1
|
Boschiero C, Beshah E, Zhu X, Tuo W, Liu GE. Profiling Genome-Wide Methylation Patterns in Cattle Infected with Ostertagia ostertagi. Int J Mol Sci 2024; 26:89. [PMID: 39795948 PMCID: PMC11719486 DOI: 10.3390/ijms26010089] [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: 11/15/2024] [Revised: 12/13/2024] [Accepted: 12/17/2024] [Indexed: 01/13/2025] Open
Abstract
DNA methylation (DNAm) regulates gene expression and genomic imprinting. This study aimed to investigate the effect of gastrointestinal (GI) nematode infection on host DNAm. Helminth-free Holstein steers were either infected with Ostertagia ostertagi (the brown stomach worm) or given tap water only as a control. Animals were euthanized 30 days post-infection, and tissues were collected at necropsy. We conducted epigenome-wide profiling using a mammalian methylation array to explore the impact of infection on methylation patterns in the mucosa from abomasal fundus (FUN), pylorus (PYL), draining lymph nodes (dLNs), and the duodenum (DUO). The analysis covered 31,107 cattle CpGs of 5082 genes and revealed infection-driven, tissue-specific, differential methylation patterns. A total of 389 shared and 2770 tissue-specific, differentially methylated positions (DMPs) were identified in dLN and FUN, particularly in genes associated with immune responses. The shared DMPs were found in 263 genes, many of which are involved in immune responses. Furthermore, 282, 244, 52, and 24 differentially methylated regions (DMRs) were observed in dLN, FUN, PYL, and DUO, respectively. More hypomethylated DMRs were detected in dLN and FUN, while more hypermethylated DMRs were found in PYL and DUO. Genes carrying DMPs and DMRs and enriched pathways relating to immune functions/responses were detected in infected animals, indicating a link between DNA methylation and the infection. The data may implicate a crucial role of DNAm in regulating the nature/strength of host immunity to infection and contribute to a deeper understanding of the epigenetic regulatory landscape in cattle infected by GI nematodes.
Collapse
Affiliation(s)
- Clarissa Boschiero
- Animal Parasitic Diseases Laboratory, Beltsville Agricultural Research Center, Agricultural Research Service, United States Department of Agriculture, Beltsville, MD 20705, USA
- Department of Veterinary Medicine, University of Maryland, College Park, MD 20742, USA
| | - Ethiopia Beshah
- Animal Parasitic Diseases Laboratory, Beltsville Agricultural Research Center, Agricultural Research Service, United States Department of Agriculture, Beltsville, MD 20705, USA
| | - Xiaoping Zhu
- Department of Veterinary Medicine, University of Maryland, College Park, MD 20742, USA
| | - Wenbin Tuo
- Animal Parasitic Diseases Laboratory, Beltsville Agricultural Research Center, Agricultural Research Service, United States Department of Agriculture, Beltsville, MD 20705, USA
| | - George E. Liu
- Animal Genomics and Improvement Laboratory, Beltsville Agricultural Research Center, Agricultural Research Service, United States Department of Agriculture, Beltsville, MD 20705, USA
| |
Collapse
|
2
|
The Study of Microbe–Host Two-Way Communication. Microorganisms 2022; 10:microorganisms10020408. [PMID: 35208862 PMCID: PMC8875206 DOI: 10.3390/microorganisms10020408] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 02/02/2022] [Accepted: 02/07/2022] [Indexed: 12/22/2022] Open
Abstract
Back-and-forth intercommunication in host–pathogen interactions has long been recognized to play an important role in commensalism and microbial pathogenesis. For centuries, we have studied these microbes in our surroundings, yet many questions about the evolutionary cross-talk between host and microbe remain unanswered. With the recent surge in research interest in the commensal microbiome, basic immunological questions have returned to the fore, such as, how are vast numbers of microbes capable of coexisting within animals and humans while also maintaining a healthy functional immune system? How is the evasion and subversion of the immune system achieved by some microbes but not others? The intricate and important-to-remember two-way interaction and coevolution of host and microbe is the communication network we must tap into as researchers to answer these questions.
Collapse
|
3
|
Zhang Z, Liu S, Huang J, Cui Y, Liu Y, Zhou Y, Zhu Z. Phloretin is protective in a murine salmonella enterica serovar typhimurium infection model. Microb Pathog 2021; 161:105298. [PMID: 34801645 DOI: 10.1016/j.micpath.2021.105298] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 11/14/2021] [Accepted: 11/14/2021] [Indexed: 11/28/2022]
Abstract
Salmonella, an important zoonotic pathogen, causes significant morbidity and mortality in both humans and animals. Phloretin mainly isolated from strawberries and apples has the effects of treating inflammation and pathogenic bacteria, but its protective efficacy and mechanism of action against Salmonella spp. are less clear. In this study, we found that phloretin alleviated body weight loss, colon length shortening, and colonic pathological damage caused by S. Typhimurium. Phloretin also decreased S. Typhimurium translocation to the mesenteric lymph nodes (MLN) and spleen. Further mechanism studies showed that phloretin significantly inhibited inflammation and oxidative stress levels in the colonic tissue. Phloretin also prevented S. Typhimurium-mediated impairment in the colon epithelium barrier by the regulation ZO-1 and occludin levels. Interestingly, phloretin did not inhibit S. typhimurium growth in vitro, but reduced the internalization of S. Typhimurium into Caco-2 cells. Taken together, these findings indicated that phloretin may be a new dietary strategy to combat the disease.
Collapse
Affiliation(s)
- Zecai Zhang
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, China; Heilongjiang Provincial Technology Innovation Center for Bovine Disease Control and Prevention, Daqing, 163319, China; Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, Daqing, 163319, China; Heilongjiang Province Cultivating Collaborative Innovation Center for the Beidahuang Modern Agricultural Industry Technology, Daqing, 163319, China
| | - Siyu Liu
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, China; Heilongjiang Provincial Technology Innovation Center for Bovine Disease Control and Prevention, Daqing, 163319, China
| | - Jiang Huang
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, China; Heilongjiang Provincial Technology Innovation Center for Bovine Disease Control and Prevention, Daqing, 163319, China
| | - Yueqi Cui
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, China; Heilongjiang Provincial Technology Innovation Center for Bovine Disease Control and Prevention, Daqing, 163319, China
| | - Yu Liu
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, China; Heilongjiang Provincial Technology Innovation Center for Bovine Disease Control and Prevention, Daqing, 163319, China; Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, Daqing, 163319, China; Heilongjiang Province Cultivating Collaborative Innovation Center for the Beidahuang Modern Agricultural Industry Technology, Daqing, 163319, China
| | - Yulong Zhou
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, China; Heilongjiang Provincial Technology Innovation Center for Bovine Disease Control and Prevention, Daqing, 163319, China; Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, Daqing, 163319, China; Heilongjiang Province Cultivating Collaborative Innovation Center for the Beidahuang Modern Agricultural Industry Technology, Daqing, 163319, China
| | - Zhanbo Zhu
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, China; Heilongjiang Provincial Technology Innovation Center for Bovine Disease Control and Prevention, Daqing, 163319, China; Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, Daqing, 163319, China; Heilongjiang Province Cultivating Collaborative Innovation Center for the Beidahuang Modern Agricultural Industry Technology, Daqing, 163319, China.
| |
Collapse
|
4
|
Ząbek T, Semik-Gurgul E, Ropka-Molik K, Szmatoła T, Kawecka-Grochocka E, Zalewska M, Kościuczuk E, Wnuk M, Bagnicka E. Short communication: Locus-specific interrelations between gene expression and DNA methylation patterns in bovine mammary gland infected by coagulase-positive and coagulase-negative staphylococci. J Dairy Sci 2020; 103:10689-10695. [PMID: 32952032 DOI: 10.3168/jds.2020-18404] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 06/25/2020] [Indexed: 11/19/2022]
Abstract
Pathogens are able to alter the cell cycle program and immune response of the host by changing the transcription and epigenetics of genes responsible for cell cycle control and inflammation. In this regard, we evaluated interrelations between DNA methylation and expression of autophagy, apoptosis, and lipid metabolism-related genes in a sample set of mammary gland secretory tissue sections derived from bovine mammary glands infected with coagulase-negative and coagulase-positive staphylococci. We assessed relative transcript abundance and DNA bisulfite sequencing in loci of the ATG5, IGF1R, TERT, and DGAT1 genes. Lack of DNA methylation in ATG5 and DGAT1 loci might be associated with maintenance of ATG5 and DGAT1 expression regardless of the health status of bovine mammary gland. Complete methylation of intragenic CpG regions in the IGF1R locus was apparently not related to the presence of its transcript in the investigated udder parenchyma samples. Detected hypermethylation of the TERT upstream element was associated with a small amount of TERT mRNA in bovine mammary gland, regardless of the presence, or absence, of the pathogen. A significant decrease in TERT gene expression in tissue sections of mammary gland free of bacteria and in those infected with coagulase-positive staphylococci was observed in parenchyma samples infected with coagulase-negative staphylococci. Two possible explanations are the direct involvement of the TERT gene in the etiology of bovine mastitis or the increase of TERT mRNA due to activation of the MAPK signaling pathway in response to release of exotoxins by coagulase-negative bacteria in the bovine mammary gland.
Collapse
Affiliation(s)
- T Ząbek
- National Research Institute of Animal Production, Krakowska 1, 32-083 Balice, Poland.
| | - E Semik-Gurgul
- National Research Institute of Animal Production, Krakowska 1, 32-083 Balice, Poland
| | - K Ropka-Molik
- National Research Institute of Animal Production, Krakowska 1, 32-083 Balice, Poland
| | - T Szmatoła
- National Research Institute of Animal Production, Krakowska 1, 32-083 Balice, Poland; University of Agriculture in Krakow, University Centre of Veterinary Medicine Krakow, Al. Mickiewicza 24/28, 30-059 Krakow, Poland
| | - E Kawecka-Grochocka
- Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Postępu 36A, Jastrzębiec, 05-552 Magdalenka, Poland
| | - M Zalewska
- Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Postępu 36A, Jastrzębiec, 05-552 Magdalenka, Poland; Department of Applied Microbiology, Institute of Microbiology, Faculty of Biology, University of Warsaw, Miecznikowa 1, 02-096 Warsaw
| | - E Kościuczuk
- Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Postępu 36A, Jastrzębiec, 05-552 Magdalenka, Poland
| | - M Wnuk
- Department of Biotechnology, University of Rzeszow, Pigonia 1, 35-310 Rzeszow, Poland
| | - E Bagnicka
- Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Postępu 36A, Jastrzębiec, 05-552 Magdalenka, Poland
| |
Collapse
|
5
|
Curto P, Riley SP, Simões I, Martinez JJ. Macrophages Infected by a Pathogen and a Non-pathogen Spotted Fever Group Rickettsia Reveal Differential Reprogramming Signatures Early in Infection. Front Cell Infect Microbiol 2019; 9:97. [PMID: 31024862 PMCID: PMC6467950 DOI: 10.3389/fcimb.2019.00097] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 03/22/2019] [Indexed: 12/22/2022] Open
Abstract
Despite their high degree of genomic similarity, different spotted fever group (SFG) Rickettsia are often associated with very different clinical presentations. For example, Rickettsia conorii causes Mediterranean spotted fever, a life-threatening disease for humans, whereas Rickettsia montanensis is associated with limited or no pathogenicity to humans. However, the molecular basis responsible for the different pathogenicity attributes are still not understood. Although killing microbes is a critical function of macrophages, the ability to survive and/or proliferate within phagocytic cells seems to be a phenotypic feature of several intracellular pathogens. We have previously shown that R. conorii and R. montanensis exhibit different intracellular fates within macrophage-like cells. By evaluating early macrophage responses upon insult with each of these rickettsial species, herein we demonstrate that infection with R. conorii results in a profound reprogramming of host gene expression profiles. Transcriptional programs generated upon infection with this pathogenic bacteria point toward a sophisticated ability to evade innate immune signals, by modulating the expression of several anti-inflammatory molecules. Moreover, R. conorii induce the expression of several pro-survival genes, which may result in the ability to prolong host cell survival, thus protecting its replicative niche. Remarkably, R. conorii-infection promoted a robust modulation of different transcription factors, suggesting that an early manipulation of the host gene expression machinery may be key to R. conorii proliferation in THP-1 macrophages. This work provides new insights into the early molecular processes hijacked by a pathogenic SFG Rickettsia to establish a replicative niche in macrophages, opening several avenues of research in host-rickettsiae interactions.
Collapse
Affiliation(s)
- Pedro Curto
- Ph.D. Programme in Experimental Biology and Biomedicine, Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
- CNC-Center for Neuroscience and Cell Biology, Coimbra, Portugal
- Vector Borne Disease Laboratories, Department of Pathobiological Sciences, LSU School of Veterinary Medicine, Baton Rouge, LA, United States
| | - Sean P. Riley
- Vector Borne Disease Laboratories, Department of Pathobiological Sciences, LSU School of Veterinary Medicine, Baton Rouge, LA, United States
| | - Isaura Simões
- CNC-Center for Neuroscience and Cell Biology, Coimbra, Portugal
- Vector Borne Disease Laboratories, Department of Pathobiological Sciences, LSU School of Veterinary Medicine, Baton Rouge, LA, United States
| | - Juan J. Martinez
- Vector Borne Disease Laboratories, Department of Pathobiological Sciences, LSU School of Veterinary Medicine, Baton Rouge, LA, United States
| |
Collapse
|
6
|
Eshwar AK, Wolfrum N, Stephan R, Fanning S, Lehner A. Interaction of matrix metalloproteinase-9 and Zpx inCronobacter turicensisLMG 23827Tmediated infections in the zebrafish model. Cell Microbiol 2018; 20:e12888. [DOI: 10.1111/cmi.12888] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 06/19/2018] [Accepted: 06/27/2018] [Indexed: 12/18/2022]
Affiliation(s)
| | - Nina Wolfrum
- Institute for Veterinary Bacteriology; University of Zurich; Zurich Switzerland
| | - Roger Stephan
- Institute for Food Safety and Hygiene; University of Zurich; Zurich Switzerland
| | - Séamus Fanning
- UCD-Centre for Food Safety, Science Centre South; University College Dublin; Dublin Ireland
| | - Angelika Lehner
- Institute for Food Safety and Hygiene; University of Zurich; Zurich Switzerland
| |
Collapse
|
7
|
Comparative Gene Expression Analysis within Mouse Macrophage for Identifying Critical Pathways in Macrophage and Brucella suis Interaction. Jundishapur J Microbiol 2017. [DOI: 10.5812/jjm.59275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
8
|
Disease and Carrier Isolates of Neisseria meningitidis Cause G1 Cell Cycle Arrest in Human Epithelial Cells. Infect Immun 2016; 84:2758-70. [PMID: 27430269 DOI: 10.1128/iai.00296-16] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 07/06/2016] [Indexed: 12/30/2022] Open
Abstract
Microbial pathogens have developed several mechanisms to modulate and interfere with host cell cycle progression. In this study, we analyzed the effect of the human pathogen Neisseria meningitidis on the cell cycle of epithelial cells. Two pathogenic isolates, as well as two carrier isolates, were tested for their ability to adhere to and invade into the epithelial cell lines Detroit 562 and NP69 and to modulate the cell cycle. We found that all isolates adhered equally well to both Detroit 562 and NP69 cells, whereas the carrier isolates were significantly less invasive. Using propidium iodide staining and 5-ethynyl-2'-deoxyuridine pulse-labeling, we provide evidence that meningococcal infection arrested cells in the G1 phase of the cell cycle at 24 h postinfection. In parallel, a significant decrease of cells in the S phase was observed. Interestingly, G1-phase arrest was only induced after infection with live bacteria but not with heat-killed bacteria. By Western blotting we demonstrate that bacterial infection resulted in a decreased protein level of the cell cycle regulator cyclin D1, whereas cyclin E expression levels were increased. Furthermore, N. meningitidis infection induced an accumulation of the cyclin-dependent kinase inhibitor (CKI) p21(WAF1/CIP1) that was accompanied by a redistribution of this CKI to the cell nucleus, as shown by immunofluorescence analysis. Moreover, the p27(CIP1) CKI was redistributed and showed punctate foci in infected cells. In summary, we present data that N. meningitidis can interfere with the processes of host cell cycle regulation.
Collapse
|
9
|
Ehrlichia chaffeensis TRP32 is a Nucleomodulin that Directly Regulates Expression of Host Genes Governing Differentiation and Proliferation. Infect Immun 2016; 84:3182-3194. [PMID: 27572329 DOI: 10.1128/iai.00657-16] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Ehrlichia chaffeensis is an obligately intracellular bacterium that reprograms the mononuclear phagocyte through diverse effector-host interactions to modulate numerous host cell processes, including transcription. In a previous study, we reported that E. chaffeensis TRP32, a type 1 secreted effector, interacts with multiple host nucleus-associated proteins and also auto-activates reporter gene expression in yeast. In this study, we demonstrate that TRP32 is a nucleomodulin that binds host DNA and alters host gene transcription. TRP32 enters the host cell nucleus via a noncanonical translocation mechanism that involves phosphorylation of Y179 located in a C-terminal tri-tyrosine motif. Both genistein and mutation of Y179 inhibited TRP32 nuclear entry. An electromobility shift assay (EMSA) demonstrated TRP32 host DNA binding via its tandem repeat domain. TRP32 DNA binding and motif preference were further confirmed by supershift assays, as well as competition and mutant probe analyses. Using ChIP-Seq, we determined that TRP32 binds a G-rich motif primarily within ±500 bp of the gene transcription start site. An ontology analysis identified genes involved in processes such as immune cell differentiation, chromatin remodeling, and RNA transcription and processing, as primary TRP32 targets. TRP32 bound genes (n=1223) were distributed on all chromosomes and included several global regulators of proliferation and inflammation such as FOS and JUN, AKT3 and NRAS, and non-coding RNA genes, miRNA 21 and miRNA 142. TRP32 target genes were differentially regulated during infection, the majority of which were repressed, and direct repression/activation of these genes by TRP32 was confirmed in vitro with a cellular luciferase reporter assay.
Collapse
|
10
|
Ruan H, Zhang Z, Tian L, Wang S, Hu S, Qiao JJ. The Salmonella effector SopB prevents ROS-induced apoptosis of epithelial cells by retarding TRAF6 recruitment to mitochondria. Biochem Biophys Res Commun 2016; 478:618-23. [PMID: 27473656 DOI: 10.1016/j.bbrc.2016.07.116] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 07/26/2016] [Indexed: 11/18/2022]
Abstract
Microbial pathogens enter host cells by injecting effector proteins of the Type III secretion system (T3SS), which facilitate pathogen translocation across the host cell membrane. These effector proteins exert their effects by modulating a variety of host innate immune responses, thereby facilitating bacterial replication and systemic infection. Salmonella enterica serovar typhimurium (S.typhimurium) is a clinically important pathogen that causes food poisoning and gastroenteritis. The SopB effector protein of S. typhimurium, encoded by Salmonella pathogenicity islands (SPI)-1 T3SS, protects host epithelial cells from infection-induced apoptosis. However, how SopB influences apoptosis induction remains unclear. Here, we investigated the mechanism of SopB action in host cells. We found that SopB inhibits infection-induced apoptosis by attenuating the production of reactive oxygen species (ROS) in mitochondria, the crucial organelles for apoptosis initiation. Further investigation revealed that SopB binds to cytosolic tumor necrosis factor receptor associated factor 6 (TRAF6) and forms a trap preventing the mitochondrial recruitment of TRAF6, an essential event for ROS generation within mitochondria. By studying the response of Traf6(+/+) and Traf6(-/-)mouse embryonic fibroblasts to S. typhimurium infection, we found that TRAF6 promoted apoptosis by increasing ROS accumulation, which led to increased Bax/Bcl-2 ratio, Bax recruitment to mitochondrial membrane, and release of Cyt c into the cytoplasm. These findings show that SopB suppresses host cell apoptosis by binding to TRAF6 and preventing mitochondrial ROS generation.
Collapse
Affiliation(s)
- Haihua Ruan
- Tianjin Key Laboratory of Food Science and Biotechnology, College of Biotechnology and Food Science, Tianjin University of Commerce, Tianjin, 300134, China.
| | - Zhen Zhang
- Tianjin Key Laboratory of Food Science and Biotechnology, College of Biotechnology and Food Science, Tianjin University of Commerce, Tianjin, 300134, China
| | - Li Tian
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300192, China
| | - Suying Wang
- Tianjin Key Laboratory of Food Science and Biotechnology, College of Biotechnology and Food Science, Tianjin University of Commerce, Tianjin, 300134, China
| | - Shuangyan Hu
- Tianjin Key Laboratory of Food Science and Biotechnology, College of Biotechnology and Food Science, Tianjin University of Commerce, Tianjin, 300134, China
| | - Jian-Jun Qiao
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300192, China.
| |
Collapse
|
11
|
Oosthuysen WF, Mueller T, Dittrich MT, Schubert-Unkmeir A. Neisseria meningitidiscauses cell cycle arrest of human brain microvascular endothelial cells at S phase via p21 and cyclin G2. Cell Microbiol 2015; 18:46-65. [DOI: 10.1111/cmi.12482] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2015] [Revised: 06/30/2015] [Accepted: 07/01/2015] [Indexed: 12/19/2022]
Affiliation(s)
| | - Tobias Mueller
- Department of Bioinformatics; University of Wuerzburg; Wuerzburg Germany
| | - Marcus T. Dittrich
- Department of Bioinformatics; University of Wuerzburg; Wuerzburg Germany
- Institute of Human Genetics; University of Wuerzburg; Germany
| | | |
Collapse
|
12
|
Eisenreich W, Heesemann J, Rudel T, Goebel W. Metabolic host responses to infection by intracellular bacterial pathogens. Front Cell Infect Microbiol 2013; 3:24. [PMID: 23847769 PMCID: PMC3705551 DOI: 10.3389/fcimb.2013.00024] [Citation(s) in RCA: 135] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Accepted: 06/11/2013] [Indexed: 12/12/2022] Open
Abstract
The interaction of bacterial pathogens with mammalian hosts leads to a variety of physiological responses of the interacting partners aimed at an adaptation to the new situation. These responses include multiple metabolic changes in the affected host cells which are most obvious when the pathogen replicates within host cells as in case of intracellular bacterial pathogens. While the pathogen tries to deprive nutrients from the host cell, the host cell in return takes various metabolic countermeasures against the nutrient theft. During this conflicting interaction, the pathogen triggers metabolic host cell responses by means of common cell envelope components and specific virulence-associated factors. These host reactions generally promote replication of the pathogen. There is growing evidence that pathogen-specific factors may interfere in different ways with the complex regulatory network that controls the carbon and nitrogen metabolism of mammalian cells. The host cell defense answers include general metabolic reactions, like the generation of oxygen- and/or nitrogen-reactive species, and more specific measures aimed to prevent access to essential nutrients for the respective pathogen. Accurate results on metabolic host cell responses are often hampered by the use of cancer cell lines that already exhibit various de-regulated reactions in the primary carbon metabolism. Hence, there is an urgent need for cellular models that more closely reflect the in vivo infection conditions. The exact knowledge of the metabolic host cell responses may provide new interesting concepts for antibacterial therapies.
Collapse
Affiliation(s)
- Wolfgang Eisenreich
- Lehrstuhl für Biochemie, Center of Isotopologue Profiling, Technische Universität München Garching, Germany
| | | | | | | |
Collapse
|