Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Article in English | MEDLINE | ID: mdl-38744621

ABSTRACT

INTRODUCTION: Lung cancer is one of the most prevalent malignancies worldwide. Substantial research has illuminated the intricate interplay between microorganisms and human health, revealing their role in disease regulation. Trichomonads is a flagellated protozoan in the human cavity and have been previously identified as a pathogen associated with pneumonia, contributing to tissue chronic inflammation and carcinogenesis. METHODS: Nested polymerase chain reaction methods were employed to scrutinize the prevalence of trichomonads in the bronchovesicular fluid of patients diagnosed with lung cancer. Subsequently, the influence of Trichomonas tenax invasion on lung cancer cells was elucidated through proliferation assays, migration assays, and transcription analysis. RESULTS: Bronchoalveolar fluid samples from lung cancer patients yielded positive nested PCR results for eight out of twenty-seven samples. Seven of these samples were identified as Trichomonas tenax, while one was identified as Tetratrichomonas spp. Our findings revealed a significant upregulation of pathways associated with carcinogenesis, including cellular proliferation, migration, and drug resistance, in response to T. tenax invasion. CONCLUSIONS: This study underscores the importance of recognizing the presence of trichomonads and the influence of T. tenax invasion on host responses to respiratory diseases. The identified pathways implicated in cancer development may pave the way for developing targeted treatment strategies for pulmonary diseases. These findings hold promise for informing and improving the precision of therapeutic interventions in the context of pulmonary ailments.

2.
Cell Rep Med ; 5(1): 101360, 2024 01 16.
Article in English | MEDLINE | ID: mdl-38232694

ABSTRACT

The failure of the Staphylococcus aureus (SA) IsdB vaccine trial can be explained by the recall of non-protective immune imprints from prior SA exposure. Here, we investigate natural human SA humoral imprints to understand their broader impact on SA immunizations. We show that antibody responses against SA cell-wall-associated antigens (CWAs) are non-opsonic, while antibodies against SA toxins are neutralizing. Importantly, the protective characteristics of the antibody imprints accurately predict the failure of corresponding vaccines against CWAs and support vaccination against toxins. In passive immunization platforms, natural anti-SA human antibodies reduce the efficacy of the human monoclonal antibodies suvratoxumab and tefibazumab, consistent with the results of their respective clinical trials. Strikingly, in the absence of specific humoral memory responses, active immunizations are efficacious in both naive and SA-experienced mice. Overall, our study points to a practical and predictive approach to evaluate and develop SA vaccines based on pre-existing humoral imprint characteristics.


Subject(s)
Methicillin-Resistant Staphylococcus aureus , Staphylococcal Vaccines , Vaccines , Animals , Humans , Mice , Immunization , Staphylococcus aureus , Clinical Trials as Topic
3.
mBio ; 15(1): e0022523, 2024 Jan 16.
Article in English | MEDLINE | ID: mdl-38112465

ABSTRACT

IMPORTANCE: The prevalence of multidrug-resistant Staphylococcus aureus is of global concern, and vaccines are urgently needed. The iron-regulated surface determinant protein B (IsdB) of S. aureus was investigated as a vaccine candidate because of its essential role in bacterial iron acquisition but failed in clinical trials despite strong immunogenicity. Here, we reveal an unexpected second function for IsdB in pathogen-host interaction: the bacterial fitness factor IsdB triggers a strong inflammatory response in innate immune cells via Toll-like receptor 4 and the inflammasome, thus acting as a novel pathogen-associated molecular pattern of S. aureus. Our discovery contributes to a better understanding of how S. aureus modulates the immune response, which is necessary for vaccine development against the sophisticated pathogen.


Subject(s)
Bacterial Proteins , Cation Transport Proteins , Cytokines , Methicillin-Resistant Staphylococcus aureus , NLR Family, Pyrin Domain-Containing 3 Protein , Staphylococcal Infections , Toll-Like Receptor 4 , Humans , Bacterial Proteins/immunology , Caspase 1/metabolism , Cation Transport Proteins/immunology , Cytokines/metabolism , Inflammasomes/metabolism , Iron/metabolism , Methicillin-Resistant Staphylococcus aureus/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Staphylococcal Infections/immunology , Toll-Like Receptor 4/metabolism
4.
Nat Commun ; 14(1): 8061, 2023 Dec 05.
Article in English | MEDLINE | ID: mdl-38052825

ABSTRACT

Acne is a dermatologic disease with a strong pathologic association with human commensal Cutibacterium acnes. Conspicuously, certain C. acnes phylotypes are associated with acne, whereas others are associated with healthy skin. Here we investigate if the evolution of a C. acnes enzyme contributes to health or acne. Two hyaluronidase variants exclusively expressed by C. acnes strains, HylA and HylB, demonstrate remarkable clinical correlation with acne or health. We show that HylA is strongly pro-inflammatory, and HylB is modestly anti-inflammatory in a murine (female) acne model. Structural and phylogenic studies suggest that the enzymes evolved from a common hyaluronidase that acquired distinct enzymatic activity. Health-associated HylB degrades hyaluronic acid (HA) exclusively to HA disaccharides leading to reduced inflammation, whereas HylA generates large-sized HA fragments that drive robust TLR2-dependent pathology. Replacing an amino acid, Serine to Glycine near the HylA catalytic site enhances the enzymatic activity of HylA and produces an HA degradation pattern intermediate to HylA and HylB. Selective targeting of HylA using peptide vaccine or inhibitors alleviates acne pathology. We suggest that the functional divergence of HylA and HylB is a major driving force behind C. acnes health- and acne- phenotype and propose targeting of HylA as an approach for acne therapy.


Subject(s)
Acne Vulgaris , Hyaluronoglucosaminidase , Humans , Female , Animals , Mice , Skin/microbiology , Propionibacterium acnes/genetics , Amino Acids
5.
BMC Microbiol ; 23(1): 198, 2023 07 26.
Article in English | MEDLINE | ID: mdl-37495951

ABSTRACT

BACKGROUND: Acanthamoeba castellanii is a free-living protist that feeds on diverse bacteria. A. castellanii has frequently been utilized in studies on microbial interactions. Grazing bacteria also exhibit diverse effects on the physiological characteristics of amoebae, such as their growth, encystation, and cytotoxicity. Since the composition of amoebae amino acids is closely related to cellular activities, it can indicate the overall responses of A. castellanii to various stimuli. METHOD: A. castellanii was exposed to different culture conditions in low-nutrient medium with heat-killed DH5α to clarify their effects. A targeted metabolomic technique was utilized to evaluate the concentration of cellular amino acids. The amino acid composition and pathways were analyzed by two web-based tools: MetaboAnalyst and Pathview. Then, long-term exposure to A. castellanii was investigated through in silico and in vitro methods to elucidate the homeostasis of amino acids and the growth of A. castellanii. RESULTS: Under short-term exposure, all kinds of amino acids were enriched in all exposed groups. In contrast to the presence of heat-killed bacteria, the medium exhibited obvious effects on the amino acid composition of A. castellanii. After long-term exposure, the amino acid composition was more similar to that of the control group. A. castellanii may achieve amino acid homeostasis through pathways related to alanine, aspartate, citrulline, and serine. DISCUSSION: Under short-term exposure, compared to the presence of bacteria, the type of medium exerted a more powerful effect on the amino acid composition of the amoeba. Previous studies focused on the interaction of the amoeba and bacteria with effective secretion systems and effectors. This may have caused the effects of low-nutrient environments to be overlooked. CONCLUSION: When A. castellanii was stimulated in the coculture system through various methods, such as the presence of bacteria and a low-nutrient environment, it accumulated intracellular amino acids within a short period. However, different stimulations correspond to different amino acid compositions. After long-term exposure, A. castellanii achieved an amino acid equilibrium by downregulating the biosynthesis of several amino acids.


Subject(s)
Acanthamoeba castellanii , Amino Acids , Escherichia coli , Acanthamoeba castellanii/chemistry , Acanthamoeba castellanii/growth & development , Acanthamoeba castellanii/physiology , Coculture Techniques , Amino Acids/analysis , Acclimatization , Hot Temperature , Culture Media
6.
Proc Natl Acad Sci U S A ; 120(30): e2301538120, 2023 07 25.
Article in English | MEDLINE | ID: mdl-37459522

ABSTRACT

Pseudomonas aeruginosa (PA) CbpD belongs to the lytic polysaccharide monooxygenases (LPMOs), a family of enzymes that cleave chitin or related polysaccharides. Here, we demonstrate a virulence role of CbpD in PA pneumonia linked to impairment of host complement function and opsonophagocytic clearance. Following intratracheal challenge, a PA ΔCbpD mutant was more easily cleared and produced less mortality than the wild-type parent strain. The x-ray crystal structure of the CbpD LPMO domain was solved to subatomic resolution (0.75Å) and its two additional domains modeled by small-angle X-ray scattering and Alphafold2 machine-learning algorithms, allowing structure-based immune epitope mapping. Immunization of naive mice with recombinant CbpD generated high IgG antibody titers that promoted human neutrophil opsonophagocytic killing, neutralized enzymatic activity, and protected against lethal PA pneumonia and sepsis. IgG antibodies generated against full-length CbpD or its noncatalytic M2+CBM73 domains were opsonic and protective, even in previously PA-exposed mice, while antibodies targeting the AA10 domain were not. Preexisting antibodies in PA-colonized cystic fibrosis patients primarily target the CbpD AA10 catalytic domain. Further exploration of LPMO family proteins, present across many clinically important and antibiotic-resistant human pathogens, may yield novel and effective vaccine antigens.


Subject(s)
Mixed Function Oxygenases , Pneumonia , Humans , Mice , Animals , Mixed Function Oxygenases/metabolism , Pseudomonas aeruginosa/metabolism , Polysaccharides/metabolism , Immunization
8.
Cell Host Microbe ; 30(8): 1163-1172.e6, 2022 08 10.
Article in English | MEDLINE | ID: mdl-35803276

ABSTRACT

Humans frequently encounter Staphylococcus aureus (SA) throughout life. Animal studies have yielded SA candidate vaccines, yet all human SA vaccine trials have failed. One notable vaccine "failure" targeted IsdB, critical for host iron acquisition. We explored a fundamental difference between humans and laboratory animals-natural SA exposure. Recapitulating the failed phase III IsdB vaccine trial, mice previously infected with SA do not mount protective antibody responses to vaccination, unlike naive animals. Non-protective antibodies exhibit increased α2,3 sialylation that blunts opsonophagocytosis and preferentially targets a non-protective IsdB domain. IsdB vaccination of SA-infected mice recalls non-neutralizing humoral responses, further reducing vaccine efficacy through direct antibody competition. IsdB vaccine interference was overcome by immunization against the IsdB heme-binding domain. Purified human IsdB-specific antibodies also blunt IsdB passive immunization, and additional SA vaccines are susceptible to SA pre-exposure. Thus, failed anti-SA immunization trials could be explained by non-protective imprint from prior host-SA interaction.


Subject(s)
Cation Transport Proteins , Staphylococcal Infections , Vaccines , Animals , Humans , Mice , Phagocytosis , Staphylococcal Infections/prevention & control , Staphylococcus aureus
9.
Exp Parasitol ; 239: 108312, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35738459

ABSTRACT

Acanthamoeba castellanii is a free-living protozoan that causes several severe human parasitic diseases such as Acanthamoeba keratitis and granulomatous encephalitis. A. castellanii feeds on bacteria, yeasts, and other organic particles as food sources, but the mechanisms of digestion in acanthamoebal cells are unclear. Rab GTPases participate in endosomal delivery in eukaryotes after phagocytosis. This study aimed to determine the potential functions of A. castellanii Rab7 (AcRab7), which is involved in phagocytosis, and the relationship between AcRab7 and further cellular physiological phenomena. In this study, the inhibitor CID1067700 (CID) was used to specifically inhibit the binding of nucleotides to confirm the potential functions of AcRab7. Cellular proliferation and ATP assays were also used to detect underlying cellular physiological functions after blocking the phagocytosis pathway. We found that AcRab7 expression increased as the co-culture time with Escherichia coli increased. Immunofluorescence staining showed that AcRab7 colocalized with lysosomes in its GTP-activating form. In addition, AcRab7 inhibition resulted in a reduction in cell proliferation and ATP levels. Our results suggest that AcRab7 participates in endosomal delivery and dominates energy production and cell growth.


Subject(s)
Acanthamoeba Keratitis , Acanthamoeba castellanii , Acanthamoeba Keratitis/parasitology , Acanthamoeba castellanii/physiology , Adenosine Triphosphate , Escherichia coli , Humans , Phagocytosis
10.
Cell Chem Biol ; 29(5): 730-740, 2022 05 19.
Article in English | MEDLINE | ID: mdl-35594849

ABSTRACT

Staphylococcus aureus (SA) is a leading cause of bacterial infection and antibiotic resistance globally. Therefore, development of an effective vaccine has been a major goal of the SA field for the past decades. With the wealth of understanding of pathogenesis, the failure of all SA vaccine trials has been a surprise. We argue that experimental SA vaccines have not worked because vaccines have been studied in naive laboratory animals, whereas clinical vaccine efficacy is tested in immune environments reprogrammed by SA. Here, we review the failed SA vaccines that have seemingly defied all principles of vaccinology. We describe major SA evasion strategies and suggest that they reshape the immune environment in a way that makes vaccines prone to failures. We propose that appropriate integration of concepts of host-pathogen interaction into vaccine study designs could lead to insight critical for the development of an effective SA vaccine.


Subject(s)
Methicillin-Resistant Staphylococcus aureus , Staphylococcal Infections , Vaccines , Animals , Host-Pathogen Interactions , Staphylococcal Infections/microbiology , Staphylococcal Infections/prevention & control , Staphylococcus aureus
11.
J Infect Dis ; 223(7): 1222-1231, 2021 04 08.
Article in English | MEDLINE | ID: mdl-32990305

ABSTRACT

A successful Staphylococcus aureus vaccine remains elusive, and one controversy in the field is whether humans generate a protective adaptive immune response to infection. We developed a bacterial challenge murine assay that directly assesses the protective capacity of adoptively transferred human serum samples. We first validated the model by showing that postpneumococcal vaccine serum samples from humans induced effective clearance of Streptococcus pneumoniae in mice. We then found that human serum samples adoptively transferred from children with invasive S. aureus infections exhibited protection from disease in a murine model, with some samples conferring near complete protection. These findings demonstrate that human serum samples are capable of conferring a protective adaptive response generated by humans during invasive staphylococcal disease, allowing for the study of protective factors in a murine model. Identification of the protective factors present in the most efficacious serum samples would be of high interest as potential staphylococcal vaccine candidates or passive therapeutics.


Subject(s)
Adoptive Transfer , Antibodies, Bacterial/immunology , Sepsis , Staphylococcal Infections , Animals , Child , Disease Models, Animal , Humans , Mice , Sepsis/prevention & control , Staphylococcal Infections/prevention & control , Staphylococcus aureus
12.
Cell ; 182(5): 1311-1327.e14, 2020 09 03.
Article in English | MEDLINE | ID: mdl-32888495

ABSTRACT

Staphylococcus aureus bacteremia (SaB) causes significant disease in humans, carrying mortality rates of ∼25%. The ability to rapidly predict SaB patient responses and guide personalized treatment regimens could reduce mortality. Here, we present a resource of SaB prognostic biomarkers. Integrating proteomic and metabolomic techniques enabled the identification of >10,000 features from >200 serum samples collected upon clinical presentation. We interrogated the complexity of serum using multiple computational strategies, which provided a comprehensive view of the early host response to infection. Our biomarkers exceed the predictive capabilities of those previously reported, particularly when used in combination. Last, we validated the biological contribution of mortality-associated pathways using a murine model of SaB. Our findings represent a starting point for the development of a prognostic test for identifying high-risk patients at a time early enough to trigger intensive monitoring and interventions.


Subject(s)
Bacteremia/blood , Bacteremia/mortality , Staphylococcal Infections/blood , Staphylococcal Infections/mortality , Staphylococcus aureus/pathogenicity , Animals , Bacteremia/metabolism , Biomarkers/blood , Biomarkers/metabolism , Disease Models, Animal , Female , Humans , Male , Metabolomics/methods , Mice , Middle Aged , Prognosis , Proteomics/methods , Risk Factors , Staphylococcal Infections/metabolism
13.
Exp Parasitol ; 218: 107985, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32918877

ABSTRACT

Acanthamoeba castellanii is a protist that has a high predation efficiency for bacteria in a number of monoxenic culture experiments. However, the role of A. castellanii in the microbial community is still unknown because of the lack of studies on multiple-species interactions. The aim of this study was to investigate the change of bacterial composition after A. castellanii emerges in a water environment. We added A. castellanii to an environmental water sample and incubated it for two days. Then, we performed 16S ribosomal RNA sequencing techniques to analyze the changes in bacterial composition. In this study, A. castellanii slightly increased the relative abundance of a few opportunistic pathogens, such as Legionella, Roseomonas, and Haemophilus. This result may be related to the training ground hypothesis. On the other hand, the growth of some bacteria was inhibited, such as Cyanobacteria and Firmicutes. Although A. castellanii did not drastically change the whole bacterial community, we surprisingly found the dissolved oxygen concentration was increased after incubation with A. castellanii. We applied environmental water at the laboratory scale to investigate the interactions among A. castellanii, complex microbial communities and the environment. We identified the bacteria that are sensitive to A. castellanii and further found the novel relationship between dissolved oxygen and microbial interaction. Our results helped to clarify the role of A. castellanii in microbial communities.


Subject(s)
Acanthamoeba castellanii/physiology , Bdellovibrio/isolation & purification , Legionella/isolation & purification , Microbiota/physiology , Oxygen/metabolism , Acanthamoeba castellanii/genetics , Bdellovibrio/genetics , Bdellovibrio/physiology , DNA/isolation & purification , Legionella/genetics , Legionella/pathogenicity , Legionella/physiology , Ponds/microbiology , Ponds/parasitology , RNA, Ribosomal, 16S/chemistry , Real-Time Polymerase Chain Reaction , Reproducibility of Results , Virulence
14.
PLoS Pathog ; 16(8): e1008733, 2020 08.
Article in English | MEDLINE | ID: mdl-32817694

ABSTRACT

Staphylococcus aureus (S. aureus) is one of the most common bacterial infections worldwide, and antibiotic resistant strains such as Methicillin-Resistant S. aureus (MRSA) are a major threat and burden to public health. MRSA not only infects immunocompromised patients but also healthy individuals and has rapidly spread from the healthcare setting to the outside community. However, all vaccines tested in clinical trials to date have failed. Immunocompromised individuals such as patients with HIV or decreased levels of CD4+ T cells are highly susceptible to S. aureus infections, and they are also at increased risk of developing fungal infections. We therefore wondered whether stimulation of antifungal immunity might promote the type of immune responses needed for effective host defense against S. aureus. Here we show that vaccination of mice with a fungal ß-glucan particle (GP) loaded with S. aureus antigens provides protective immunity to S. aureus. We generated glucan particles loaded with the four S. aureus proteins ClfA, IsdA, MntC, and SdrE, creating the 4X-SA-GP vaccine. Vaccination of mice with three doses of 4X-SA-GP promoted protection in a systemic model of S. aureus infection with a significant reduction in the bacterial burden in the spleen and kidneys. 4X-SA-GP vaccination induced antigen-specific Th1 and Th17 CD4+ T cell and antibody responses and provided long-term protection. This work suggests that the GP vaccine system has potential as a novel approach to developing vaccines for S. aureus.


Subject(s)
Saccharomyces cerevisiae/immunology , Staphylococcal Infections/immunology , Staphylococcal Vaccines/immunology , Staphylococcus aureus/immunology , Animals , Antibodies, Bacterial/immunology , Coagulase/administration & dosage , Coagulase/genetics , Coagulase/immunology , Female , Humans , Mice , Mice, Inbred C57BL , Saccharomyces cerevisiae/chemistry , Staphylococcal Infections/microbiology , Staphylococcal Vaccines/administration & dosage , Staphylococcal Vaccines/genetics , Staphylococcus aureus/genetics , Th1 Cells/immunology , Th17 Cells/immunology , Vaccination , beta-Glucans/administration & dosage , beta-Glucans/immunology
15.
J Innate Immun ; 12(4): 333-343, 2020.
Article in English | MEDLINE | ID: mdl-31805552

ABSTRACT

Pathogenic microorganisms are sensed by the inflammasome, resulting in the release of the pro-immune and proinflammatory cytokine interleukin-1ß (IL-1ß). In humans, the paired sialic acid-binding Ig-like lectin receptors Siglec-5 (inhibitory) and Siglec-14 (activating) have been shown to have reciprocal roles in regulating macrophage immune responses, but their interaction with IL-1ß signaling and the inflammasome has not been characterized. Here we show that in response to known inflammasome activators (ATP, nigericin) or the sialic acid-expressing human bacterial pathogen group B Streptococcus (GBS), the presence of Siglec-14 enhances, whereas Siglec-5 reduces, inflammasome activation and macrophage IL-1ß release. Human THP-1 macrophages stably transfected with Siglec-14 exhibited increased caspase-1 activation, IL-1ß release and pyroptosis after GBS infection, in a manner blocked by a specific inhibitor of nucleotide-binding domain leucine-rich repeat protein 3 (NLRP3), a protein involved in inflammasome assembly. Another leading pathogen, Streptococcus pneumoniae, lacks sialic acid but rather prominently expresses a sialidase, which cleaves sialic acid from macrophages, eliminating cis- interactions with the lectin receptor, thus attenuating Siglec-14 induced IL-1ß secretion. Vimentin, a cytoskeletal protein released during macrophage inflammatory activation is known to induce the inflammasome. We found that vimentin has increased interaction with Siglec-14 compared to Siglec-5, and this interaction heightened IL-1ß production by Siglec-14-expressing cells. Siglec-14 is absent from some humans because of a SIGLEC5/14 fusion polymorphism, and we found increased IL-1ß expression in primary macrophages from SIGLEC14+/+ individuals compared to those with the SIGLEC14-/+ and SIGLEC14-/- genotypes. Collectively, our results identify a new immunoregulatory role of Siglec-14 as a positive regulator of NLRP3 inflammasome activation.


Subject(s)
Inflammasomes/immunology , Lectins/immunology , Macrophages/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Receptors, Cell Surface/immunology , Humans , Inflammasomes/genetics , Lectins/genetics , Macrophages/microbiology , Macrophages/pathology , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Receptors, Cell Surface/genetics , Streptococcal Infections/genetics , Streptococcal Infections/immunology , Streptococcal Infections/pathology , Streptococcus agalactiae/immunology , THP-1 Cells
16.
JCI Insight ; 4(5)2019 03 07.
Article in English | MEDLINE | ID: mdl-30843879

ABSTRACT

Genomic studies revealed the existence of health- and acne-associated P. acnes strains and suggested novel approaches for broadening understanding of acne vulgaris. However, clinical association of P. acnes with disease or health has yet to be corroborated experimentally. Current animal models of acne do not closely mimic human disease and have unclear translational value. We have developed a murine model of acne by combining P. acnes inoculation with topical application of a synthetic human sebum. We showed that human sebum promoted persistence of intradermally injected P. acnes with little loss of viability after 1 week and permitted use of more physiologic inoculums. Application of acne-associated P. acnes RT4/5 strains led to development of moderate to severe skin pathology compared with application of health-associated type II P. acnes strains (RT2/6). RT4/5 P. acnes strains uniformly induced higher levels of KC (IL-8), IL-1α, IL-1ß, and IL-6 in vitro and in vivo compared with type II P. acnes strains. Overall, our data provide immunopathologic corroboration of health and disease association of clinical P. acnes strains and inform on a platform to query putative virulence factors uncovered by genomic studies.


Subject(s)
Acne Vulgaris/immunology , Acne Vulgaris/microbiology , Gram-Positive Bacterial Infections/immunology , Propionibacterium acnes/immunology , Skin/immunology , Skin/pathology , Acne Vulgaris/genetics , Acne Vulgaris/pathology , Animals , Bone Marrow Cells , Cell Line , Disease Models, Animal , Female , Humans , Interleukin-1alpha/metabolism , Interleukin-6 , Interleukin-8/metabolism , Mice , Mice, Inbred C57BL , Propionibacterium acnes/pathogenicity , Skin/microbiology , Virulence Factors
17.
EMBO J ; 38(8)2019 04 15.
Article in English | MEDLINE | ID: mdl-30918008

ABSTRACT

Long noncoding RNAs (lncRNAs) can regulate target gene expression by acting in cis (locally) or in trans (non-locally). Here, we performed genome-wide expression analysis of Toll-like receptor (TLR)-stimulated human macrophages to identify pairs of cis-acting lncRNAs and protein-coding genes involved in innate immunity. A total of 229 gene pairs were identified, many of which were commonly regulated by signaling through multiple TLRs and were involved in the cytokine responses to infection by group B Streptococcus We focused on elucidating the function of one lncRNA, named lnc-MARCKS or ROCKI (Regulator of Cytokines and Inflammation), which was induced by multiple TLR stimuli and acted as a master regulator of inflammatory responses. ROCKI interacted with APEX1 (apurinic/apyrimidinic endodeoxyribonuclease 1) to form a ribonucleoprotein complex at the MARCKS promoter. In turn, ROCKI-APEX1 recruited the histone deacetylase HDAC1, which removed the H3K27ac modification from the promoter, thus reducing MARCKS transcription and subsequent Ca2+ signaling and inflammatory gene expression. Finally, genetic variants affecting ROCKI expression were linked to a reduced risk of certain inflammatory and infectious disease in humans, including inflammatory bowel disease and tuberculosis. Collectively, these data highlight the importance of cis-acting lncRNAs in TLR signaling, innate immunity, and pathophysiological inflammation.


Subject(s)
Gene Expression Regulation , Immunity, Innate/immunology , Inflammation/immunology , Macrophages/immunology , RNA, Long Noncoding/metabolism , Streptococcal Infections/microbiology , Toll-Like Receptors/metabolism , Cells, Cultured , Cytokines/metabolism , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , Genome, Human , Histone Deacetylase 1/genetics , Histone Deacetylase 1/metabolism , Humans , Inflammation/genetics , Inflammation/microbiology , Macrophages/metabolism , Macrophages/microbiology , Myristoylated Alanine-Rich C Kinase Substrate/genetics , Myristoylated Alanine-Rich C Kinase Substrate/metabolism , Promoter Regions, Genetic , RNA, Long Noncoding/genetics , Streptococcal Infections/immunology , Streptococcus agalactiae/isolation & purification , Toll-Like Receptors/genetics
18.
J Innate Immun ; 11(1): 86-98, 2019.
Article in English | MEDLINE | ID: mdl-30391945

ABSTRACT

Trichomonas vaginalis is a sexually transmitted, eukaryotic parasite that causes trichomoniasis, the most common nonviral, sexually transmitted disease in the USA and worldwide. Little is known about the molecular mechanisms involved in the host immune response to this widespread parasite. Here we report that T. vaginalis induces NLRP3 inflammasome activation in human macrophages, leading to caspase-1 activation and the processing of pro-IL-1ß to the mature and bioactive form of the cytokine. Using inhibitor-based approaches, we show that NLRP3 activation by T. vaginalis involves host cell detection of extracellular ATP via P2X7 receptors and potassium efflux. In addition, our data reveal that T. vaginalis inflammasome activation induces macrophage inflammatory cell death by pyroptosis, known to occur via caspase-1 cleavage of the gasdermin D protein, which assembles to form pores in the host cell membrane. We found that T. vaginalis-induced cytolysis of macrophages is attenuated in gasdermin D knockout cells. Lastly, in a murine challenge model, we detected IL-1ß production in vaginal fluids in response to T. vaginalis infection in vivo. Together, our findings mechanistically dissect how T. vaginalis contributes to the production of the proinflammatory IL-1ß cytokine and uncover pyroptosis as a mechanism by which the parasite can trigger host macrophage cell death.


Subject(s)
Inflammasomes , Intracellular Signaling Peptides and Proteins/metabolism , Macrophages/metabolism , Macrophages/parasitology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Phosphate-Binding Proteins/metabolism , Pyroptosis , Trichomonas vaginalis , Animals , Caspase 1/metabolism , Humans , Interleukin-1beta/metabolism , Macrophages/pathology , Mice , THP-1 Cells
19.
Proc Natl Acad Sci U S A ; 115(41): 10410-10415, 2018 10 09.
Article in English | MEDLINE | ID: mdl-30254166

ABSTRACT

Natural killer (NK) cells are innate immune lymphocytes that recognize and destroy abnormal host cells, such as tumor cells or those infected by viral pathogens. To safely accomplish these functions, NK cells display activating receptors that detect stress molecules or viral ligands displayed at the cell surface, balanced by inhibitory receptors that bind to self-molecules. To date, such activating and inhibitory receptors on NK cells are not known to recognize bacterial determinants. Moreover, NK cell responses to direct interactions with extracellular bacteria are poorly explored. In this study, we observed the human neonatal pathogen group B Streptococcus (GBS) can directly engage human NK cells. The interaction was mediated through the B6N segment of streptococcal ß-protein, binding to the inhibitory receptor Siglec-7 via its amino-terminal V-set domain. Unlike classical Siglec binding, the interaction is also independent of its sialic acid recognition property. In contrast to WT GBS, mutants lacking ß-protein induced efficient pyroptosis of NK cells through the NLRP3 inflammasome, with production and secretion of the proinflammatory cytokine IL-1ß and dissemination of the cytotoxic molecule granzyme B. We postulate that GBS evolved ß-protein engagement of inhibitory human Siglec-7 to suppress the pyroptotic response of NK cells and thereby block recruitment of a broader innate immune response, i.e., by "silencing the sentinel."


Subject(s)
Antigens, Differentiation, Myelomonocytic/metabolism , DNA-Binding Proteins/metabolism , Immunity, Innate/immunology , Inflammation Mediators/metabolism , Killer Cells, Natural/pathology , Lectins/metabolism , Pyroptosis , Antigens, Differentiation, Myelomonocytic/genetics , Cells, Cultured , DNA-Binding Proteins/genetics , Humans , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Lectins/genetics
20.
Front Microbiol ; 9: 262, 2018.
Article in English | MEDLINE | ID: mdl-29515544

ABSTRACT

Staphylococcus aureus produces membrane-derived vesicles (MVs), which share functional properties to outer membrane vesicles. Atomic force microscopy revealed that S. aureus-derived MVs are associated with the bacterial surface or released into the surrounding environment depending on bacterial growth conditions. By using a comparative proteomic approach, a total of 131 and 617 proteins were identified in MVs isolated from S. aureus grown in Luria-Bertani and brain-heart infusion broth, respectively. Purified S. aureus MVs derived from the bacteria grown in either media induced comparable levels of cytotoxicity and neutrophil-activation. Administration of exogenous MVs increased the resistance of S. aureus to killing by whole blood or purified human neutrophils ex vivo and increased S. aureus survival in vivo. Finally, immunization of mice with S. aureus-derived MVs induced production of IgM, total IgG, IgG1, IgG2a, and IgG2b resulting in protection against subcutaneous and systemic S. aureus infection. Collectively, our results suggest S. aureus MVs can influence bacterial-host interactions during systemic infections and provide protective immunity in murine models of infection.

SELECTION OF CITATIONS
SEARCH DETAIL
...