Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 72
1.
Cell Rep ; 37(5): 109916, 2021 11 02.
Article En | MEDLINE | ID: mdl-34731608

Intestinal epithelial cells (IECs) have long been understood to express high levels of major histocompatibility complex class II (MHC class II) molecules but are not considered canonical antigen-presenting cells, and the impact of IEC-MHC class II signaling on gut homeostasis remains enigmatic. As IECs serve as the primary barrier between underlying host immune cells, we reasoned that IEC-intrinsic antigen presentation may play a role in responses toward the microbiota. Mice with an IEC-intrinsic deletion of MHC class II (IECΔMHC class II) are healthy but have fewer microbial-bound IgA, regulatory T cells (Tregs), and immune repertoire selection. This was associated with increased interindividual microbiota variation and altered proportions of two taxa in the ileum where MHC class II on IECs is highest. Intestinal mononuclear phagocytes (MNPs) have similar MHC class II transcription but less surface MHC class II and are capable of acquiring MHC class II from IECs. Thus, epithelial-myeloid interactions mediate development of adaptive responses to microbial antigens within the gastrointestinal tract.


Adaptive Immunity , Bacteria/immunology , Epithelial Cells/immunology , Gastrointestinal Microbiome , Histocompatibility Antigens Class II/immunology , Ileum/microbiology , Immunity, Mucosal , Mononuclear Phagocyte System/immunology , Myeloid Cells/immunology , Animals , Antigens, Bacterial/immunology , Antigens, Bacterial/metabolism , Bacteria/growth & development , Bacteria/metabolism , Cell Line , Colitis/immunology , Colitis/metabolism , Colitis/microbiology , Disease Models, Animal , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Female , Histocompatibility Antigens Class II/metabolism , Host-Pathogen Interactions , Ileum/immunology , Ileum/metabolism , Immunoglobulin A/immunology , Immunoglobulin A/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Mononuclear Phagocyte System/metabolism , Mononuclear Phagocyte System/microbiology , Myeloid Cells/metabolism , Myeloid Cells/microbiology , Signal Transduction , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism
2.
EMBO J ; 40(23): e108605, 2021 12 01.
Article En | MEDLINE | ID: mdl-34622466

The immune cells of the central nervous system (CNS) comprise parenchymal microglia and at the CNS border regions meningeal, perivascular, and choroid plexus macrophages (collectively called CNS-associated macrophages, CAMs). While previous work has shown that microglial properties depend on environmental signals from the commensal microbiota, the effects of microbiota on CAMs are unknown. By combining several microbiota manipulation approaches, genetic mouse models, and single-cell RNA-sequencing, we have characterized CNS myeloid cell composition and function. Under steady-state conditions, the transcriptional profiles and numbers of choroid plexus macrophages were found to be tightly regulated by complex microbiota. In contrast, perivascular and meningeal macrophages were affected to a lesser extent. An acute perturbation through viral infection evoked an attenuated immune response of all CAMs in germ-free mice. We further assessed CAMs in a more chronic pathological state in 5xFAD mice, a model for Alzheimer's disease, and found enhanced amyloid beta uptake exclusively by perivascular macrophages in germ-free 5xFAD mice. Our results aid the understanding of distinct microbiota-CNS macrophage interactions during homeostasis and disease, which could potentially be targeted therapeutically.


Alzheimer Disease/immunology , Bacteria/growth & development , Central Nervous System/immunology , Homeostasis , Macrophages/immunology , Myeloid Cells/immunology , Alzheimer Disease/genetics , Alzheimer Disease/microbiology , Alzheimer Disease/pathology , Animals , Bacteria/classification , Bacteria/metabolism , Central Nervous System/metabolism , Central Nervous System/microbiology , Central Nervous System/pathology , Female , Macrophages/metabolism , Macrophages/microbiology , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Microbiota , Myeloid Cells/metabolism , Myeloid Cells/microbiology , Myeloid Cells/pathology , Transcriptome
3.
Nat Immunol ; 22(11): 1382-1390, 2021 11.
Article En | MEDLINE | ID: mdl-34663978

Intergenerational inheritance of immune traits linked to epigenetic modifications has been demonstrated in plants and invertebrates. Here we provide evidence for transmission of trained immunity across generations to murine progeny that survived a sublethal systemic infection with Candida albicans or a zymosan challenge. The progeny of trained mice exhibited cellular, developmental, transcriptional and epigenetic changes associated with the bone marrow-resident myeloid effector and progenitor cell compartment. Moreover, the progeny of trained mice showed enhanced responsiveness to endotoxin challenge, alongside improved protection against systemic heterologous Escherichia coli and Listeria monocytogenes infections. Sperm DNA of parental male mice intravenously infected with the fungus C. albicans showed DNA methylation differences linked to immune gene loci. These results provide evidence for inheritance of trained immunity in mammals, enhancing protection against infections.


Candida albicans/immunology , Candidiasis/immunology , Escherichia coli Infections/immunology , Escherichia coli/immunology , Heredity , Immunity, Innate/genetics , Listeria monocytogenes/immunology , Listeriosis/immunology , Myeloid Cells/immunology , Animals , Candida albicans/pathogenicity , Candidiasis/genetics , Candidiasis/metabolism , Candidiasis/microbiology , Cells, Cultured , DNA Methylation , Disease Models, Animal , Epigenesis, Genetic , Escherichia coli/pathogenicity , Escherichia coli Infections/genetics , Escherichia coli Infections/metabolism , Escherichia coli Infections/microbiology , Host-Pathogen Interactions , Listeria monocytogenes/pathogenicity , Listeriosis/genetics , Listeriosis/metabolism , Listeriosis/microbiology , Male , Mice, Transgenic , Myeloid Cells/metabolism , Myeloid Cells/microbiology , Spermatozoa/immunology , Spermatozoa/metabolism , Transcription, Genetic
4.
Nat Commun ; 12(1): 5454, 2021 09 15.
Article En | MEDLINE | ID: mdl-34526512

Chlamydia trachomatis infection causes severe inflammatory disease resulting in blindness and infertility. The pathophysiology of these diseases remains elusive but myeloid cell-associated inflammation has been implicated. Here we show NLRP3 inflammasome activation is essential for driving a macrophage-associated endometritis resulting in infertility by using a female mouse genital tract chlamydial infection model. We find the chlamydial parasitophorous vacuole protein CT135 triggers NLRP3 inflammasome activation via TLR2/MyD88 signaling as a pathogenic strategy to evade neutrophil host defense. Paradoxically, a consequence of CT135 mediated neutrophil killing results in a submucosal macrophage-associated endometritis driven by ATP/P2X7R induced NLRP3 inflammasome activation. Importantly, macrophage-associated immunopathology occurs independent of macrophage infection. We show chlamydial infection of neutrophils and epithelial cells produce elevated levels of extracellular ATP. We propose this source of ATP serves as a DAMP to activate submucosal macrophage NLRP3 inflammasome that drive damaging immunopathology. These findings offer a paradigm of sterile inflammation in infectious disease pathogenesis.


Chlamydia Infections/immunology , Chlamydia/immunology , Inflammation/immunology , Myeloid Cells/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Neutrophils/immunology , Receptors, Purinergic P2X7/immunology , Adenosine Triphosphate/immunology , Adenosine Triphosphate/metabolism , Animals , Cells, Cultured , Chlamydia/physiology , Chlamydia Infections/metabolism , Chlamydia Infections/microbiology , Disease Models, Animal , Female , HeLa Cells , Host-Pathogen Interactions/immunology , Humans , Immune Evasion/immunology , Inflammation/metabolism , Inflammation/microbiology , Macrophages/immunology , Macrophages/metabolism , Macrophages/microbiology , Mice, Inbred C57BL , Mice, Knockout , Myeloid Cells/metabolism , Myeloid Cells/microbiology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Neutrophils/metabolism , Neutrophils/microbiology , Receptors, Purinergic P2X7/genetics , Receptors, Purinergic P2X7/metabolism
5.
Methods Mol Biol ; 2314: 167-182, 2021.
Article En | MEDLINE | ID: mdl-34235652

Mycobacterium tuberculosis colonizes, survives, and grows inside macrophages. In vitro macrophage infection models, using both primary macrophages and cell lines, enable the characterization of the pathogen response to macrophage immune pressure and intracellular environmental cues. We describe methods to propagate and infect primary murine bone marrow-derived macrophages, HoxB8 conditionally immortalized myeloid cells, Max Planck Institute alveolar macrophage-like cells, and J774 and THP-1 macrophage-like cell lines. We also present methods on the characterization of M. tuberculosis intracellular survival and the preparation of infected macrophages for imaging.


Macrophages, Alveolar/microbiology , Macrophages/microbiology , Molecular Imaging/methods , Mycobacterium tuberculosis/growth & development , Myeloid Cells/microbiology , Animals , Cells, Cultured , Humans , In Vitro Techniques , Macrophages/pathology , Macrophages, Alveolar/pathology , Mice , Mycobacterium tuberculosis/pathogenicity , Myeloid Cells/pathology
6.
Infect Immun ; 89(10): e0012621, 2021 09 16.
Article En | MEDLINE | ID: mdl-34097504

Whooping cough (pertussis) is a severe pulmonary infectious disease caused by the bacteria Bordetella pertussis. Pertussis infects an estimated 24 million people annually, resulting in >150,000 deaths. The NIH placed pertussis on the list of emerging pathogens in 2015. Antibiotics are ineffective unless administered before the onset of the disease characteristic cough. Therefore, there is an urgent need for novel pertussis therapeutics. We have shown that sphingosine-1-phosphate receptor (S1PR) agonists reduce pertussis inflammation without increasing bacterial burden. Transcriptomic studies were performed to identify this mechanism and allow for the development of pertussis therapeutics that specifically target problematic inflammation without sacrificing bacterial control. These data suggested a role for triggering receptor expressed on myeloid cells-1 (TREM-1). TREM-1 cell surface receptor functions as an amplifier of inflammatory responses. Expression of TREM-1 is increased in response to bacterial infection of mucosal surfaces. In mice, B. pertussis infection results in Toll-like receptor 9 (TLR9)-dependent increased expression of TREM-1 and its associated cytokines. Interestingly, S1PR agonists dampen pulmonary inflammation and TREM-1 expression. Mice challenged intranasally with B. pertussis and treated with ligand-dependent (LP17) and ligand-independent (GF9) TREM-1 inhibitors showed no differences in bacterial burden and significantly reduced tumor necrosis factor-α (TNF-α) and C-C motif chemokine ligand 2 (CCL-2) expression compared to controls. Mice receiving TREM-1 inhibitors showed reduced pulmonary inflammation compared to controls, indicating that TREM-1 promotes inflammatory pathology, but not bacterial control, during pertussis infection. This implicates TREM-1 as a potential therapeutic target for the treatment of pertussis.


Bordetella pertussis/immunology , Inflammation/immunology , Inflammation/metabolism , Triggering Receptor Expressed on Myeloid Cells-1/metabolism , Animals , Disease Models, Animal , Inflammation/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Cells/metabolism , Myeloid Cells/microbiology , Tumor Necrosis Factor-alpha/metabolism , Whooping Cough/immunology , Whooping Cough/metabolism , Whooping Cough/microbiology
7.
Cancer Res ; 81(10): 2745-2759, 2021 05 15.
Article En | MEDLINE | ID: mdl-34003774

Porphyromonas gingivalis (P. gingivalis) is a keystone periodontal pathogen associated with various digestive cancers. However, whether P. gingivalis can promote colorectal cancer and the underlying mechanism associated with such promotion remains unclear. In this study, we found that P. gingivalis was enriched in human feces and tissue samples from patients with colorectal cancer compared with those from patients with colorectal adenoma or healthy subjects. Cohort studies demonstrated that P. gingivalis infection was associated with poor prognosis in colorectal cancer. P. gingivalis increased tumor counts and tumor volume in the ApcMin/+ mouse model and increased tumor growth in orthotopic rectal and subcutaneous carcinoma models. Furthermore, orthotopic tumors from mice exposed to P. gingivalis exhibited tumor-infiltrating myeloid cell recruitment and a proinflammatory signature. P. gingivalis promoted colorectal cancer via NLRP3 inflammasome activation in vitro and in vivo. NLRP3 chimeric mice harboring orthotopic tumors showed that the effect of NLRP3 on P. gingivalis pathogenesis was mediated by hematopoietic sources. Collectively, these data suggest that P. gingivalis contributes to colorectal cancer neoplasia progression by activating the hematopoietic NLRP3 inflammasome. SIGNIFICANCE: This study demonstrates that the periodontal pathogen P. gingivalis can promote colorectal tumorigenesis by recruiting myeloid cells and creating a proinflammatory tumor microenvironment. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/81/10/2745/F1.large.jpg.


Carcinogenesis/pathology , Colorectal Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Inflammasomes/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/physiology , Neoplastic Stem Cells/pathology , Porphyromonas gingivalis/pathogenicity , Animals , Apoptosis , Bacteroidaceae Infections/complications , Bacteroidaceae Infections/immunology , Bacteroidaceae Infections/microbiology , Bacteroidaceae Infections/pathology , Carcinogenesis/immunology , Carcinogenesis/metabolism , Cell Proliferation , Colorectal Neoplasms/immunology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/microbiology , Humans , Inflammasomes/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , Myeloid Cells/immunology , Myeloid Cells/metabolism , Myeloid Cells/microbiology , Myeloid Cells/pathology , Neoplastic Stem Cells/immunology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/microbiology , Prognosis , Survival Rate , Tumor Cells, Cultured , Tumor Microenvironment/immunology , Xenograft Model Antitumor Assays
8.
Cell Mol Life Sci ; 78(5): 1887-1907, 2021 Mar.
Article En | MEDLINE | ID: mdl-33125509

Various factors of the tissue microenvironment such as the oxygen concentration influence the host-pathogen interaction. During the past decade, hypoxia-driven signaling via hypoxia-inducible factors (HIF) has emerged as an important factor that affects both the pathogen and the host. In this chapter, we will review the current knowledge of this complex interplay, with a particular emphasis given to the impact of hypoxia and HIF on the inflammatory and antimicrobial activity of myeloid cells, the bacterial responses to hypoxia and the containment of bacterial infections under oxygen-limited conditions. We will also summarize how low oxygen concentrations influence the metabolism of neutrophils, macrophages and dendritic cells. Finally, we will discuss the consequences of hypoxia and HIFα activation for the invading pathogen, with a focus on Pseudomonas aeruginosa, Mycobacterium tuberculosis, Coxiella burnetii, Salmonella enterica and Staphylococcus aureus. This includes a description of the mechanisms and microbial factors, which the pathogens use to sense and react to hypoxic conditions.


Mycobacterium tuberculosis/physiology , Myeloid Cells/microbiology , Pseudomonas aeruginosa/physiology , Staphylococcus aureus/physiology , Animals , Cell Hypoxia , Host-Pathogen Interactions , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Myeloid Cells/metabolism , Oxygen/metabolism
9.
Med Sci Monit ; 26: e921886, 2020 Aug 01.
Article En | MEDLINE | ID: mdl-32737964

BACKGROUND An increasing number of studies have demonstrated that Streptococcus bovis and its concomitant inflammatory factors concentrate in the intestine in colorectal cancer (CRC). However, the molecular mechanism of S. bovis on colorectal tumorigenesis remains unclear. This study aimed to explore the role of S. bovis in carcinogenesis and its potential mechanism in CRC of mice orally pretreated with S. bovis. MATERIAL AND METHODS The colons of experimental mice were collected and evaluated for the extent of neoplasm. In addition, comparative feces DNA sequencing was adopted to verify the abundance change of S. bovis during the progression of CRC in patients. RESULTS The results of this study found that S. bovis is more likely to be present at higher levels in patients with progressive colorectal carcinoma compared to those adenoma patients and healthy volunteers (P<0.05). Pretreatment with S. bovis aggravated tumor formation in mice, resulting in more substantial and a higher number of tumor nodes (P<0.05). A cytokine expression pattern with increased levels of IL-6, Scyb1, Ptgs2, IL-1ß, TNF, and Ccl2 was detected in S. bovis pretreated CRC mice (all P<0.05). Furthermore, S. bovis recruited myeloid cells, especially CD11b⁺TLR-4⁺ cells, which could promote pro-tumor immunity in the tumor microenvironment (P<0.05). CONCLUSIONS Collectively, our study indicates that S. bovis may induce a suppressive immunity that is conducive to CRC by recruiting tumor-infiltrating CD11b⁺TLR-4⁺ cells. In conclusion, S. bovis contributes to colorectal tumorigenesis via recruiting CD11b⁺TLR-4⁺ cells.


Adenoma/microbiology , Carcinogenesis/immunology , Colonic Neoplasms/microbiology , Colorectal Neoplasms/microbiology , Gene Expression Regulation, Neoplastic , Streptococcus bovis/pathogenicity , Adenoma/genetics , Adenoma/immunology , Adenoma/pathology , Aged , Animals , Bacterial Load , CD11b Antigen/genetics , CD11b Antigen/immunology , Carcinogenesis/genetics , Carcinogenesis/pathology , Case-Control Studies , Chemokine CCL2/genetics , Chemokine CCL2/immunology , Chemokine CXCL1/genetics , Chemokine CXCL1/immunology , Colonic Neoplasms/genetics , Colonic Neoplasms/immunology , Colonic Neoplasms/pathology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/immunology , Colorectal Neoplasms/pathology , Cyclooxygenase 2/genetics , Cyclooxygenase 2/immunology , Feces/microbiology , Female , Humans , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Interleukin-6/genetics , Interleukin-6/immunology , Lymphatic Metastasis , Male , Mice , Mice, Inbred C57BL , Middle Aged , Myeloid Cells/immunology , Myeloid Cells/microbiology , Streptococcus bovis/growth & development , Streptococcus bovis/immunology , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/immunology , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
10.
Article En | MEDLINE | ID: mdl-32391286

Monocytes play a critical role during infection with Mycobacterium tuberculosis (Mtb). They are recruited to the lung, where they participate in the control of infection during active tuberculosis (TB). Alternatively, inflammatory monocytes may participate in inflammation or serve as niches for Mtb infection. Monocytes response to infection may vary depending on the particularities of the clinical isolate of Mtb from which they are infected. In this pilot study, we have examined the baseline mRNA profiles of circulating human monocytes from patients with active TB (MoTB) compared with monocytes from healthy individuals (MoCT). Circulating MoTB displayed a pro-inflammatory transcriptome characterized by increased gene expression of genes associated with cytokines, monocytopoiesis, and down-regulation of MHC class II gene expression. In response to in vitro infection with two clinical isolates of the LAM family of Mtb (UT127 and UT205), MoTB displayed an attenuated inflammatory mRNA profile associated with down-regulation the TREM1 signaling pathway. Furthermore, the gene expression signature induced by Mtb UT205 clinical strain was characterized by the enrichment of genes in pathways and biological processes mainly associated with a signature of IFN-inducible genes and the inhibition of cell death mechanisms compared to MoTB-127, which could favor the establishment and survival of Mtb within the monocytes. These results suggest that circulating MoTB have an altered transcriptome that upon infection with Mtb may help to maintain chronic inflammation and infection. Moreover, this functional abnormality of monocytes may also depend on potential differences in virulence of circulating clinical strains of Mtb.


Monocytes/microbiology , Mycobacterium tuberculosis , Myeloid Cells/microbiology , Tuberculosis , Cells, Cultured , Cytokines , Humans , Pilot Projects
11.
Sci Rep ; 10(1): 7340, 2020 04 30.
Article En | MEDLINE | ID: mdl-32355205

Microbiota derived metabolites act as chemical messengers that elicit a profound impact on host physiology. Vitamin D receptor (VDR) is a key genetic factor for shaping the host microbiome. However, it remains unclear how microbial metabolites are altered in the absence of VDR. We investigated metabolites from mice with tissue-specific deletion of VDR in intestinal epithelial cells or myeloid cells. Conditional VDR deletion severely changed metabolites specifically produced from carbohydrate, protein, lipid, and bile acid metabolism. Eighty-four out of 765 biochemicals were significantly altered due to the Vdr status, and 530 significant changes were due to the high-fat diet intervention. The impact of diet was more prominent due to loss of VDR as indicated by the differences in metabolites generated from energy expenditure, tri-carboxylic acid cycle, tocopherol, polyamine metabolism, and bile acids. The effect of HFD was more pronounced in female mice after VDR deletion. Interestingly, the expression levels of farnesoid X receptor in liver and intestine were significantly increased after intestinal epithelial VDR deletion and were further increased by the high-fat diet. Our study highlights the gender differences, tissue specificity, and potential gut-liver-microbiome axis mediated by VDR that might trigger downstream metabolic disorders.


Gastrointestinal Microbiome , Myeloid Cells/microbiology , Receptors, Calcitriol/genetics , Animals , Bile Acids and Salts/metabolism , Carbohydrate Metabolism , Diet, High-Fat , Energy Metabolism , Female , Gene Deletion , Intestines , Lipid Metabolism/genetics , Liver/metabolism , Male , Metabolic Syndrome/genetics , Mice , Mice, Knockout , Polyamines/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Sex Factors , Signal Transduction , Tocopherols/metabolism
12.
Immunology ; 159(1): 121-129, 2020 01.
Article En | MEDLINE | ID: mdl-31606895

The transcription factor hypoxia-inducible factor-1 alpha (HIF-1α) is a key regulator of the response and function of myeloid cells in hypoxic and inflammatory microenvironments. To define the role of HIF-1α in tuberculosis, the progression of aerosol Mycobacterium tuberculosis infection was analysed in mice deficient in HIF-1α in the myeloid lineage (mHIF-1α-/- ). We show that myeloid HIF-1α is not required for the containment of the infection, as both wild-type (WT) and mHIF-1α-/- mice mounted normal Th1 responses and maintained control of bacterial growth throughout infection. However, during chronic infection mHIF-1α-/- mice developed extensive lymphocytic inflammatory involvement of the interstitial lung tissue and died earlier than WT mice. These data support the hypothesis that HIF-1α activity coordinates the response of myeloid cells during M. tuberculosis infection to prevent excessive leucocyte recruitment and immunopathological consequences to the host.


Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lung/metabolism , Mycobacterium tuberculosis/growth & development , Myeloid Cells/metabolism , Pneumonia/metabolism , Tuberculosis, Pulmonary/metabolism , Animals , Bacterial Load , Cells, Cultured , Disease Models, Animal , Disease Progression , Host-Pathogen Interactions , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Lung/immunology , Lung/microbiology , Mice, Inbred C57BL , Mice, Knockout , Mycobacterium tuberculosis/immunology , Myeloid Cells/immunology , Myeloid Cells/microbiology , Pneumonia/genetics , Pneumonia/immunology , Pneumonia/microbiology , Signal Transduction , Tuberculosis, Pulmonary/genetics , Tuberculosis, Pulmonary/immunology , Tuberculosis, Pulmonary/microbiology
13.
Helicobacter ; 24 Suppl 1: e12644, 2019 Sep.
Article En | MEDLINE | ID: mdl-31486236

Chronic inflammation induced by Helicobacter pylori infection is a critical factor in the development of peptic ulcer disease and gastric cancer. Central to this inflammation is the initiation of pro-inflammatory signaling cascades within epithelial cells, in particular those mediated by two sensors of bacterial cell wall components, nucleotide-binding oligomerization domain-containing protein 1 (NOD1) and alpha-protein kinase 1 (ALPK1). H pylori is, however, also highly adept at mitigating inflammation in the host, thereby restricting tissue damage and favoring bacterial persistence. H pylori modulates host immune responses by altering cytokine signaling in epithelial and myeloid cells, which results in increased proliferation of regulatory T cells and downregulation of effector T-cell responses. H pylori vacuolating cytotoxin A (VacA) has been shown to play an important role in the dampening of immune responses and induction of immune tolerance capable of protecting against asthma. It is also possible to generate protective immune responses by immunization with various H pylori antigens or their epitopes, in combination with an adjuvant, though this for now has only been shown in mouse models. Novel non-toxic adjuvants, consisting of modified bacterial enterotoxins or nanoparticles, have recently been developed that may not only enhance vaccine efficacy, but also help translate candidate vaccines to the clinic. This review will summarize the main discoveries in the past year regarding host immune responses to H pylori infection, as well as the design of new vaccine approaches against this infection.


Bacterial Vaccines/immunology , Helicobacter Infections/immunology , Helicobacter Infections/pathology , Helicobacter pylori/immunology , Helicobacter pylori/pathogenicity , Host-Pathogen Interactions , Bacterial Vaccines/isolation & purification , Biomedical Research/trends , Epithelial Cells/immunology , Epithelial Cells/microbiology , Helicobacter Infections/prevention & control , Humans , Immune Tolerance , Inflammation/immunology , Inflammation/pathology , Inflammation/prevention & control , Myeloid Cells/immunology , Myeloid Cells/microbiology , T-Lymphocytes, Regulatory/immunology
14.
J Immunol ; 203(6): 1480-1492, 2019 09 15.
Article En | MEDLINE | ID: mdl-31413108

Bony fish represent the most basal vertebrate branch with a dedicated mucosal immune system, which comprises immunologically heterogeneous microenvironments armed with innate and adaptive components. In rainbow trout (Oncorhynchus mykiss), a nasopharynx-associated lymphoid tissue (NALT) was recently described as a diffuse network of myeloid and lymphoid cells located in the olfactory organ of fish. Several studies have demonstrated high levels of protection conferred by nasal vaccines against viral and bacterial pathogens; however, the mechanisms underlying the observed protection are not well understood. We applied 5'RACE and a deep sequencing-based approach to investigate the clonal structure of the systemic and mucosal rainbow trout B cell repertoire. The analysis of Ig repertoire in control trout suggests different structures of IgM and IgT spleen and NALT repertoires, with restricted repertoire diversity in NALT. Nasal and injection vaccination with a bacterial vaccine revealed unique dynamics of IgM and IgT repertoires at systemic and mucosal sites and the remarkable ability of nasal vaccines to induce spleen Ig responses. Our findings provide an important immunological basis for the effectiveness of nasal vaccination in fish and other vertebrate animals and will help the design of future nasal vaccination strategies.


Antibodies/immunology , Bacterial Vaccines/immunology , Nose/immunology , Oncorhynchus mykiss/immunology , Animals , Fish Diseases/immunology , Fish Diseases/microbiology , Immunity, Mucosal/immunology , Immunoglobulin M/immunology , Lymphocytes/immunology , Lymphocytes/microbiology , Myeloid Cells/immunology , Myeloid Cells/microbiology , Nose/microbiology , Oncorhynchus mykiss/microbiology , Spleen/immunology , Spleen/microbiology , Vaccination/methods
15.
Eur J Immunol ; 49(9): 1380-1390, 2019 09.
Article En | MEDLINE | ID: mdl-31365119

CD200 receptor 1(CD200R1) signalling limits myeloid cell responses and reduces autoimmunity, alloimmunity and viral-mediated immunopathology, but has never been examined in the context of eosinophilic inflammation. Susceptibility to lung fungal infection is associated with T-helper 2 (Th2) cytokine dominated responses and strong eosinophilic pathology. Blockade of CD200R1 enhances type I cytokine responses in many infectious and non-infectious settings and so may promote a more protective response to fungal infection. By contrast, we demonstrate that, rather than promoting type I cytokine responses, CD200R1 blockade enhanced eosinophilia in a mouse model of Cryptococcus neoformans infection, whereas CD200R1 agonism reduced lung eosinophilia - with neither strategy completely altering fungal burden. Thus, we reveal a surprising disconnect between pulmonary eosinophilia and cryptococcal burden and dissemination. This research has 2 important implications. Firstly, a lack of CD200R1 signalling enhances immune responses regardless of cytokine polarisation, and secondly reducing eosinophils does not allow protective immunity to develop in susceptible fungal system. Therefore, agonists of CD200R1 may be beneficial for eosinophilic pathologies.


Lung Diseases, Fungal/immunology , Orexin Receptors/immunology , Pulmonary Eosinophilia/immunology , Animals , Cryptococcosis/immunology , Cryptococcosis/microbiology , Cryptococcus neoformans/immunology , Cytokines/immunology , Disease Models, Animal , Inflammation/immunology , Inflammation/microbiology , Lung , Lung Diseases, Fungal/microbiology , Mice , Myeloid Cells/immunology , Myeloid Cells/microbiology , Pulmonary Eosinophilia/microbiology , Th2 Cells/immunology , Th2 Cells/microbiology
16.
Cell Mol Life Sci ; 76(20): 3917-3937, 2019 Oct.
Article En | MEDLINE | ID: mdl-31250035

The gastrointestinal tract is the site of nutrient digestion and absorption and is also colonized by diverse, highly mutualistic microbes. The intestinal microbiota has diverse effects on the development and function of the gut-specific immune system, and provides some protection from infectious pathogens. However, interactions between intestinal immunity and microorganisms are very complex, and recent studies have revealed that this intimate crosstalk may depend on the production and sensing abilities of multiple bioactive small molecule metabolites originating from direct produced by the gut microbiota or by the metabolism of dietary components. Here, we review the interplay between the host immune system and the microbiota, how commensal bacteria regulate the production of metabolites, and how these microbiota-derived products influence the function of several major innate and adaptive immune cells involved in modulating host immune homeostasis.


Adaptive Immunity , Dysbiosis/metabolism , Gastrointestinal Microbiome/immunology , Immunity, Innate , Intestinal Mucosa/metabolism , Metabolome/immunology , Amino Acids/immunology , Amino Acids/metabolism , Animals , Bile Acids and Salts/immunology , Bile Acids and Salts/metabolism , Dysbiosis/immunology , Dysbiosis/microbiology , Dysbiosis/therapy , Fatty Acids/immunology , Fatty Acids/metabolism , Fecal Microbiota Transplantation , Germ-Free Life/immunology , Homeostasis/immunology , Humans , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Lymphocytes/immunology , Lymphocytes/metabolism , Lymphocytes/microbiology , Myeloid Cells/immunology , Myeloid Cells/metabolism , Myeloid Cells/microbiology , Symbiosis/immunology
17.
PLoS One ; 13(12): e0208940, 2018.
Article En | MEDLINE | ID: mdl-30550567

RATIONALE: HSP90B1, also known as gp96, is a chaperone for multiple Toll-like receptors (TLRs) and is necessary for TLR-mediated inflammatory responses in murine myeloid cells. The molecule is also expressed in T-cells though its specific role is unknown. We hypothesized that human HSP90B1 regulates monocyte and T-cell responses to Mycobacterium tuberculosis (Mtb) and bacilli Calmette-Guerin (BCG) and that its variants are associated with susceptibility to TB disease. METHODS: We screened 17 haplotype-tagging SNPs in the HSP90B1 gene region for association with BCG-induced T-cell cytokine responses using both an ex-vivo whole blood assay (N = 295) and an intracellular cytokine staining assay (N = 180) on samples collected 10 weeks after birth. Using a case-control study design, we evaluated the same SNPs for association with TB disease in a South African pediatric cohort (N = 217 cases, 604 controls). A subset of these SNPs was evaluated for association with HSP90B1 expression in human monocytes, monocyte-derived dendritic cells, and T-cells using RT-PCR. Lastly, we used CRISPR/Cas9 gene editing to knock down HSP90B1 expression in a human monocyte cell line (U937). Knockdown and control cell lines were tested for TLR surface expression and control of Mtb replication. RESULTS: We identified three SNPs, rs10507172, rs10507173 and rs1920413, that were associated with BCG-induced IL-2 secretion (p = 0.017 for rs10507172 and p = 0.03 for rs10507173 and rs1920413, Mann-Whitney, dominant model). SNPs rs10507172 and rs10507173 were associated with TB disease in an unadjusted analysis (p = 0.036 and 0.025, respectively, dominant model) that strengthened with sensitivity analysis of the definite TB cases, which included only those patients with microbiologically confirmed Mtb (p = 0.007 and 0.012, respectively). Knockdowns of HSP90B1 in monocyte cell lines with CRISPR did not alter TLR2 surface expression nor influence Mtb replication relative to controls. CONCLUSION: Among infants, an HSP90B1 gene-region variant is associated with BCG-induced IL-2 production and may be associated with protection from TB disease. HSP90B1 knockdown in human monocyte-like cell lines did not influence TLR2 surface localization nor Mtb replication. Together, these data suggest that HSP90B1 regulates T-cell, but not monocyte, responses to mycobacteria in humans.


Interleukin-2/genetics , Membrane Glycoproteins/genetics , Mycobacterium tuberculosis/genetics , Toll-Like Receptor 2/genetics , Tuberculosis/genetics , Animals , Female , Gene Editing , Gene Expression Regulation/genetics , Gene Expression Regulation/immunology , Genetic Predisposition to Disease/genetics , Haplotypes , Humans , Infant , Interleukin-2/immunology , Membrane Glycoproteins/immunology , Mice , Mycobacterium bovis/genetics , Mycobacterium bovis/pathogenicity , Mycobacterium tuberculosis/pathogenicity , Myeloid Cells/immunology , Myeloid Cells/microbiology , Polymorphism, Single Nucleotide/genetics , T-Lymphocytes/immunology , T-Lymphocytes/microbiology , Toll-Like Receptor 2/immunology , Tuberculosis/microbiology
18.
Biochem Biophys Res Commun ; 506(4): 907-911, 2018 12 02.
Article En | MEDLINE | ID: mdl-30392911

Metagenomic analyses indicate that streptococcus gallolyticus is enriched at carcinoma in colitis associated colorectal cancer compared with sporadic colorectal cancer. But the particular mechanism of streptococcus gallolyticus in Inflammatory Bowel Disease malignant progression remains undefined yet. We aim to explore the biological carcinogenesis efficacy of streptococcus gallolyticus and its potential mechanism in azoxymethane and dextran sodium sulphate-induced colitis associated colorectal cancer in mice. Oral pretreatment of streptococcus gallolyticus was adopted to evaluate its detrimental effect. The colorectums of experimental C57BL/6 mice were collected and examined for the degree of tumorigenesis. Comparative 16S rRNA sequencing was carried out to observe streptococcus gallolyticus alterations in abundance. We found that streptococcus gallolyticus are enriched in colonic carcinoma compared to adenoma and healthy mice. Pretreatment of Streptococcus gallolyticus aggravated tumor formation, with more colonic obstruction, larger number and diameter of tumor node. Furthermore, Streptococcus gallolyticus selectively recruits tumor-infiltrating myeloid cells but not mast cells, including marrow-derived suppressor cells, tumor-associated macrophages and dendritic cells, which can inhibit competence of T cells. Moreover, several myeloid-cell-derived proinflammatory cytokines (IL-6, IL-1ß, IL-8, CCL2, COX-2, TNF-α) were increased with the formation of carcinoma in IBD. Collectively, these data suggest that, through recruitment of tumor-infiltrating immune cells, Streptococcus gallolyticus generate an immune suppressive microenvironment that is conducive for neoplasia progression of Inflammatory Bowel Disease.


Carcinogenesis/pathology , Inflammatory Bowel Diseases/pathology , Myeloid Cells/microbiology , Myeloid Cells/pathology , Streptococcus gallolyticus/physiology , Adenoma/pathology , Animals , CD11b Antigen/metabolism , Colorectal Neoplasms/pathology , Female , Intestinal Mucosa/pathology , Mice, Inbred C57BL
19.
J Exp Med ; 215(11): 2936-2954, 2018 11 05.
Article En | MEDLINE | ID: mdl-30355616

The foodborne pathogen Listeria monocytogenes (Lm) crosses the intestinal villus epithelium via goblet cells (GCs) upon the interaction of Lm surface protein InlA with its receptor E-cadherin. Here, we show that Lm infection accelerates intestinal villus epithelium renewal while decreasing the number of GCs expressing luminally accessible E-cadherin, thereby locking Lm portal of entry. This novel innate immune response to an enteropathogen is triggered by the infection of Peyer's patch CX3CR1+ cells and the ensuing production of IL-23. It requires STAT3 phosphorylation in epithelial cells in response to IL-22 and IL-11 expressed by lamina propria gp38+ stromal cells. Lm-induced IFN-γ signaling and STAT1 phosphorylation in epithelial cells is also critical for Lm-associated intestinal epithelium response. GC depletion also leads to a decrease in colon mucus barrier thickness, thereby increasing host susceptibility to colitis. This study unveils a novel innate immune response to an enteropathogen, which implicates gp38+ stromal cells and locks intestinal villus invasion, but favors colitis.


Colitis/immunology , Intestinal Mucosa/immunology , Listeria monocytogenes/immunology , Listeriosis/immunology , Membrane Glycoproteins/immunology , Myeloid Cells/immunology , Peyer's Patches/immunology , Animals , Colitis/genetics , Colitis/microbiology , Colitis/pathology , Cytokines/genetics , Cytokines/immunology , Immunity, Innate/genetics , Immunity, Mucosal/genetics , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Listeriosis/genetics , Listeriosis/pathology , Membrane Glycoproteins/genetics , Mice , Mice, Knockout , Myeloid Cells/microbiology , Myeloid Cells/pathology , Peyer's Patches/microbiology , Peyer's Patches/pathology , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/immunology , Stromal Cells/immunology , Stromal Cells/microbiology , Stromal Cells/pathology
20.
PLoS One ; 13(8): e0203395, 2018.
Article En | MEDLINE | ID: mdl-30169526

IL-6 is required for the response of mice against Listeria monocytogenes. Control of infection depends on classical IL-6 signaling via membrane IL-6Rα, but IL-6 target cells and protective mechanisms remain unclear. We used mice with IL-6Rα-deficiency in T cells (Il6rafl/fl×CD4cre) or myeloid cells (Il6rafl/fl×LysMcre) to define the role of these cells in IL-6-mediated protection. Abrogation of IL-6Rα in T cells did not interfere with bacteria control and induction of TH1 and CD8+ T-cell responses. IL-6Rα-deficiency in myeloid cells caused significant defects in listeria control. This defect was not associated with reduced recruitment of granulocytes and inflammatory monocytes, and both cell populations were activated and not impaired in cytokine production. However, IL-6Rα-deficient inflammatory monocytes displayed diminished expression of IL-4Rα and of CD38, a protein required for phagocytosis and innate control of listeria. In vitro studies revealed that IL-4 and IL-6 cooperated in induction of CD38. In listeria-infected mice, phagocytic activity of inflammatory monocytes correlated with CD38 expression levels on cells and inflammatory monocytes of Il6rafl/fl×LysMcre mice were significantly impaired in phagocytosis. In conclusion, we demonstrate that inhibition of classical IL-6 signaling in myeloid cells causes alterations in differentiation and function of these cells, which subsequently prevent effective control of L. monocytogenes.


Interleukin-6/metabolism , Listeria monocytogenes/immunology , Listeriosis/immunology , Listeriosis/metabolism , Myeloid Cells/immunology , Signal Transduction/immunology , ADP-ribosyl Cyclase 1/metabolism , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/microbiology , Inflammation/immunology , Inflammation/metabolism , Inflammation/microbiology , Mice , Mice, Inbred C57BL , Monocytes/immunology , Monocytes/metabolism , Monocytes/microbiology , Myeloid Cells/metabolism , Myeloid Cells/microbiology , Phagocytosis/immunology , Receptors, Interleukin-4/metabolism , Receptors, Interleukin-6/metabolism
...