1
|
Al-Nasser MM, Al-Saeedi MJ, Alhowaiti SA, Shinwari Z, Alhamlan FS, Alothaid H, Alkahtani S, Al-Qahtani AA. Combination of Methotrexate and Resveratrol Reduces Pro-Inflammatory Chemokines in Human THP-1 Cells. J Inflamm Res 2024; 17:8085-8098. [PMID: 39507267 PMCID: PMC11539838 DOI: 10.2147/jir.s482503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 10/08/2024] [Indexed: 11/08/2024] Open
Abstract
Introduction Methotrexate (MTX) is a widely used anti-metabolite drug in cancer therapy, but its efficacy is often hindered by reactive oxygen species (ROS)-induced cellular toxicity. Resveratrol, a natural polyphenol, possesses antioxidant and anticancer properties. Therefore, this in vitro study aimed to investigate the synergistic anti-proliferative and anti-inflammatory effects of MTX and resveratrol in human THP-1 cells. Methods THP-1 cells were differentiated into macrophage-like cells using phorbol 12-myristate 13-acetate. In vitro experiments assessed the impact of various concentrations of MTX and resveratrol on cell viability and proliferation using the MTT assay. Concentration-effect curves were generated to elucidate their relationship. Gene expression was analyzed by RT-qPCR, while chemokine expression was measured via ELISA. Phagocytic and migratory activities were also evaluated. Results Differentiated THP-1 cells were treated with MTX and resveratrol and stimulated with dimethyl sulfoxide (DMSO) and lipopolysaccharide (LPS) as inflammatory stimuli. The combination of MTX and resveratrol exhibited an anti-proliferative effect in THP-1 cells (p = 0.03). The concentration-effect curve revealed IC50 values of 49.15 µg at 24 hours (R = 0.8236) and 2.029e-005 µg at 48 hours (R = 0.97) for MTX, and 20.17 µg at 48 hours (R = 1.000) and 55.38 µg at 96 hours (R = 0.9666) for resveratrol. Co-treatment with MTX and resveratrol significantly reduced mRNA and chemokine expression of CCL2, CCL3, CCL4, CCL5, and CXCL10 (p < 0.05). Moreover, decreased phagocytic and migratory activities were confirmed by phagocytosis and migration assays (p < 0.05). Conclusion The combination of MTX and resveratrol effectively attenuated pro-inflammatory activity in THP-1 cells, as evidenced by the downregulation of mRNA and chemokine expression. These findings suggest that the synergistic effects of MTX and resveratrol hold promise for enhancing cancer therapeutics.
Collapse
Affiliation(s)
- Moonerah M Al-Nasser
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Mashael J Al-Saeedi
- Department of Infection and Immunity, Research Centre, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia
| | - Saltana A Alhowaiti
- Department of Infection and Immunity, Research Centre, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia
| | - Zakia Shinwari
- Therapeutics & Biomarker Discovery for Clinical Applications, Stem Cell & Tissue Re-Engineering Program, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Fatimah S Alhamlan
- Department of Infection and Immunity, Research Centre, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia
- Department of Microbiology and Immunology, College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Hani Alothaid
- Department of Basic Sciences, Faculty of Applied Medical Sciences, Al-Baha University, Al-Baha, Saudi Arabia
| | - Saad Alkahtani
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Ahmed A Al-Qahtani
- Department of Infection and Immunity, Research Centre, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia
- Department of Microbiology and Immunology, College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| |
Collapse
|
2
|
In Vitro Studies on the Immunomodulatory Effects of Pulicaria crispa Extract on Human THP-1 Monocytes. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2020:7574606. [PMID: 33628359 PMCID: PMC7895603 DOI: 10.1155/2020/7574606] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 06/29/2020] [Accepted: 07/21/2020] [Indexed: 12/02/2022]
Abstract
Background Pulicaria crispa (P. crispa) is a plant from the Compositae family that exhibits antioxidant, anti-inflammatory, antibacterial, and cytotoxic activities. Objective The current study aimed at investigating the immunomodulatory effects of P. crispa extract in lipopolysaccharide- (LPS-) stimulated human monocytic THP-1 cells. Methods To induce macrophage differentiation, THP-1 cell lines were treated with phorbol-12-myristate 13-acetate, followed by exposure to LPS with or without 50 or 100 μg/ml of P. crispa extract. The following tests were employed to test the immunomodulatory effects of the extract: MTT assay, ELISA, Western blotting analysis, cell migration and phagocytosis assays, and Annexin V staining method. Results Exposure to 100 μg/ml P. crispa extract significantly reduced THP-1 cell proliferation, migration, and phagocytosis (in LPS-stimulated cells, but not in unstimulated cells). Moreover, the extract alone significantly reduced the rate of THP-1 cell apoptosis, while it increased the rate of late apoptosis. Molecular investigations showed that treatment with P. crispa extract significantly upregulated the expression of ERK1, p-MAPK, P-P38, and Bcl2, while it significantly reduced the expression of ERK5, Bax, NF-κB, P-NF-κB, CCL1, CCL2, CCL5, CCL22, CXCL1, and CXCL10. Conclusion Pulicaria crispa extract exhibited anti-inflammatory, antiproliferative, antimigratory, and antiphagocytic effects in LPS-stimulated THP-1 cells. Future studies should investigate these mechanisms in animal models with chronic inflammatory diseases.
Collapse
|
3
|
Abstract
Germination of Clostridium difficile spores is a crucial early requirement for colonization of the gastrointestinal tract. Likewise, C. difficile cannot cause disease pathologies unless its spores germinate into metabolically active, toxin-producing cells. Recent advances in our understanding of C. difficile spore germination mechanisms indicate that this process is both complex and unique. This review defines unique aspects of the germination pathways of C. difficile and compares them to those of two other well-studied organisms, Bacillus anthracis and Clostridium perfringensC. difficile germination is unique, as C. difficile does not contain any orthologs of the traditional GerA-type germinant receptor complexes and is the only known sporeformer to require bile salts in order to germinate. While recent advances describing C. difficile germination mechanisms have been made on several fronts, major gaps in our understanding of C. difficile germination signaling remain. This review provides an updated, in-depth summary of advances in understanding of C. difficile germination and potential avenues for the development of therapeutics, and discusses the major discrepancies between current models of germination and areas of ongoing investigation.
Collapse
|
4
|
Animal Models for the Pathogenesis, Treatment, and Prevention of Infection by Bacillus anthracis. Microbiol Spectr 2016; 3:TBS-0001-2012. [PMID: 26104551 DOI: 10.1128/microbiolspec.tbs-0001-2012] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
This article reviews the characteristics of the major animal models utilized for studies on Bacillus anthracis and highlights their contributions to understanding the pathogenesis and host responses to anthrax and its treatment and prevention. Advantages and drawbacks associated with each model, to include the major models (murine, guinea pig, rabbit, nonhuman primate, and rat), and other less frequently utilized models, are discussed. Although the three principal forms of anthrax are addressed, the main focus of this review is on models for inhalational anthrax. The selection of an animal model for study is often not straightforward and is dependent on the specific aims of the research or test. No single animal species provides complete equivalence to humans; however, each species, when used appropriately, can contribute to a more complete understanding of anthrax and its etiologic agent.
Collapse
|
5
|
do Vale A, Cabanes D, Sousa S. Bacterial Toxins as Pathogen Weapons Against Phagocytes. Front Microbiol 2016; 7:42. [PMID: 26870008 PMCID: PMC4734073 DOI: 10.3389/fmicb.2016.00042] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 01/11/2016] [Indexed: 12/31/2022] Open
Abstract
Bacterial toxins are virulence factors that manipulate host cell functions and take over the control of vital processes of living organisms to favor microbial infection. Some toxins directly target innate immune cells, thereby annihilating a major branch of the host immune response. In this review we will focus on bacterial toxins that act from the extracellular milieu and hinder the function of macrophages and neutrophils. In particular, we will concentrate on toxins from Gram-positive and Gram-negative bacteria that manipulate cell signaling or induce cell death by either imposing direct damage to the host cells cytoplasmic membrane or enzymatically modifying key eukaryotic targets. Outcomes regarding pathogen dissemination, host damage and disease progression will be discussed.
Collapse
Affiliation(s)
- Ana do Vale
- Host Interaction and Response, Instituto de Investigação e Inovação em Saúde, Universidade do PortoPorto, Portugal; Group of Fish Immunology and Vaccinology, Instituto de Biologia Molecular e Celular, Universidade do PortoPorto, Portugal
| | - Didier Cabanes
- Host Interaction and Response, Instituto de Investigação e Inovação em Saúde, Universidade do PortoPorto, Portugal; Group of Molecular Microbiology, Instituto de Biologia Molecular e Celular, Universidade do PortoPorto, Portugal
| | - Sandra Sousa
- Host Interaction and Response, Instituto de Investigação e Inovação em Saúde, Universidade do PortoPorto, Portugal; Group of Molecular Microbiology, Instituto de Biologia Molecular e Celular, Universidade do PortoPorto, Portugal
| |
Collapse
|
6
|
Cote CK, Welkos SL. Anthrax Toxins in Context of Bacillus anthracis Spores and Spore Germination. Toxins (Basel) 2015; 7:3167-78. [PMID: 26287244 PMCID: PMC4549744 DOI: 10.3390/toxins7083167] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Revised: 08/08/2015] [Accepted: 08/11/2015] [Indexed: 11/18/2022] Open
Abstract
The interaction of anthrax toxin or toxin components with B. anthracis spores has been demonstrated. Germinating spores can produce significant amounts of toxin components very soon after the initiation of germination. In this review, we will summarize the work performed that has led to our understanding of toxin and spore interactions and discuss the complexities associated with these interactions.
Collapse
Affiliation(s)
- Christopher K Cote
- United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Bacteriology Division, 1425 Porter Street, Fort Detrick, Frederick, MD 21702-5011, USA.
| | - Susan L Welkos
- United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Bacteriology Division, 1425 Porter Street, Fort Detrick, Frederick, MD 21702-5011, USA.
| |
Collapse
|
7
|
The impact of "omic" and imaging technologies on assessing the host immune response to biodefence agents. J Immunol Res 2014; 2014:237043. [PMID: 25333059 PMCID: PMC4182007 DOI: 10.1155/2014/237043] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Revised: 07/23/2014] [Accepted: 08/05/2014] [Indexed: 01/08/2023] Open
Abstract
Understanding the interactions between host and pathogen is important for the development and assessment of medical countermeasures to infectious agents, including potential biodefence pathogens such as Bacillus anthracis, Ebola virus, and Francisella tularensis. This review focuses on technological advances which allow this interaction to be studied in much greater detail. Namely, the use of “omic” technologies (next generation sequencing, DNA, and protein microarrays) for dissecting the underlying host response to infection at the molecular level; optical imaging techniques (flow cytometry and fluorescence microscopy) for assessing cellular responses to infection; and biophotonic imaging for visualising the infectious disease process. All of these technologies hold great promise for important breakthroughs in the rational development of vaccines and therapeutics for biodefence agents.
Collapse
|
8
|
Effects of experimental exclusion of scavengers from carcasses of anthrax-infected herbivores on Bacillus anthracis sporulation, survival, and distribution. Appl Environ Microbiol 2013; 79:3756-61. [PMID: 23584788 DOI: 10.1128/aem.00181-13] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Scavenging of anthrax carcasses has long been hypothesized to play a critical role in the production of the infectious spore stage of Bacillus anthracis after host death, though empirical studies assessing this are lacking. We compared B. anthracis spore production, distribution, and survival at naturally occurring anthrax herbivore carcasses that were either experimentally caged to exclude vertebrate scavengers or left unmanipulated. We found no significant effect of scavengers on soil spore density (P > 0.05). Soil stained with terminally hemorrhaged blood and with nonhemorrhagic fluids exhibited high levels of B. anthracis spore contamination (ranging from 10(3) to 10(8) spores/g), even in the absence of vertebrate scavengers. At most of the carcass sites, we also found that spore density in samples taken from hemorrhagic-fluid-stained soil continued to increase for >4 days after host death. We conclude that scavenging by vertebrates is not a critical factor in the life cycle of B. anthracis and that anthrax control measures relying on deterrence or exclusion of vertebrate scavengers to prevent sporulation are unlikely to be effective.
Collapse
|
9
|
Bensman MD, Mackie RS, Minter ZA, Gutting BW. Effect of animal sera on Bacillus anthracis Sterne spore germination and vegetative cell growth. J Appl Microbiol 2012; 113:276-83. [PMID: 22515644 DOI: 10.1111/j.1365-2672.2012.05314.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
AIMS The aims of this work were to investigate the effects of sera on B. anthracis Sterne germination and growth. Sera examined included human, monkey and rabbit sera, as well as sera from eight other species. METHODS AND RESULTS Standard dilution plate assay (with and without heat kill) was used as a measure of germination, and spectroscopy was used to measure growth. In addition, a Coulter Counter particle counter was used to monitor germination and growth based on bacterial size. Spores germinated best in foetal bovine and monkey sera, moderately with human sera and showed limited germination in the presence of rabbit or rat sera. Vegetative bacteria grew best in foetal bovine sera and moderately in rabbit sera. Human and monkey sera supported little growth of vegetative bacteria. CONCLUSION The data suggested sera can have a significant impact on germination and growth of Sterne bacteria. SIGNIFICANCE AND IMPACT OF THE STUDY These data should be considered when conducting in vitro cell culture studies and may aid in interpreting in vivo infection studies.
Collapse
Affiliation(s)
- M D Bensman
- Dahlgren Division, CBR Concepts and Experimentation Branch-Z21, Naval Surface Warfare Center, Dahlgren, VA, USA
| | | | | | | |
Collapse
|
10
|
Tsai P, Cao GL, Tomczuk B, Suzdak PD, Cross AS, Shapiro P, Rosen GM. Effect of the mammalian arginase inhibitor 2(S)-amino-6-boronohexanoic acid on Bacillus anthracis arginase. Curr Microbiol 2012; 64:379-84. [PMID: 22271269 DOI: 10.1007/s00284-012-0084-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2011] [Accepted: 01/04/2012] [Indexed: 10/14/2022]
Abstract
Macrophages, upon phagocytosing endospores of Bacillus anthracis, up-regulate the expression of the immunological isoform of nitric oxide synthase, NOS 2, concomitant with production of nitric oxide (NO•) from metabolism of L -arginine. We have previously demonstrated that macrophages that secrete NO• kill the bacilli of B. anthracis. To circumvent this microbicidal activity of NO•, B. anthracis has evolved pathways that include the enzyme arginase, which metabolizes L: -arginine to ornithine and urea. Compounds that inhibit arginase might, therefore, offer a therapeutic approach to controlling B. anthracis infection. 2(S)-Amino-6-boronohexanoic acid (ABH) has been reported to be an inhibitor of mammalian arginase. In this study, we explore the inhibitory effect of ABH against B. anthracis arginase and its potential for future development, as an effective therapeutic agent against microbial infection. We found that ABH is an inhibitor of bacterial arginase in several different endospore strains of B. anthracis. Further, ABH inhibits neither the phagocytosis of these endospores nor the up-regulation of NOS 2 concomitant with secretion of NO•. These findings set the stage to determine how efficacious ABH will be in promoting NO•-mediating killing of B. anthracis.
Collapse
Affiliation(s)
- Pei Tsai
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD 21201, USA
| | | | | | | | | | | | | |
Collapse
|
11
|
Rubert Pérez C, López-Pérez D, Chmielewski J, Lipton M. Small molecule inhibitors of anthrax toxin-induced cytotoxicity targeted against protective antigen. Chem Biol Drug Des 2012; 79:260-9. [PMID: 22146079 DOI: 10.1111/j.1747-0285.2011.01285.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Two molecular scaffolds were designed using the CAVEAT molecular design package to inhibit the oligomerization of protective antigen (PA(63) ), a key protein component of anthrax toxin. The inhibitors were designed to prevent heptamerization of PA(63) by mimicking key residues of PA(63) needed for the intermolecular interactions that stabilize the heptamer. Using the scaffolds identified by CAVEAT, seven candidate inhibitors were synthesized and tested for their ability to inhibit anthrax toxin-induced cytotoxicity, with three of the agents demonstrating modest inhibition in murine J774A.1 macrophage cells.
Collapse
|
12
|
Terra JK, France B, Cote CK, Jenkins A, Bozue JA, Welkos SL, Bhargava R, Ho CL, Mehrabian M, Pan C, Lusis AJ, Davis RC, LeVine SM, Bradley KA. Allelic variation on murine chromosome 11 modifies host inflammatory responses and resistance to Bacillus anthracis. PLoS Pathog 2011; 7:e1002469. [PMID: 22241984 PMCID: PMC3248472 DOI: 10.1371/journal.ppat.1002469] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2011] [Accepted: 11/16/2011] [Indexed: 01/23/2023] Open
Abstract
Anthrax is a potentially fatal disease resulting from infection with Bacillus anthracis. The outcome of infection is influenced by pathogen-encoded virulence factors such as lethal toxin (LT), as well as by genetic variation within the host. To identify host genes controlling susceptibility to anthrax, a library of congenic mice consisting of strains with homozygous chromosomal segments from the LT-responsive CAST/Ei strain introgressed on a LT-resistant C57BL/6 (B6) background was screened for response to LT. Three congenic strains containing CAST/Ei regions of chromosome 11 were identified that displayed a rapid inflammatory response to LT similar to, but more severe than that driven by a LT-responsive allele of the inflammasome constituent NRLP1B. Importantly, increased response to LT in congenic mice correlated with greater resistance to infection by the Sterne strain of B. anthracis. The genomic region controlling the inflammatory response to LT was mapped to 66.36–74.67 Mb on chromosome 11, a region that encodes the LT-responsive CAST/Ei allele of Nlrp1b. However, known downstream effects of NLRP1B activation, including macrophage pyroptosis, cytokine release, and leukocyte infiltration could not fully explain the response to LT or the resistance to B. anthracis Sterne in congenic mice. Further, the exacerbated response in congenic mice is inherited in a recessive manner while the Nlrp1b-mediated response to LT is dominant. Finally, congenic mice displayed increased responsiveness in a model of sepsis compared with B6 mice. In total, these data suggest that allelic variation of one or more chromosome 11 genes in addition to Nlrp1b controls the severity of host response to multiple inflammatory stimuli and contributes to resistance to B. anthracis Sterne. Expression quantitative trait locus analysis revealed 25 genes within this region as high priority candidates for contributing to the host response to LT. We show that genetic variation within an 8.3 Mb region on mouse chromosome 11 controls host response to anthrax lethal toxin (LT) and resistance to infection by the Sterne strain of Bacillus anthracis. Specifically, congenic C57BL/6 mice in which this region of chromosome 11 is derived from a genetically divergent CAST/Ei strain presented with a rapid and strong innate immune response to LT and displayed increased survival following infection with Sterne spores. CAST/Ei chromosome 11 encodes a dominant LT-responsive allele of Nlrp1b that may partially account for the severe response to LT. However, the strength of this response was attenuated in mice with only one copy of chromosome 11 derived from CAST/Ei indicating the existence of a recessive modifier of the inflammatory response to LT. In addition, congenic mice displayed a pronounced immune response using an experimental model of sepsis, indicating that one or more genes within the chromosome 11 region control host response to multiple inflammatory stimuli. Analyzing the influence of allelic variation on gene expression identified 25 genes as candidates for controlling these responses. In summary, we report a genetic model to study inflammatory responses beneficial to the host during anthrax.
Collapse
Affiliation(s)
- Jill K Terra
- Department of Microbiology, Immunology, and Molecular Genetics, University of California at Los Angeles, Los Angeles, California, United States of America
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Bacillus anthracis spore interactions with mammalian cells: relationship between germination state and the outcome of in vitro. BMC Microbiol 2011; 11:46. [PMID: 21356113 PMCID: PMC3060849 DOI: 10.1186/1471-2180-11-46] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2010] [Accepted: 02/28/2011] [Indexed: 11/10/2022] Open
Abstract
Background During inhalational anthrax, internalization of Bacillus anthracis spores by host cells within the lung is believed to be a key step for initiating the transition from the localized to disseminated stages of infection. Despite compelling in vivo evidence that spores remain dormant within the bronchioalveolar spaces of the lungs, and germinate only after uptake into host cells, most in vitro studies of infection have been conducted under conditions that promote rapid germination of spores within the culture medium. Results Using an in vitro model of infection, we evaluated the influence of the germination state of B. anthracis spores, as controlled by defined culture conditions, on the outcome of infection. Spores prepared from B. anthracis Sterne 7702 germinated in a variety of common cell culture media supplemented with fetal bovine serum (FBS) while, in the absence of FBS, germination was strictly dependent on medium composition. RAW264.7 macrophage-like cells internalized spores to the same extent in either germinating or non-germinating media. However, significantly more viable, intracellular B. anthracis were recovered from cells infected under non-germinating conditions compared to germinating conditions. At the same time, RAW264.7 cells demonstrated a significant loss in viability when infected under non-germinating conditions. Conclusions These results suggest that the outcome of host cell infection is sensitive to the germination state of spores at the time of uptake. Moreover, this study demonstrates the efficacy of studying B. anthracis spore infection of host cells within a defined, non-germinating, in vitro environment.
Collapse
|
14
|
Porasuphatana S, Cao GL, Tsai P, Tavakkoli F, Huwar T, Baillie L, Cross AS, Shapiro P, Rosen GM. Bacillus anthracis endospores regulate ornithine decarboxylase and inducible nitric oxide synthase through ERK1/2 and p38 mitogen-activated protein kinases. Curr Microbiol 2010; 61:567-73. [PMID: 20440620 DOI: 10.1007/s00284-010-9654-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2010] [Accepted: 04/12/2010] [Indexed: 11/26/2022]
Abstract
Interactions between Bacillus anthracis (B. anthracis) and host cells are of particular interest given the implications of anthrax as a biological weapon. Inhaled B. anthracis endospores encounter alveolar macrophages as the first line of defense in the innate immune response. Yet, the consequences of this interaction remain unclear. We have demonstrated that B. anthracis uses arginase, inherent in the endospores, to reduce the ability of macrophages to produce nitric oxide ((•)NO) from inducible nitric oxide synthase (NOS2) by competing for L-arginine, producing L-ornithine at the expense of (•)NO. In the current study, we used genetically engineered B. anthracis endospores to evaluate the contribution of germination and the lethal toxin (LT) in mediating signaling pathways responsible for the induction of NOS2 and ornithine decarboxylase (ODC), which is the rate-limiting enzyme in the conversion of L-ornithine into polyamines. We found that induction of NOS2 and ODC expression in macrophages exposed to B. anthracis occurs through the activation of p38 and ERK1/2 MAP kinases, respectively. Optimal induction of NOS2 was observed following exposure to germination-competent endospores, whereas ODC induction occurred irrespective of the endospores' germination capabilities and was more prominent in macrophages exposed to endospores lacking LT. Our findings suggest that activation of kinase signaling cascades that determine macrophage defense responses against B. anthracis infection occurs through distinct mechanisms.
Collapse
Affiliation(s)
- Supatra Porasuphatana
- Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen, 40002, Thailand
| | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Averette KM, Pratt MR, Yang Y, Bassilian S, Whitelegge JP, Loo JA, Muir TW, Bradley KA. Anthrax lethal toxin induced lysosomal membrane permeabilization and cytosolic cathepsin release is Nlrp1b/Nalp1b-dependent. PLoS One 2009; 4:e7913. [PMID: 19924255 PMCID: PMC2775945 DOI: 10.1371/journal.pone.0007913] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2009] [Accepted: 10/18/2009] [Indexed: 12/15/2022] Open
Abstract
NOD-like receptors (NLRs) are a group of cytoplasmic molecules that recognize microbial invasion or 'danger signals'. Activation of NLRs can induce rapid caspase-1 dependent cell death termed pyroptosis, or a caspase-1 independent cell death termed pyronecrosis. Bacillus anthracis lethal toxin (LT), is recognized by a subset of alleles of the NLR protein Nlrp1b, resulting in pyroptotic cell death of macrophages and dendritic cells. Here we show that LT induces lysosomal membrane permeabilization (LMP). The presentation of LMP requires expression of an LT-responsive allele of Nlrp1b, and is blocked by proteasome inhibitors and heat shock, both of which prevent LT-mediated pyroptosis. Further the lysosomal protease cathepsin B is released into the cell cytosol and cathepsin inhibitors block LT-mediated cell death. These data reveal a role for lysosomal membrane permeabilization in the cellular response to bacterial pathogens and demonstrate a shared requirement for cytosolic relocalization of cathepsins in pyroptosis and pyronecrosis.
Collapse
Affiliation(s)
- Kathleen M. Averette
- Department of Microbiology, Immunology & Molecular Genetics, University of California Los Angeles, Los Angeles, California, United States of America
| | - Matthew R. Pratt
- Laboratory of Synthetic Protein Chemistry, The Rockefeller University, New York, New York, United States of America
| | - Yanan Yang
- Department of Chemistry and Biochemistry, University of California Los Angeles, Los Angeles, California, United States of America
| | - Sara Bassilian
- The Pasarow Mass Spectrometry Laboratory, The NPI-Semel Institute, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Julian P. Whitelegge
- The Pasarow Mass Spectrometry Laboratory, The NPI-Semel Institute, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Joseph A. Loo
- Department of Chemistry and Biochemistry, University of California Los Angeles, Los Angeles, California, United States of America
| | - Tom W. Muir
- Laboratory of Synthetic Protein Chemistry, The Rockefeller University, New York, New York, United States of America
| | - Kenneth A. Bradley
- Department of Microbiology, Immunology & Molecular Genetics, University of California Los Angeles, Los Angeles, California, United States of America
- * E-mail:
| |
Collapse
|
16
|
Hughes MA, Burns DL, Juris SJ, Tang WJ, Clement KH, Eaton LJ, Kelly-Cirino CD, McKee ML, Powell BS, Bishop BL, Rudge TL, Shine N, Verma A, Willis MS, Morse SA. The case for developing consensus standards for research in microbial pathogenesis: Bacillus anthracis toxins as an example. Infect Immun 2009; 77:4182-6. [PMID: 19651858 PMCID: PMC2747970 DOI: 10.1128/iai.00368-09] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Affiliation(s)
- Molly A Hughes
- Department of Medicine, Division of Infectious Diseases and International Health, University of Virginia Health System, P.O. Box 800513, Charlottesville, VA 22908, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Global transcriptional response to spermine, a component of the intramacrophage environment, reveals regulation of Francisella gene expression through insertion sequence elements. J Bacteriol 2009; 191:6855-64. [PMID: 19749055 DOI: 10.1128/jb.00995-09] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Tularemia is caused by the category A biodefense agent Francisella tularensis. This bacterium is associated with diverse environments and a plethora of arthropod and mammalian hosts. How F. tularensis adapts to these different conditions, particularly the eukaryotic intracellular environment in which it replicates, is poorly understood. Here, we demonstrate that the polyamines spermine and spermidine are environmental signals that alter bacterial stimulation of host cells. Genomewide analysis showed that F. tularensis LVS undergoes considerable changes in gene expression in response to spermine. Unexpectedly, analysis of gene expression showed that multiple members of two classes of Francisella insertion sequence (IS) elements, ISFtu1 and ISFtu2, and the genes adjacent to these elements were induced by spermine. Spermine was sufficient to activate transcription of these IS elements and of nearby genes in broth culture and in macrophages. Importantly, the virulent strain of F. tularensis, Schu S4, exhibited similar phenotypes of cytokine induction and gene regulation in response to spermine. Distinctions in gene expression changes between Schu S4 and LVS at one orthologous locus, however, correlated with differences in IS element location. Our results indicate that spermine and spermidine are novel triggers to alert F. tularensis of its eukaryotic host environment. The results reported here also identify an unexpected mechanism of gene regulation controlled by a spermine-responsive promoter contained within IS elements. Different arrangements of these mobile genetic elements among Francisella strains may contribute to virulence by conveying new expression patterns for genes from different strains.
Collapse
|
18
|
Dozmorov M, Wu W, Chakrabarty K, Booth JL, Hurst RE, Coggeshall KM, Metcalf JP. Gene expression profiling of human alveolar macrophages infected by B. anthracis spores demonstrates TNF-alpha and NF-kappab are key components of the innate immune response to the pathogen. BMC Infect Dis 2009; 9:152. [PMID: 19744333 PMCID: PMC2752459 DOI: 10.1186/1471-2334-9-152] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2009] [Accepted: 09/10/2009] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Bacillus anthracis, the etiologic agent of anthrax, has recently been used as an agent of bioterrorism. The innate immune system initially appears to contain the pathogen at the site of entry. Because the human alveolar macrophage (HAM) plays a key role in lung innate immune responses, studying the HAM response to B. anthracis is important in understanding the pathogenesis of the pulmonary form of this disease. METHODS In this paper, the transcriptional profile of B. anthracis spore-treated HAM was compared with that of mock-infected cells, and differentially expressed genes were identified by Affymetrix microarray analysis. A portion of the results were verified by Luminex protein analysis. RESULTS The majority of genes modulated by spores were upregulated, and a lesser number were downregulated. The differentially expressed genes were subjected to Ingenuity Pathway analysis, the Database for Annotation, Visualization and Integrated Discovery (DAVID) analysis, the Promoter Analysis and Interaction Network Toolset (PAINT) and Oncomine analysis. Among the upregulated genes, we identified a group of chemokine ligand, apoptosis, and, interestingly, keratin filament genes. Central hubs regulating the activated genes were TNF-alpha, NF-kappaB and their ligands/receptors. In addition to TNF-alpha, a broad range of cytokines was induced, and this was confirmed at the level of translation by Luminex multiplex protein analysis. PAINT analysis revealed that many of the genes affected by spores contain the binding site for c-Rel, a member of the NF-kappaB family of transcription factors. Other transcription regulatory elements contained in many of the upregulated genes were c-Myb, CP2, Barbie Box, E2F and CRE-BP1. However, many of the genes are poorly annotated, indicating that they represent novel functions. Four of the genes most highly regulated by spores have only previously been associated with head and neck and lung carcinomas. CONCLUSION The results demonstrate not only that TNF-alpha and NF-kappab are key components of the innate immune response to the pathogen, but also that a large part of the mechanisms by which the alveolar macrophage responds to B. anthracis are still unknown as many of the genes involved are poorly annotated.
Collapse
Affiliation(s)
- Mikhail Dozmorov
- Pulmonary and Critical Care Division, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| | | | | | | | | | | | | |
Collapse
|
19
|
Bacillus anthracis capsule activates caspase-1 and induces interleukin-1beta release from differentiated THP-1 and human monocyte-derived dendritic cells. Infect Immun 2009; 78:387-92. [PMID: 19737897 DOI: 10.1128/iai.00956-09] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The poly-gamma-d-glutamic acid (PGA) capsule is one of the major virulence factors of Bacillus anthracis, which causes a highly lethal infection. The antiphagocytic PGA capsule disguises the bacilli from immune surveillance and allows unimpeded growth of bacilli in the host. Recently, efforts have been made to include PGA as a component of anthrax vaccine; however, the innate immune response of PGA itself has been poorly investigated. In this study, we characterized the innate immune response elicited by PGA in the human monocytic cell line THP-1, which was differentiated into macrophages with phorbol 12-myristate 13-acetate (PMA) and human monocyte-derived dendritic cells (hMoDCs). PGA capsules were isolated from the culture supernatant of either the pXO1-cured strain of B. anthracis H9401 or B. licheniformis ATCC 9945a. PGA treatment of differentiated THP-1 cells and hMoDCs led to the specific extracellular release of interleukin-1beta (IL-1beta) in a dose-dependent manner. Evaluation of IL-1beta processing by Western blotting revealed that cleaved IL-1beta increased in THP-1 cells and hMoDCs after PGA treatment. Enhanced processing of IL-1beta directly correlated with increased activation of its upstream regulator, caspase-1, also known as IL-1beta-converting enzyme (ICE). The extracellular release of IL-1beta in response to PGA was ICE dependent, since the administration of an ICE inhibitor prior to PGA treatment blocked induction of IL-1beta. These results demonstrate that B. anthracis PGA elicits IL-1beta production through activation of ICE in PMA-differentiated THP-1 cells and hMoDCs, suggesting the potential for PGA as a therapeutic target for anthrax.
Collapse
|
20
|
Popova T, Espina V, Bailey C, Liotta L, Petricoin E, Popov S. Anthrax infection inhibits the AKT signaling involved in the E-cadherin-mediated adhesion of lung epithelial cells. ACTA ACUST UNITED AC 2009; 56:129-42. [PMID: 19416348 PMCID: PMC2734923 DOI: 10.1111/j.1574-695x.2009.00558.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The effect of anthrax infection on phosphoprotein signaling was studied in human small airway lung epithelial cells exposed to B. anthracis spores of the plasmidless dSterne strain in comparison with the Sterne strain containing the toxigenic plasmid (pXO1). The differential regulation of phosphorylation was found in the mitogen-activated protein kinase cascade (ERK, p38, and P90RSK), the PI3K cascade (AKT, GSK-3alpha/beta), and downstream in the case of the proapoptotic BAD and the transcription factor STAT3. Both strains stimulate phosphorylation of CREB and inhibit phosphorylation of 4E-BP1 required for activation of cap-dependent translation. Downregulation of the survival AKT phosphorylation by the Sterne strain inhibits the process of Ca(2+)-dependent homophilic interaction of E-cadherin (EC) upon formation or repair of cell-cell contacts. Both lethal and edema toxins produced by the Sterne strain inhibit the AKT phosphorylation induced during the EC-mediated signaling. Activity of ERK1/2 and p38 inhibitors indicates that inhibition of AKT phosphorylation takes place through the ERK1/2-PI3K crosstalk. In Sterne spore-challenged mice, a specific inhibitor of PI3K/AKT, wortmannin, accelerates the lethal outcome, and reduction of AKT phosphorylation in the circulating blood cells coincides with the death of animals. We conclude that the PI3K/AKT pathway controlling the integrity of epithelium plays an important survival role in anthrax infection.
Collapse
Affiliation(s)
- Taissia Popova
- National Center for Biodefense and Infectious Diseases, George Mason University, Manassas, VA 20110, USA
| | | | | | | | | | | |
Collapse
|
21
|
Chou PJJ, Newton CA, Perkins I, Friedman H, Klein TW. Suppression of dendritic cell activation by anthrax lethal toxin and edema toxin depends on multiple factors including cell source, stimulus used, and function tested. DNA Cell Biol 2009; 27:637-48. [PMID: 18821847 DOI: 10.1089/dna.2008.0760] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Bacillus anthracis produces lethal toxin (LT) and edema toxin (ET), and they suppress the function of LPS-stimulated dendritic cells (DCs). Because DCs respond differently to various microbial stimuli, we compared toxin effects in bone marrow DCs stimulated with either LPS or Legionella pneumophila (Lp). LT, not ET, was more toxic for cells from BALB/c than from C57BL/6 (B6) as measured by 7-AAD uptake; however, ET suppressed CD11c expression. LT suppressed IL-12, IL-6, and TNF-alpha in cells from BALB/c and B6 mice but increased IL-1beta in LPS-stimulated cultures. ET also suppressed IL-12 and TNF-alpha, but increased IL-6 and IL-1beta in Lp-stimulated cells from B6. Regarding maturation marker expression, LT increased MHCII and CD86 while suppressing CD40 and CD80; ET generally decreased marker expression across all groups. We conclude that the suppression of cytokine production by anthrax toxins is dependent on variables, including the source of the DCs, the type of stimulus and cytokine measured, and the individual toxin tested. However, LT and ET enhancement or suppression of maturation marker expression is more related to the marker studied than the stimuli or cell source. Anthrax toxins are not uniformly suppressive of DC function but instead can increase function under defined conditions.
Collapse
Affiliation(s)
- Ping-Jen Joe Chou
- Department of Molecular Medicine, University of South Florida College of Medicine, Tampa, Florida 33612, USA.
| | | | | | | | | |
Collapse
|
22
|
Bradburne C, Chung MC, Zong Q, Schlauch K, Liu D, Popova T, Popova A, Bailey C, Soppet D, Popov S. Transcriptional and apoptotic responses of THP-1 cells to challenge with toxigenic, and non-toxigenic Bacillus anthracis. BMC Immunol 2008; 9:67. [PMID: 19014542 PMCID: PMC2613145 DOI: 10.1186/1471-2172-9-67] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2007] [Accepted: 11/13/2008] [Indexed: 11/10/2022] Open
Abstract
Background Bacillus anthracis secretes several virulence factors targeting different host organs and cell types during inhalational anthrax infection. The bacterial expression of a key virulence factor, lethal toxin (LeTx) is closely tied to another factor, edema toxin (EdTx). Both are transcribed on the same virulence plasmid (pXO1) and both have been the subject of much individual study. Their combined effect during virulent anthrax likely modulates both the global transcriptional and the phenotypic response of macrophages and phagocytes. In fact, responses brought about by the toxins may be different than each of their individual effects. Results Here we report the transcriptional and apoptotic responses of the macrophage-like phagocytic cell line THP-1 exposed to B. anthracis Sterne (pXO1+) spores, and B. anthracis Δ Sterne (pXO1-) spores. These cells are resistant to LeTx-induced cytolysis, a phenotype seen in macrophages from several mouse strains which are sensitive to toxigenic anthrax infection. Our results indicate that the pXO1-containing strain induces higher pro-inflammatory transcriptional responses during the first 4 hours of interaction with bacterium, evident in the upregulation of several genes relevant to Nf-κB, phosphatases, prostaglandins, and TNF-α, along with decreases in expression levels of genes for mitochondrial components. Both bacterial strains induce apoptosis, but in the toxigenic strain-challenged cells, apoptosis is delayed. Conclusion This delay in apoptosis occurs despite the much higher level of TNF-α secretion induced by the toxigenic-strain challenge. Interestingly, CFLAR, an important apoptotic inhibitor which blocks apoptosis induced by large amounts of extracellular TNF-α, is upregulated significantly during toxigenic-strain infection, but not at all during non-toxigenic-strain infection, indicating that it may play a role in blocking or delaying TNF-α-mediated apoptosis. The suppression of apoptosis by the toxigenic anthrax strain is consistent with the notion that apoptosis itself may represent a protective host cell response.
Collapse
Affiliation(s)
- Christopher Bradburne
- Center for Bio/Molecular Science and Engineering Code 6900, US Naval Research Laboratory, Washington DC, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Zhang C, Crasta O, Cammer S, Will R, Kenyon R, Sullivan D, Yu Q, Sun W, Jha R, Liu D, Xue T, Zhang Y, Moore M, McGarvey P, Huang H, Chen Y, Zhang J, Mazumder R, Wu C, Sobral B. An emerging cyberinfrastructure for biodefense pathogen and pathogen-host data. Nucleic Acids Res 2008; 36:D884-91. [PMID: 17984082 PMCID: PMC2239001 DOI: 10.1093/nar/gkm903] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2007] [Revised: 10/04/2007] [Accepted: 10/05/2007] [Indexed: 01/07/2023] Open
Abstract
The NIAID-funded Biodefense Proteomics Resource Center (RC) provides storage, dissemination, visualization and analysis capabilities for the experimental data deposited by seven Proteomics Research Centers (PRCs). The data and its publication is to support researchers working to discover candidates for the next generation of vaccines, therapeutics and diagnostics against NIAID's Category A, B and C priority pathogens. The data includes transcriptional profiles, protein profiles, protein structural data and host-pathogen protein interactions, in the context of the pathogen life cycle in vivo and in vitro. The database has stored and supported host or pathogen data derived from Bacillus, Brucella, Cryptosporidium, Salmonella, SARS, Toxoplasma, Vibrio and Yersinia, human tissue libraries, and mouse macrophages. These publicly available data cover diverse data types such as mass spectrometry, yeast two-hybrid (Y2H), gene expression profiles, X-ray and NMR determined protein structures and protein expression clones. The growing database covers over 23 000 unique genes/proteins from different experiments and organisms. All of the genes/proteins are annotated and integrated across experiments using UniProt Knowledgebase (UniProtKB) accession numbers. The web-interface for the database enables searching, querying and downloading at the level of experiment, group and individual gene(s)/protein(s) via UniProtKB accession numbers or protein function keywords. The system is accessible at http://www.proteomicsresource.org/.
Collapse
Affiliation(s)
- C. Zhang
- Virginia Bioinformatics Institute at Virginia Polytechnic Institute and State University, Washington Street (0477), Blacksburg, VA 24061, Social & Scientific Systems, Inc., 8757 Georgia Avenue, 12th Floor Silver Spring, MD 20910 and Protein Information Resource, Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, 3300 Whitehaven Street NW, Suite 1200, Washington, DC 20007, USA
| | - O. Crasta
- Virginia Bioinformatics Institute at Virginia Polytechnic Institute and State University, Washington Street (0477), Blacksburg, VA 24061, Social & Scientific Systems, Inc., 8757 Georgia Avenue, 12th Floor Silver Spring, MD 20910 and Protein Information Resource, Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, 3300 Whitehaven Street NW, Suite 1200, Washington, DC 20007, USA
| | - S. Cammer
- Virginia Bioinformatics Institute at Virginia Polytechnic Institute and State University, Washington Street (0477), Blacksburg, VA 24061, Social & Scientific Systems, Inc., 8757 Georgia Avenue, 12th Floor Silver Spring, MD 20910 and Protein Information Resource, Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, 3300 Whitehaven Street NW, Suite 1200, Washington, DC 20007, USA
| | - R. Will
- Virginia Bioinformatics Institute at Virginia Polytechnic Institute and State University, Washington Street (0477), Blacksburg, VA 24061, Social & Scientific Systems, Inc., 8757 Georgia Avenue, 12th Floor Silver Spring, MD 20910 and Protein Information Resource, Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, 3300 Whitehaven Street NW, Suite 1200, Washington, DC 20007, USA
| | - R. Kenyon
- Virginia Bioinformatics Institute at Virginia Polytechnic Institute and State University, Washington Street (0477), Blacksburg, VA 24061, Social & Scientific Systems, Inc., 8757 Georgia Avenue, 12th Floor Silver Spring, MD 20910 and Protein Information Resource, Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, 3300 Whitehaven Street NW, Suite 1200, Washington, DC 20007, USA
| | - D. Sullivan
- Virginia Bioinformatics Institute at Virginia Polytechnic Institute and State University, Washington Street (0477), Blacksburg, VA 24061, Social & Scientific Systems, Inc., 8757 Georgia Avenue, 12th Floor Silver Spring, MD 20910 and Protein Information Resource, Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, 3300 Whitehaven Street NW, Suite 1200, Washington, DC 20007, USA
| | - Q. Yu
- Virginia Bioinformatics Institute at Virginia Polytechnic Institute and State University, Washington Street (0477), Blacksburg, VA 24061, Social & Scientific Systems, Inc., 8757 Georgia Avenue, 12th Floor Silver Spring, MD 20910 and Protein Information Resource, Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, 3300 Whitehaven Street NW, Suite 1200, Washington, DC 20007, USA
| | - W. Sun
- Virginia Bioinformatics Institute at Virginia Polytechnic Institute and State University, Washington Street (0477), Blacksburg, VA 24061, Social & Scientific Systems, Inc., 8757 Georgia Avenue, 12th Floor Silver Spring, MD 20910 and Protein Information Resource, Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, 3300 Whitehaven Street NW, Suite 1200, Washington, DC 20007, USA
| | - R. Jha
- Virginia Bioinformatics Institute at Virginia Polytechnic Institute and State University, Washington Street (0477), Blacksburg, VA 24061, Social & Scientific Systems, Inc., 8757 Georgia Avenue, 12th Floor Silver Spring, MD 20910 and Protein Information Resource, Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, 3300 Whitehaven Street NW, Suite 1200, Washington, DC 20007, USA
| | - D. Liu
- Virginia Bioinformatics Institute at Virginia Polytechnic Institute and State University, Washington Street (0477), Blacksburg, VA 24061, Social & Scientific Systems, Inc., 8757 Georgia Avenue, 12th Floor Silver Spring, MD 20910 and Protein Information Resource, Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, 3300 Whitehaven Street NW, Suite 1200, Washington, DC 20007, USA
| | - T. Xue
- Virginia Bioinformatics Institute at Virginia Polytechnic Institute and State University, Washington Street (0477), Blacksburg, VA 24061, Social & Scientific Systems, Inc., 8757 Georgia Avenue, 12th Floor Silver Spring, MD 20910 and Protein Information Resource, Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, 3300 Whitehaven Street NW, Suite 1200, Washington, DC 20007, USA
| | - Y. Zhang
- Virginia Bioinformatics Institute at Virginia Polytechnic Institute and State University, Washington Street (0477), Blacksburg, VA 24061, Social & Scientific Systems, Inc., 8757 Georgia Avenue, 12th Floor Silver Spring, MD 20910 and Protein Information Resource, Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, 3300 Whitehaven Street NW, Suite 1200, Washington, DC 20007, USA
| | - M. Moore
- Virginia Bioinformatics Institute at Virginia Polytechnic Institute and State University, Washington Street (0477), Blacksburg, VA 24061, Social & Scientific Systems, Inc., 8757 Georgia Avenue, 12th Floor Silver Spring, MD 20910 and Protein Information Resource, Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, 3300 Whitehaven Street NW, Suite 1200, Washington, DC 20007, USA
| | - P. McGarvey
- Virginia Bioinformatics Institute at Virginia Polytechnic Institute and State University, Washington Street (0477), Blacksburg, VA 24061, Social & Scientific Systems, Inc., 8757 Georgia Avenue, 12th Floor Silver Spring, MD 20910 and Protein Information Resource, Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, 3300 Whitehaven Street NW, Suite 1200, Washington, DC 20007, USA
| | - H. Huang
- Virginia Bioinformatics Institute at Virginia Polytechnic Institute and State University, Washington Street (0477), Blacksburg, VA 24061, Social & Scientific Systems, Inc., 8757 Georgia Avenue, 12th Floor Silver Spring, MD 20910 and Protein Information Resource, Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, 3300 Whitehaven Street NW, Suite 1200, Washington, DC 20007, USA
| | - Y. Chen
- Virginia Bioinformatics Institute at Virginia Polytechnic Institute and State University, Washington Street (0477), Blacksburg, VA 24061, Social & Scientific Systems, Inc., 8757 Georgia Avenue, 12th Floor Silver Spring, MD 20910 and Protein Information Resource, Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, 3300 Whitehaven Street NW, Suite 1200, Washington, DC 20007, USA
| | - J. Zhang
- Virginia Bioinformatics Institute at Virginia Polytechnic Institute and State University, Washington Street (0477), Blacksburg, VA 24061, Social & Scientific Systems, Inc., 8757 Georgia Avenue, 12th Floor Silver Spring, MD 20910 and Protein Information Resource, Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, 3300 Whitehaven Street NW, Suite 1200, Washington, DC 20007, USA
| | - R. Mazumder
- Virginia Bioinformatics Institute at Virginia Polytechnic Institute and State University, Washington Street (0477), Blacksburg, VA 24061, Social & Scientific Systems, Inc., 8757 Georgia Avenue, 12th Floor Silver Spring, MD 20910 and Protein Information Resource, Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, 3300 Whitehaven Street NW, Suite 1200, Washington, DC 20007, USA
| | - C. Wu
- Virginia Bioinformatics Institute at Virginia Polytechnic Institute and State University, Washington Street (0477), Blacksburg, VA 24061, Social & Scientific Systems, Inc., 8757 Georgia Avenue, 12th Floor Silver Spring, MD 20910 and Protein Information Resource, Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, 3300 Whitehaven Street NW, Suite 1200, Washington, DC 20007, USA
| | - B. Sobral
- Virginia Bioinformatics Institute at Virginia Polytechnic Institute and State University, Washington Street (0477), Blacksburg, VA 24061, Social & Scientific Systems, Inc., 8757 Georgia Avenue, 12th Floor Silver Spring, MD 20910 and Protein Information Resource, Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, 3300 Whitehaven Street NW, Suite 1200, Washington, DC 20007, USA
| |
Collapse
|
24
|
Passalacqua KD, Bergman NH. Bacillus anthracis: interactions with the host and establishment of inhalational anthrax. Future Microbiol 2007; 1:397-415. [PMID: 17661631 DOI: 10.2217/17460913.1.4.397] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Due to its potential as a bioweapon, Bacillus anthracis has received a great deal of attention in recent years, and a significant effort has been devoted to understanding how this organism causes anthrax. There has been a particular focus on the inhalational form of the disease, and studies over the past several years have painted an increasingly complex picture of how B. anthracis enters the mammalian host, survives the host's defense mechanisms, disseminates throughout the body and causes death. This article reviews recent advances in these areas, with a focus on how the bacterium interacts with its host in establishing infection and causing anthrax.
Collapse
Affiliation(s)
- Karla D Passalacqua
- University of Michigan Medical School, Department of Microbiology & Immunology, Ann Arbor, MI 48109, USA.
| | | |
Collapse
|
25
|
Ojha S, Kostrzynska M. Examination of animal and zoonotic pathogens using microarrays. Vet Res 2007; 39:4. [DOI: 10.1051/vetres:2007042] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2006] [Accepted: 07/27/2007] [Indexed: 01/13/2023] Open
|
26
|
Moen ST, Yeager LA, Lawrence WS, Ponce C, Galindo CL, Garner HR, Baze WB, Suarez G, Peterson JW, Chopra AK. Transcriptional profiling of murine organ genes in response to infection with Bacillus anthracis Ames spores. Microb Pathog 2007; 44:293-310. [PMID: 18037264 DOI: 10.1016/j.micpath.2007.10.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2007] [Revised: 10/11/2007] [Accepted: 10/11/2007] [Indexed: 10/22/2022]
Abstract
Bacillus anthracis is the Gram-positive, spore-forming etiological agent of anthrax, an affliction studied because of its importance as a potential bioweapon. Although in vitro transcriptional responses of macrophages to either spore or anthrax toxins have been previously reported, little is known regarding the impact of infection on gene expression in host tissues. We infected Swiss-Webster mice intranasally with 5 LD(50) of B. anthracis-virulent Ames spores and observed the global transcriptional profiles of various tissues over a 48 h time period. RNA was extracted from spleen, lung, and heart tissues of infected and control mice and examined by Affymetrix GeneChip analysis. Approximately 580 host genes were significantly over or under expressed among the lung, spleen, and heart tissues at 8 and 48 h time points. Expression of genes encoding for surfactant and major histocompatibility complex (MHC) presentation was diminished during the early phase of infection in lungs. By 48 h, a significant number of genes were modulated in the heart, including up-regulation of calcium-binding-related gene expression, and down-regulation of multiple genes related to cell adhesion, formation of the extracellular matrix, and the cell cytoskeleton. Interestingly, the spleen 8h post-infection showed striking increases in the expression of genes that encode hydrolytic enzymes, and these levels remained elevated throughout infection. Further, genes involving antigen presentation and interferon responses were down-regulated in the spleen at 8 h. In late stages of infection, splenic genes related to the inflammatory response were up-regulated. This study is the first to describe the in vivo global transcriptional response of multiple organs during inhalational anthrax. Although numerous genes related to the host immunological response and certain protection mechanisms were up-regulated in these organs, a vast list of genes important for fully developing and maintaining this response were decreased. Additionally, the lung, spleen, and heart showed differential responses to the infection, further validating the demand for a better understanding of anthrax pathogenesis in order to design therapies against novel targets.
Collapse
Affiliation(s)
- Scott T Moen
- Department of Microbiology and Immunology, Medical Research Building, 301 University Boulevard, University of Texas Medical Branch, Galveston, TX 77555-1070, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Wheeler JX, Whiting G, Rijpkema S. Proteomic analysis of the response of the human neutrophil-like cell line NB-4 after exposure to anthrax lethal toxin. Proteomics Clin Appl 2007; 1:1266-79. [PMID: 21136624 DOI: 10.1002/prca.200700074] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2007] [Indexed: 12/19/2022]
Abstract
We used 2-D DIGE to analyze the early response of NB-4 cells, a human promyelotic leukemia cell line, exposed to lethal toxin from Bacillus anthracis at the proteome level. After a 2 h exposure, cells were still viable and 43% of spots (n = 1042) showed a significant change in protein level. We identified 59 spots whose expression had changed significantly, and these reflected cytoskeleton damage, mitochondrial lysis and endoplasmic reticulum stress. Actin filament assembly was disrupted as evidenced by an increase in both actin subunits and phosphorylated cofilin, whilst levels of tropomyosin, tropomodulin and actin related protein 2/3 complex subunit decreased. Lower levels of ATP synthase subunits and mitochondrial inner membrane protein were identified as markers of mitochondrial lysis. Levels of various stress response proteins rose and, uniquely, levels of Ca(2+) binding proteins such as translationally controlled tumor protein rose and hippocalcin-like protein 1 decreased. This response may have mitigated effects brought about by mitochondrial lysis and endoplasmic reticulum stress, and delayed or prevented apoptosis in NB-4 cells. These results resemble findings of similar proteomics studies in murine macrophages, although quantitative differences were observed.
Collapse
Affiliation(s)
- Jun X Wheeler
- Laboratory of Molecular Structure, National Institute for Biological Standards and Control, Potters Bar, UK
| | | | | |
Collapse
|
28
|
Bergman NH, Anderson EC, Swenson EE, Janes BK, Fisher N, Niemeyer MM, Miyoshi AD, Hanna PC. Transcriptional profiling of Bacillus anthracis during infection of host macrophages. Infect Immun 2007; 75:3434-44. [PMID: 17470545 PMCID: PMC1932917 DOI: 10.1128/iai.01345-06] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2006] [Revised: 10/03/2006] [Accepted: 04/18/2007] [Indexed: 01/27/2023] Open
Abstract
The interaction between Bacillus anthracis and the mammalian phagocyte is one of the central stages in the progression of inhalational anthrax, and it is commonly believed that the host cell plays a key role in facilitating germination and dissemination of inhaled B. anthracis spores. Given this, a detailed definition of the survival strategies used by B. anthracis within the phagocyte is critical for our understanding of anthrax. In this study, we report the first genome-wide analysis of B. anthracis gene expression during infection of host phagocytes. We developed a technique for specific isolation of bacterial RNA from within infected murine macrophages, and we used custom B. anthracis microarrays to characterize the expression patterns occurring within intracellular bacteria throughout infection of the host phagocyte. We found that B. anthracis adapts very quickly to the intracellular environment, and our analyses identified metabolic pathways that appear to be important to the bacterium during intracellular growth, as well as individual genes that show significant induction in vivo. We used quantitative reverse transcription-PCR to verify that the expression trends that we observed by microarray analysis were valid, and we chose one gene (GBAA1941, encoding a putative transcriptional regulator) for further characterization. A deletion strain missing this gene showed no phenotype in vitro but was significantly attenuated in a mouse model of inhalational anthrax, suggesting that the microarray data described here provide not only the first comprehensive view of how B. anthracis survives within the host cell but also a number of promising leads for further research in anthrax.
Collapse
Affiliation(s)
- Nicholas H Bergman
- Bioinformatics Program and Department of Microbiology & Immunology, University of Michigan Medical School, 6706 Medical Sciences Bldg. II, 1150 W. Medical Center Drive, Ann Arbor, MI 48109-0620, USA.
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Habib F, Johnson AD, Bundschuh R, Janies D. Large scale genotype-phenotype correlation analysis based on phylogenetic trees. Bioinformatics 2007; 23:785-8. [PMID: 17267431 DOI: 10.1093/bioinformatics/btm003] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
We provide two methods for identifying changes in genotype that are correlated with changes in a phenotype implied by phylogenetic trees. The first method, VENN, works when the number of branches over which the change occurred are modest. VENN looks for genetic changes that are completely penetrant with phenotype changes on a tree. The second method, CCTSWEEP, allows for a partial matching between changes in phenotypes and genotypes and provides a score for each change using Maddison's concentrated changes test. The mutations that are highly correlated with phenotypic change can be ranked by score. We use these methods to find SNPs correlated with resistance to Bacillus anthracis in inbred mouse strains. Our findings are consistent with the current biological literature, and also suggest potential novel candidate genes.
Collapse
Affiliation(s)
- Farhat Habib
- Department of Physics, The Ohio State University, Columbus, OH 43210, USA.
| | | | | | | |
Collapse
|
30
|
Tournier JN, Quesnel-Hellmann A, Cleret A, Vidal DR. Contribution of toxins to the pathogenesis of inhalational anthrax. Cell Microbiol 2007; 9:555-65. [PMID: 17223930 DOI: 10.1111/j.1462-5822.2006.00866.x] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Inhalational anthrax is a life-threatening infectious disease of considerable concern, especially as a potential bioterrorism agent. Progress is gradually being made towards understanding the mechanisms used by Bacillus anthracis to escape the immune system and to induce severe septicaemia associated with toxaemia and leading to death. Recent advances in fundamental research have revealed previously unsuspected roles for toxins in various cell types. We summarize here pathological data for animal models and macroscopic histological examination data from recent clinical records, which we link to the effects of toxins. We describe three major steps in infection: (i) an invasion phase in the lung, during which toxins have short-distance effects on lung phagocytes; (ii) a phase of bacillus proliferation in the mediastinal lymph nodes, with local effects of toxins; and (iii) a terminal, diffusion phase, characterized by a high blood bacterial load and by long-distance effects of toxins, leading to host death. The pathophysiology of inhalational anthrax thus involves interactions between toxins and various cell partners, throughout the course of infection.
Collapse
Affiliation(s)
- Jean-Nicolas Tournier
- Pôle interactions hôte-pathogènes, Département de biologie des agents transmissibles, CRSSA, F-38702 La Tronche cedex, France.
| | | | | | | |
Collapse
|
31
|
Baldari CT, Tonello F, Paccani SR, Montecucco C. Anthrax toxins: a paradigm of bacterial immune suppression. Trends Immunol 2006; 27:434-40. [PMID: 16861036 DOI: 10.1016/j.it.2006.07.002] [Citation(s) in RCA: 136] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2006] [Revised: 06/20/2006] [Accepted: 07/07/2006] [Indexed: 12/17/2022]
Abstract
Several species of microorganism have developed immune evasion and/or immunosuppression strategies. Bacillus anthracis secretes two toxins, edema toxin and lethal toxin, that enter the cytosol of almost every cell type, including the cells of the innate and adaptive immune systems, and subvert cell signaling. Edema toxin causes a consistent elevation of cyclic adenosine monophosphate, whereas lethal toxin cleaves most isoforms of mitogen-activated protein kinase kinases. In a concerted manner, these toxins alter major signaling pathways involved in the development of immune-cell effector functions, with the inhibition of bacterial clearance by phagocytes and of B. anthracis-specific responses. Thus, B. anthracis can invade the host, with ensuing massive bacteremia and toxemia. Here, we review the specific effects of B. anthracis on neutrophils, macrophages, dendritic cells, T- and B-lymphocytes.
Collapse
Affiliation(s)
- Cosima T Baldari
- Department of Evolutionary Biology, University of Siena, 53100, Siena, Italy
| | | | | | | |
Collapse
|
32
|
Chakrabarty K, Wu W, Booth JL, Duggan ES, Coggeshall KM, Metcalf JP. Bacillus anthracis spores stimulate cytokine and chemokine innate immune responses in human alveolar macrophages through multiple mitogen-activated protein kinase pathways. Infect Immun 2006; 74:4430-8. [PMID: 16861629 PMCID: PMC1539616 DOI: 10.1128/iai.00446-06] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Contact with the human alveolar macrophage plays a key role in the innate immune response to Bacillus anthracis spores. Because there is a significant delay between the initial contact of the spore with the host and clinical evidence of disease, there appears to be temporary containment of the pathogen by the innate immune system. Therefore, the early macrophage response to Bacillus anthracis exposure is important in understanding the pathogenesis of this disease. In this paper, we studied the initial events after exposure to spores, beginning with the rapid internalization of spores by the macrophages. Spore exposure rapidly activated the mitogen-activated protein kinase signaling pathways extracellular signal-regulated kinase, c-Jun-NH2-terminal kinase, and p38. This was followed by the transcriptional activation of cytokine and primarily monocyte chemokine genes as determined by RNase protection assays. Transcriptional induction is reflected at the translational level, as interleukin-1alpha (IL-1alpha), IL-1beta, IL-6, and tumor necrosis factor alpha (TNF-alpha) cytokine protein levels were markedly elevated as determined by enzyme-linked immunosorbent assay. Induction of IL-6 and TNF-alpha, and, to a lesser extent, IL-1alpha and IL-1beta, was partially inhibited by the blockade of individual mitogen-activated protein kinases, while the complete inhibition of cytokine induction was achieved when multiple signaling pathway inhibitors were used. Taken together, these data clearly show activation of the innate immune system in human alveolar macrophages by Bacillus anthracis spores. The data also show that multiple signaling pathways are involved in this cytokine response. This report is the first comprehensive examination of this process in primary human alveolar macrophages.
Collapse
Affiliation(s)
- Kaushik Chakrabarty
- Pulmonary and Critical Care Division, Department of Medicine, University of Oklahoma Health Sciences Center, RM 425, RP1, 800 N. Research Pkwy., Oklahoma City, Oklahoma 73104, USA
| | | | | | | | | | | |
Collapse
|
33
|
Comer JE, Galindo CL, Zhang F, Wenglikowski AM, Bush KL, Garner HR, Peterson JW, Chopra AK. Murine macrophage transcriptional and functional responses to Bacillus anthracis edema toxin. Microb Pathog 2006; 41:96-110. [PMID: 16846716 DOI: 10.1016/j.micpath.2006.05.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2006] [Revised: 05/02/2006] [Accepted: 05/02/2006] [Indexed: 01/29/2023]
Abstract
Edema toxin (EdTx), which is a combination of edema factor and a binding moiety (protective antigen), is produced by Bacillus anthracis, the etiological agent of anthrax. EdTx is an adenylyl cyclase enzyme that converts adenosine triphosphate to adenosine-3',5'-monophosphate, resulting in interstitial edema seen in anthrax patients. We used GeneChip analysis to examine global transcriptional profiles of EdTx-treated RAW 264.7 murine macrophage-like cells and identified 71 and 259 genes whose expression was significantly altered by the toxin at 3 and 6h, respectively. Alteration in the expression levels of selected genes was confirmed by real time-reverse transcriptase polymerase chain reaction. The genes with up-regulated expression in macrophages in response to EdTx-treatment were known to be involved in inflammatory responses, regulation of apoptosis, adhesion, immune cell activation, and transcription regulation. Additionally, GeneChip analysis results implied that EdTx-induced activation of activator protein-1 (AP-1) and CAAAT/enhancer-binding protein-beta (C/EBP-beta). Gel shift assays were therefore performed, and an increase in the activities of both of these transcription factors was observed within 30 min. EdTx also inhibited tumor necrosis factor alpha production and crippled the phagocytic ability of the macrophages. This is the first report detailing the host cell global transcriptional responses to EdTx.
Collapse
Affiliation(s)
- Jason E Comer
- Department of Microbiology and Immunology, The University of Texas Medical Branch, Galveston, TX 77555-1070, USA
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Li T, Wang HL, Shi ZX, Feng EL, Liu RY, Huang LY. Germination process of Bacillus anthracis endospores within macrophage RAW264.7. Shijie Huaren Xiaohua Zazhi 2005; 13:1540-1543. [DOI: 10.11569/wcjd.v13.i13.1540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To confirm the germination process of Bacillus anthracis A16R endospores within murine macrophage RAW264.7
METHODS: Macrophage RAW264.7 cells were infected by Bacillus anthrax A16R (pXO2-) spores at a multiplicity of infection (MOI) of 20∶1. Then the cells were harvested at different time points (1, 2, 2.5, 3, 3.5, 4, 4.5, 5, 5.5, 6, 6.5, 7, 7.5 and 8 h after infection). The growth of infected cells was observed under light microscope by fuchsin basic methylene blue staining.
RESULTS: The endospores began to germinate and develop into vegetative bodies 2 to 2.5 h after infection. The vegetative bodies entered the phase of binary fission 3.5 to 4 h after infection. At 5 to 5.5 h, the bacillus proliferated into exponential phase andthe macrophages began to lyse 7 to 8 h after infected.
CONCLUSION: For the fisrt time, fuchsin basic methylene blue staining is used to study the germination process of Bacillus anthracis endospores within macrophage RAW264.7.
Collapse
|
35
|
Current Awareness on Comparative and Functional Genomics. Comp Funct Genomics 2005. [PMCID: PMC2447509 DOI: 10.1002/cfg.490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
|