valderrama
- Division of Immunobiology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA.,Department of Microbiology, PhD Biomedical Sciences Program, School of Medicine, Universidad Nacional de Colombia, Bogotá, Colombia
| | - Amy Clark
- Division of Endocrinology, Metabolism and Lipid Research, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Fumihiko Urano
- Division of Endocrinology, Metabolism and Lipid Research, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Emil R Unanue
- Division of Immunobiology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Javier A Carrero
- Division of Immunobiology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
42
|
Listeriolysin S Is a Streptolysin S-Like Virulence Factor That Targets Exclusively Prokaryotic Cells In Vivo. mBio 2017; 8:mBio.00259-17. [PMID: 28377528 PMCID: PMC5380841 DOI: 10.1128/mbio.00259-17] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Streptolysin S (SLS)-like virulence factors from clinically relevant Gram-positive pathogens have been proposed to behave as potent cytotoxins, playing key roles in tissue infection. Listeriolysin S (LLS) is an SLS-like hemolysin/bacteriocin present among Listeria monocytogenes strains responsible for human listeriosis outbreaks. As LLS cytotoxic activity has been associated with virulence, we investigated the LLS-specific contribution to host tissue infection. Surprisingly, we first show that LLS causes only weak red blood cell (RBC) hemolysis in vitro and neither confers resistance to phagocytic killing nor favors survival of L. monocytogenes within the blood cells or in the extracellular space (in the plasma). We reveal that LLS does not elicit specific immune responses, is not cytotoxic for eukaryotic cells, and does not impact cell infection by L. monocytogenes. Using in vitro cell infection systems and a murine intravenous infection model, we actually demonstrate that LLS expression is undetectable during infection of cells and murine inner organs. Importantly, upon intravenous animal inoculation, L. monocytogenes is found in the gastrointestinal system, and only in this environment LLS expression is detected in vivo. Finally, we confirm that LLS production is associated with destruction of target bacteria. Our results demonstrate therefore that LLS does not contribute to L. monocytogenes tissue injury and virulence in inner host organs as previously reported. Moreover, we describe that LlsB, a putative posttranslational modification enzyme encoded in the LLS operon, is necessary for murine inner organ colonization. Overall, we demonstrate that LLS is the first SLS-like virulence factor targeting exclusively prokaryotic cells during in vivo infections. The most severe human listeriosis outbreaks are caused by L. monocytogenes strains harboring listeriolysin S (LLS), previously described as a cytotoxin that plays a critical role in host inner tissue infection. Cytotoxic activities have been proposed as a general mode of action for streptolysin S (SLS)-like toxins, including clostridiolysin S and LLS. We now challenge this dogma by demonstrating that LLS does not contribute to virulence in vivo once the intestinal barrier has been crossed. Importantly, we show that intravenous L. monocytogenes inoculation leads to bacterial translocation to the gastrointestinal system, where LLS is specifically expressed, targeting the host gut microbiota. Our study highlights the heterogeneous modes of action of SLS-like toxins, and we demonstrate for the first time a further level of complexity for SLS-like biosynthetic clusters as we reveal that the putative posttranslational modification enzyme LlsB is actually required for inner organ colonization, independently of the LLS activity.
Collapse
|
43
|
Gagnaire A, Nadel B, Raoult D, Neefjes J, Gorvel JP. Collateral damage: insights into bacterial mechanisms that predispose host cells to cancer. Nat Rev Microbiol 2017; 15:109-128. [DOI: 10.1038/nrmicro.2016.171] [Citation(s) in RCA: 114] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
44
|
Pinheiro J, Reis O, Vieira A, Moura IM, Zanolli Moreno L, Carvalho F, Pucciarelli MG, García-Del Portillo F, Sousa S, Cabanes D. Listeria monocytogenes encodes a functional ESX-1 secretion system whose expression is detrimental to in vivo infection. Virulence 2016; 8:993-1004. [PMID: 27723420 DOI: 10.1080/21505594.2016.1244589] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023] Open
Abstract
Bacterial pathogenicity deeply depends on the ability to secrete virulence factors that bind specific targets on host cells and manipulate host responses. The Gram-positive bacterium Listeria monocytogenes is a human foodborne pathogen that remains a serious public health concern. To transport proteins across its cell envelope, this facultative intracellular pathogen engages a set of specialized secretion systems. Here we show that L. monocytogenes EGDe uses a specialized secretion system, named ESX-1, to secrete EsxA, a homolog of the virulence determinants ESAT-6 and EsxA of Mycobacterium tuberculosis and Staphylococcus aureus, respectively. Our data show that the L. monocytogenes ESX-1 secretion system and its substrates are dispensable for bacterial invasion and intracellular multiplication in eukaryotic cell lines. Surprisingly, we found that the EssC-dependent secretion of EsxA has a detrimental effect on L. monocytogenes in vivo infection.
Collapse
Affiliation(s)
- Jorge Pinheiro
- a Instituto de Investigação e Inovação em Saúde - i3S, Universidade do Porto , Porto , Portugal.,b Group of Molecular Microbiology , Instituto de Biologia Molecular e Celular - IBMC , Porto , Portugal.,c Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto , Porto , Portugal
| | - Olga Reis
- a Instituto de Investigação e Inovação em Saúde - i3S, Universidade do Porto , Porto , Portugal.,b Group of Molecular Microbiology , Instituto de Biologia Molecular e Celular - IBMC , Porto , Portugal.,c Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto , Porto , Portugal
| | - Ana Vieira
- a Instituto de Investigação e Inovação em Saúde - i3S, Universidade do Porto , Porto , Portugal.,b Group of Molecular Microbiology , Instituto de Biologia Molecular e Celular - IBMC , Porto , Portugal
| | - Ines M Moura
- a Instituto de Investigação e Inovação em Saúde - i3S, Universidade do Porto , Porto , Portugal.,b Group of Molecular Microbiology , Instituto de Biologia Molecular e Celular - IBMC , Porto , Portugal
| | - Luisa Zanolli Moreno
- a Instituto de Investigação e Inovação em Saúde - i3S, Universidade do Porto , Porto , Portugal.,b Group of Molecular Microbiology , Instituto de Biologia Molecular e Celular - IBMC , Porto , Portugal.,d Laboratório de Saúde Pública , Faculdade de Saúde Pública, Universidade de São Paulo , São Paulo , Brazil
| | - Filipe Carvalho
- a Instituto de Investigação e Inovação em Saúde - i3S, Universidade do Porto , Porto , Portugal.,b Group of Molecular Microbiology , Instituto de Biologia Molecular e Celular - IBMC , Porto , Portugal.,c Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto , Porto , Portugal
| | - M Graciela Pucciarelli
- e Centro Nacional de Biotecnología-CSIC (CNB-CSIC) , Madrid , Spain.,f Departamento de Biología Molecular , Universidad Autónoma de Madrid, Centro de Biología Molecular "Severo Ochoa" (CBMSO-CSIC) , Madrid , Spain
| | | | - Sandra Sousa
- a Instituto de Investigação e Inovação em Saúde - i3S, Universidade do Porto , Porto , Portugal.,b Group of Molecular Microbiology , Instituto de Biologia Molecular e Celular - IBMC , Porto , Portugal
| | - Didier Cabanes
- a Instituto de Investigação e Inovação em Saúde - i3S, Universidade do Porto , Porto , Portugal.,b Group of Molecular Microbiology , Instituto de Biologia Molecular e Celular - IBMC , Porto , Portugal
| |
Collapse
|
45
|
Dinner S, Kaltschmidt J, Stump-Guthier C, Hetjens S, Ishikawa H, Tenenbaum T, Schroten H, Schwerk C. Mitogen-activated protein kinases are required for effective infection of human choroid plexus epithelial cells by Listeria monocytogenes. Microbes Infect 2016; 19:18-33. [PMID: 27671041 DOI: 10.1016/j.micinf.2016.09.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 08/30/2016] [Accepted: 09/14/2016] [Indexed: 01/01/2023]
Abstract
Listeria monocytogenes, a Gram-positive bacterium, can cause meningitis after invading the human central nervous system. The blood-cerebrospinal fluid barrier (BCSFB), located at the epithelium of the choroid plexus, is a possible entry site for L. monocytogenes into the brain, and in vitro L. monocytogenes invades human choroid plexus epithelial papilloma (HIBCPP) cells. Although host cell signal transduction subsequent to infection by L. monocytogenes has been investigated, the role of mitogen-activated protein kinases (MAPK) is not clarified yet. We show that infection with L. monocytogenes causes activation of the MAPKs Erk1/2 and p38 preferentially when bacteria are added to the physiologically more relevant basolateral side of HIBCPP cells. Deletion of the listerial virulence factors Internalin (InlA) and InlB reduces MAPK activation. Whereas inhibition of either Erk1/2 or p38 signaling significantly attenuates infection of HIBCPP cells with L. monocytogenes, simultaneous inhibition of both MAPK pathways shows an additive effect, and Erk1/2 and p38 are involved in regulation of cytokine and chemokine expression following infection. Blocking of endocytosis with the synthetic dynamin inhibitor dynasore strongly abrogates infection of HIBCPP cells with L. monocytogenes. Concurrent inhibition of MAPK signaling further reduces infection, suggesting MAPKs mediate infection with L. monocytogenes during inhibition of dynamin-mediated endocytosis.
Collapse
Affiliation(s)
- Stefanie Dinner
- Pediatric Infectious Diseases, Department of Pediatrics, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Julian Kaltschmidt
- Pediatric Infectious Diseases, Department of Pediatrics, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Carolin Stump-Guthier
- Pediatric Infectious Diseases, Department of Pediatrics, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Svetlana Hetjens
- Institute of Medical Statistics and Biomathematics, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Hiroshi Ishikawa
- Department of NDU Life Sciences, Nippon Dental University, School of Life Dentistry, Chyoda-ku, Tokyo, Japan
| | - Tobias Tenenbaum
- Pediatric Infectious Diseases, Department of Pediatrics, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Horst Schroten
- Pediatric Infectious Diseases, Department of Pediatrics, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Christian Schwerk
- Pediatric Infectious Diseases, Department of Pediatrics, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
| |
Collapse
|
46
|
Wang S, Liu P, Wei J, Zhu Z, Shi Z, Shao D, Ma Z. Tumor suppressor p53 protects mice against Listeria monocytogenes infection. Sci Rep 2016; 6:33815. [PMID: 27644341 PMCID: PMC5028743 DOI: 10.1038/srep33815] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 09/02/2016] [Indexed: 02/07/2023] Open
Abstract
Tumor suppressor p53 is involved in regulating immune responses, which contribute to antitumor and antiviral activity. However, whether p53 has anti-bacterial functions remains unclear. Listeria monocytogenes (LM) causes listeriosis in humans and animals, and it is a powerful model for studying innate and adaptive immunity. In the present study, we illustrate an important regulatory role of p53 during LM infection. p53 knockout (p53KO) mice were more susceptible to LM infection, which was manifested by a shorter survival time and lower survival rate. p53KO mice showed significant impairments in LM eradication. Knockdown of p53 in RAW264.7 and HeLa cells resulted in increased invasion and intracellular survival of LM. Furthermore, the invasion and intracellular survival of LM was inhibited in p53-overexpressing RAW264.7 and HeLa cells. LM-infected p53KO mice exhibited severe clinical symptoms and organ injury, presumably because of the abnormal production of the pro-inflammatory cytokines TNF-α, IL-6, IL-12, and IL-18. Decreased IFN-γ and GBP1 productions were observed in LM-infected p53-deficient mice or cells. The combination of these defects likely resulted in the overwhelming LM infection in the p53KO mice. These observations indicate that p53 serves as an important regulator of the host innate immune that protects against LM infection.
Collapse
Affiliation(s)
- Shaohui Wang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, No. 518, Ziyue Road, Shanghai 200241, China
| | - Pingping Liu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, No. 518, Ziyue Road, Shanghai 200241, China
| | - Jianchao Wei
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, No. 518, Ziyue Road, Shanghai 200241, China
| | - Zixiang Zhu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, No. 518, Ziyue Road, Shanghai 200241, China.,State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China
| | - Zixue Shi
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, No. 518, Ziyue Road, Shanghai 200241, China
| | - Donghua Shao
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, No. 518, Ziyue Road, Shanghai 200241, China
| | - Zhiyong Ma
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, No. 518, Ziyue Road, Shanghai 200241, China
| |
Collapse
|
47
|
Westermann C, Gleinser M, Corr SC, Riedel CU. A Critical Evaluation of Bifidobacterial Adhesion to the Host Tissue. Front Microbiol 2016; 7:1220. [PMID: 27547201 PMCID: PMC4974247 DOI: 10.3389/fmicb.2016.01220] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 07/22/2016] [Indexed: 01/15/2023] Open
Abstract
Bifidobacteria are common inhabitants of the human gastrointestinal tract that, despite a long history of research, have not shown any pathogenic potential whatsoever. By contrast, some bifidobacteria are associated with a number of health-related benefits for the host. The reported beneficial effects of bifidobacteria include competitive exclusion of pathogens, alleviation of symptoms of irritable bowel syndrome and inflammatory bowel disease, and modulation of intestinal and systemic immune responses. Based on these effects, bifidobacteria are widely used as probiotics by pharmaceutical and dairy industries. In order to exert a beneficial effect bifidobacteria have to, at least transiently, colonize the host in a sufficient population size. Besides other criteria such as resistance to manufacturing processes and intestinal transit, potential probiotic bacteria are tested for adhesion to the host structures including intestinal epithelial cells, mucus, and extracellular matrix components. In the present review article, we summarize the current knowledge on bifidobacterial structures that mediate adhesion to host tissue and compare these to similar structures of pathogenic bacteria. This reveals that most of the adhesive structures and mechanisms involved in adhesion of bifidobacteria to host tissue are similar or even identical to those employed by pathogens to cause disease. It is thus reasonable to assume that these structures and mechanisms are equally important for commensal or probiotic bacteria and play a similar role in the beneficial effects exerted by bifidobacteria.
Collapse
Affiliation(s)
| | - Marita Gleinser
- Institute of Microbiology and Biotechnology, University of Ulm Ulm, Germany
| | - Sinéad C Corr
- Department of Microbiology, Moyne Institute of Preventative Medicine, School of Genetics and Microbiology, Trinity College Dublin Dublin, Ireland
| | - Christian U Riedel
- Institute of Microbiology and Biotechnology, University of Ulm Ulm, Germany
| |
Collapse
|
48
|
Identification of a Lipoteichoic Acid Glycosyltransferase Enzyme Reveals that GW-Domain-Containing Proteins Can Be Retained in the Cell Wall of Listeria monocytogenes in the Absence of Lipoteichoic Acid or Its Modifications. J Bacteriol 2016; 198:2029-42. [PMID: 27185829 PMCID: PMC4944223 DOI: 10.1128/jb.00116-16] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 05/10/2016] [Indexed: 12/04/2022] Open
Abstract
Listeria monocytogenes is a foodborne Gram-positive bacterial pathogen, and many of its virulence factors are either secreted proteins or proteins covalently or noncovalently attached to the cell wall. Previous work has indicated that noncovalently attached proteins with GW (glycine-tryptophan) domains are retained in the cell wall by binding to the cell wall polymer lipoteichoic acid (LTA). LTA is a glycerol phosphate polymer, which is modified in L. monocytogenes with galactose and d-alanine residues. We identified Lmo0933 as the cytoplasmic glycosyltransferase required for the LTA glycosylation process and renamed the protein GtlA, for glycosyltransferase LTA A. Using L. monocytogenes mutants lacking galactose or d-alanine modifications or the complete LTA polymer, we show that GW domain proteins are retained within the cell wall, indicating that other cell wall polymers are involved in the retention of GW domain proteins. Further experiments revealed peptidoglycan as the binding receptor as a purified GW domain fusion protein can bind to L. monocytogenes cells lacking wall teichoic acid (WTA) as well as purified peptidoglycan derived from a wild-type or WTA-negative strain. With this, we not only identify the first enzyme involved in the LTA glycosylation process, but we also provide new insight into the binding mechanism of noncovalently attached cell wall proteins.
IMPORTANCE Over the past 20 years, a large number of bacterial genome sequences have become available. Computational approaches are used for the genome annotation and identification of genes and encoded proteins. However, the function of many proteins is still unknown and often cannot be predicted bioinformatically. Here, we show that the previously uncharacterized Listeria monocytogenes gene lmo0933 likely codes for a glycosyltransferase required for the decoration of the cell wall polymer lipoteichoic acid (LTA) with galactose residues. Using L. monocytogenes mutants lacking LTA modifications or the complete polymer, we show that specific cell wall proteins, often associated with virulence, are retained within the cell wall, indicating that additional cell wall polymers are involved in their retention.
Collapse
|
49
|
TLR and TNF-R1 activation of the MKK3/MKK6-p38α axis in macrophages is mediated by TPL-2 kinase. Biochem J 2016; 473:2845-61. [PMID: 27402796 PMCID: PMC5095906 DOI: 10.1042/bcj20160502] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 07/11/2016] [Indexed: 01/08/2023]
Abstract
Previous studies suggested that Toll-like receptor (TLR) stimulation of the p38α MAP kinase (MAPK) is mediated by transforming growth factor-β-activated kinase 1 (TAK1) activation of MAPK kinases, MKK3, MKK4 and MKK6. We used quantitative mass spectrometry to monitor tumour progression locus 2 (TPL-2)-dependent protein phosphorylation following TLR4 stimulation with lipopolysaccharide, comparing macrophages from wild-type mice and Map3k8(D270A/D270A) mice expressing catalytically inactive TPL-2 (MAP3K8). In addition to the established TPL-2 substrates MKK1/2, TPL-2 kinase activity was required to phosphorylate the activation loops of MKK3/6, but not of MKK4. MKK3/6 activation required IκB kinase (IKK) phosphorylation of the TPL-2 binding partner nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB1) p105, similar to MKK1/2 activation. Tumour necrosis factor (TNF) stimulation of MKK3/6 phosphorylation was similarly dependent on TPL-2 catalytic activity and IKK phosphorylation of NF-κB1 p105. Owing to redundancy of MKK3/6 with MKK4, Map3k8(D270A) mutation only fractionally decreased lipopolysaccharide activation of p38α. TNF activation of p38α, which is mediated predominantly via MKK3/6, was substantially reduced. TPL-2 catalytic activity was also required for MKK3/6 and p38α activation following macrophage stimulation with Mycobacterium tuberculosis and Listeria monocytogenes Our experiments demonstrate that the IKK/NF-κB1 p105/TPL-2 signalling pathway, downstream of TAK1, regulates MKK3/6 and p38α activation in macrophages in inflammation.
Collapse
|
50
|
Margaroli C, Oberle S, Lavanchy C, Scherer S, Rosa M, Strasser A, Pellegrini M, Zehn D, Acha-Orbea H, Ehirchiou D. Role of proapoptotic BH3-only proteins inListeria monocytogenesinfection. Eur J Immunol 2016; 46:1427-37. [DOI: 10.1002/eji.201545857] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 01/25/2016] [Accepted: 03/29/2016] [Indexed: 01/09/2023]
Affiliation(s)
- Camilla Margaroli
- Department of Biochemistry CIIL; University of Lausanne; Epalinges Switzerland
| | - Susanne Oberle
- Swiss Vaccine Research Institute; Centre des laboratoires d'Epalinges; Epalinges Switzerland
- Division of Immunology and Allergy; Department of Medicine; Lausanne University Hospital; Lausanne Switzerland
| | - Christine Lavanchy
- Department of Biochemistry CIIL; University of Lausanne; Epalinges Switzerland
| | - Stefanie Scherer
- Swiss Vaccine Research Institute; Centre des laboratoires d'Epalinges; Epalinges Switzerland
- Division of Immunology and Allergy; Department of Medicine; Lausanne University Hospital; Lausanne Switzerland
| | - Muriel Rosa
- Department of Biochemistry CIIL; University of Lausanne; Epalinges Switzerland
| | - Andreas Strasser
- The Walter and Eliza Hall Institute of Medical Research; Melbourne Australia
- The Department of Medical Biology; University of Melbourne; Melbourne Australia
| | - Marc Pellegrini
- The Walter and Eliza Hall Institute of Medical Research; Melbourne Australia
- The Department of Medical Biology; University of Melbourne; Melbourne Australia
| | - Dietmar Zehn
- Technische Universität München; Weihenstephaner Berg 3; 85354 Freising-Weihenstephan Germany
| | - Hans Acha-Orbea
- Department of Biochemistry CIIL; University of Lausanne; Epalinges Switzerland
| | - Driss Ehirchiou
- Department of Biochemistry CIIL; University of Lausanne; Epalinges Switzerland
| |
Collapse
|