1
|
Casanova JL, Abel L. The Microbe, the Infection Enigma, and the Host. Annu Rev Microbiol 2024; 78:103-124. [PMID: 38986133 DOI: 10.1146/annurev-micro-092123-022855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/12/2024]
Abstract
Human infectious diseases are unique in that the discovery of their environmental trigger, the microbe, was sufficient to drive the development of extraordinarily effective principles and tools for their prevention or cure. This unique medical prowess has outpaced, and perhaps even hindered, the development of scientific progress of equal magnitude in the biological understanding of infectious diseases. Indeed, the hope kindled by the germ theory of disease was rapidly subdued by the infection enigma, in need of a host solution, when it was realized that most individuals infected with most infectious agents continue to do well. The root causes of disease and death in the unhappy few remained unclear. While canonical approaches in vitro (cellular microbiology), in vivo (animal models), and in natura (clinical studies) analyzed the consequences of infection with a microbe, considered to be the cause of disease, in cells, tissues, or organisms seen as a uniform host, alternative approaches searched for preexisting causes of disease, particularly human genetic and immunological determinants in populations of diverse individuals infected with a trigger microbe.
Collapse
Affiliation(s)
- Jean-Laurent Casanova
- Howard Hughes Medical Institute, New York, NY, USA
- Department of Pediatrics, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
- Laboratory of Human Genetics of Infectious Diseases, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA;
| | - Laurent Abel
- Paris Cité University, Imagine Institute, Paris, France
- Laboratory of Human Genetics of Infectious Diseases, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA;
| |
Collapse
|
2
|
Kumadaki K, Suzuki N, Tatematsu K, Doi Y, Tsukamoto K. Comparison of Biological Activities of BafA Family Autotransporters within Bartonella Species Derived from Cats and Rodents. Infect Immun 2023; 91:e0018622. [PMID: 36744895 PMCID: PMC10016083 DOI: 10.1128/iai.00186-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Bartonella species are hemotropic, facultative intracellular bacteria, some of which cause zoonoses, that are widely disseminated among many mammals, including humans. During infection in humans, vascular endothelial cells play a crucial role as a replicative niche for Bartonella, and some are capable of promoting vascular proliferation. Along with well-studied pathogenic factors such as a trimeric autotransporter adhesin BadA or VirB/D4 type IV secretion system, bacteria-secreted protein BafA is also involved in Bartonella-induced vasoproliferation. Genes encoding BafA orthologs have been found in the genomes of most Bartonella species, but their functionality remains unclear. In this study, we focused on three cat-derived zoonotic species (B. henselae, B. koehlerae, and B. clarridgeiae) and two rodent-derived species (B. grahamii and B. doshiae) and compared the activity of BafA derived from each species. Recombinant BafA proteins of B. henselae, B. koehlerae, B. clarridgeiae, and B. grahamii, species that also cause human disease, induced cell proliferation and tube formation in cultured endothelial cells, while BafA derived from B. doshiae, a species that is rarely found in humans, showed neither activity. Additionally, treatment of cells with these BafA proteins increased phosphorylation of both vascular endothelial growth factor receptor 2 and extracellular signal-regulated kinase 1/2, with the exception of B. doshiae BafA. Differential bafA mRNA expression and BafA secretion among the species likely contributed to the differences in the cell proliferation phenotype of the bacteria-infected cells. These findings suggest that the biological activity of BafA may be involved in the infectivity or pathogenicity of Bartonella species in humans.
Collapse
Affiliation(s)
- Kayo Kumadaki
- Department of Microbiology, Fujita Health University School of Medicine, Toyoake, Aichi, Japan
| | - Natsumi Suzuki
- Department of Microbiology, Fujita Health University School of Medicine, Toyoake, Aichi, Japan
| | - Kaoru Tatematsu
- Department of Microbiology, Fujita Health University School of Medicine, Toyoake, Aichi, Japan
| | - Yohei Doi
- Department of Microbiology, Fujita Health University School of Medicine, Toyoake, Aichi, Japan
- Department of Infectious Diseases, Fujita Health University School of Medicine, Toyoake, Aichi, Japan
- Division of Infectious Diseases, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Kentaro Tsukamoto
- Department of Microbiology, Fujita Health University School of Medicine, Toyoake, Aichi, Japan
| |
Collapse
|
3
|
Increased IL-17A Serum Levels and Gastric Th17 Cells in Helicobacter pylori-Infected Patients with Gastric Premalignant Lesions. Cancers (Basel) 2023; 15:cancers15061662. [PMID: 36980548 PMCID: PMC10046233 DOI: 10.3390/cancers15061662] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 02/28/2023] [Accepted: 03/06/2023] [Indexed: 03/11/2023] Open
Abstract
Background: Helicobacter pylori infection is characterized by an inflammatory infiltrate that might be an important antecedent of gastric cancer. The purpose of this study was to evaluate whether interleukin (IL)-17 inflammation is elicited by gastric T cells in Helicobacter pylori patients with gastric intestinal metaplasia and dysplasia (IM/DYS). We also investigated the serum IL-17A levels in Helicobacter pylori patients with gastric intestinal metaplasia and dysplasia, and patients with Helicobacter pylori non-atrophic gastritis (NAG). Methods: the IL-17 cytokine profile of gastric T cells was investigated in six patients with IM/DYS and Helicobacter pylori infection. Serum IL-17A levels were measured in 45 Helicobacter pylori-infected IM/DYS patients, 45 Helicobacter pylori-infected patients without IM/DYS and in 45 healthy controls (HC). Results: gastric T cells from all IM/DYS patients with Helicobacter pylori were able to proliferate in response to Helicobacter pylori and to produce IL-17A. The Luminex analysis revealed that IL-17A levels were significantly increased in Helicobacter pylori IM/DYS patients compared to healthy controls and to Helicobacter pylori gastritis patients without IM/DYS (452.34 ± 369.13 pg/mL, 246.82 ± 156.06 pg/mL, 169.26 ± 73.82 pg/mL, respectively; p < 0.01, p < 0.05). Conclusions: the results obtained indicate that Helicobacter pylori is able to drive gastric IL-17 inflammation in IM/DYS Helicobacter pylori-infected patients, and that IL-17A serum levels are significantly increased in Helicobacter pylori-infected patients with IM/DYS.
Collapse
|
4
|
Delaney SL, Murray LA, Fallon BA. Neuropsychiatric Symptoms and Tick-Borne Diseases. Curr Top Behav Neurosci 2023; 61:279-302. [PMID: 36512289 DOI: 10.1007/7854_2022_406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
In North America, Lyme disease (LD) is primarily caused by the spirochetal bacterium Borrelia burgdorferi, transmitted to humans by Ixodes species tick bites, at an estimated rate of 476,000 patients diagnosed per year. Acute LD often manifests with flu-like symptoms and an expanding rash known as erythema migrans (EM) and less often with neurologic, neuropsychiatric, arthritic, or cardiac features. Most acute cases of Lyme disease are effectively treated with antibiotics, but 10-20% of individuals may experience recurrent or persistent symptoms. This chapter focuses on the neuropsychiatric aspects of Lyme disease, as these are less widely recognized by physicians and often overlooked. Broader education about the potential complexity, severity, and diverse manifestations of tick-borne diseases is needed.
Collapse
Affiliation(s)
- Shannon L Delaney
- Lyme and Tick-Borne Diseases Research Center at Columbia University Irving Medical Center, New York, NY, USA.
| | - Lilly A Murray
- Lyme and Tick-Borne Diseases Research Center at Columbia University Irving Medical Center, New York, NY, USA
| | - Brian A Fallon
- Lyme and Tick-Borne Diseases Research Center at Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
5
|
Tena-Garitaonaindia M, Ceacero-Heras D, Montoro MDMM, de Medina FS, Martínez-Augustin O, Daddaoua A. A Standardized Extract of Lentinula edodes Cultured Mycelium Inhibits Pseudomonas aeruginosa Infectivity Mechanisms. Front Microbiol 2022; 13:814448. [PMID: 35369436 PMCID: PMC8966770 DOI: 10.3389/fmicb.2022.814448] [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: 11/13/2021] [Accepted: 02/14/2022] [Indexed: 12/03/2022] Open
Abstract
The priority pathogen list of the World Health Organization classified Pseudomonas aeruginosa as the second top critical pathogen. Hence, the development of novel antibacterial strategies to tackle this bacterium is highly necessary. Herein we explore the potential antibacterial effect of a standardized extract of cultured mycelium of Lentinula edodes (AHCC®) on P. aeruginosa. AHCC® was found to inhibit the growth rate and biofilm formation of strain PAO1. No change in swarming was observed, but AHCC® hampered swimming and twitching motility. In accordance, a decreased expression of metabolism, growth, and biofilm formation genes was shown. AHCC® also diminished the levels of exotoxin A and bacteria inside IEC18 cells and the secretion of IL-6, IL-10 and TNF by infected macrophages. This effect was related to a reduced phosphorylation of MAPKs and to bacteria internalization. Taken together, our data suggest that AHCC® has a potential role to prevent P. aeruginosa infections and may lead to the development of new therapies.
Collapse
Affiliation(s)
- Mireia Tena-Garitaonaindia
- Department of Biochemistry and Molecular Biology II, Pharmacy School, University of Granada, Granada, Spain
| | - Diego Ceacero-Heras
- Department of Biochemistry and Molecular Biology II, Pharmacy School, University of Granada, Granada, Spain
| | - María Del Mar Maldonado Montoro
- Clinical Analysis Service, Hospital Campus de la Salud, Granada, Spain.,Instituto de Investigación Biosanitaria (IBS), Granada, Spain
| | - Fermín Sánchez de Medina
- Department of Pharmacology, School of Pharmacy, University of Granada, Granada, Spain.,Department of Pharmacology, Pharmacy School, University of Granada, Granada, Spain
| | - Olga Martínez-Augustin
- Department of Biochemistry and Molecular Biology II, Pharmacy School, University of Granada, Granada, Spain.,Instituto de Investigación Biosanitaria (IBS), Granada, Spain.,Department of Pharmacology, Pharmacy School, University of Granada, Granada, Spain.,Institute of Nutrition and Food Technology "José Mataix," Center of Biomedical Research, University of Granada, Granada, Spain
| | - Abdelali Daddaoua
- Department of Biochemistry and Molecular Biology II, Pharmacy School, University of Granada, Granada, Spain.,Instituto de Investigación Biosanitaria (IBS), Granada, Spain.,Institute of Nutrition and Food Technology "José Mataix," Center of Biomedical Research, University of Granada, Granada, Spain
| |
Collapse
|
6
|
Liu Z, Huo JH, Dong WT, Sun GD, Li FJ, Zhang YN, Qin ZW, Pengna J, Wang WM. A Study Based on Metabolomics, Network Pharmacology, and Experimental Verification to Explore the Mechanism of Qinbaiqingfei Concentrated Pills in the treatment of Mycoplasma Pneumonia. Front Pharmacol 2021; 12:761883. [PMID: 34803705 PMCID: PMC8599429 DOI: 10.3389/fphar.2021.761883] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 09/27/2021] [Indexed: 12/13/2022] Open
Abstract
Qinbaiqingfei concentrated pills (QB) are a commonly used medicine for the treatment of mycoplasma pneumonia in China, and the mechanism of action of QB needs to be studied further. Therefore, we use a combination of metabolomics and network pharmacology to clarify the mechanism of QB. Nontarget metabolomics studies were performed on rat serum, urine, and lung tissues, and 56 therapeutic biomarkers were found. Subsequently, the components of QB absorbed into the blood and lung tissues were clarified, and based on this finding, the core target of network pharmacology was predicted. The enrichment analysis of biomarkers–genes finally confirmed their close relationship with the NF-κB signaling pathway. By western blotting expression of the proteins in the lung tissue–related signaling pathways, it is finally confirmed that QB inhibits the NF-κB signaling pathway through SIRT1, IL-10 and MMP9, CTNNB1, EGFR, and other targets. It plays a role in regulating immunity, regulating metabolism, and treating diseases.
Collapse
Affiliation(s)
- Zheng Liu
- Heilongjiang Academy of Chinese Medicine, Institute of Chinese Materia Medica, Harbin, China
| | - Jin-Hai Huo
- Heilongjiang Academy of Chinese Medicine, Institute of Chinese Materia Medica, Harbin, China
| | - Wen-Ting Dong
- Heilongjiang Academy of Chinese Medicine, Institute of Chinese Materia Medica, Harbin, China
| | - Guo-Dong Sun
- Heilongjiang Academy of Chinese Medicine, Institute of Chinese Materia Medica, Harbin, China
| | - Feng-Jin Li
- Heilongjiang Academy of Chinese Medicine, Institute of Chinese Materia Medica, Harbin, China
| | - Ya-Nan Zhang
- Heilongjiang Academy of Chinese Medicine, Institute of Chinese Materia Medica, Harbin, China
| | - Zhi-Wei Qin
- Heilongjiang Academy of Chinese Medicine, Institute of Chinese Materia Medica, Harbin, China
| | - Jiang Pengna
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Wei-Ming Wang
- Heilongjiang Academy of Chinese Medicine, Institute of Chinese Materia Medica, Harbin, China
| |
Collapse
|
7
|
Abstract
PURPOSE OF REVIEW The purpose of the review is to summarize recent advances in understanding the origins, drivers and clinical context of zoonotic disease epidemics and pandemics. In addition, we aimed to highlight the role of clinicians in identifying sentinel cases of zoonotic disease outbreaks. RECENT FINDINGS The majority of emerging infectious disease events over recent decades, including the COVID-19 pandemic, have been caused by zoonotic viruses and bacteria. In particular, coronaviruses, haemorrhagic fever viruses, arboviruses and influenza A viruses have caused significant epidemics globally. There have been recent advances in understanding the origins and drivers of zoonotic epidemics, yet there are gaps in diagnostic capacity and clinical training about zoonoses. SUMMARY Identifying the origins of zoonotic pathogens, understanding factors influencing disease transmission and improving the diagnostic capacity of clinicians will be crucial to early detection and prevention of further epidemics of zoonoses.
Collapse
Affiliation(s)
| | - Peter M Rabinowitz
- Department of Medicine
- Department of Environmental and Occupational Health Sciences, Department of Global Health, University of Washington, Seattle, Washington, USA
| |
Collapse
|
8
|
Effect of Helicobacter pylori and Helminth Coinfection on the Immune Response to Mycobacterium tuberculosis. Curr Microbiol 2021; 78:3351-3371. [PMID: 34251513 DOI: 10.1007/s00284-021-02604-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Accepted: 06/30/2021] [Indexed: 02/07/2023]
Abstract
Tuberculosis remains one of the main causes of morbidity and mortality worldwide despite decades of efforts to eradicate the disease. Although the immune response controls the infection in most infected individuals (90%), the ability of the bacterium to persist throughout the host's life leads to a risk of reactivation. Underlying conditions including human immunodeficiency virus (HIV) infection, organ transplantation, and immunosuppressive therapies are considered risk factors for progression to active disease. However, many individuals infected with Mycobacterium tuberculosis may develop clinical disease in the absence of underlying immunosuppression. It is also possible that unknown conditions may drive the progression to disease. The human microbiota can be an important modulator of the immune system; it can not only trigger inflammatory disorders, but also drive the response to other infectious diseases. In developing countries, chronic mucosal infections with Helicobacter pylori and helminths may be particularly important, as these infections frequently coexist throughout the host's life. However, little is known about the interactions of these pathogens with the immune system and their effects on M. tuberculosis clinical disease, if any. In this review, we discuss the potential effects of H. pylori and helminth co-infections on the immune response to M. tuberculosis. This may contribute to our understanding of host-pathogen interactions and in designing new strategies for the prevention and control of tuberculosis.
Collapse
|
9
|
Wu Y, Yang D, Liu R, Wang L, Chen GY. Selective Response to Bacterial Infection by Regulating Siglec-E Expression. iScience 2020; 23:101473. [PMID: 32889432 PMCID: PMC7479279 DOI: 10.1016/j.isci.2020.101473] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 07/10/2020] [Accepted: 08/11/2020] [Indexed: 12/24/2022] Open
Abstract
Interactions between microbes and hosts can be a benign, deleterious, or even fatal, resulting in death of the host, the microbe, or both. Sialic acid-binding immunoglobulin-like lectins (Siglecs) suppress infection responses to sialylated pathogens. However, most pathogens are nonsialylated. Here we determined Siglecs respond to nonsialylated Gram-negative bacteria (Escherichia coli 25922 and DH5α) and Gram-positive bacteria (Staphylococcus aureus and Listeria monocytogenes). We found that Siglece-/- mice had higher mortality than wild-type mice following Gram-negative but not Gram-positive bacterial infection. Better survival in wild-type mice depended on more efficient clearance of Gram-negative than Gram-positive bacteria. Gram-negative bacteria upregulated Siglec-E, thus increasing reactive oxygen species (ROS); Tyr432 in the ITIM domain of Siglec-E was required to increase ROS. Moreover, Gram-negative bacteria upregulated Siglec-E via TLR4/MyD88/JNK/NF-κB/AP-1, whereas Gram-positive bacteria downregulated Siglec-E via TLR2/RANKL/TRAF6/Syk. Thus, our study describes a fundamentally new role for Siglec-E during infection.
Collapse
Affiliation(s)
- Yin Wu
- Children's Foundation Research Institute at Le Bonheur Children's Hospital, Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Darong Yang
- Children's Foundation Research Institute at Le Bonheur Children's Hospital, Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Runhua Liu
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, USA
- Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Lizhong Wang
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, USA
- Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Guo-Yun Chen
- Children's Foundation Research Institute at Le Bonheur Children's Hospital, Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| |
Collapse
|
10
|
Josenhans C, Müthing J, Elling L, Bartfeld S, Schmidt H. How bacterial pathogens of the gastrointestinal tract use the mucosal glyco-code to harness mucus and microbiota: New ways to study an ancient bag of tricks. Int J Med Microbiol 2020; 310:151392. [DOI: 10.1016/j.ijmm.2020.151392] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 11/28/2019] [Accepted: 12/06/2019] [Indexed: 12/13/2022] Open
|
11
|
Song M, Rabkin CS, Torres J, Kemp TJ, Zabaleta J, Pinto LA, Hildesheim A, Sánchez-Figueroa L, Guarner J, Herrera-Goepfert R, Parsonnet J, Camargo MC. Circulating inflammation-related markers and advanced gastric premalignant lesions. J Gastroenterol Hepatol 2019; 34:852-856. [PMID: 30357905 DOI: 10.1111/jgh.14518] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 09/26/2018] [Accepted: 10/15/2018] [Indexed: 12/28/2022]
Abstract
BACKGROUND AND AIM Chronic Helicobacter pylori infection causes gastric mucosal inflammation as an important antecedent of gastric cancer. We aimed to evaluate associations of blood markers of inflammation with gastric intestinal metaplasia and dysplasia in H. pylori-infected individuals. METHODS We compared pre-treatment serum levels of immune-related and inflammation-related markers between 99 individuals with intestinal metaplasia or dysplasia and 75 control individuals with non-atrophic gastritis within an H. pylori eradication trial in Mexico. Serum levels of 28 markers measured with Luminex bead-based assays were categorized in tertiles as low (T1), middle (T2), and high (T3). Logistic regression models were used to calculate age-adjusted and sex-adjusted odds ratios and 95% confidence intervals. All statistical tests were two-sided, and significance values were adjusted for multiple comparisons using false discovery rate methods. RESULTS Five markers were nominally associated (Ptrend < 0.05) with the presence of advanced premalignant gastric lesions. Adjusted odds ratios (95% confidence interval) of T2 and T3 versus T1 were 4.09 (1.65-10.17) and 3.08 (1.23-7.68) for CCL3/MIP1A, 3.21 (1.33-7.75) and 2.69 (1.10-6.57) for CCL20/MIP3A levels, 1.79 (0.77-4.18) and 2.39 (1.02-5.60) for IL-1β, 1.34 (0.56-3.19) and 3.02 (1.29-7.12) for IL-4, and 1.07 (0.44-2.59) and 3.07 (1.32-7.14) for IL-5, respectively. Two (IL-4 and IL-5) of the five markers had false discovery rate adjusted Ptrend < 0.2. CONCLUSIONS Our results suggest that certain Th2 and other cytokines may have a role in promoting carcinogenesis in the setting of H. pylori infection. Additional research is needed to replicate these findings, extend to pre-diagnostic samples, and elucidate the underlying mechanisms.
Collapse
Affiliation(s)
- Minkyo Song
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland, USA
| | - Charles S Rabkin
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland, USA
| | - Javier Torres
- Unidad de Investigación en Enfermedades Infecciosas, UMAE Pediatría, CMN SXXI, Instituto Mexicano del Seguro Social, México City, Mexico
| | - Troy J Kemp
- HPV Immunology Laboratory, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, Maryland, USA
| | - Jovanny Zabaleta
- Department of Pediatrics and Stanley S. Scott Cancer Center, Louisiana Cancer Research Center, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| | - Ligia A Pinto
- HPV Immunology Laboratory, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, Maryland, USA
| | - Allan Hildesheim
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland, USA
| | - Luz Sánchez-Figueroa
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford School of Medicine, Stanford, California, USA
| | - Jeannette Guarner
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia, USA
| | | | - Julie Parsonnet
- Division of Infectious Diseases, Department of Medicine, Stanford University, Stanford, California, USA
| | - Maria Constanza Camargo
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland, USA
| |
Collapse
|
12
|
Model of bacterial toxin-dependent pathogenesis explains infective dose. Proc Natl Acad Sci U S A 2018; 115:10690-10695. [PMID: 30279184 DOI: 10.1073/pnas.1721061115] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The initial amount of pathogens required to start an infection within a susceptible host is called the infective dose and is known to vary to a large extent between different pathogen species. We investigate the hypothesis that the differences in infective doses are explained by the mode of action in the underlying mechanism of pathogenesis: Pathogens with locally acting mechanisms tend to have smaller infective doses than pathogens with distantly acting mechanisms. While empirical evidence tends to support the hypothesis, a formal theoretical explanation has been lacking. We give simple analytical models to gain insight into this phenomenon and also investigate a stochastic, spatially explicit, mechanistic within-host model for toxin-dependent bacterial infections. The model shows that pathogens secreting locally acting toxins have smaller infective doses than pathogens secreting diffusive toxins, as hypothesized. While local pathogenetic mechanisms require smaller infective doses, pathogens with distantly acting toxins tend to spread faster and may cause more damage to the host. The proposed model can serve as a basis for the spatially explicit analysis of various virulence factors also in the context of other problems in infection dynamics.
Collapse
|
13
|
Cohen C, Shemesh M, Garrido M, Messika I, Einav M, Khokhlova I, Tasker S, Hawlena H. Haemoplasmas in wild rodents: Routes of transmission and infection dynamics. Mol Ecol 2018; 27:3714-3726. [PMID: 30074652 DOI: 10.1111/mec.14826] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 07/04/2018] [Accepted: 07/19/2018] [Indexed: 01/06/2023]
Abstract
The way that some parasites and pathogens persist in the hostile environment of their host for long periods remains to be resolved. Here, longitudinal field surveys were combined with laboratory experiments to investigate the routes of transmission and infection dynamics of such a pathogen-a wild rodent haemotropic bacterium, specifically a Mycoplasma haemomuris-like bacterium. Fleaborne transmission, direct rodent-to-rodent transmission and vertical transmission from fleas or rodents to their offspring were experimentally quantified, and indications were found that the main route of bacterial transmission is direct, although its rate of successful transmission is low (~20%). The bacterium's temporal dynamics was then compared in the field to that observed under a controlled infection experiment in field-infected and laboratory-infected rodents, and indications were found, under all conditions, that the bacterium reached its peak infection level after 25-45 days and then decreased to low bacterial loads, which persist for the rodent's lifetime. These findings suggest that the bacterium relies on persistency with low bacterial loads for long-term coexistence with its rodent host, having both conceptual and applied implications.
Collapse
Affiliation(s)
- Carmit Cohen
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Merav Shemesh
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Mario Garrido
- Mitrani Department of Desert Ecology, Jacob Blaustein Institutes for Desert Research, Ben-Gurion University of the Negev, Midreshet Ben-Gurion, Israel
| | - Irit Messika
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Monica Einav
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Irina Khokhlova
- Wyler Department of Dryland Agriculture, French Associates Institute for Agriculture and Biotechnology of Drylands, Jacob Blaustein Institutes for Desert Research, Ben-Gurion University of the Negev, Midreshet Ben-Gurion, Israel
| | - Séverine Tasker
- School of Veterinary Sciences, University of Bristol, Langford, UK
| | - Hadas Hawlena
- Mitrani Department of Desert Ecology, Jacob Blaustein Institutes for Desert Research, Ben-Gurion University of the Negev, Midreshet Ben-Gurion, Israel
| |
Collapse
|
14
|
Totten AH, Xiao L, Luo D, Briles D, Hale JY, Crabb DM, Schoeb TR, Alishlash AS, Waites KB, Atkinson TP. Allergic airway sensitization impairs antibacterial IgG antibody responses during bacterial respiratory tract infections. J Allergy Clin Immunol 2018; 143:1183-1197.e7. [PMID: 30092287 DOI: 10.1016/j.jaci.2018.07.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 06/02/2018] [Accepted: 07/24/2018] [Indexed: 11/17/2022]
Abstract
BACKGROUND Mycoplasma pneumoniae, an atypical human pathogen, has been associated with asthma initiation and exacerbation. Asthmatic patients have been reported to have higher carriage rates of M pneumoniae compared with nonasthmatic subjects and are at greater risk for invasive respiratory infections. OBJECTIVE We sought to study whether prior allergen sensitization affects the host response to chronic bacterial infection. METHODS BALB/cJ and IL-4 receptor α-/- mice were sensitized with ovalbumin (OVA) and then infected with M pneumoniae or Streptococcus pneumoniae. Immune parameters were analyzed at 30 days postinfection and included cellular profiles in bronchoalveolar lavage fluid (BALF) and serum IgG and IgE antibody levels to whole bacterial lysate, recombinant P1 adhesin, and OVA. Total lung RNA was examined for transcript levels, and BALF was examined for cytokine protein profiles. RESULTS Anti-M pneumoniae antibody responses were decreased in allergen-sensitized, M pneumoniae-infected animals compared with control animals, but OVA-specific IgG responses were unaffected. Similar decreases in anti-S pneumoniae antibody levels were found in OVA-sensitized animals. However, M pneumoniae, but not S pneumoniae, infection augmented anti-OVA IgE antibody responses. Loss of IL-4 receptor signaling partially restored anti-M pneumoniae antibody responses in IgG2a and IgG2b subclasses. Inflammatory cytokine levels in BALF from OVA-sensitized, M pneumoniae-infected or S pneumoniae-infected animals were reduced compared with those in uninfected OVA-sensitized control animals. Unexpectedly, airway hyperreactivity to methacholine was essentially ablated in M pneumoniae-infected, OVA-sensitized animals. CONCLUSIONS An established type 2-biased host immune response impairs the host immune response to respiratory bacterial infection in a largely pathogen-independent manner. Some pathogens, such as M pneumoniae, can augment ongoing allergic responses and inhibit pulmonary type 2 cytokine responses and allergic airway hyperreactivity.
Collapse
Affiliation(s)
- Arthur H Totten
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, Ala
| | - Li Xiao
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Ala
| | - Danlin Luo
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, Ala
| | - David Briles
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Ala
| | - Joanetha Y Hale
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Ala
| | - Donna M Crabb
- Department of Pathology, Diagnostic Mycoplasma Laboratory, University of Alabama at Birmingham, Birmingham, Ala
| | - Trenton R Schoeb
- Department of Genetics, University of Alabama at Birmingham, Birmingham, Ala
| | | | - Ken B Waites
- Department of Pathology, Diagnostic Mycoplasma Laboratory, University of Alabama at Birmingham, Birmingham, Ala
| | - T Prescott Atkinson
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, Ala.
| |
Collapse
|
15
|
Donovan TA, Balakrishnan N, Carvalho Barbosa I, McCoy T, Breitschwerdt EB, Fox PR. Bartonella spp. as a Possible Cause or Cofactor of Feline Endomyocarditis-Left Ventricular Endocardial Fibrosis Complex. J Comp Pathol 2018; 162:29-42. [PMID: 30060840 DOI: 10.1016/j.jcpa.2018.05.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 05/02/2018] [Accepted: 05/16/2018] [Indexed: 10/28/2022]
Abstract
Endomyocarditis is a commonly detected post-mortem finding in domestic cats presenting for sudden onset cardiovascular death, yet the aetiology remains unresolved. Cats are documented reservoir hosts for Bartonella henselae, the infectious cause of cat scratch disease in man. Various Bartonella spp. have been associated with culture-negative endocarditis, myocarditis and sudden death in man and animals. We hypothesized that Bartonella spp. DNA could be amplified more often from the hearts of cats with feline endomyocarditis-left ventricular endocardial fibrosis (FEMC-LVEF) complex compared with cats with hypertrophic cardiomyopathy (HCM) or cats with grossly and microscopically unremarkable hearts (designated non-cardiac disease controls). Formalin-fixed and paraffin wax-embedded, cardiac tissues from 60 domestic and purebred cats aged 3 months to 18 years were examined, and histological features were recorded. Cardiac tissue sections were tested for Bartonella DNA using multiple 16-23S intergenic transcribed spacer region polymerase chain reaction (PCR) primer sets, including two Bartonella genera, a Bartonella koehlerae species-specific and a Bartonella vinsonii subsp. berkhoffii-specific assay, followed by DNA sequence confirmation of the species or genotype. Special precautions were taken to avoid DNA cross-contamination between tissues. Bartonella spp. DNA was amplified by PCR and sequenced from 18 of 36 cats (50%) with FEMC-LVEF and 1/12 (8.3%) cats with HCM. Bartonella spp. DNA was not amplified from any non-cardiac disease control hearts. Based on PCR/DNA sequencing, one Bartonella spp. was amplified from 10 cats, while the remaining eight were coinfected with more than one Bartonella spp. To our knowledge, this study represents the first documentation of B. vinsonii subsp. berkhoffii genotype I infection in cats (n = 11). Fluorescence in-situ hybridization testing facilitated visualization of Bartonella bacteria within the myocardium of four of seven PCR-positive FEMC-LVEF hearts. Collectively, these findings support the hypothesis that Bartonella spp. may play a primary role or act as a cofactor in the pathogenesis of FEMC-LVEF. Studies involving cats from other geographical regions and definitive demonstration of Bartonella spp. within regions of inflammation are needed to confirm an association between Bartonella spp. and FEMC-LVEF induced morbidity and mortality in cats.
Collapse
Affiliation(s)
- T A Donovan
- Department of Anatomic Pathology, The Animal Medical Center, New York, New York, USA.
| | - N Balakrishnan
- Clinical Microbiology Unit, State Laboratory of Public Health, North Carolina Department of Health and Human Services, Raleigh, North Carolina, USA
| | - I Carvalho Barbosa
- Department of Clinical Sciences, and the Intracellular Pathogens Research Laboratory, Comparative Medicine Institute, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA
| | - T McCoy
- Department of Clinical Sciences, and the Intracellular Pathogens Research Laboratory, Comparative Medicine Institute, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA
| | - E B Breitschwerdt
- Department of Clinical Sciences, and the Intracellular Pathogens Research Laboratory, Comparative Medicine Institute, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA
| | - P R Fox
- Department of Cardiology, The Animal Medical Center, New York, New York, USA
| |
Collapse
|
16
|
IcmF and DotU are required for the virulence of Acidovorax oryzae strain RS-1. Arch Microbiol 2018; 200:897-910. [DOI: 10.1007/s00203-018-1497-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Revised: 02/06/2018] [Accepted: 02/16/2018] [Indexed: 10/18/2022]
|
17
|
Ferraris DM, Miggiano R, Rossi F, Rizzi M. Mycobacterium tuberculosis Molecular Determinants of Infection, Survival Strategies, and Vulnerable Targets. Pathogens 2018; 7:E17. [PMID: 29389854 PMCID: PMC5874743 DOI: 10.3390/pathogens7010017] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Revised: 01/26/2018] [Accepted: 01/28/2018] [Indexed: 12/13/2022] Open
Abstract
Mycobacterium tuberculosis is the causative agent of tuberculosis, an ancient disease which, still today, represents a major threat for the world population. Despite the advances in medicine and the development of effective antitubercular drugs, the cure of tuberculosis involves prolonged therapies which complicate the compliance and monitoring of drug administration and treatment. Moreover, the only available antitubercular vaccine fails to provide an effective shield against adult lung tuberculosis, which is the most prevalent form. Hence, there is a pressing need for effective antitubercular drugs and vaccines. This review highlights recent advances in the study of selected M. tuberculosis key molecular determinants of infection and vulnerable targets whose structures could be exploited for the development of new antitubercular agents.
Collapse
Affiliation(s)
- Davide M Ferraris
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale "A. Avogadro", Largo Donegani 2, 28100 Novara, Italy.
| | - Riccardo Miggiano
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale "A. Avogadro", Largo Donegani 2, 28100 Novara, Italy.
| | - Franca Rossi
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale "A. Avogadro", Largo Donegani 2, 28100 Novara, Italy.
| | - Menico Rizzi
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale "A. Avogadro", Largo Donegani 2, 28100 Novara, Italy.
| |
Collapse
|
18
|
Costales MG, Alam MS, Cavanaugh C, Williams KM. Extracellular adenosine produced by ecto-5'-nucleotidase (CD73) regulates macrophage pro-inflammatory responses, nitric oxide production, and favors Salmonella persistence. Nitric Oxide 2017; 72:7-15. [PMID: 29108754 DOI: 10.1016/j.niox.2017.11.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 09/21/2017] [Accepted: 11/02/2017] [Indexed: 12/24/2022]
Abstract
Surface enzymes CD39 (nucleoside triphosphate dephosphorylase) and CD73 (ecto-5'-nucleotidase) mediate the synthesis of extracellular adenosine that can regulate immune responses. Adenosine produced by CD39/CD73 acts via adenosine receptors (ARs). CD73 is expressed by a variety of cell types and mediates anti-inflammatory responses. Because efficient innate immune responses are required for clearance of Salmonella infection, we investigated the role of CD73 in macrophage function, including phagocytosis, intracellular killing of Salmonella, and anti-bacterial pro-inflammatory responses to Salmonella-whole cell lysate (ST-WCL) or Salmonella infection. Additionally, RAW 264.7 macrophage mRNA expression of CD39, CD73, and all ARs were measured by qPCR after ST-WCL treatment. Pro-inflammatory cytokine mRNA and nitric oxide (NO) production were quantitated in the ST-WCL treated macrophage with and without CD73-inhibitor (APCP) treatment. Phagocytosis and intracellular killing by peritoneal macrophages from CD73-deficent mice were also evaluated using E. coli BioParticles® and GFP-Salmonella infection, respectively. CD73, CD39, and A2BAR mRNA were predominantly expressed in RAW cells. ST-WCL treatment significantly reduced CD73 expression, suggesting endogenous down-regulation of CD73, and an enhanced pro-inflammatory response. ST-WCL treated and CD73-inhibited macrophages produced more NO and a higher level of pro-inflammatory cytokines than CD73-competent macrophages (e.g. IL-1β, TNF-α). Phagocytosis of E. coli BioParticles® was significantly higher in the macrophages treated with APCP and in the peritoneal macrophages from CD73-deficent mice as compared to APCP-untreated, and CD73-competent macrophages. Internalized bacteria were more efficiently cleared from macrophages in the absence of CD73, as observed by fluorescence-microscopy and Salmonella-DNA measurement by qPCR from the infected cells. CD73 down-regulation or CD73-inhibition of macrophages during Salmonella infection can enhance the production of pro-inflammatory cytokines and NO production, improving intracellular killing and host survivability. Extracellular adenosine synthesized by CD73 suppresses antibacterial responses of macrophages, which may weaken macrophage function and impair innate immune responses to Salmonella infection.
Collapse
Affiliation(s)
- Matthew G Costales
- Immunobiology Branch, Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, US Food and Drug Administration (FDA), Laurel, MD 20708, USA
| | - Mohammad Samiul Alam
- Immunobiology Branch, Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, US Food and Drug Administration (FDA), Laurel, MD 20708, USA.
| | - Christopher Cavanaugh
- Immunobiology Branch, Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, US Food and Drug Administration (FDA), Laurel, MD 20708, USA
| | - Kristina M Williams
- Immunobiology Branch, Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, US Food and Drug Administration (FDA), Laurel, MD 20708, USA
| |
Collapse
|
19
|
Fila G, Kawiak A, Grinholc MS. Blue light treatment of Pseudomonas aeruginosa: Strong bactericidal activity, synergism with antibiotics and inactivation of virulence factors. Virulence 2017; 8:938-958. [PMID: 27763824 PMCID: PMC5626244 DOI: 10.1080/21505594.2016.1250995] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 10/12/2016] [Accepted: 10/15/2016] [Indexed: 01/16/2023] Open
Abstract
Pseudomonas aeruginosa is among the most common pathogens responsible for both acute and chronic infections of high incidence and severity. Additionally, P. aeruginosa resistance to conventional antimicrobials has increased rapidly over the past decade. Therefore, it is crucial to explore new therapeutic options, particularly options that specifically target the pathogenic mechanisms of this microbe. The ability of a pathogenic bacterium to cause disease is dependent upon the production of agents termed 'virulence factors', and approaches to mitigate these agents have gained increasing attention as new antibacterial strategies. Although blue light irradiation is a promising alternative approach, only limited and preliminary studies have described its effect on virulence factors. The current study aimed to investigate the effects of lethal and sub-lethal doses of blue light treatment (BLT) on P. aeruginosa virulence factors. We analyzed the inhibitory effects of blue light irradiation on the production/activity of several virulence factors. Lethal BLT inhibited the activity of pyocyanin, staphylolysin, pseudolysin and other proteases, but sub-lethal BLT did not affect the production/expression of proteases, phospholipases, and flagella- or type IV pili-associated motility. Moreover, a eukaryotic cytotoxicity test confirmed the decreased toxicity of blue light-treated extracellular P. aeruginosa fractions. Finally, the increased antimicrobial susceptibility of P. aeruginosa treated with sequential doses of sub-lethal BLT was demonstrated with a checkerboard test. Thus, this work provides evidence-based proof of the susceptibility of drug-resistant P. aeruginosa to BLT-mediated killing, accompanied by virulence factor reduction, and describes the synergy between antibiotics and sub-lethal BLT.
Collapse
Affiliation(s)
- Grzegorz Fila
- Laboratory of Molecular Diagnostics, Department of Biotechnology, Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Gdansk, Poland
| | - Anna Kawiak
- Division of Plant Protection and Biotechnology, Department of Biotechnology, Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Gdansk, Poland
- Laboratory of Human Physiology, Medical University of Gdansk, Gdansk, Poland
| | - Mariusz Stanislaw Grinholc
- Laboratory of Molecular Diagnostics, Department of Biotechnology, Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Gdansk, Poland
| |
Collapse
|
20
|
Weitzman CL, Sandmeier FC, Tracy CR. Prevalence and Diversity of the Upper Respiratory Pathogen Mycoplasma agassizii in Mojave Desert Tortoises (Gopherus agassizii). HERPETOLOGICA 2017. [DOI: 10.1655/herpetologica-d-16-00079.1] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Chava L. Weitzman
- Program in Ecology, Evolution, and Conservation Biology, University of Nevada, Reno, NV 89557, USA
| | | | - C. Richard Tracy
- Program in Ecology, Evolution, and Conservation Biology, University of Nevada, Reno, NV 89557, USA
| |
Collapse
|
21
|
Santosh ABR, Reddy BVR. Oral Mucosal Infections: Insights into Specimen Collection and Medication Management. Dent Clin North Am 2017; 61:283-304. [PMID: 28317567 DOI: 10.1016/j.cden.2016.12.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Oral mucosal infections appear as localized or generalized lesions. Symptoms range from almost unnoticeable lesions to severe pain. Systemic disease, age, immunocompromised condition, and medication use are common causes. Local causes include dentures, poor oral hygiene, traumatized epithelium, ulcerations, dentures, implants, oral piercing, and reduced salivary secretion. Oral mucosal infections are underdiagnosed and microbiological diagnosis should be more frequently used. Candidiasis is most frequently diagnosed. Clinical appearances are not always clear and are varied, creating a diagnostic challenge. Thorough understanding of clinical appearance and updated information on diagnostic and therapeutic management are essential for successful patient outcome.
Collapse
Affiliation(s)
| | - Baddam Venkat Ramana Reddy
- Department of Oral and Maxillofacial Pathology, SIBAR Institute of Dental Sciences, Takkellapadu, Guntur, Andhra Pradesh 522601, India
| |
Collapse
|
22
|
Scorpio DG. Do cats serve as good sentinels for Bartonella species infection risk in people and animals? Vet Rec 2017; 180:322-324. [PMID: 28364072 DOI: 10.1136/vr.j1549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Affiliation(s)
- Diana G Scorpio
- Animal Program Director, National Institutes of Health, Vaccine Research Center, Building 40 Bethesda, Maryland, USA; e-mail:
| |
Collapse
|
23
|
Ahn D, Prince A. Host-Pathogen Interface: Progress in Understanding the Pathogenesis of Infection Due to Multidrug-Resistant Bacteria in the Intensive Care Unit. J Infect Dis 2017; 215:S1-S8. [PMID: 28375516 PMCID: PMC5853223 DOI: 10.1093/infdis/jiw405] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The diverse responses of critically ill patients to infection with multi-drug resistant (MDR) bacteria are determined by many complex factors. These include the nature of the immune response activated by specific organisms. Properties unique to each organism such as adherence proteins, microvesicle formation, toxin production and the propensity to form biofilms are important factors in pathogenesis. Equally important is the variability in the host immune response, whether due to genetic or iatrogenic factors, including the presence of major comorbidities, treatment with immunomodulatory therapy and disruption of the microbiome. Future approaches in treating infections caused by MDR bacteria will be heavily influenced by a precision medicine approach, with rapid diagnostic techniques of both bacterial and host factors and high throughput screening of novel therapeutics becoming the mainstay of treatment.
Collapse
Affiliation(s)
- Danielle Ahn
- Department of Pediatrics, Columbia University Medical Center, New York
| | - Alice Prince
- Department of Pediatrics, Columbia University Medical Center, New York
| |
Collapse
|
24
|
Rickettsia and Rickettsia-Like Organisms. Infect Dis (Lond) 2017. [DOI: 10.1016/b978-0-7020-6285-8.00187-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
|
25
|
|
26
|
Culture, PCR, DNA sequencing, and second harmonic generation (SHG) visualization of Bartonella henselae from a surgically excised human femoral head. Clin Rheumatol 2016; 36:1669-1675. [PMID: 28028681 DOI: 10.1007/s10067-016-3524-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 12/16/2016] [Accepted: 12/18/2016] [Indexed: 10/20/2022]
|
27
|
Donovan TA, Fox PR, Balakrishnan N, Ericson M, Hooker V, Breitschwerdt EB. Pyogranulomatous Pancarditis with Intramyocardial Bartonella henselae San Antonio 2 (BhSA2) in a Dog. J Vet Intern Med 2016; 31:142-148. [PMID: 27883248 PMCID: PMC5259629 DOI: 10.1111/jvim.14609] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Revised: 09/21/2016] [Accepted: 10/18/2016] [Indexed: 12/27/2022] Open
Affiliation(s)
- T A Donovan
- Department of Pathology, The Animal Medical Center, New York, NY
| | - P R Fox
- Department of Cardiology, The Animal Medical Center, New York, NY
| | - N Balakrishnan
- Department of Clinical Sciences, Intracellular Pathogens Research Laboratory, Center for Comparative Medicine and Translational Research, College of Veterinary Medicine, North Carolina State University, Raleigh, NC
| | - M Ericson
- University of Minnesota Imaging Center, Minneapolis, MN
| | - V Hooker
- The Animal Medical Center, New York, NY
| | - E B Breitschwerdt
- Department of Clinical Sciences, Intracellular Pathogens Research Laboratory, Center for Comparative Medicine and Translational Research, College of Veterinary Medicine, North Carolina State University, Raleigh, NC
| |
Collapse
|
28
|
Kogut MH, Swaggerty CL, Byrd JA, Selvaraj R, Arsenault RJ. Chicken-Specific Kinome Array Reveals that Salmonella enterica Serovar Enteritidis Modulates Host Immune Signaling Pathways in the Cecum to Establish a Persistence Infection. Int J Mol Sci 2016; 17:ijms17081207. [PMID: 27472318 PMCID: PMC5000605 DOI: 10.3390/ijms17081207] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2016] [Revised: 06/15/2016] [Accepted: 07/08/2016] [Indexed: 02/07/2023] Open
Abstract
Non-typhoidal Salmonella enterica induces an early, short-lived pro-inflammatory response in chickens that is asymptomatic of clinical disease and results in a persistent colonization of the gastrointestinal (GI) tract that transmits infections to naïve hosts via fecal shedding of bacteria. The underlying mechanisms that control this persistent colonization of the ceca of chickens by Salmonella are only beginning to be elucidated. We hypothesize that alteration of host signaling pathways mediate the induction of a tolerance response. Using chicken-specific kinomic immune peptide arrays and quantitative RT-PCR of infected cecal tissue, we have previously evaluated the development of disease tolerance in chickens infected with Salmonella enterica serovar Enteritidis (S. Enteritidis) in a persistent infection model (4-14 days post infection). Here, we have further outlined the induction of an tolerance defense strategy in the cecum of chickens infected with S. Enteritidis beginning around four days post-primary infection. The response is characterized by alterations in the activation of T cell signaling mediated by the dephosphorylation of phospholipase c-γ1 (PLCG1) that inhibits NF-κB signaling and activates nuclear factor of activated T-cells (NFAT) signaling and blockage of interferon-γ (IFN-γ) production through the disruption of the JAK-STAT signaling pathway (dephosphorylation of JAK2, JAK3, and STAT4). Further, we measured a significant down-regulation reduction in IFN-γ mRNA expression. These studies, combined with our previous findings, describe global phenotypic changes in the avian cecum of Salmonella Enteritidis-infected chickens that decreases the host responsiveness resulting in the establishment of persistent colonization. The identified tissue protein kinases also represent potential targets for future antimicrobial compounds for decreasing Salmonella loads in the intestines of food animals before going to market.
Collapse
Affiliation(s)
- Michael H Kogut
- Southern Plains Agricultural Resarch Center, United States Department of Agriculture, Agricultural Research Service, College Station, TX 77845, USA.
| | - Christina L Swaggerty
- Southern Plains Agricultural Resarch Center, United States Department of Agriculture, Agricultural Research Service, College Station, TX 77845, USA.
| | - James Allen Byrd
- Southern Plains Agricultural Resarch Center, United States Department of Agriculture, Agricultural Research Service, College Station, TX 77845, USA.
| | - Ramesh Selvaraj
- Ohio Agricultural Research Center, The Ohio State University, Wooster, OH 44691, USA.
| | - Ryan J Arsenault
- Department of Animal and Food Sciences, University of Delaware, Newark, DE 19716, USA.
| |
Collapse
|
29
|
Harms A, Stanger FV, Dehio C. Biological Diversity and Molecular Plasticity of FIC Domain Proteins. Annu Rev Microbiol 2016; 70:341-60. [PMID: 27482742 DOI: 10.1146/annurev-micro-102215-095245] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The ubiquitous proteins with FIC (filamentation induced by cyclic AMP) domains use a conserved enzymatic machinery to modulate the activity of various target proteins by posttranslational modification, typically AMPylation. Following intensive study of the general properties of FIC domain catalysis, diverse molecular activities and biological functions of these remarkably versatile proteins are now being revealed. Here, we review the biological diversity of FIC domain proteins and summarize the underlying structure-function relationships. The original and most abundant genuine bacterial FIC domain proteins are toxins that use diverse molecular activities to interfere with bacterial physiology in various, yet ill-defined, biological contexts. Host-targeted virulence factors have evolved repeatedly out of this pool by exaptation of the enzymatic FIC domain machinery for the manipulation of host cell signaling in favor of bacterial pathogens. The single human FIC domain protein HypE (FICD) has a specific function in the regulation of protein stress responses.
Collapse
Affiliation(s)
- Alexander Harms
- Focal Area Infection Biology, Biozentrum, University of Basel, CH-4056 Basel, Switzerland; , ,
| | - Frédéric V Stanger
- Focal Area Infection Biology, Biozentrum, University of Basel, CH-4056 Basel, Switzerland; , , .,Focal Area Structural Biology and Biophysics, Biozentrum, University of Basel, CH-4056 Basel, Switzerland.,*Current address: Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Christoph Dehio
- Focal Area Infection Biology, Biozentrum, University of Basel, CH-4056 Basel, Switzerland; , ,
| |
Collapse
|
30
|
Lääveri T, Sterne J, Rombo L, Kantele A. Systematic review of loperamide: No proof of antibiotics being superior to loperamide in treatment of mild/moderate travellers' diarrhoea. Travel Med Infect Dis 2016; 14:299-312. [PMID: 27363327 DOI: 10.1016/j.tmaid.2016.06.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 06/19/2016] [Accepted: 06/20/2016] [Indexed: 12/18/2022]
Abstract
Looking at the worldwide emergency of antimicrobial resistance, international travellers appear to have a central role in spreading the bacteria across the globe. Travellers' diarrhoea (TD) is the most common disease encountered by visitors to the (sub)tropics. Both TD and its treatment with antibiotics have proved significant independent risk factors of colonization by resistant intestinal bacteria while travelling. Travellers should therefore be given preventive advice regarding TD and cautioned about taking antibiotics: mild or moderate TD does not require antibiotics. Logical alternatives are medications with effects on gastrointestinal function, such as loperamide. The present review explores literature on loperamide in treating TD. Adhering to manufacturer's dosage recommendations, loperamide offers a safe and effective alternative for relieving mild and moderate symptoms. Moreover, loperamide taken singly does no predispose to contracting MDR bacteria. Most importantly, we found no proof that would show antibiotics to be significantly more effective than loperamide in treating mild/moderate TD.
Collapse
Affiliation(s)
- Tinja Lääveri
- Inflammation Center, Division of Infectious Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, POB 348, FIN-00029 HUS, Finland.
| | - Jesper Sterne
- Centre for Clinical Research, Sörmland County Council, Eskilstuna and University of Uppsala, SE 631 88 Eskilstuna, Sweden.
| | - Lars Rombo
- Centre for Clinical Research, Sörmland County Council, Eskilstuna and University of Uppsala, SE 631 88 Eskilstuna, Sweden; Karolinska Institutet, Department of Medicine/Solna, Unit for Infectious Diseases, SE 17176 Stockholm, Sweden.
| | - Anu Kantele
- Inflammation Center, Division of Infectious Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, POB 348, FIN-00029 HUS, Finland; Karolinska Institutet, Department of Medicine/Solna, Unit for Infectious Diseases, SE 17176 Stockholm, Sweden; Department of Medicine, University of Helsinki, Finland.
| |
Collapse
|
31
|
Vinther AML, Heegaard PMH, Skovgaard K, Buhl R, Andreassen SM, Andersen PH. Characterization and differentiation of equine experimental local and early systemic inflammation by expression responses of inflammation-related genes in peripheral blood leukocytes. BMC Vet Res 2016; 12:83. [PMID: 27250718 PMCID: PMC4888743 DOI: 10.1186/s12917-016-0706-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Accepted: 05/22/2016] [Indexed: 01/17/2023] Open
Abstract
Background Local inflammation may progress into systemic inflammation. To increase our understanding of the basic immunological processes during transition of equine local inflammation into a systemic state, investigation into the equine systemic immune response to local inflammation is warranted. Therefore, the aim of this study was to investigate the innate peripheral blood leukocyte (PBL) immune response to local inflammation in horses, and to compare this response with the PBL immune response during the early phase of acute systemic inflammation. Expression of 22 selected inflammation-related genes was measured in whole blood leukocytes from 6 horses in an experimental cross-over model of lipopolysaccharide- (LPS-) induced acute synovitis (3 μg LPS intraarticularly; locally inflamed [LI] horses) and endotoxemia (1 μg LPS/kg intravenously; systemically inflamed [SI] horses). Multiple clinical and hematological/biochemical examinations were performed, and serial blood samples were analyzed by reverse transcription quantitative real-time PCR. Post-induction expression profiles of all genes were compared between study groups using principal component analysis (PCA) and hierarchical clustering. Results Moderate synovitis and mild systemic inflammation of approximately 24 h duration was confirmed by clinical and paraclinical observations in LI and SI horses, respectively. In the LI group, samples obtained 3–16 h post-injection showed distinct clustering in the PCA compared with baseline levels, indicating a transcriptional response to local inflammation in PBLs in this time interval. There was no clinical or hematological indication of actual systemic inflammation. There was a clear separation of all LI samples from all SI samples in two distinct clusters, indicating that expression profiles in the two study groups were different, independent of time since LPS injection. Co-regulated genes formed four clusters across study groups which were distinctly differently regulated. Only few of individual genes displayed different expression between the study groups at all times after LPS injection. Conclusions Local inflammation in horses initiated an innate transcriptional response in PBLs, which differed from the transcriptional response during the early phase of systemic inflammation. This study may provide new insights into the immunobiology of PBLs during the transition of local inflammation into a systemic state. Electronic supplementary material The online version of this article (doi:10.1186/s12917-016-0706-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Anne Mette L Vinther
- Department of Large Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Taastrup, Denmark.
| | - Peter M H Heegaard
- Innate Immunology Group, Section for Immunology and Vaccinology, National Veterinary Institute, Technical University of Denmark, Frederiksberg, Denmark
| | - Kerstin Skovgaard
- Innate Immunology Group, Section for Immunology and Vaccinology, National Veterinary Institute, Technical University of Denmark, Frederiksberg, Denmark
| | - Rikke Buhl
- Department of Large Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Taastrup, Denmark
| | - Stine M Andreassen
- Department of Large Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Taastrup, Denmark
| | - Pia H Andersen
- Department of Clinical Sciences, Faculty of Veterinary Medicine and Animal Science, Swedish University of Agricultural Sciences, Uppsala, Sweden
| |
Collapse
|
32
|
Balakrishnan N, Ericson M, Maggi R, Breitschwerdt EB. Vasculitis, cerebral infarction and persistent Bartonella henselae infection in a child. Parasit Vectors 2016; 9:254. [PMID: 27161220 PMCID: PMC4862072 DOI: 10.1186/s13071-016-1547-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Accepted: 04/27/2016] [Indexed: 12/05/2022] Open
Abstract
Background The genus Bartonella is comprised of a rapidly increasing number of pathogenic species that induce a seemingly diverse spectrum of neurological symptoms. During the 12 year period that followed the initial onset of neurological and gastrointestinal symptoms, an 11 year-old girl experienced a spectrum of neurological complaints including frequent headaches, visual and auditory hallucinations, anxiety, vision loss involving the lower left quadrant of both eyes, episodic bouts of generalized paralysis, facial palsy, chronic insomnia, seizures, dizziness, cognitive dysfunction, and memory loss. PCR assays targeting Bartonella spp. were used to test formalin-fixed, paraffin embedded brain tissue, patient blood specimens and Bartonella alpha Proteobacteria growth medium (BAPGM) enrichment blood cultures. PCR positive amplicons were sequenced directly and compared to GenBank sequences. Bartonella spp. serology was performed by indirect fluorescent antibody testing and confocal laser scanning microscopy was used to visualize B. henselae organisms in resected brain. Results Bartonella henselae DNA was independently PCR amplified and sequenced from the girl’s right parietal lobe, surgically resected in 2000 and from a blood specimen collected in 2012. Although causation cannot be established by a case report, prior diagnostic testing resulted in findings that were either inconclusive or within normal reference ranges and no etiological diagnosis had been obtained to explain the patient’s initial or progressive neurological symptoms. Conclusions As intravascular, intra-erythrocytic and endotheliotropic bacteria, it is possible that B. henselae initially induced a vasculitis, resulting in secondary cerebral infarction, tissue necrosis and surgical resection. Bartonella bacteremia, potentially spanning a 12-year time frame, in conjunction with the therapeutic administration of immunosuppressive drugs may have resulted in a progression and potentiation of the neurological disease that was partially reversible following antibiotic administration.
Collapse
Affiliation(s)
- Nandhakumar Balakrishnan
- Department of Clinical Sciences and the Intracellular Pathogens Research Laboratory, Center for Comparative Medicine and Translational Research, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, USA
| | - Marna Ericson
- Cutaneous Imaging Center, Department of Dermatology, University of Minnesota, Minnesota, USA
| | - Ricardo Maggi
- Department of Clinical Sciences and the Intracellular Pathogens Research Laboratory, Center for Comparative Medicine and Translational Research, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, USA
| | - Edward B Breitschwerdt
- Department of Clinical Sciences and the Intracellular Pathogens Research Laboratory, Center for Comparative Medicine and Translational Research, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, USA.
| |
Collapse
|
33
|
Wagner S, Sommer R, Hinsberger S, Lu C, Hartmann RW, Empting M, Titz A. Novel Strategies for the Treatment of Pseudomonas aeruginosa Infections. J Med Chem 2016; 59:5929-69. [DOI: 10.1021/acs.jmedchem.5b01698] [Citation(s) in RCA: 176] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Stefanie Wagner
- Chemical
Biology of Carbohydrates, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), D-66123 Saarbrücken, Germany
- Deutsches Zentrum für Infektionsforschung (DZIF), 30625 Standort Hannover-Braunschweig, Germany
| | - Roman Sommer
- Chemical
Biology of Carbohydrates, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), D-66123 Saarbrücken, Germany
- Deutsches Zentrum für Infektionsforschung (DZIF), 30625 Standort Hannover-Braunschweig, Germany
| | - Stefan Hinsberger
- Deutsches Zentrum für Infektionsforschung (DZIF), 30625 Standort Hannover-Braunschweig, Germany
- Drug
Design and Optimization, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), D-66123 Saarbrücken, Germany
| | - Cenbin Lu
- Deutsches Zentrum für Infektionsforschung (DZIF), 30625 Standort Hannover-Braunschweig, Germany
- Drug
Design and Optimization, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), D-66123 Saarbrücken, Germany
| | - Rolf W. Hartmann
- Deutsches Zentrum für Infektionsforschung (DZIF), 30625 Standort Hannover-Braunschweig, Germany
- Drug
Design and Optimization, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), D-66123 Saarbrücken, Germany
| | - Martin Empting
- Deutsches Zentrum für Infektionsforschung (DZIF), 30625 Standort Hannover-Braunschweig, Germany
- Drug
Design and Optimization, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), D-66123 Saarbrücken, Germany
| | - Alexander Titz
- Chemical
Biology of Carbohydrates, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), D-66123 Saarbrücken, Germany
- Deutsches Zentrum für Infektionsforschung (DZIF), 30625 Standort Hannover-Braunschweig, Germany
| |
Collapse
|
34
|
Spínola-Amilibia M, Davó-Siguero I, Ruiz FM, Santillana E, Medrano FJ, Romero A. The structure of VgrG1 fromPseudomonas aeruginosa, the needle tip of the bacterial type VI secretion system. ACTA CRYSTALLOGRAPHICA SECTION D-STRUCTURAL BIOLOGY 2016; 72:22-33. [DOI: 10.1107/s2059798315021142] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 11/06/2015] [Indexed: 11/10/2022]
Abstract
The type VI secretion system (T6SS) is a mechanism that is commonly used by pathogenic bacteria to infect host cells and for survival in competitive environments. This system assembles on a core baseplate and elongates like a phage puncturing device; it is thought to penetrate the target membrane and deliver effectors into the host or competing bacteria. Valine–glycine repeat protein G1 (VgrG1) forms the spike at the tip of the elongating tube formed by haemolysin co-regulated protein 1 (Hcp1); it is structurally similar to the T4 phage (gp27)3–(gp5)3puncturing complex. Here, the crystal structure of full-length VgrG1 fromPseudomonas aeruginosais reported at a resolution of 2.0 Å, which through a trimeric arrangement generates a needle-like shape composed of two main parts, the head and the spike, connectedviaa small neck region. The structure reveals several remarkable structural features pointing to the possible roles of the two main segments of VgrG1: the head as a scaffold cargo domain and the β-roll spike with implications in the cell-membrane puncturing process and as a carrier of cognate toxins.
Collapse
|
35
|
Ruwandeepika HAD, Karunasagar I, Bossier P, Defoirdt T. Expression and Quorum Sensing Regulation of Type III Secretion System Genes of Vibrio harveyi during Infection of Gnotobiotic Brine Shrimp. PLoS One 2015; 10:e0143935. [PMID: 26636765 PMCID: PMC4670211 DOI: 10.1371/journal.pone.0143935] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Accepted: 11/11/2015] [Indexed: 12/24/2022] Open
Abstract
Type III secretion systems enable pathogens to inject their virulence factors directly into the cytoplasm of the host cells. The type III secretion system of Vibrio harveyi, a major pathogen of aquatic organisms and a model species in quorum sensing studies, is repressed by the quorum sensing master regulator LuxR. In this study, we found that during infection of gnotobiotic brine shrimp larvae, the expression levels of three type III secretion operons in V. harveyi increased within the first 12h after challenge and decreased again thereafter. The in vivo expression levels were highest in a mutant with a quorum sensing system that is locked in low cell density configuration (minimal LuxR levels) and lowest in a mutant with a quorum sensing system that is locked in the high cell density configuration (maximal LuxR levels), which is consistent with repression of type III secretion by LuxR. Remarkably, in vivo expression levels of the type III secretion system genes were much (> 1000 fold) higher than the in vitro expression levels, indicating that (currently unknown) host factors significantly induce the type III secretion system. Given the fact that type III secretion is energy-consuming, repression by the quorum sensing master regulators might be a mechanism to save energy under conditions where it does not provide an advantage to the cells.
Collapse
Affiliation(s)
- H. A. Darshanee Ruwandeepika
- Department of Livestock Production, Faculty of Agricultural Sciences, Sabaragamuwa University of Sri Lanka, Belihuloya, Sri Lanka
- Centre for Science Education and Research, UNESCO MIRCEN for Medical and Marine Biotechnology, Nitte University, Mangalore, India
| | - Indrani Karunasagar
- Centre for Science Education and Research, UNESCO MIRCEN for Medical and Marine Biotechnology, Nitte University, Mangalore, India
| | - Peter Bossier
- Laboratory of Aquaculture & Artemia Reference Center, Ghent University, Ghent, Belgium
| | - Tom Defoirdt
- Laboratory of Aquaculture & Artemia Reference Center, Ghent University, Ghent, Belgium
- * E-mail:
| |
Collapse
|
36
|
Friedenberg SG, Balakrishnan N, Guillaumin J, Cooper ES, Lewis K, Russell DS, Breitschwerdt EB. Splenic vasculitis, thrombosis, and infarction in a febrile dog infected withBartonella henselae. J Vet Emerg Crit Care (San Antonio) 2015; 25:789-94. [DOI: 10.1111/vec.12367] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Revised: 03/04/2014] [Accepted: 07/22/2015] [Indexed: 11/29/2022]
Affiliation(s)
- Steven G. Friedenberg
- Department of Veterinary Clinical Sciences; College of Veterinary Medicine, The Ohio State University, Columbus; OH 43210
| | - Nandhakumar Balakrishnan
- the Intracellular Pathogens Research Laboratory, Center for Comparative Medicine and Translational Research, College of Veterinary Medicine, North Carolina State University; Raleigh NC 27607
| | - Julien Guillaumin
- Department of Veterinary Clinical Sciences; College of Veterinary Medicine, The Ohio State University, Columbus; OH 43210
| | - Edward S. Cooper
- Department of Veterinary Clinical Sciences; College of Veterinary Medicine, The Ohio State University, Columbus; OH 43210
| | - Kristin Lewis
- the Department of Veterinary Biosciences; College of Veterinary Medicine, The Ohio State University, Columbus; OH 43210
| | - Duncan S. Russell
- the Department of Veterinary Biosciences; College of Veterinary Medicine, The Ohio State University, Columbus; OH 43210
| | - Edward B. Breitschwerdt
- the Intracellular Pathogens Research Laboratory, Center for Comparative Medicine and Translational Research, College of Veterinary Medicine, North Carolina State University; Raleigh NC 27607
| |
Collapse
|
37
|
Dolgova EV, Efremov YR, Taranov OS, Potter EA, Nikolin VP, Popova NA, Omigov VV, Chernykh ER, Proskurina AS, Bogachev SS. Comparative analysis of pathologic processes developing in mice housed in SPF vs non-SPF conditions and treated with cyclophosphamide and dsDNA preparation. Pathol Res Pract 2015; 211:754-8. [PMID: 26293796 DOI: 10.1016/j.prp.2015.07.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Revised: 06/16/2015] [Accepted: 07/03/2015] [Indexed: 12/28/2022]
Abstract
In our earlier studies, we observed that when mice are treated with cyclophosphamide and fragmented exogenous dsDNA (18-30 h post cytostatic treatment), they develop a very characteristic set of symptoms and 80-90% of such animals succumb within 6-25 days. This was called "delayed death" phenomenon, and the gap between cyclophosphamide and DNA injections required for such phenotype to develop was termed "death window". We established that mice succumbed to multi-organ failure, which was caused by systemic inflammation and sepsis. These processes unfolded along with accidental involution of lymphoid organs, which resulted from the failure of CD34(+) hematopoietic stem cells to differentiate into lymphoid lineage progenitors. Here we compare SPF and non-SPF animals, and demonstrate that the major cause of systemic inflammation and sepsis observed upon such treatments is activation of an opportunistic infection. Mice of the same strain (CBA) housed under SPF conditions do not develop the characteristic symptoms, nor do they become moribund. Yet, regardless of the breeding conditions, upon synergistic action of cyclophosphamide and dsDNA, CD34(+) hematopoietic stem cells consistently fail to give rise to lymphoid lineage progenitors. We demonstrate that this differentiation defect is reversible and that population of lymphoid progenitors is restored by day 29 after cyclophosphamide injection.
Collapse
Affiliation(s)
- Evgeniya V Dolgova
- Institute of Cytology and Genetics, Siberian Branch, Russian Academy of Sciences, Novosibirsk 630090, Russia.
| | - Yaroslav R Efremov
- Institute of Cytology and Genetics, Siberian Branch, Russian Academy of Sciences, Novosibirsk 630090, Russia
| | - Oleg S Taranov
- The State Research Center of Virology and Biotechnology VECTOR, Koltsovo, Novosibirsk Region 630559, Russia
| | - Ekaterina A Potter
- Institute of Cytology and Genetics, Siberian Branch, Russian Academy of Sciences, Novosibirsk 630090, Russia
| | - Valeriy P Nikolin
- Institute of Cytology and Genetics, Siberian Branch, Russian Academy of Sciences, Novosibirsk 630090, Russia
| | - Nelly A Popova
- Institute of Cytology and Genetics, Siberian Branch, Russian Academy of Sciences, Novosibirsk 630090, Russia; Novosibirsk State University, Novosibirsk 630090, Russia
| | - Vladimir V Omigov
- The State Research Center of Virology and Biotechnology VECTOR, Koltsovo, Novosibirsk Region 630559, Russia
| | - Elena R Chernykh
- Institute of Clinical Immunology, Siberian Branch, Russian Academy of Medical Sciences, Novosibirsk 630099, Russia
| | - Anastasia S Proskurina
- Institute of Cytology and Genetics, Siberian Branch, Russian Academy of Sciences, Novosibirsk 630090, Russia
| | - Sergey S Bogachev
- Institute of Cytology and Genetics, Siberian Branch, Russian Academy of Sciences, Novosibirsk 630090, Russia
| |
Collapse
|
38
|
Multiple Functions of Glutamate Uptake via Meningococcal GltT-GltM L-Glutamate ABC Transporter in Neisseria meningitidis Internalization into Human Brain Microvascular Endothelial Cells. Infect Immun 2015; 83:3555-67. [PMID: 26099588 DOI: 10.1128/iai.00654-15] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 06/17/2015] [Indexed: 12/30/2022] Open
Abstract
We previously reported that Neisseria meningitidis internalization into human brain microvasocular endothelial cells (HBMEC) was triggered by the influx of extracellular L-glutamate via the GltT-GltM L-glutamate ABC transporter, but the underlying mechanism remained unclear. We found that the ΔgltT ΔgltM invasion defect in assay medium (AM) was alleviated in AM without 10% fetal bovine serum (FBS) [AM(-S)]. The alleviation disappeared again in AM(-S) supplemented with 500 μM glutamate. Glutamate uptake by the ΔgltT ΔgltM mutant was less efficient than that by the wild-type strain, but only upon HBMEC infection. We also observed that both GltT-GltM-dependent invasion and accumulation of ezrin, a key membrane-cytoskeleton linker, were more pronounced when N. meningitidis formed larger colonies on HBMEC under physiological glutamate conditions. These results suggested that GltT-GltM-dependent meningococcal internalization into HBMEC might be induced by the reduced environmental glutamate concentration upon infection. Furthermore, we found that the amount of glutathione within the ΔgltT ΔgltM mutant was much lower than that within the wild-type N. meningitidis strain only upon HBMEC infection and was correlated with intracellular survival. Considering that the L-glutamate obtained via GltT-GltM is utilized as a nutrient in host cells, l-glutamate uptake via GltT-GltM plays multiple roles in N. meningitidis internalization into HBMEC.
Collapse
|
39
|
Shek WR, Smith AL, Pritchett-Corning KR. Microbiological Quality Control for Laboratory Rodents and Lagomorphs. LABORATORY ANIMAL MEDICINE 2015. [PMCID: PMC7150201 DOI: 10.1016/b978-0-12-409527-4.00011-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Mice (Mus musculus), rats (Rattus norvegicus), other rodent species, and domestic rabbits (Oryctolagus cuniculus) have been used in research for over 100 years. During the first half of the 20th century, microbiological quality control of lab animals was at best rudimentary as colonies were conventionally housed and little or no diagnostic testing was done. Hence, animal studies were often curtailed and confounded by infectious disease (Mobraaten and Sharp, 1999; Morse, 2007; Weisbroth, 1999). By the 1950s, it became apparent to veterinarians in the nascent field of comparative medicine that disease-free animals suitable for research could not be produced by standard veterinary disease control measures (e.g., improved sanitation and nutrition, antimicrobial treatments) in conventional facilities. Henry Foster, the veterinarian who founded Charles River Breeding Laboratories in 1948 and a pioneer in the large-scale production of laboratory rodents, stated in a seminar presented at the 30th anniversary of AALAS, “After a variety of frustrating health-related problems, it was decided that a major change in the company’s philosophy was required and an entirely different approach was essential”. Consequently, he and others developed innovative biosecurity systems to eliminate and exclude pathogens (Allen, 1999). In 1958, Foster reported on the Cesarean-originated barrier-sustained (COBS) process for the large-scale production of specific pathogen-free (SPF) laboratory rodents (Foster, 1958). To eliminate horizontally transmitted pathogens, a hysterectomy was performed on a near-term dam from a contaminated or conventionally housed colony. The gravid uterus was pulled through a disinfectant solution into a sterile flexible film isolator where the pups were removed from the uterus and suckled on axenic (i.e., germ-free) foster dams. After being mated to expand their number and associated with a cocktail of nonpathogenic bacteria to normalize their physiology and prime their immune system, rederived rodents were transferred to so-called barrier rooms for large-scale production. The room-level barrier to adventitious infection entailed disinfection of the room, equipment, and supplies, limiting access to trained and properly gowned personnel, and the application of new technologies such as high-efficiency particulate air-filtration of incoming air (Dubos and Schaedler, 1960; Foster, 1980; Schaedler and Orcutt, 1983; Trexler and Orcutt, 1999). The axenic and associated rodents mentioned in the COBS process are collectively classified as gnotobiotic to indicate that they have a completely known microflora. By contrast, barrier-reared rodent colonies are not gnotobiotic because they are housed in uncovered cages and thus acquire a complex microflora from the environment, supplies, personnel, and other sources. Instead, they are described as SPF to indicate that according to laboratory testing, they are free from infection with a defined list of infectious agents, commonly known as an ‘exclusion’ list.
Collapse
|
40
|
Girard A, Roques E, Massie B, Archambault D. Flagellin in fusion with human rotavirus structural proteins exerts an adjuvant effect when delivered with replicating but non-disseminating adenovectors through the intrarectal route. Mol Biotechnol 2014; 56:394-407. [PMID: 24271565 DOI: 10.1007/s12033-013-9723-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Human rotavirus (HRV) is the worldwide leading cause of gastroenteritis in young children. Two live attenuated HRV vaccines have been approved since 2006. However, these live vaccines still have potential risks including reversion of virulence. Adenoviruses are suitable vectors for mucosal administration of subunit vaccines. In addition to the adjuvant effect of certain adenovirus components, the use of an adjuvant like flagellin is also another means to increase the immune response to the immunogen. The aim of this study was to determine whether flagellin in fusion with HRV structural proteins stimulates the innate immune response and enhances the HRV-specific immune response when delivered through the intrarectal route with replicating but non-disseminating adenovector (R-AdV). Salmonella typhimurium flagellin B (FljB) in fusion with HRV VP4Δ::VP7 protein induced IL-1β production in J774A.1 macrophages exposed to the R-AdV. Intrarectal administration of R-AdVs expressing either VP4Δ::VP7 or VP4Δ::VP7::FljB in BALB/c mice resulted in HRV-specific mixed Th1/Th2 immune responses. The HRV-specific antibody response elicited with the use of R-AdV expressing VP4Δ::VP7::FljB was higher than that with R-AdV expressing VP4Δ::VP7. The results also show that the replication capability of R-AdVs contributed to enhance the HRV-specific immune response as compared with that obtained with non-replicative AdVs. This work lays the foundation for using the R-AdV system and FljB-adjuvanted formulation to elicit a mucosal immune response specific to HRV.
Collapse
Affiliation(s)
- Aurélie Girard
- Department of Biological Sciences, University of Québec at Montréal, P.O. Box 8888, Succursale Centre-Ville, Montreal, QC, H3C 3P8, Canada
| | | | | | | |
Collapse
|
41
|
Wigley P. Salmonella enterica in the Chicken: How it has Helped Our Understanding of Immunology in a Non-Biomedical Model Species. Front Immunol 2014; 5:482. [PMID: 25346731 PMCID: PMC4193332 DOI: 10.3389/fimmu.2014.00482] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Accepted: 09/20/2014] [Indexed: 11/13/2022] Open
Abstract
Salmonella infection of the chicken is important both as a source of foodborne human salmonellosis and as a source of disease in the chicken itself. Vaccination and other control strategies require an understanding of the immune response and as such have been important in understanding both mucosal immunity and more generally the response to bacterial infection. In this review, we discuss the contribution the study of avian salmonellosis has made to understanding innate immunity including the function of phagocytic cells, pattern recognition receptors, and defensins. The mucosal response to Salmonella infection and its regulation and the contribution this makes in protection against infection and persistence within the gut and future directions in better understanding the role of TH17 and Tregs in this response. Finally, we discuss the role of the immune system and its modulation in persistent infection and infection of the reproductive tract. We also outline key areas of research required to fully understand the interaction between the chicken immune system and Salmonella and how infection is maintained in the absence of substantive gastrointestinal disease.
Collapse
Affiliation(s)
- Paul Wigley
- Department of Infection Biology, School of Veterinary Science and Institute for Infection and Global Health, University of Liverpool, Liverpool, UK
| |
Collapse
|
42
|
Talebi Bezmin Abadi A. Helicobacter pylori: A Beneficial Gastric Pathogen? Front Med (Lausanne) 2014; 1:26. [PMID: 25593901 PMCID: PMC4291894 DOI: 10.3389/fmed.2014.00026] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2014] [Accepted: 08/12/2014] [Indexed: 12/24/2022] Open
Affiliation(s)
- Amin Talebi Bezmin Abadi
- Department of Medical Microbiology, University Medical Center Utrecht , Utrecht , Netherlands ; Department of Bacteriology, Faculty of Medical Sciences, Tarbiat Modares University , Tehran , Iran
| |
Collapse
|
43
|
Schoen C, Kischkies L, Elias J, Ampattu BJ. Metabolism and virulence in Neisseria meningitidis. Front Cell Infect Microbiol 2014; 4:114. [PMID: 25191646 PMCID: PMC4138514 DOI: 10.3389/fcimb.2014.00114] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Accepted: 07/31/2014] [Indexed: 01/14/2023] Open
Abstract
A longstanding question in infection biology addresses the genetic basis for invasive behavior in commensal pathogens. A prime example for such a pathogen is Neisseria meningitidis. On the one hand it is a harmless commensal bacterium exquisitely adapted to humans, and on the other hand it sometimes behaves like a ferocious pathogen causing potentially lethal disease such as sepsis and acute bacterial meningitis. Despite the lack of a classical repertoire of virulence genes in N. meningitidis separating commensal from invasive strains, molecular epidemiology suggests that carriage and invasive strains belong to genetically distinct populations. In recent years, it has become increasingly clear that metabolic adaptation enables meningococci to exploit host resources, supporting the concept of nutritional virulence as a crucial determinant of invasive capability. Here, we discuss the contribution of core metabolic pathways in the context of colonization and invasion with special emphasis on results from genome-wide surveys. The metabolism of lactate, the oxidative stress response, and, in particular, glutathione metabolism as well as the denitrification pathway provide examples of how meningococcal metabolism is intimately linked to pathogenesis. We further discuss evidence from genome-wide approaches regarding potential metabolic differences between strains from hyperinvasive and carriage lineages and present new data assessing in vitro growth differences of strains from these two populations. We hypothesize that strains from carriage and hyperinvasive lineages differ in the expression of regulatory genes involved particularly in stress responses and amino acid metabolism under infection conditions.
Collapse
Affiliation(s)
- Christoph Schoen
- Institute for Hygiene and Microbiology, University of Würzburg Würzburg, Germany ; Research Center for Infectious Diseases (ZINF), University of Würzburg Würzburg, Germany
| | - Laura Kischkies
- Institute for Hygiene and Microbiology, University of Würzburg Würzburg, Germany
| | - Johannes Elias
- Institute for Hygiene and Microbiology, University of Würzburg Würzburg, Germany ; National Reference Centre for Meningococci and Haemophilus influenzae (NRZMHi), University of Würzburg Würzburg, Germany
| | - Biju Joseph Ampattu
- Institute for Hygiene and Microbiology, University of Würzburg Würzburg, Germany
| |
Collapse
|
44
|
Angelakis E, Raoult D. Pathogenicity and treatment of Bartonella infections. Int J Antimicrob Agents 2014; 44:16-25. [DOI: 10.1016/j.ijantimicag.2014.04.006] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Accepted: 04/30/2014] [Indexed: 10/25/2022]
|
45
|
Ecto-5'-nucleotidase (CD73) regulates host inflammatory responses and exacerbates murine salmonellosis. Sci Rep 2014; 4:4486. [PMID: 24670982 PMCID: PMC3967249 DOI: 10.1038/srep04486] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Accepted: 03/10/2014] [Indexed: 01/13/2023] Open
Abstract
Food-borne Salmonella spp., are a major cause of hospitalization and death. Adenosine, an important immune regulator of inflammation, limits tissue damage during infection. CD39 (nucleoside triphosphate dephosphorylase) combined with ecto-5′-nucleotidase (CD73) metabolizes ATP to adenosine. We studied the expressions of CD39 and CD73 in tissues, and T helper cells in mice after Salmonella infection and evaluated the role of CD73 in regulating immune responses and bacterial clearance in wild-type and CD73-deficient (CD73−/−) mice. Both CD39 and CD73 transcript levels declined in the infected wild-type mice. Compared to wild-type mice, tissues from infected CD73−/− mice had significantly higher expression of pro-inflammatory cytokines and reduced anti-inflammatory responses. CD73−/− mice were more resistant to infection and had a greater inflammatory responses and a significantly lower bacterial load in the liver compared to wild-type mice. Thus, CD73 expression attenuates inflammation during murine Salmonellosis and impairs immunity, leading to increased bacterial colonization and prolonged infection.
Collapse
|
46
|
Abstract
The pathogenetic mechanisms leading to asthma are likely to be diverse, influenced by multiple genetic polymorphisms as well as elements of the environment. Recent data on the microbiome of the airway have revealed intriguing differences between the number and diversity of microbial populations in healthy persons and asthmatics. There is convincing evidence that early viral infections, particularly with human rhinovirus and respiratory syncytial virus, are often associated with the development of chronic asthma and with exacerbations. Recent studies suggest that two unrelated types of atypical bacteria, Mycoplasma pneumoniae (Mpn) and Chlamydia pneumoniae, are present in the airways of a substantial proportion of the population, bringing up the possibility that the persistent presence of the organism may contribute to the asthmatic phenotype in a subset of patients. This review will examine the current data regarding a possible role for infection in chronic asthma with a particular focus on atypical bacterial infections.
Collapse
Affiliation(s)
- T Prescott Atkinson
- Children's of Alabama CPP M220, 1601 4th Ave South, Birmingham, AL, 35233, USA,
| |
Collapse
|
47
|
Ramachandran G. Gram-positive and gram-negative bacterial toxins in sepsis: a brief review. Virulence 2014; 5:213-8. [PMID: 24193365 PMCID: PMC3916377 DOI: 10.4161/viru.27024] [Citation(s) in RCA: 247] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Revised: 10/28/2013] [Accepted: 10/31/2013] [Indexed: 12/31/2022] Open
Abstract
Bacterial sepsis is a major cause of fatality worldwide. Sepsis is a multi-step process that involves an uncontrolled inflammatory response by the host cells that may result in multi organ failure and death. Both gram-negative and gram-positive bacteria play a major role in causing sepsis. These bacteria produce a range of virulence factors that enable them to escape the immune defenses and disseminate to remote organs, and toxins that interact with host cells via specific receptors on the cell surface and trigger a dysregulated immune response. Over the past decade, our understanding of toxins has markedly improved, allowing for new therapeutic strategies to be developed. This review summarizes some of these toxins and their role in sepsis.
Collapse
Affiliation(s)
- Girish Ramachandran
- Center for Vaccine Development; Department of Medicine; University of Maryland School of Medicine; Baltimore, MD USA
| |
Collapse
|
48
|
Vaumourin E, Gasqui P, Buffet JP, Chapuis JL, Pisanu B, Ferquel E, Vayssier-Taussat M, Vourc'h G. A probabilistic model in cross-sectional studies for identifying interactions between two persistent vector-borne pathogens in reservoir populations. PLoS One 2013; 8:e66167. [PMID: 23840418 PMCID: PMC3688727 DOI: 10.1371/journal.pone.0066167] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2013] [Accepted: 05/03/2013] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND In natural populations, individuals are infected more often by several pathogens than by just one. In such a context, pathogens can interact. This interaction could modify the probability of infection by subsequent pathogens. Identifying when pathogen associations correspond to biological interactions is a challenge in cross-sectional studies where the sequence of infection cannot be demonstrated. METHODOLOGY/PRINCIPAL FINDINGS Here we modelled the probability of an individual being infected by one and then another pathogen, using a probabilistic model and maximum likelihood statistics. Our model was developed to apply to cross-sectional data, vector-borne and persistent pathogens, and to take into account confounding factors. Our modelling approach was more powerful than the commonly used Chi-square test of independence. Our model was applied to detect potential interaction between Borrelia afzelii and Bartonella spp. that infected a bank vole population at 11% and 57% respectively. No interaction was identified. CONCLUSIONS/SIGNIFICANCE The modelling approach we proposed is powerful and can identify the direction of potential interaction. Such an approach can be adapted to other types of pathogens, such as non-persistents. The model can be used to identify when co-occurrence patterns correspond to pathogen interactions, which will contribute to understanding how organism communities are assembled and structured. In the long term, the model's capacity to better identify pathogen interactions will improve understanding of infectious risk.
Collapse
Affiliation(s)
- Elise Vaumourin
- INRA, UR346 Epidémiologie Animale, Saint Genès Champanelle, France.
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Abstract
The immune response to sepsis can be seen as a pattern recognition receptor-mediated dysregulation of the immune system following pathogen invasion in which a careful balance between inflammatory and anti-inflammatory responses is vital. Invasive infection triggers both pro-inflammatory and anti-inflammatory host responses, the magnitude of which depends on multiple factors, including pathogen virulence, site of infection, host genetics, and comorbidities. Toll-like receptors, the inflammasomes, and other pattern recognition receptors initiate the immune response after recognition of danger signals derived from microorganisms, so-called pathogen-associated molecular patterns or derived from the host, so-called danger-associated molecular patterns. Further dissection of the role of host–pathogen interactions, the cytokine response, the coagulation cascade, and their multidirectional interactions in sepsis should lead toward the development of new therapeutic strategies in sepsis.
Collapse
Affiliation(s)
- Willem Joost Wiersinga
- Center for Infection and Immunity Amsterdam (CINIMA); Center for Experimental and Molecular Medicine (CEMM); Division of Infectious Diseases; Academic Medical Center; University of Amsterdam; Amsterdam, The Netherlands
| | - Stije J Leopold
- Center for Infection and Immunity Amsterdam (CINIMA); Center for Experimental and Molecular Medicine (CEMM); Division of Infectious Diseases; Academic Medical Center; University of Amsterdam; Amsterdam, The Netherlands
| | - Duncan R Cranendonk
- Center for Infection and Immunity Amsterdam (CINIMA); Center for Experimental and Molecular Medicine (CEMM); Division of Infectious Diseases; Academic Medical Center; University of Amsterdam; Amsterdam, The Netherlands
| | - Tom van der Poll
- Center for Infection and Immunity Amsterdam (CINIMA); Center for Experimental and Molecular Medicine (CEMM); Division of Infectious Diseases; Academic Medical Center; University of Amsterdam; Amsterdam, The Netherlands
| |
Collapse
|
50
|
Sriramulu D. Evolution and impact of bacterial drug resistance in the context of cystic fibrosis disease and nosocomial settings. Microbiol Insights 2013; 6:29-36. [PMID: 24826072 PMCID: PMC3987750 DOI: 10.4137/mbi.s10792] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The use of antibiotics is unavoidable in trying to treat acute infections and in the prevention and control of chronic infections. Over the years, an ever increasing number of infections has escalated the use of antibiotics, which has necessitated action against an emerging bacterial resistance. There seems to be a continuous acquisition of new resistance mechanisms among bacteria that switch niches between human, animals, and the environment. An antibiotic resistant strain emerges when it acquires the DNA that confers the added capacity needed to survive in an unusual niche. Once acquired, a new resistance mechanism evolves according to the dynamics of the microenvironment; there is then a high probability that it is transferred to other species or to an avirulent strain of the same species. A well understood model for studying emerging antibiotic resistance and its impact is Pseudomonas aeruginosa, an opportunistic pathogen which is able to cause acute and chronic infections in nosocomial settings. This bacterium has a huge genetic repertoire consisting of genes that encode both innate and acquired antibiotic resistance traits. Besides acute infections, chronic colonization of P. aeruginosa in the lungs of cystic fibrosis (CF) patients plays a significant role in morbidity and mortality. Antibiotics used in the treatment of such infections has increased the longevity of patients over the last several decades. However, emerging multidrug resistant strains and the eventual increase in the dosage of antibiotic(s) is of major concern. Though there are various infections that are treated by single/combined antibiotics, the particular case of P. aeruginosa infection in CF patients serves as a reference for understanding the impact of overuse of antibiotics and emerging antibiotic resistant strains. This mini review presents the need for judicious use of antibiotics to treat various types of infections, protecting patients and the environment, as well as achieving a better treatment outcome.
Collapse
Affiliation(s)
- Dinesh Sriramulu
- Shres Consultancy, Aparna Towers, Near Lakshmi Hospital, Chittur Road, Palakkad, Kerala, India
| |
Collapse
|