Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
Infect Control Hosp Epidemiol ; : 1-5, 2024 Apr 02.
Article in English | MEDLINE | ID: mdl-38561197

ABSTRACT

Using statewide surveillance, we describe candidemia in Connecticut during 2019-2020. Mortality was high among individuals with candidemia, and the readmission rate was high among survivors. Mortality and readmission were associated with hospital-onset candidemia. Understanding risk factors for mortality and readmission can optimize prevention strategies to reduce mortality and readmissions.

2.
Arthritis Care Res (Hoboken) ; 76(1): 120-130, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37605835

ABSTRACT

OBJECTIVE: The objective of this study was to assess the SARS-CoV-2-specific humoral and T cell response after a two-dose regimen of SARS-CoV-2 vaccine in patients with rheumatoid arthritis (RA). METHODS: In this observational study, patients with RA who are ≥18 years of age and vaccinated for SARS-CoV-2 according to the Argentine National Health Ministry's vaccination strategy were included. Anti-SARS-CoV-2 immunoglobulin G (IgG) antibodies (ELISA-COVIDAR test), neutralizing activity (cytotoxicity in VERO cells), and specific T cell response (IFN-γ ELISpot Assay) were assessed after the first and second dose. RESULTS: A total of 120 patients with RA were included. Mostly, homologous regimens were used, including Gam-COVID-Vac (27.5%), ChAdOx1 (24.2%), and BBIBP-CorV (22.5%). The most frequent combination was Gam-COVID-Vac/mRNA-1273 (21.7%). After the second dose, 81.7% presented with anti-SARS-CoV-2 antibodies, 70.0% presented with neutralizing activity, and 65.3% presented with specific T cell response. The use of BBIBP-CorV and treatment with abatacept (ABA) and rituximab (RTX) were associated with undetectable antibodies and no neutralizing activity after two doses. BBIBP-CorV was also associated with the absence of T cell response. The total incidence of adverse events was 357.1 events per 1,000 doses, significantly lower with BBIBP-CorV (166.7 events per 1,000 doses, P < 0.02). CONCLUSION: In this RA cohort vaccinated with homologous and heterologous regimens against COVID-19, 2 out of 10 patients did not develop anti-SARS-CoV-2 IgG, 70% presented with neutralizing activity, and 65% presented with specific T cell response. The use of BBIBP-CorV was associated with deficient humoral and cellular response, whereas treatment with ABA and RTX resulted in an impaired anti-SARS-CoV-2 IgG formation and neutralizing activity.


Subject(s)
Arthritis, Rheumatoid , COVID-19 , Chlorocebus aethiops , Animals , Humans , COVID-19 Vaccines , SARS-CoV-2 , Vero Cells , COVID-19/prevention & control , T-Lymphocytes , Arthritis, Rheumatoid/drug therapy , Abatacept , Rituximab , Vaccination , Antibodies, Viral , Immunoglobulin G
3.
J Extracell Vesicles ; 12(1): e12298, 2023 01.
Article in English | MEDLINE | ID: mdl-36604533

ABSTRACT

Over the last decade, research interest in defining how extracellular vesicles (EVs) shape cross-species communication has grown rapidly. Parasitic helminths, worm species found in the phyla Nematoda and Platyhelminthes, are well-recognised manipulators of host immune function and physiology. Emerging evidence supports a role for helminth-derived EVs in these processes and highlights EVs as an important participant in cross-phylum communication. While the mammalian EV field is guided by a community-agreed framework for studying EVs derived from model organisms or cell systems [e.g., Minimal Information for Studies of Extracellular Vesicles (MISEV)], the helminth community requires a supplementary set of principles due to the additional challenges that accompany working with such divergent organisms. These challenges include, but are not limited to, generating sufficient quantities of EVs for descriptive or functional studies, defining pan-helminth EV markers, genetically modifying these organisms, and identifying rigorous methodologies for in vitro and in vivo studies. Here, we outline best practices for those investigating the biology of helminth-derived EVs to complement the MISEV guidelines. We summarise community-agreed standards for studying EVs derived from this broad set of non-model organisms, raise awareness of issues associated with helminth EVs and provide future perspectives for how progress in the field will be achieved.


Subject(s)
Extracellular Vesicles , Helminths , Animals , Humans , Extracellular Vesicles/physiology , Reproducibility of Results , Mammals
4.
Front Cell Infect Microbiol ; 12: 976017, 2022.
Article in English | MEDLINE | ID: mdl-36034712

ABSTRACT

Gastrointestinal nematodes are a diverse class of pathogens that colonise a quarter of the world's human population and nearly all grazing livestock. These macroparasites establish, and some migrate, within host gastrointestinal niches during their life cycles and release molecules that condition the host mucosa to enable chronic infections. Understanding how helminths do this, and defining the molecules and mechanisms involved in host modulation, holds promise for novel strategies of anthelmintics and vaccines, as well as new knowledge of immune regulation and tissue repair. Yet the size and complexity of these multicellular parasites, coupled with the reliance on hosts to maintain their life cycles, present obstacles to interrogate how they interact with the gastric and intestinal epithelium, stroma and immune cells during infection, and also to develop protocols to genetically modify these parasites. Gastrointestinal organoids have transformed research on gastric and gut physiology during homeostasis and disease, including investigations on host-pathogen interactions with viruses, bacteria, protozoa and more recently, parasitic nematodes. Here we outline applications and important considerations for the best use of organoids to study gastrointestinal nematode development and interactions with their hosts. The careful use of different organoid culture configurations in order to achieve a closer replication of the in vivo infection context will lead not only to new knowledge on gastrointestinal nematode infection biology, but also towards the replication of their life cycles in vitro, and the development of valuable experimental tools such as genetically modified parasites.


Subject(s)
Gastrointestinal Diseases , Nematoda , Nematode Infections , Parasites , Animals , Host-Parasite Interactions , Humans , Organoids
5.
Nat Commun ; 13(1): 1725, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35365634

ABSTRACT

Whipworms are large metazoan parasites that inhabit multi-intracellular epithelial tunnels in the large intestine of their hosts, causing chronic disease in humans and other mammals. How first-stage larvae invade host epithelia and establish infection remains unclear. Here we investigate early infection events using both Trichuris muris infections of mice and murine caecaloids, the first in-vitro system for whipworm infection and organoid model for live helminths. We show that larvae degrade mucus layers to access epithelial cells. In early syncytial tunnels, larvae are completely intracellular, woven through multiple live dividing cells. Using single-cell RNA sequencing of infected mouse caecum, we reveal that progression of infection results in cell damage and an expansion of enterocytes expressing of Isg15, potentially instigating the host immune response to the whipworm and tissue repair. Our results unravel intestinal epithelium invasion by whipworms and reveal specific host-parasite interactions that allow the whipworm to establish its multi-intracellular niche.


Subject(s)
Helminths , Trichuriasis , Animals , Intestinal Mucosa , Intestines/parasitology , Mammals , Mice , Trichuris/physiology
6.
Trends Parasitol ; 38(2): 174-187, 2022 02.
Article in English | MEDLINE | ID: mdl-34538735

ABSTRACT

Although hatching from eggs is fundamental for nematode biology it remains poorly understood. For animal-parasitic nematodes in particular, advancement has been slow since the 1980s. Understanding such a crucial life-cycle process would greatly improve the tractability of parasitic nematodes as experimental systems, advance fundamental knowledge, and enable translational research. Here, we review the role of eggs in the nematode life cycle and the current knowledge on the hatching cascade, including the different inducing and contributing factors, and highlight specific areas of the field that remain unknown. We examine how these knowledge gaps could be addressed and discuss their potential impact and application in nematode parasite research, treatment, and control.


Subject(s)
Nematoda , Parasites , Animals , Host-Parasite Interactions
7.
Sci Rep ; 10(1): 10880, 2020 07 02.
Article in English | MEDLINE | ID: mdl-32616765

ABSTRACT

The microbiome is an assemblage of microorganisms living in association with a multicellular host. Numerous studies have identified a role for the microbiome in host physiology, development, immunity, and behaviour. The generation of axenic (germ-free) and gnotobiotic model systems has been vital to dissecting the role of the microbiome in host biology. We have previously reported the generation of axenic Aedes aegypti mosquitoes, the primary vector of several human pathogenic viruses, including dengue virus and Zika virus. In order to better understand the influence of the microbiome on mosquitoes, we examined the transcriptomes of axenic and conventionally reared Ae. aegypti before and after a blood meal. Our results suggest that the microbiome has a much lower effect on the mosquito's gene expression than previously thought with only 170 genes influenced by the axenic state, while in contrast, blood meal status influenced 809 genes. The pattern of expression influenced by the microbiome is consistent with transient changes similar to infection rather than sweeping physiological changes. While the microbiome does seem to affect some pathways such as immune function and metabolism, our data suggest the microbiome is primarily serving a nutritional role in development with only minor effects in the adult.


Subject(s)
Aedes/microbiology , Microbiota , Mosquito Vectors/microbiology , Transcriptome , Aedes/genetics , Aedes/growth & development , Aedes/metabolism , Animals , Axenic Culture , Blood , Diet , Drosophila melanogaster/metabolism , Female , Gene Ontology , Germ-Free Life , Larva , Mosquito Vectors/genetics , Mosquito Vectors/growth & development , Sugars , Transcription, Genetic
8.
Int J Parasitol ; 50(9): 707-718, 2020 08.
Article in English | MEDLINE | ID: mdl-32659277

ABSTRACT

The caecum, an intestinal appendage in the junction of the small and large intestines, displays a unique epithelium that serves as an exclusive niche for a range of pathogens including whipworms (Trichuris spp.). While protocols to grow organoids from small intestine (enteroids) and colon (colonoids) exist, the conditions to culture organoids from the caecum have yet to be described. Here, we report methods to grow, differentiate and characterise mouse adult stem cell-derived caecal organoids, termed caecaloids. We compare the cellular composition of caecaloids with that of enteroids, identifying differences in intestinal epithelial cell populations that mimic those found in the caecum and small intestine. The remarkable similarity in the intestinal epithelial cell composition and spatial conformation of caecaloids and their tissue of origin enables their use as an in vitro model to study host interactions with important caecal pathogens. Thus, exploiting this system, we investigated the responses of caecal intestinal epithelial cells to extracellular vesicles secreted/excreted by the intracellular helminth Trichuris muris. Our findings reveal novel immunoregulatory effects of whipworm extracellular vesicles on the caecal epithelium, including the downregulation of responses to nucleic acid recognition and type-I interferon signalling.


Subject(s)
Cecum/parasitology , Extracellular Vesicles/metabolism , Host-Parasite Interactions , Organoids , Trichuriasis/parasitology , Trichuris/metabolism , Animals , Mice , Mice, Inbred C57BL , Organoids/metabolism , Organoids/parasitology
9.
Int J Parasitol ; 50(9): 719-729, 2020 08.
Article in English | MEDLINE | ID: mdl-32659276

ABSTRACT

Extracellular vesicles (EVs) have emerged as a ubiquitous component of helminth excretory-secretory products that can deliver parasite molecules to host cells to elicit immunomodulatory effects. RNAs are one type of cargo molecule that can underpin EV functions, hence there is extensive interest in characterising the RNAs that are present in EVs from different helminth species. Here we outline methods for identifying all of the small RNAs (sRNA) in helminth EVs and address how different methodologies may influence the sRNAs detected. We show that different EV purification methods introduce relatively little variation in the sRNAs that are detected, and that different RNA library preparation methods yielded larger differences. We compared the EV sRNAs in the gastrointestinal nematode Heligmosomoides bakeri with those in EVs from the distantly related gastrointestinal nematode Trichuris muris, and found that many of the sRNAs in both organisms derive from repetitive elements or intergenic regions. However, only in H. bakeri do these RNAs contain a 5' triphosphate, and Guanine (G) starting nucleotide, consistent with their biogenesis by RNA-dependent RNA polymerases (RdRPs). Distinct microRNA (miRNA) families are carried in EVs from each parasite, with H. bakeri EVs specific for miR-71, miR-49, miR-63, miR-259 and miR-240 gene families, and T. muris EVs specific for miR-1, miR-1822 and miR-252, and enriched for miR-59, miR-72 and miR-44 families, with the miR-9, miR-10, miR-80 and let-7 families abundant in both. We found a larger proportion of miRNA reads derive from the mouse host in T. muris EVs, compared with H. bakeri EVs. Our report underscores potential biases in the sRNAs sequenced based on library preparation methods, suggests specific nematode lineages have evolved distinct sRNA synthesis/export pathways, and highlights specific differences in EV miRNAs from H. bakeri and T. muris that may underpin functional adaptation to their host niches.


Subject(s)
Extracellular Vesicles/metabolism , MicroRNAs , RNA, Helminth , RNA, Small Interfering , Trichuris/metabolism , Animals , MicroRNAs/isolation & purification , MicroRNAs/metabolism , RNA, Helminth/isolation & purification , RNA, Helminth/metabolism , RNA, Small Interfering/isolation & purification , RNA, Small Interfering/metabolism
10.
PLoS Negl Trop Dis ; 14(5): e0007754, 2020 05.
Article in English | MEDLINE | ID: mdl-32421713

ABSTRACT

Macroautophagy is an evolutionarily conserved cellular process critical for maintaining cellular homeostasis. It can additionally function as an innate immune response to viral infection as has been demonstrated for a number of arthropod-borne (arbo-) viruses. Arboviruses are maintained in a transmission cycle between vertebrate hosts and invertebrate vectors yet the majority of studies assessing autophagy-arbovirus interactions have been limited to the mammalian host. Therefore we evaluated the role of autophagy during arbovirus infection of the invertebrate vector using the tractable Aag2 Aedes aegypti mosquito cell culture system. Our data demonstrates that autophagy is significantly induced in mosquito cells upon infection with two divergent arboviruses: dengue virus-2 (DENV-2; Flaviviridae, Flavivirus) and chikungunya virus (CHIKV; Togaviridae, Alphavirus). While assessing the role of autophagy during arbovirus infection, we observed a somewhat paradoxical outcome. Both induction and suppression of autophagy via torin-1 and spautin-1, respectively, resulted in increased viral titers for both viruses, yet suppression of autophagy-related genes had no effect. Interestingly, chemical modulators of autophagy had either no effect or opposite effects in another widely used mosquito cell line, C6/36 Aedes albopictus cells. Together, our data reveals a limited role for autophagy during arbovirus infection of mosquito cells. Further, our findings suggest that commonly used chemical modulators of autophagy alter mosquito cells in such a way as to promote viral replication; however, it is unclear if this occurs directly through autophagic manipulation or other means.


Subject(s)
Aedes/physiology , Aedes/virology , Arboviruses/physiology , Autophagy , Aedes/genetics , Animals , Cell Line , Insect Proteins/genetics , Insect Proteins/metabolism , Virus Replication
11.
Nat Immunol ; 21(1): 86-100, 2020 01.
Article in English | MEDLINE | ID: mdl-31844327

ABSTRACT

By developing a high-density murine immunophenotyping platform compatible with high-throughput genetic screening, we have established profound contributions of genetics and structure to immune variation (http://www.immunophenotype.org). Specifically, high-throughput phenotyping of 530 unique mouse gene knockouts identified 140 monogenic 'hits', of which most had no previous immunologic association. Furthermore, hits were collectively enriched in genes for which humans show poor tolerance to loss of function. The immunophenotyping platform also exposed dense correlation networks linking immune parameters with each other and with specific physiologic traits. Such linkages limit freedom of movement for individual immune parameters, thereby imposing genetically regulated 'immunologic structures', the integrity of which was associated with immunocompetence. Hence, we provide an expanded genetic resource and structural perspective for understanding and monitoring immune variation in health and disease.


Subject(s)
Enterobacteriaceae Infections/immunology , Genetic Variation/genetics , High-Throughput Screening Assays/methods , Immunophenotyping/methods , Salmonella Infections/immunology , Animals , Citrobacter/immunology , Enterobacteriaceae Infections/microbiology , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Animal , Salmonella/immunology , Salmonella Infections/microbiology
12.
Trends Parasitol ; 36(2): 170-181, 2020 02.
Article in English | MEDLINE | ID: mdl-31791691

ABSTRACT

Organoids are multicellular culture systems that replicate tissue architecture and function, and are increasingly used as models of viral, bacterial, and protozoan infections. Organoids have great potential to improve our current understanding of helminth interactions with their hosts and to replace or reduce the dependence on using animal models. In this review, we discuss the applicability of this technology to helminth infection research, including strategies of co-culture of helminths or their products with organoids and the challenges, advantages, and drawbacks of the use of organoids for these studies. We also explore how complementing organoid systems with other cell types and components may allow more complex models to be generated in the future to further investigate helminth-host interactions.


Subject(s)
Helminths/physiology , Organoids/parasitology , Animals , Helminthiasis/parasitology , Humans , Parasitology/trends , Research/trends
13.
Front Genet ; 10: 826, 2019.
Article in English | MEDLINE | ID: mdl-31616465

ABSTRACT

Whole-genome sequencing is being rapidly applied to the study of helminth genomes, including de novo genome assembly, population genetics, and diagnostic applications. Although late-stage juvenile and adult parasites typically produce sufficient DNA for molecular analyses, these parasitic stages are almost always inaccessible in the live host; immature life stages found in the environment for which samples can be collected non-invasively offer a potential alternative; however, these samples typically yield very low quantities of DNA, can be environmentally resistant, and are susceptible to contamination, often from bacterial or host DNA. Here, we have tested five low-input DNA extraction protocols together with a low-input sequencing library protocol to assess the feasibility of whole-genome sequencing of individual immature helminth samples. These approaches do not use whole-genome amplification, a common but costly approach to increase the yield of low-input samples. We first tested individual parasites from two species spotted onto FTA cards-egg and L1 stages of Haemonchus contortus and miracidia of Schistosoma mansoni-before further testing on an additional five species-Ancylostoma caninum, Ascaridia dissimilis, Dirofilaria immitis, Strongyloides stercoralis, and Trichuris muris-with an optimal protocol. A sixth species-Dracunculus medinensis-was included for comparison. Whole-genome sequencing followed by analyses to determine the proportion of on- and off-target mapping revealed successful sample preparations for six of the eight species tested with variation both between species and between different life stages from some species described. These results demonstrate the feasibility of whole-genome sequencing of individual parasites, and highlight a new avenue toward generating sensitive, specific, and information-rich data for the diagnosis and surveillance of helminths.

14.
PLoS Pathog ; 15(1): e1007265, 2019 01.
Article in English | MEDLINE | ID: mdl-30640950

ABSTRACT

The whipworm Trichuris trichiura is a soil-transmitted helminth that dwells in the epithelium of the caecum and proximal colon of their hosts causing the human disease, trichuriasis. Trichuriasis is characterized by colitis attributed to the inflammatory response elicited by the parasite while tunnelling through intestinal epithelial cells (IECs). The IL-10 family of receptors, comprising combinations of subunits IL-10Rα, IL-10Rß, IL-22Rα and IL-28Rα, modulates intestinal inflammatory responses. Here we carefully dissected the role of these subunits in the resistance of mice to infection with T. muris, a mouse model of the human whipworm T. trichiura. Our findings demonstrate that whilst IL-22Rα and IL-28Rα are dispensable in the host response to whipworms, IL-10 signalling through IL-10Rα and IL-10Rß is essential to control caecal pathology, worm expulsion and survival during T. muris infections. We show that deficiency of IL-10, IL-10Rα and IL-10Rß results in dysbiosis of the caecal microbiota characterised by expanded populations of opportunistic bacteria of the families Enterococcaceae and Enterobacteriaceae. Moreover, breakdown of the epithelial barrier after whipworm infection in IL-10, IL-10Rα and IL-10Rß-deficient mice, allows the translocation of these opportunistic pathogens or their excretory products to the liver causing organ failure and lethal disease. Importantly, bone marrow chimera experiments indicate that signalling through IL-10Rα and IL-10Rß in haematopoietic cells, but not IECs, is crucial to control worm expulsion and immunopathology. These findings are supported by worm expulsion upon infection of conditional mutant mice for the IL-10Rα on IECs. Our findings emphasize the pivotal and complex role of systemic IL-10Rα signalling on immune cells in promoting microbiota homeostasis and maintaining the intestinal epithelial barrier, thus preventing immunopathology during whipworm infections.


Subject(s)
Interleukin-10/metabolism , Receptors, Interleukin-10/metabolism , Trichuris/immunology , Animals , Cytokines/metabolism , Disease Models, Animal , Gastrointestinal Microbiome/immunology , Homeostasis , Interleukins/metabolism , Intestines/microbiology , Intestines/pathology , Mice , Mice, Inbred C57BL , Signal Transduction , Trichuriasis/immunology , Trichuris/parasitology , Interleukin-22
15.
Nat Commun ; 9(1): 4464, 2018 10 26.
Article in English | MEDLINE | ID: mdl-30367055

ABSTRACT

The mosquito gut microbiome plays an important role in mosquito development and fitness, providing a promising avenue for novel mosquito control strategies. Here we present a method for rearing axenic (bacteria free) Aedes aegypti mosquitoes, consisting of feeding sterilized larvae on agar plugs containing a high concentration of liver and yeast extract. This approach allows for the complete development to adulthood while maintaining sterility; however, axenic mosquito's exhibit delayed development time and stunted growth in comparison to their bacterially colonized cohorts. These data challenge the notion that live microorganisms are required for mosquito development, and suggest that the microbiota's main role is nutritional. Furthermore, we colonize axenic mosquitoes with simplified microbial communities ranging from a single bacterial species to a three-member community, demonstrating the ability to control the composition of the microbiota. This axenic system will allow the systematic manipulation of the mosquito microbiome for a deeper understanding of microbiota-host interactions.


Subject(s)
Aedes/growth & development , Aedes/microbiology , Germ-Free Life , Animal Feed/analysis , Animal Feed/microbiology , Animals , Bacteria/genetics , Larva/growth & development , Larva/microbiology , Microbiota/genetics , Mosquito Control , RNA, Ribosomal, 16S/genetics
16.
Sci Rep ; 7(1): 8853, 2017 08 18.
Article in English | MEDLINE | ID: mdl-28821804

ABSTRACT

During active TB in humans a spectrum of pulmonary granulomas with central necrosis and hypoxia exists. BALB/c mice, predominantly used in TB drug development, do not reproduce this complex pathology thereby inaccurately predicting clinical outcome. We found that Nos2 -/- mice incapable of NO-production in immune cells as microbial defence uniformly develop hypoxic necrotizing lung lesions, widely observed in human TB. To study the impact of hypoxic necrosis on the efficacy of antimycobacterials and drug candidates, we subjected Nos2 -/- mice with TB to monotherapy before or after establishment of human-like pathology. Isoniazid induced a drug-tolerant persister population only when necrotic lesions were present. Rifapentine was more potent than rifampin prior to development of human-like pathology and equally potent thereafter, in agreement with recent clinical trials. Pretomanid, delamanid and the pre-clinical candidate BTZ043 were bactericidal independent of pulmonary pathology. Linezolid was bacteriostatic in TB-infected Nos2 -/- mice but significantly improved lung pathology. Hypoxic necrotizing lesions rendered moxifloxacin less active. In conclusion, Nos2 -/- mice are a predictive TB drug development tool owing to their consistent development of human-like pathology.


Subject(s)
Hypoxia/metabolism , Necrosis/genetics , Necrosis/metabolism , Nitric Oxide Synthase Type II/deficiency , Tuberculosis, Pulmonary/etiology , Tuberculosis, Pulmonary/metabolism , Animals , Antitubercular Agents/pharmacology , Disease Models, Animal , Fibrosis , Foam Cells/immunology , Foam Cells/metabolism , Foam Cells/pathology , Humans , Hypoxia/pathology , Isoniazid/pharmacology , Mice , Mice, Knockout , Necrosis/pathology , Rifampin/analogs & derivatives , Rifampin/pharmacology , Treatment Outcome , Tuberculosis, Pulmonary/drug therapy , Tuberculosis, Pulmonary/pathology
17.
mBio ; 7(3)2016 06 21.
Article in English | MEDLINE | ID: mdl-27329747

ABSTRACT

UNLABELLED: Mice harboring a mutation in the gene encoding gastric intrinsic factor (Gif), a protein essential for the absorption of vitamin B12/cobalamin (Cbl), have potential as a model to explore the role of vitamins in infection. The levels of Cbl in the blood of Gif(tm1a/tm1a) mutant mice were influenced by the maternal genotype, with offspring born to heterozygous (high Cbl, F1) mothers exhibiting a significantly higher serum Cbl level than those born to homozygous (low Cbl, F2) equivalents. Low Cbl levels correlated with susceptibility to an infectious challenge with Salmonella enterica serovar Typhimurium or Citrobacter rodentium, and this susceptibility phenotype was moderated by Cbl administration. Transcriptional and metabolic profiling revealed that Cbl deficient mice exhibited a bioenergetic shift similar to a metabolic phenomenon commonly found in cancerous cells under hypoxic conditions known as the Warburg effect, with this metabolic effect being exacerbated further by infection. Our findings demonstrate a role for Cbl in bacterial infection, with potential general relevance to dietary deficiency and infection susceptibility. IMPORTANCE: Malnutrition continues to be a major public health problem in countries with weak infrastructures. In communities with a high prevalence of poor diet, malnourishment and infectious disease can impact vulnerable individuals such as pregnant women and children. Here, we describe a highly flexible murine model for monitoring maternal and environmental influences of vitamin B12 metabolism. We also demonstrate the potential importance of vitamin B12 in controlling susceptibility to bacterial pathogens such as C. rodentium and S Typhimurium. We postulate that this model, along with similarly vitamin deficient mice, could be used to further explore the mechanisms associated with micronutrients and susceptibility to diseases, thereby increasing our understanding of disease in the malnourished.


Subject(s)
Disease Susceptibility , Enterobacteriaceae Infections/immunology , Vitamin B 12 Deficiency/complications , Animals , Citrobacter rodentium/immunology , Disease Models, Animal , Gene Expression Profiling , Metabolome , Mice , Mice, Knockout , Salmonella typhimurium/immunology
18.
J Exp Med ; 213(6): 1061-77, 2016 05 30.
Article in English | MEDLINE | ID: mdl-27217537

ABSTRACT

Enteric fever, caused by Salmonella enterica serovar Typhi, is an important public health problem in resource-limited settings and, despite decades of research, human responses to the infection are poorly understood. In 41 healthy adults experimentally infected with wild-type S. Typhi, we detected significant cytokine responses within 12 h of bacterial ingestion. These early responses did not correlate with subsequent clinical disease outcomes and likely indicate initial host-pathogen interactions in the gut mucosa. In participants developing enteric fever after oral infection, marked transcriptional and cytokine responses during acute disease reflected dominant type I/II interferon signatures, which were significantly associated with bacteremia. Using a murine and macrophage infection model, we validated the pivotal role of this response in the expression of proteins of the host tryptophan metabolism during Salmonella infection. Corresponding alterations in tryptophan catabolites with immunomodulatory properties in serum of participants with typhoid fever confirmed the activity of this pathway, and implicate a central role of host tryptophan metabolism in the pathogenesis of typhoid fever.


Subject(s)
Immunomodulation , Interferon Type I/immunology , Interferon-gamma/immunology , Salmonella typhi/immunology , Tryptophan/immunology , Typhoid Fever/immunology , Adult , Female , Humans , Interferon Type I/blood , Interferon-gamma/blood , Male , Middle Aged , Salmonella typhi/metabolism , Tryptophan/biosynthesis , Typhoid Fever/blood , Typhoid Fever/pathology
19.
Immunity ; 42(1): 108-22, 2015 Jan 20.
Article in English | MEDLINE | ID: mdl-25607461

ABSTRACT

The probabilistic expression of cytokine genes in differentiated T helper (Th) cell populations remains ill defined. By single-cell analyses and mathematical modeling, we show that one stimulation featured stable cytokine nonproducers as well as stable producers with wide cell-to-cell variability in the magnitude of expression. Focusing on interferon-γ (IFN-γ) expression by Th1 cells, mathematical modeling predicted that this behavior reflected different cell-intrinsic capacities and not mere gene-expression noise. In vivo, Th1 cells sort purified by secreted IFN-γ amounts preserved a quantitative memory for both probability and magnitude of IFN-γ re-expression for at least 1 month. Mechanistically, this memory resulted from quantitatively distinct transcription of individual alleles and was controlled by stable expression differences of the Th1 cell lineage-specifying transcription factor T-bet. Functionally, Th1 cells with graded IFN-γ production competence differentially activated Salmonella-infected macrophages for bacterial killing. Thus, individual Th cells commit to produce distinct amounts of a given cytokine, thereby generating functional intrapopulation heterogeneity.


Subject(s)
Interferon-gamma/metabolism , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Macrophages/immunology , Salmonella Infections/immunology , Salmonella typhimurium/immunology , Th1 Cells/immunology , Animals , Cell Differentiation , Cell Lineage , Cells, Cultured , Colony Count, Microbial , Gene Expression Regulation , Immunologic Memory , Interferon-gamma/genetics , Interferon-gamma/immunology , Lymphocyte Activation , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Models, Theoretical , Receptors, Interferon/genetics , Single-Cell Analysis , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Th1 Cells/virology , Viral Load , Interferon gamma Receptor
20.
Proc Natl Acad Sci U S A ; 111(38): E4024-32, 2014 Sep 23.
Article in English | MEDLINE | ID: mdl-25201986

ABSTRACT

Lung granulomas develop upon Mycobacterium tuberculosis (Mtb) infection as a hallmark of human tuberculosis (TB). They are structured aggregates consisting mainly of Mtb-infected and -uninfected macrophages and Mtb-specific T cells. The production of NO by granuloma macrophages expressing nitric oxide synthase-2 (NOS2) via l-arginine and oxygen is a key protective mechanism against mycobacteria. Despite this protection, TB granulomas are often hypoxic, and bacterial killing via NOS2 in these conditions is likely suboptimal. Arginase-1 (Arg1) also metabolizes l-arginine but does not require oxygen as a substrate and has been shown to regulate NOS2 via substrate competition. However, in other infectious diseases in which granulomas occur, such as leishmaniasis and schistosomiasis, Arg1 plays additional roles such as T-cell regulation and tissue repair that are independent of NOS2 suppression. To address whether Arg1 could perform similar functions in hypoxic regions of TB granulomas, we used a TB murine granuloma model in which NOS2 is absent. Abrogation of Arg1 expression in macrophages in this setting resulted in exacerbated lung granuloma pathology and bacterial burden. Arg1 expression in hypoxic granuloma regions correlated with decreased T-cell proliferation, suggesting that Arg1 regulation of T-cell immunity is involved in disease control. Our data argue that Arg1 plays a central role in the control of TB when NOS2 is rendered ineffective by hypoxia.


Subject(s)
Arginase/metabolism , Granuloma/enzymology , Hypoxia/enzymology , Macrophages/enzymology , Mycobacterium tuberculosis , Tuberculosis, Pulmonary/enzymology , Animals , Arginase/genetics , Arginase/immunology , Arginine/genetics , Arginine/immunology , Arginine/metabolism , Cell Proliferation/genetics , Disease Models, Animal , Granuloma/genetics , Granuloma/immunology , Granuloma/pathology , Humans , Hypoxia/genetics , Hypoxia/immunology , Hypoxia/pathology , Lung/enzymology , Lung/immunology , Lung/pathology , Macrophages/immunology , Macrophages/pathology , Mice , Mice, Knockout , Nitric Oxide/genetics , Nitric Oxide/immunology , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/immunology , Nitric Oxide Synthase Type II/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Tuberculosis, Pulmonary/genetics , Tuberculosis, Pulmonary/immunology , Tuberculosis, Pulmonary/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...