Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
1.
Elife ; 122024 Jan 09.
Article in English | MEDLINE | ID: mdl-38193896

ABSTRACT

The white-footed deermouse Peromyscus leucopus, a long-lived rodent, is a key reservoir in North America for agents of several zoonoses, including Lyme disease, babesiosis, anaplasmosis, and a viral encephalitis. While persistently infected, this deermouse is without apparent disability or diminished fitness. For a model for inflammation elicited by various pathogens, the endotoxin lipopolysaccharide (LPS) was used to compare genome-wide transcription in blood by P. leucopus, Mus musculus, and Rattus norvegicus and adjusted for white cell concentrations. Deermice were distinguished from the mice and rats by LPS response profiles consistent with non-classical monocytes and alternatively-activated macrophages. LPS-treated P. leucopus, in contrast to mice and rats, also displayed little transcription of interferon-gamma and lower magnitude fold-changes in type 1 interferon-stimulated genes. These characteristics of P. leucopus were also noted in a Borrelia hermsii infection model. The phenomenon was associated with comparatively reduced transcription of endogenous retrovirus sequences and cytoplasmic pattern recognition receptors in the deermice. The results reveal a mechanism for infection tolerance in this species and perhaps other animal reservoirs for agents of human disease.


Lyme disease is an illness caused by bacteria that spread from infected animals to humans through tick bites. While most people fully recover after a week or two of antibiotic treatments, some will continue to experience debilitating symptoms due, potentially, to the way their immune system responded to the infection. In North America, the white-footed deermouse is one of the most common hosts of the Lyme disease bacteria. Despite its name, this rodent is more closely related to hamsters than to the mice or rats most often used in laboratory studies. Unlike mice and humans, however, deermice carrying Lyme disease bacteria do not get sick; in fact, most deermice living in a Lyme disease region will acquire the infection during their lifetimes, but it has little apparent effect on population numbers. These animals can also better tolerate infection from other microbes. To investigate why this is the case, Milovic et al. exposed mice, rats and deermice to a bacterial toxin that triggers inflammation common to encounters with many kinds of microbes. While all species exhibited physical symptoms as a result, blood samples revealed that mice and rats, but not deermice, reacted as if they were infected with viruses as well as bacteria. This was particularly the case for interferons, a group of hormone-like proteins that protect against viruses but can also lead to harmful long-term inflammatory effects. The deermice controlled their interferon responses to the bacterial substance in a way that mice and rats could not. Milovic et al. also checked which genes each species switched on after exposure to the toxin. This revealed that, unlike deer mice, rats and mice turned on some DNA sequences called endogenous retroviruses, which have no role in fighting infection from bacteria but can lead to harmful persistent inflammation. These results provide elements to better understand why recovery from Lyme disease may differ between people, with some patients retaining symptoms long after their infection has abated. They could also help to better grasp why other diseases, such as COVID-19, can be followed by fatigue and other symptoms of ongoing inflammation.


Subject(s)
Endotoxins , Interferon Type I , Humans , Mice , Animals , Rats , Lipopolysaccharides , Interferon-gamma , Zoonoses
2.
bioRxiv ; 2023 Oct 12.
Article in English | MEDLINE | ID: mdl-37745581

ABSTRACT

The white-footed deermouse Peromyscus leucopus, a long-lived rodent, is a key reservoir for agents of several zoonoses, including Lyme disease. While persistently infected, this deermouse is without apparent disability or diminished fitness. For a model for inflammation elicited by various pathogens, the endotoxin lipopolysaccharide (LPS) was used to compare genome-wide transcription in blood by P. leucopus, Mus musculus and Rattus norvegicus and adjusted for white cell concentrations. Deermice were distinguished from the mice and rats by LPS response profiles consistent with non-classical monocytes and alternatively-activated macrophages. LPS-treated P. leucopus, in contrast to mice and rats, also displayed little transcription of interferon-gamma and lower magnitude fold-changes in type 1 interferon-stimulated genes. This was associated with comparatively reduced transcription of endogenous retrovirus sequences and cytoplasmic pattern recognition receptors in the deermice. The results reveal a mechanism for infection tolerance in this species and perhaps other animal reservoirs for agents of human disease.

3.
mBio ; 12(2)2021 04 13.
Article in English | MEDLINE | ID: mdl-33849979

ABSTRACT

Animals that are competent reservoirs of zoonotic pathogens commonly suffer little morbidity from the infections. To investigate mechanisms of this tolerance of infection, we used single-dose lipopolysaccharide (LPS) as an experimental model of inflammation and compared the responses of two rodents: Peromyscus leucopus, the white-footed deermouse and reservoir for the agents of Lyme disease and other zoonoses, and the house mouse Mus musculus Four hours after injection with LPS or saline, blood, spleen, and liver samples were collected and subjected to transcriptome sequencing (RNA-seq), metabolomics, and specific reverse transcriptase quantitative PCR (RT-qPCR). Differential expression analysis was at the gene, pathway, and network levels. LPS-treated deermice showed signs of sickness similar to those of exposed mice and had similar increases in corticosterone levels and expression of interleukin 6 (IL-6), tumor necrosis factor, IL-1ß, and C-reactive protein. By network analysis, the M. musculus response to LPS was characterized as cytokine associated, while the P. leucopus response was dominated by neutrophil activity terms. In addition, dichotomies in the expression levels of arginase 1 and nitric oxide synthase 2 and of IL-10 and IL-12 were consistent with type M1 macrophage responses in mice and type M2 responses in deermice. Analysis of metabolites in plasma and RNA in organs revealed species differences in tryptophan metabolism. Two genes in particular signified the different phenotypes of deermice and mice: the Slpi and Ibsp genes. Key RNA-seq findings for P. leucopus were replicated in older animals, in a systemic bacterial infection, and with cultivated fibroblasts. The findings indicate that P. leucopus possesses several adaptive traits to moderate inflammation in its balancing of infection resistance and tolerance.IMPORTANCE Animals that are natural carriers of pathogens that cause human diseases commonly manifest little or no sickness as a consequence of infection. Examples include the deermouse, Peromyscus leucopus, which is a reservoir for Lyme disease and several other disease agents in North America, and some types of bats, which are carriers of viruses with pathogenicity for humans. Mechanisms of this phenomenon of infection tolerance and entailed trade-off costs are poorly understood. Using a single injection of lipopolysaccharide (LPS) endotoxin as a proxy for infection, we found that deermice differed from the mouse (Mus musculus) in responses to LPS in several diverse pathways, including innate immunity, oxidative stress, and metabolism. Features distinguishing the deermice cumulatively would moderate downstream ill effects of LPS. Insights gained from the P. leucopus model in the laboratory have implications for studying infection tolerance in other important reservoir species, including bats and other types of wildlife.


Subject(s)
Disease Reservoirs/microbiology , Endotoxins/administration & dosage , Inflammation/genetics , Peromyscus/microbiology , Zoonoses/immunology , Zoonoses/microbiology , Animals , Disease Susceptibility/etiology , Disease Susceptibility/immunology , Endotoxins/immunology , Female , Gene Expression Profiling , Inflammation/immunology , Lyme Disease/microbiology , Male , Metabolomics , Mice , Mice, Inbred BALB C , Peromyscus/immunology , Sequence Analysis, RNA
4.
PLoS One ; 15(8): e0231801, 2020.
Article in English | MEDLINE | ID: mdl-32817657

ABSTRACT

The cricetine rodent Peromyscus leucopus is an important reservoir for several human zoonoses, including Lyme disease, in North America. Akin to hamsters, the white-footed deermouse has been unevenly characterized in comparison to the murid Mus musculus. To further understanding of P. leucopus' total genomic content, we investigated gut microbiomes of an outbred colony of P. leucopus, inbred M. musculus, and a natural population of P. leucopus. Metagenome and whole genome sequencing were combined with microbiology and microscopy approaches. A focus was the genus Lactobacillus, four diverse species of which were isolated from forestomach and feces of colony P. leucopus. Three of the species-L. animalis, L. reuteri, and provisionally-named species "L. peromysci"-were identified in fecal metagenomes of wild P. leucopus but not discernibly in samples from M. musculus. L. johnsonii, the fourth species, was common in M. musculus but absent or sparse in wild P. leucopus. Also identified in both colony and natural populations were a Helicobacter sp. in feces but not stomach, and a Tritrichomonas sp. protozoan in cecum or feces. The gut metagenomes of colony P. leucopus were similar to those of colony M. musculus at the family or higher level and for major subsystems. But there were multiple differences between species and sexes within each species in their gut metagenomes at orthologous gene level. These findings provide a foundation for hypothesis-testing of functions of individual microbial species and for interventions, such as bait vaccines based on an autochthonous bacterium and targeting P. leucopus for transmission-blocking.


Subject(s)
Gastrointestinal Microbiome/genetics , Peromyscus/microbiology , Zoonoses/microbiology , Animals , Humans , Lactobacillus/genetics , Lactobacillus/metabolism , Lyme Disease/epidemiology , Lyme Disease/etiology , North America , Peromyscus/genetics , Zoonoses/genetics
5.
Microbiol Resour Announc ; 8(40)2019 Oct 03.
Article in English | MEDLINE | ID: mdl-31582438

ABSTRACT

Three colony types of Lactobacillus were isolated from the stomach of LL colony stock Peromyscus leucopus deermice, a reservoir for several human zoonoses. Genome sequences revealed two isolates to be new strains of Lactobacillus animalis and Lactobacillus reuteri The third was distinct from known species and was provisionally designated Lactobacillus sp. strain LL6.

6.
N Engl J Med ; 363(11): 1005-15, 2010 Sep 09.
Article in English | MEDLINE | ID: mdl-20825313

ABSTRACT

BACKGROUND: Global control of tuberculosis is hampered by slow, insensitive diagnostic methods, particularly for the detection of drug-resistant forms and in patients with human immunodeficiency virus infection. Early detection is essential to reduce the death rate and interrupt transmission, but the complexity and infrastructure needs of sensitive methods limit their accessibility and effect. METHODS: We assessed the performance of Xpert MTB/RIF, an automated molecular test for Mycobacterium tuberculosis (MTB) and resistance to rifampin (RIF), with fully integrated sample processing in 1730 patients with suspected drug-sensitive or multidrug-resistant pulmonary tuberculosis. Eligible patients in Peru, Azerbaijan, South Africa, and India provided three sputum specimens each. Two specimens were processed with N-acetyl-L-cysteine and sodium hydroxide before microscopy, solid and liquid culture, and the MTB/RIF test, and one specimen was used for direct testing with microscopy and the MTB/RIF test. RESULTS: Among culture-positive patients, a single, direct MTB/RIF test identified 551 of 561 patients with smear-positive tuberculosis (98.2%) and 124 of 171 with smear-negative tuberculosis (72.5%). The test was specific in 604 of 609 patients without tuberculosis (99.2%). Among patients with smear-negative, culture-positive tuberculosis, the addition of a second MTB/RIF test increased sensitivity by 12.6 percentage points and a third by 5.1 percentage points, to a total of 90.2%. As compared with phenotypic drug-susceptibility testing, MTB/RIF testing correctly identified 200 of 205 patients (97.6%) with rifampin-resistant bacteria and 504 of 514 (98.1%) with rifampin-sensitive bacteria. Sequencing resolved all but two cases in favor of the MTB/RIF assay. CONCLUSIONS: The MTB/RIF test provided sensitive detection of tuberculosis and rifampin resistance directly from untreated sputum in less than 2 hours with minimal hands-on time. (Funded by the Foundation for Innovative New Diagnostics.)


Subject(s)
Antitubercular Agents/pharmacology , Drug Resistance, Bacterial , Mycobacterium tuberculosis/drug effects , Polymerase Chain Reaction/methods , Rifampin/pharmacology , Tuberculosis, Multidrug-Resistant/diagnosis , Tuberculosis/microbiology , Adolescent , Adult , Aged , Aged, 80 and over , Bacterial Proteins/genetics , DNA-Directed RNA Polymerases , Female , Humans , Male , Microbial Sensitivity Tests/methods , Middle Aged , Mycobacterium tuberculosis/genetics , Polymerase Chain Reaction/instrumentation , Prospective Studies , Reference Standards , Sensitivity and Specificity , Sputum/microbiology , Tuberculosis/diagnosis , Tuberculosis, Multidrug-Resistant/microbiology , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...