Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
J Immunol ; 213(3): 373-383, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38884660

ABSTRACT

Conventionally, immune responses are studied in the context of inflamed tissues and their corresponding draining lymph nodes (LNs). However, little is known about the effects of systemic inflammatory signals generated during local inflammation on distal tissues and nondraining LNs. Using a mouse model of cutaneous immunization, we found that systemic inflammatory stimuli triggered a rapid and selective distal response in the small intestine and the mesenteric LN (mesLN). This consisted of increased permeability of intestinal blood vessels and lymphatic drainage of bloodborne solutes into the mesLN, enhanced activation and migration of intestinal dendritic cells, as well as amplified T cell responses in the mesLNs to systemic but not orally derived Ags. Mechanistically, we found that the small intestine endothelial cells preferentially expressed molecules involved in TNF-α signaling and that TNF-α blockade markedly diminished distal intestinal responses to cutaneous immunization. Together, these findings reveal that the intestinal immune system is rapidly and selectively activated in response to inflammatory cues regardless of their origin, thus identifying an additional layer of defense and enhanced surveillance of a key barrier organ at constant risk of pathogen encounter.


Subject(s)
Immunization , Lymph Nodes , Animals , Mice , Lymph Nodes/immunology , Immunization/methods , Mice, Inbred C57BL , Cytokines/immunology , Cytokines/metabolism , Intestine, Small/immunology , Dendritic Cells/immunology , Inflammation/immunology , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism , T-Lymphocytes/immunology , Intestinal Mucosa/immunology
2.
Am J Respir Crit Care Med ; 195(1): 104-114, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27447987

ABSTRACT

RATIONALE: The potential role of the airway microbiota in dictating immune responses and infection outcomes in HIV-associated pneumonia is largely unknown. OBJECTIVES: To investigate whether microbiologically and immunologically distinct subsets of patients with HIV and pneumonia exist and are related to mortality. METHODS: Bronchoalveolar lavage samples from Ugandan patients with HIV and pneumonia (n = 182) were obtained at study enrollment (following antibiotic treatment); patient demographics including 8- and 70-day mortality were collected. Lower airway bacterial community composition was assessed via amplification and sequencing of the V4 region of the 16S ribosomal RNA gene. Host immune response gene expression profiles were generated by quantitative polymerase chain reaction using RNA extracted from bronchoalveolar lavage fluid. Liquid and gas chromatography mass spectrometry was used to profile serum metabolites. MEASUREMENTS AND MAIN RESULTS: Based on airway microbiome composition, most patients segregated into three distinct groups, each of which were predicted to encode metagenomes capable of producing metabolites characteristically enriched in paired serum samples from these patients. These three groups also exhibited differences in mortality; those with the highest rate had increased ceftriaxone administration and culturable Aspergillus, and demonstrated significantly increased induction of airway T-helper cell type 2 responses. The group with the lowest mortality was characterized by increased expression of T-cell immunoglobulin and mucin domain 3, which down-regulates T-helper cell type 1 proinflammatory responses and is associated with chronic viral infection. CONCLUSIONS: These data provide evidence that compositionally and structurally distinct lower airway microbiomes are associated with discrete local host immune responses, peripheral metabolic reprogramming, and different rates of mortality.


Subject(s)
Coinfection/mortality , HIV Infections/mortality , Lung/microbiology , Microbiota/immunology , Pneumonia, Bacterial/mortality , Bronchoalveolar Lavage Fluid/microbiology , Coinfection/immunology , Coinfection/microbiology , Female , HIV Infections/complications , HIV Infections/immunology , HIV Infections/microbiology , Humans , Male , Microbiota/genetics , Pneumonia, Bacterial/complications , Pneumonia, Bacterial/immunology , RNA, Ribosomal, 16S/genetics , Risk Factors
3.
J Immunol ; 191(4): 1586-93, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23858036

ABSTRACT

CD1 proteins evolved to present diverse lipid Ags to T cells. In comparison with MHC proteins, CD1 proteins exhibit minimal allelic diversity as a result of nonsynonymous single nucleotide polymorphisms (SNPs). However, it is unknown if common SNPs in gene regulatory regions affect CD1 expression and function. We report surprising diversity in patterns of inducible CD1a expression on human dendritic cells (DCs), spanning the full range from undetectable to high density, a finding not seen with other CD1 isoforms. CD1a-deficient DCs failed to present mycobacterial lipopeptide to T cells but had no defects in endocytosis, cytokine secretion, or expression of costimulatory molecules after LPS treatment. We identified an SNP in the 5' untranslated region (rs366316) that was common and strongly associated with low CD1a surface expression and mRNA levels (p = 0.03 and p = 0.001, respectively). Using a CD1a promoter-luciferase system in combination with mutagenesis studies, we found that the polymorphic allele reduced luciferase expression by 44% compared with the wild-type variant (p < 0.001). Genetic regulation of lipid Ag presentation by varying expression on human DCs provides a mechanism for achieving population level differences in immune responses despite limited structural variation in CD1a proteins.


Subject(s)
5' Untranslated Regions/genetics , Antigens, CD1/genetics , Dendritic Cells/immunology , Lipopeptides/immunology , Polymorphism, Single Nucleotide , Antigens, CD1/biosynthesis , Antigens, CD1/immunology , Cells, Cultured , Cytokines/metabolism , Endocytosis , Escherichia coli/immunology , Female , Genes, Reporter , Genetic Variation , Humans , Lipopolysaccharides/therapeutic use , Male , Mutagenesis, Site-Directed , Mycobacterium tuberculosis/immunology , Oxazoles/immunology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , T-Lymphocyte Subsets/immunology
4.
Cancer Res Commun ; 3(9): 1756-1769, 2023 09.
Article in English | MEDLINE | ID: mdl-37674528

ABSTRACT

Mechanisms for Helicobacter pylori (Hp)-driven stomach cancer are not fully understood. In a transgenic mouse model of gastric preneoplasia, concomitant Hp infection and induction of constitutively active KRAS (Hp+KRAS+) alters metaplasia phenotypes and elicits greater inflammation than either perturbation alone. Gastric single-cell RNA sequencing showed that Hp+KRAS+ mice had a large population of metaplastic pit cells that expressed the intestinal mucin Muc4 and the growth factor amphiregulin. Flow cytometry and IHC-based immune profiling revealed that metaplastic pit cells were associated with macrophage and T-cell inflammation. Accordingly, expansion of metaplastic pit cells was prevented by gastric immunosuppression and reversed by antibiotic eradication of Hp. Finally, MUC4 expression was significantly associated with proliferation in human gastric cancer samples. These studies identify an Hp-associated metaplastic pit cell lineage, also found in human gastric cancer tissues, whose expansion is driven by Hp-dependent inflammation. Significance: Using a mouse model, we have delineated metaplastic pit cells as a precancerous cell type whose expansion requires Hp-driven inflammation. In humans, metaplastic pit cells show enhanced proliferation as well as enrichment in precancer and early cancer tissues, highlighting an early step in the gastric metaplasia to cancer cascade.


Subject(s)
Helicobacter pylori , Stomach Neoplasms , Humans , Animals , Mice , Proto-Oncogene Proteins p21(ras) , Disease Models, Animal , Inflammation
6.
Lancet Microbe ; 3(5): e357-e365, 2022 05.
Article in English | MEDLINE | ID: mdl-35544096

ABSTRACT

BACKGROUND: Pneumonia is a leading cause of death worldwide and is a major health-care challenge in people living with HIV. Despite this, the causes of pneumonia in this population remain poorly understood. We aimed to assess the feasibility of metatranscriptomics for epidemiological surveillance of pneumonia in patients with HIV in Uganda. METHODS: We performed a retrospective observational study in patients with HIV who were admitted to Mulago Hospital, Kampala, Uganda between Oct 1, 2009, and Dec 31, 2011. Inclusion criteria were age 18 years or older, HIV-positivity, and clinically diagnosed pneumonia. Exclusion criteria were contraindication to bronchoscopy or an existing diagnosis of tuberculosis. Bronchoalveolar lavage fluid was collected within 72 h of admission and a combination of RNA sequencing and Mycobacterium tuberculosis culture plus PCR were performed. The primary outcome was detection of an established or possible respiratory pathogen in the total study population. FINDINGS: We consecutively enrolled 217 patients during the study period. A potential microbial cause for pneumonia was identified in 211 (97%) patients. At least one microorganism of established respiratory pathogenicity was identified in 113 (52%) patients, and a microbe of possible pathogenicity was identified in an additional 98 (45%). M tuberculosis was the most commonly identified established pathogen (35 [16%] patients; in whom bacterial or viral co-infections were identified in 13 [37%]). Streptococcus mitis, although not previously reported as a cause of pneumonia in patients with HIV, was the most commonly identified bacterial organism (37 [17%] patients). Haemophilus influenzae was the most commonly identified established bacterial pathogen (20 [9%] patients). Pneumocystis jirovecii was only identified in patients with a CD4 count of less than 200 cells per mL. INTERPRETATION: We show the feasibility of using metatranscriptomics for epidemiologic surveillance of pneumonia by describing the spectrum of respiratory pathogens in adults with HIV in Uganda. Applying these methods to a contemporary cohort could enable broad assessment of changes in pneumonia aetiology following the emergence of SARS-CoV-2. FUNDING: US National Institutes of Health, Chan Zuckerberg Biohub.


Subject(s)
COVID-19 , HIV Infections , Pneumonia , Adolescent , Adult , Cross-Sectional Studies , HIV Infections/complications , Humans , Pneumonia/epidemiology , SARS-CoV-2 , Uganda/epidemiology , United States
7.
Cell Host Microbe ; 27(6): 849-851, 2020 06 10.
Article in English | MEDLINE | ID: mdl-32526178

ABSTRACT

Childhood undernutrition is associated with dysbiosis and dampened vaccine responses. Understanding how nutrients influence the microbiota and immunity is critical for vaccine efficacy. In this issue of Cell Host & Microbe, Di Luccia et al. and Huus et al. reveal that nutrition affects IgA responses to the microbiota and oral vaccines.


Subject(s)
Immunity, Mucosal , Microbiota , Bacteria , Child , Dysbiosis , Humans , Immunoglobulin A
8.
Sci Rep ; 10(1): 16186, 2020 09 30.
Article in English | MEDLINE | ID: mdl-32999331

ABSTRACT

Chronic lung disease (CLD) is a common co-morbidity for HIV-positive children and adolescents on antiretroviral therapy (ART) in sub-Saharan Africa. In this population, distinct airway microbiota may differentially confer risk of CLD. In a cross-sectional study of 202 HIV-infected children aged 6-16 years in Harare, Zimbabwe, we determined the association of sputum microbiota composition (using 16S ribosomal RNA V4 gene region sequencing) with CLD defined using clinical, spirometric, or radiographic criteria. Forty-two percent of children were determined to have CLD according to our definition. Dirichlet multinomial mixtures identified four compositionally distinct sputum microbiota structures. Patients whose sputum microbiota was dominated by Haemophilus, Moraxella or Neisseria (HMN) were at 1.5 times higher risk of CLD than those with Streptococcus or Prevotella (SP)-dominated microbiota (RR = 1.48, p = 0.035). Cell-free products of HMN sputum microbiota induced features of epithelial disruption and inflammatory gene expression in vitro, indicating enhanced pathogenic potential of these CLD-associated microbiota. Thus, HIV-positive children harbor distinct sputum microbiota, with those dominated by Haemophilus, Moraxella or Neisseria associated with enhanced pathogenesis in vitro and clinical CLD.


Subject(s)
HIV Infections/complications , Lung Diseases/microbiology , Lung/microbiology , Microbiota , Sputum/microbiology , Adolescent , Child , Cross-Sectional Studies , Female , HIV Infections/microbiology , Humans , Lung/virology , Lung Diseases/virology , Male , Zimbabwe
9.
Microbiome ; 7(1): 37, 2019 03 11.
Article in English | MEDLINE | ID: mdl-30857553

ABSTRACT

Pneumonia is common and frequently fatal in HIV-infected patients, due to rampant, systemic inflammation and failure to control microbial infection. While airway microbiota composition is related to local inflammatory response, gut microbiota has been shown to correlate with the degree of peripheral immune activation (IL6 and IP10 expression) in HIV-infected patients. We thus hypothesized that both airway and gut microbiota are perturbed in HIV-infected pneumonia patients, that the gut microbiota is related to peripheral CD4+ cell counts, and that its associated products differentially program immune cell populations necessary for controlling microbial infection in CD4-high and CD4-low patients. To assess these relationships, paired bronchoalveolar lavage and stool microbiota (bacterial and fungal) from a large cohort of Ugandan, HIV-infected patients with pneumonia were examined, and in vitro tests of the effect of gut microbiome products on macrophage effector phenotypes performed. While lower airway microbiota stratified into three compositionally distinct microbiota as previously described, these were not related to peripheral CD4 cell count. In contrast, variation in gut microbiota composition significantly related to CD4 cell count, lung microbiota composition, and patient mortality. Compared with patients with high CD4+ cell counts, those with low counts possessed more compositionally similar airway and gut microbiota, evidence of microbial translocation, and their associated gut microbiome products reduced macrophage activation and IL-10 expression and increased IL-1ß expression in vitro. These findings suggest that the gut microbiome is related to CD4 status and plays a key role in modulating macrophage function, critical to microbial control in HIV-infected patients with pneumonia.


Subject(s)
Bacteria/classification , HIV Infections/microbiology , Lung/microbiology , Macrophages/immunology , Microbiota , Pneumonia/immunology , Bacteria/genetics , Bacteria/isolation & purification , Bronchoalveolar Lavage Fluid/microbiology , CD4 Lymphocyte Count , Chemokine CXCL10/metabolism , Cohort Studies , DNA, Bacterial/genetics , DNA, Ribosomal , Feces/microbiology , HIV Infections/immunology , Humans , Interleukin-6/metabolism , Lung/immunology , Pneumonia/microbiology , Pneumonia/therapy , RNA, Ribosomal, 16S/genetics , Sequence Analysis, DNA/methods , Uganda
10.
Curr Opin HIV AIDS ; 13(1): 45-52, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29035946

ABSTRACT

PURPOSE OF REVIEW: The purpose of this review is to summarize recent findings on the lung microbiome in HIV-infected patients and associated pulmonary diseases, and the relationship of airway microbial communities to metabolic and immune signatures within this patient population. RECENT FINDINGS: The lung microbiome in HIV infection is a relatively new and rapidly developing field; early studies in the field produced inconclusive evidence as to whether HIV-infection changes the lower airway microbiome. More recent microbiome investigations have addressed these inconsistencies by incorporating systems biology approaches and laboratory models. Several investigations have now identified enrichment of Prevotella, Veillonella, and Streptococcus in the lower airways as consistent correlates of advanced HIV-infection and HIV-associated pulmonary diseases. These bacteria are associated with specific metabolic and immune profiles within the lung and circulation, providing the first indication that the lung microbiome may play a functional role in the pathogenesis of HIV-infection and HIV-associated pulmonary disease. SUMMARY: This review summarizes knowledge to date on the lung microbiome in HIV infection, as well as challenges and accomplishments in the field within the last 2 years. Although the lung microbiome in HIV infection is still an emerging field, recent studies have formed a framework for future functional analysis of microbes in HIV pathogenesis.


Subject(s)
Dysbiosis , HIV Infections/complications , HIV Infections/physiopathology , Host-Pathogen Interactions , Lung/microbiology , Microbiota , Bacteria/classification , Bacteria/isolation & purification , Bacterial Load , Biomarkers/blood , Blood Chemical Analysis , Humans
11.
Cell Rep ; 14(11): 2611-23, 2016 Mar 22.
Article in English | MEDLINE | ID: mdl-26971994

ABSTRACT

Diets rich in saturated fatty acids (SFAs) produce a form of tissue inflammation driven by "metabolically activated" macrophages. We show that SFAs, when in excess, induce a unique transcriptional signature in both mouse and human macrophages that is enriched by a subset of ER stress markers, particularly IRE1α and many adaptive downstream target genes. SFAs also activate the NLRP3 inflammasome in macrophages, resulting in IL-1ß secretion. We found that IRE1α mediates SFA-induced IL-1ß secretion by macrophages and that its activation by SFAs does not rely on unfolded protein sensing. We show instead that the ability of SFAs to stimulate either IRE1α activation or IL-1ß secretion can be specifically reduced by preventing their flux into phosphatidylcholine (PC) or by increasing unsaturated PC levels. Thus, IRE1α is an unrecognized intracellular PC sensor critical to the process by which SFAs stimulate macrophages to secrete IL-1ß, a driver of diet-induced tissue inflammation.


Subject(s)
Endoribonucleases/metabolism , Fatty Acids/pharmacology , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Bone Marrow Cells/cytology , Cells, Cultured , Diet , Endoplasmic Reticulum Stress/drug effects , Humans , Interleukin-1beta/metabolism , Lipopolysaccharides/toxicity , Macrophages/cytology , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Phosphatidylcholines/metabolism , Principal Component Analysis , Real-Time Polymerase Chain Reaction , Signal Transduction/drug effects , X-Box Binding Protein 1/genetics , X-Box Binding Protein 1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL