Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
1.
Nature ; 575(7781): 224-228, 2019 11.
Article in English | MEDLINE | ID: mdl-31666699

ABSTRACT

The human gastrointestinal tract consists of a dense and diverse microbial community, the composition of which is intimately linked to health. Extrinsic factors such as diet and host immunity are insufficient to explain the constituents of this community, and direct interactions between co-resident microorganisms have been implicated as important drivers of microbiome composition. The genomes of bacteria derived from the gut microbiome contain several pathways that mediate contact-dependent interbacterial antagonism1-3. Many members of the Gram-negative order Bacteroidales encode the type VI secretion system (T6SS), which facilitates the delivery of toxic effector proteins into adjacent cells4,5. Here we report the occurrence of acquired interbacterial defence (AID) gene clusters in Bacteroidales species that reside within the human gut microbiome. These clusters encode arrays of immunity genes that protect against T6SS-mediated intra- and inter-species bacterial antagonism. Moreover, the clusters reside on mobile elements, and we show that their transfer is sufficient to confer resistance to toxins in vitro and in gnotobiotic mice. Finally, we identify and validate the protective capability of a recombinase-associated AID subtype (rAID-1) that is present broadly in Bacteroidales genomes. These rAID-1 gene clusters have a structure suggestive of active gene acquisition and include predicted immunity factors of toxins derived from diverse organisms. Our data suggest that neutralization of contact-dependent interbacterial antagonism by AID systems helps to shape human gut microbiome ecology.


Subject(s)
Bacteroidetes , Gastrointestinal Microbiome , Gastrointestinal Tract/microbiology , Microbial Interactions , Type VI Secretion Systems/antagonists & inhibitors , Animals , Bacteroidetes/genetics , Bacteroidetes/immunology , Female , Gastrointestinal Microbiome/immunology , Gastrointestinal Tract/immunology , Genes, Bacterial/genetics , Humans , Mice , Microbial Interactions/genetics , Microbial Interactions/immunology , Multigene Family/genetics , Type VI Secretion Systems/genetics , Type VI Secretion Systems/immunology
2.
Nature ; 548(7665): 43-51, 2017 08 02.
Article in English | MEDLINE | ID: mdl-28770836

ABSTRACT

The human body carries vast communities of microbes that provide many benefits. Our microbiome is complex and challenging to understand, but evolutionary theory provides a universal framework with which to analyse its biology and health impacts. Here we argue that to understand a given microbiome feature, such as colonization resistance, host nutrition or immune development, we must consider how hosts and symbionts evolve. Symbionts commonly evolve to compete within the host ecosystem, while hosts evolve to keep the ecosystem on a leash. We suggest that the health benefits of the microbiome should be understood, and studied, as an interplay between microbial competition and host control.


Subject(s)
Biological Evolution , Ecosystem , Microbial Interactions/physiology , Microbiota/physiology , Models, Biological , Symbiosis/physiology , Animals , Biodiversity , Health , Humans , Microbial Interactions/drug effects , Microbial Interactions/immunology , Microbiota/drug effects , Microbiota/immunology , Species Specificity , Symbiosis/drug effects , Symbiosis/immunology
3.
Infect Immun ; 89(9): e0066520, 2021 08 16.
Article in English | MEDLINE | ID: mdl-33526567

ABSTRACT

Immunotherapy has become a new paradigm in oncology, improving outcomes for several types of cancer. However, there are some aspects about its management that remain uncertain. One of the key points that needs better understanding is the interaction between immunotherapy and gut microbiome and how modulation of the microbiome might modify the efficacy of immunotherapy. Consequently, the negative impact of systemic antibiotics and corticosteroids on the efficacy of immunotherapy needs to be clarified.


Subject(s)
Adrenal Cortex Hormones/pharmacology , Anti-Bacterial Agents/pharmacology , Host Microbial Interactions , Immune Checkpoint Inhibitors/therapeutic use , Microbiota , Neoplasms/drug therapy , Probiotics , Adrenal Cortex Hormones/therapeutic use , Animals , Anti-Bacterial Agents/therapeutic use , Host Microbial Interactions/drug effects , Host Microbial Interactions/immunology , Humans , Immune Checkpoint Inhibitors/pharmacology , Immunomodulation/drug effects , Microbial Interactions/drug effects , Microbial Interactions/immunology , Microbiota/drug effects , Neoplasms/etiology , Treatment Outcome
4.
Int Arch Allergy Immunol ; 182(9): 807-818, 2021.
Article in English | MEDLINE | ID: mdl-33882482

ABSTRACT

Since conventional allergy medication for asthma or allergic rhinitis (AR) can cause side effects which limit the patients' quality of life, it is of interest to find other forms of therapy. In particular, probiotic bacteria, such as Lactobacillus species, have shown anti-allergic effects in various mouse and human studies. For instance, administration of some Lactobacillus species resulted in nasal and ocular symptom relief and improvement of quality of life in children and adults suffering from rhinitis. Different changes in cytokine profiles, such as elevated Th1 and decreased Th2 cytokines, reduced allergy-related immunoglobulins and cell immigration have been found in both human and murine studies. Positive effects on patients like less activity limitations or fewer rhinitis episodes and longer periods free from asthma or rhinitis were also described following oral administration of Lactobacillus bacteria. However, it is still unclear how this type of lactic acid bacteria leads to changes in the immune system and thus inhibits the development of allergies or relieves their symptoms. This review gives an overview of current studies and draws conclusions concerning the usage of probiotic Lactobacillus strains in AR.


Subject(s)
Host-Pathogen Interactions , Lactobacillus/immunology , Microbial Interactions , Probiotics , Rhinitis, Allergic/etiology , Animals , Dose-Response Relationship, Immunologic , Humans , Immunomodulation , Immunotherapy , Microbial Interactions/immunology , Probiotics/administration & dosage , Rhinitis, Allergic/diagnosis , Rhinitis, Allergic/therapy , Severity of Illness Index , Symptom Assessment
5.
J Infect Dis ; 220(8): 1367-1376, 2019 09 13.
Article in English | MEDLINE | ID: mdl-31233130

ABSTRACT

BACKGROUND: Pneumococci and nontypeable Haemophilus influenzae (NTHi) often cocolonize children. The impact of species interactions on disease risk across the upper respiratory mucosa is not known. METHODS: We analyzed data from 4104 acute conjunctivitis (AC) cases, 11 767 otitis media (OM) cases, and 1587 nasopharyngeal specimens collected from Israeli children before pneumococcal conjugate vaccine introduction. We compared pneumococcal serotype distributions with NTHi present and absent, and compared single-species and mixed-species rates of serotype-specific progression from colonization to AC and OM. RESULTS: Pneumococcal serotypes causing single-species OM (NTHi absent) were less diverse than colonizing serotypes and also less diverse than those causing mixed-species OM; colonizing and OM-causing pneumococcal serotype distributions were more similar to each other with NTHi present than with NTHi absent. In contrast, serotype diversity did not differ appreciably between colonizing and AC-causing pneumococci, regardless of NTHi co-occurrence. The similarity of colonizing and AC-causing pneumococcal serotype distributions was consistent in the presence and absence of NTHi. Differences in rates that pneumococcal serotypes progressed from colonization to disease were reduced in both AC and OM when NTHi was present. CONCLUSIONS: Interactions with NTHi may alter progression of pneumococcal serotypes to diseases of the upper respiratory mucosa in a site-specific manner.


Subject(s)
Conjunctivitis, Bacterial/immunology , Haemophilus influenzae/immunology , Microbial Interactions/immunology , Otitis Media/immunology , Pneumococcal Infections/immunology , Streptococcus pneumoniae/immunology , Carrier State/epidemiology , Carrier State/immunology , Carrier State/microbiology , Child , Conjunctivitis, Bacterial/epidemiology , Conjunctivitis, Bacterial/microbiology , Conjunctivitis, Bacterial/prevention & control , Disease Progression , Epidemiological Monitoring , Haemophilus influenzae/isolation & purification , Humans , Israel/epidemiology , Nasopharynx/immunology , Otitis Media/epidemiology , Otitis Media/microbiology , Otitis Media/prevention & control , Pneumococcal Infections/epidemiology , Pneumococcal Infections/microbiology , Pneumococcal Infections/prevention & control , Pneumococcal Vaccines/administration & dosage , Prevalence , Prospective Studies , Respiratory Mucosa/immunology , Respiratory Mucosa/microbiology , Streptococcus pneumoniae/isolation & purification
6.
J Immunol ; 198(2): 557-563, 2017 01 15.
Article in English | MEDLINE | ID: mdl-28069750

ABSTRACT

The existence of a neonatal window was first highlighted by epidemiological studies that revealed the particular importance of this early time in life for the susceptibility to immune-mediated diseases in humans. Recently, the first animal studies emerged that present examples of early-life exposure-triggered persisting immune events, allowing a detailed analysis of the factors that define this particular time period. The enteric microbiota and the innate and adaptive immune system represent prime candidates that impact on the pathogenesis of immune-mediated diseases and are known to reach a lasting homeostatic equilibrium following a dynamic priming period after birth. In this review, we outline the postnatal establishment of the microbiota and maturation of the innate and adaptive immune system and discuss examples of early-life exposure-triggered immune-mediated diseases that start to shed light on the critical importance of the early postnatal period for life-long immune homeostasis.


Subject(s)
Homeostasis/immunology , Microbiota/immunology , Animals , Disease Susceptibility/immunology , Humans , Microbial Interactions/immunology
7.
Infect Immun ; 85(11)2017 11.
Article in English | MEDLINE | ID: mdl-28808158

ABSTRACT

The Campylobacter jejuni-host interaction may be affected by the host's gut microbiota through competitive exclusion, metabolites, or modification of the immune response. To understand this interaction, C. jejuni colonization and local immune responses were compared in chickens with different gut microbiota compositions. Birds were treated with an antibiotic cocktail (AT) (experiments 1 and 2) or raised under germfree (GF) conditions (experiment 3). At 18 days posthatch (dph), they were orally inoculated either with 104 CFU of C. jejuni or with diluent. Cecal as well as systemic C. jejuni colonization, T- and B-cell numbers in the gut, and gut-associated tissue were compared between the different groups. Significantly higher numbers of CFU of C. jejuni were detected in the cecal contents of AT and GF birds, with higher colonization rates in spleen, liver, and ileum, than in birds with a conventional gut microbiota (P < 0.05). Significant upregulation of T and B lymphocyte numbers was detected in cecum, cecal tonsils, and bursa of Fabricius of AT or GF birds after C. jejuni inoculation compared to the respective controls (P < 0.05). This difference was less clear in birds with a conventional gut microbiota. Histopathological gut lesions were observed only in C. jejuni-inoculated AT and GF birds but not in microbiota-colonized C. jejuni-inoculated hatchmates. These results demonstrate that the gut microbiota may contribute to the control of C. jejuni colonization and prevent lesion development. Further studies are needed to identify key players of the gut microbiota and the mechanisms behind their protective role.


Subject(s)
Campylobacter Infections/veterinary , Campylobacter jejuni/immunology , Gastrointestinal Microbiome/immunology , Host-Pathogen Interactions/immunology , Microbial Interactions/immunology , Poultry Diseases/immunology , Animals , Anti-Bacterial Agents/pharmacology , B-Lymphocytes/immunology , B-Lymphocytes/microbiology , Bursa of Fabricius/drug effects , Bursa of Fabricius/immunology , Bursa of Fabricius/microbiology , Campylobacter Infections/immunology , Campylobacter Infections/microbiology , Campylobacter jejuni/drug effects , Campylobacter jejuni/pathogenicity , Cecum/drug effects , Cecum/immunology , Cecum/microbiology , Chickens , Colony Count, Microbial , Germ-Free Life/immunology , Ileum/drug effects , Ileum/immunology , Ileum/microbiology , Liver/drug effects , Liver/immunology , Liver/microbiology , Poultry Diseases/microbiology , Spleen/drug effects , Spleen/immunology , Spleen/microbiology , T-Lymphocytes/immunology , T-Lymphocytes/microbiology
8.
Fish Shellfish Immunol ; 52: 198-205, 2016 May.
Article in English | MEDLINE | ID: mdl-26997202

ABSTRACT

The present study evaluated the benefits of dietary administration of host-derived candidate probiotics Enterococcus casseliflavus in juvenile rainbow trout Oncorhynchus mykiss. Experimental diets were prepared by incorporating the microorganisms in the basal feed at 3 inclusion levels (i.e. 10(7) CFU g(-1) of feed [T1], 10(8) CFU g(-1) of feed [T2], 10(9) CFU g(-1) of feed [T3]). The probiotic feeds were administered for 8 weeks, with a group fed with the basal diet serving as control. The effects on growth performance, gut health, innate immunity and disease resistance were evaluated. Results showed that growth performance parameters were significantly improved in T2 and T3 groups. Activities of digestive enzymes such as trypsin and lipase were significantly higher in these two groups as well. Gut micro-ecology was influenced by probiotic feeding as shown by the significant increase in intestinal lactic acid bacteria and total viable aerobic counts in T2 and T3. Humoral immunity was impacted by dietary probiotics as total serum protein and albumin were significantly elevated in T3. The levels of serum IgM significantly increased in all probiotic fed groups at week 8; with the T3 group registering the highest increment. Respiratory burst activity of blood leukocytes were significantly improved in T2 and T3. Hematological profiling further revealed that neutrophil counts significantly increased in all probiotic fed groups. Challenge test showed that probiotic feeding significantly improved host resistance to Streptococcus iniae infection, specifically in T2 and T3 where a considerable modulation of immune responses was observed. Taken together, this study demonstrated E. casseliflavus as a potential probiotics for rainbow trout with the capability of improving growth performance and enhancing disease resistance by immunomodulation.


Subject(s)
Enterococcus/physiology , Fish Diseases , Immunomodulation/immunology , Oncorhynchus mykiss , Probiotics/metabolism , Streptococcal Infections/veterinary , Streptococcus iniae/physiology , Administration, Oral , Animal Feed/microbiology , Animals , Aquaculture , Diet/veterinary , Digestive System/microbiology , Enterococcus/immunology , Fish Diseases/immunology , Fish Diseases/microbiology , Gastrointestinal Microbiome , Immunity, Innate , Microbial Interactions/immunology , Oncorhynchus mykiss/immunology , Oncorhynchus mykiss/microbiology , Streptococcal Infections/immunology , Streptococcal Infections/microbiology
9.
Proc Natl Acad Sci U S A ; 110(7): 2623-8, 2013 Feb 12.
Article in English | MEDLINE | ID: mdl-23362380

ABSTRACT

Type VI protein secretion system (T6SS) is important for bacterial competition through contact-dependent killing of competitors. T6SS delivers effectors to neighboring cells and corresponding antagonistic proteins confer immunity against effectors that are delivered by sister cells. Although T6SS has been found in more than 100 gram-negative bacteria including many important human pathogens, few T6SS-dependent effector and immunity proteins have been experimentally determined. Here we report a high-throughput approach using transposon mutagenesis and deep sequencing (Tn-seq) to identify T6SS immunity proteins in Vibrio cholerae. Saturating transposon mutagenesis was performed in wild type and a T6SS null mutant. Genes encoding immunity proteins were predicted to be essential in the wild type but dispensable in the T6SS mutant. By comparing the relative abundance of each transposon mutant in the mutant library using deep sequencing, we identified three immunity proteins that render protection against killing by T6SS predatory cells. We also identified their three cognate T6SS-secreted effectors and show these are important for not only antibacterial and antieukaryotic activities but also assembly of T6SS apparatus. The lipase and muramidase T6SS effectors identified in this study underscore the diversity of T6SS-secreted substrates and the distinctly different mechanisms that target these for secretion by the dynamic T6SS organelle.


Subject(s)
Bacterial Proteins/genetics , Bacterial Secretion Systems/genetics , Microbial Interactions/genetics , Vibrio cholerae/genetics , Actins/metabolism , Bacterial Proteins/immunology , Bacterial Secretion Systems/immunology , Blotting, Western , DNA Transposable Elements/genetics , Dictyostelium/growth & development , Dictyostelium/microbiology , Genetic Vectors/genetics , High-Throughput Nucleotide Sequencing , Immunoprecipitation , Lipase/genetics , Microbial Interactions/immunology , Muramidase/genetics , Mutagenesis, Site-Directed , Plasmids/genetics , Stem Cells , Vibrio cholerae/immunology
10.
J Infect Dis ; 211(8): 1296-305, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25378635

ABSTRACT

BACKGROUND: Acinetobacter baumannii is one of the most antibiotic-resistant pathogens. Defining mechanisms driving pathogenesis is critical to enable new therapeutic approaches. METHODS: We studied virulence differences across a diverse panel of A. baumannii clinical isolates during murine bacteremia to elucidate host-microbe interactions that drive outcome. RESULTS: We identified hypervirulent strains that were lethal at low intravenous inocula and achieved very high early, and persistent, blood bacterial densities. Virulent strains were nonlethal at low inocula but lethal at 2.5-fold higher inocula. Finally, relatively avirulent (hypovirulent) strains were nonlethal at 20-fold higher inocula and were efficiently cleared by early time points. In vivo virulence correlated with in vitro resistance to complement and macrophage uptake. Depletion of complement, macrophages, and neutrophils each independently increased bacterial density of the hypovirulent strain but insufficiently to change lethality. However, disruption of all 3 effector mechanisms enabled early bacterial densities similar to hypervirulent strains, rendering infection 100% fatal. CONCLUSIONS: The lethality of A. baumannii strains depends on distinct stages. Strains resistant to early innate effectors are able to establish very high early bacterial blood density, and subsequent sustained bacteremia leads to Toll-like receptor 4-mediated hyperinflammation and lethality. These results have important implications for translational efforts to develop therapies that modulate host-microbe interactions.


Subject(s)
Acinetobacter Infections/immunology , Acinetobacter baumannii/immunology , Bacteremia/immunology , Immunity, Innate/immunology , Microbial Interactions/immunology , Acinetobacter Infections/microbiology , Animals , Anti-Bacterial Agents/immunology , Bacteremia/microbiology , Drug Resistance, Multiple, Bacterial/immunology , Macrophages/immunology , Macrophages/microbiology , Mice , Mice, Inbred C3H , Neutrophils/immunology , Neutrophils/microbiology , Virulence/immunology , Virulence Factors/immunology
11.
Proc Biol Sci ; 280(1771): 20131939, 2013 Nov 22.
Article in English | MEDLINE | ID: mdl-24089337

ABSTRACT

More than 90 capsular serotypes of Streptococcus pneumoniae coexist despite competing for nasopharyngeal carriage and a gradient in fitness. The underlying mechanisms for this are poorly understood and make assessment of the likely population impact of vaccination challenging. We use an individual-based simulation model to generalize widely used deterministic models for pneumococcal competition and show that in these models short-term serotype-specific and serotype non-specific immunity could constitute the mechanism governing between-host competition and coexistence. We find that non-specific immunity induces between-host competition and that serotype-specific immunity limits a type's competitive advantage and allows stable coexistence of multiple serotypes. Serotypes carried at low prevalence show high variance in carriage levels, which would result in apparent outbreaks if they were highly pathogenic. Vaccination against few serotypes can lead to elimination of the vaccine types and induces replacement by others. However, in simulations where the elimination of the targeted types is achieved only by a combination of vaccine effects and the competitive pressure of the non-vaccine types, a universal vaccine with similar-type-specific effectiveness can fail to eliminate pneumococcal carriage and offers limited herd immunity. Hence, if vaccine effects are insufficient to control the majority of serotypes at the same time, then exploiting the competitive pressure by selective vaccination can help control the most pathogenic serotypes.


Subject(s)
Microbial Interactions/immunology , Models, Biological , Nasopharynx/microbiology , Pneumococcal Infections/transmission , Streptococcus pneumoniae/immunology , Vaccination/methods , Age Factors , Computer Simulation , Humans , Species Specificity , Streptococcus pneumoniae/classification , Streptococcus pneumoniae/pathogenicity , United Kingdom
13.
Periodontol 2000 ; 62(1): 163-202, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23574466

ABSTRACT

During the last two to three decades our understanding of the immunobiology of periodontal disease has increased exponentially, both with respect to the microbial agents triggering the disease process and the molecular mechanisms of the host engagement maintaining homeostasis or leading to collateral tissue damage. These foundational scientific findings have laid the groundwork for translating cell phenotype, receptor engagement, intracellular signaling pathways and effector functions into a 'picture' of the periodontium as the host responds to the 'danger signals' of the microbial ecology to maintain homeostasis or succumb to a disease process. These findings implicate the chronicity of the local response in attempting to manage the microbial challenge, creating a 'Double Indemnity' in some patients that does not 'insure' health for the periodontium. As importantly, in reflecting the title of this volume of Periodontology 2000, this review attempts to inform the community of how the science of periodontal immunology gestated, how continual probing of the biology of the disease has led to an evolution in our knowledge base and how more recent studies in the postgenomic era are revolutionizing our understanding of disease initiation, progression and resolution. Thus, there has been substantial progress in our understanding of the molecular mechanisms of host-bacteria interactions that result in the clinical presentation and outcomes of destructive periodontitis. The science has embarked from observations of variations in responses related to disease expression with a focus for utilization of the responses in diagnosis and therapeutic outcomes, to current investigations using cutting-edge fundamental biological processes to attempt to model the initiation and progression of soft- and hard-tissue destruction of the periodontium. As importantly, the next era in the immunobiology of periodontal disease will need to engage more sophisticated experimental designs for clinical studies to enable robust translation of basic biologic processes that are in action early in the transition from health to disease, those which stimulate microenvironmental changes that select for a more pathogenic microbial ecology and those that represent a rebalancing of the complex host responses and a resolution of inflammatory tissue destruction.


Subject(s)
Host-Pathogen Interactions/immunology , Periodontal Diseases/immunology , Adaptive Immunity/immunology , Biofilms , Cytokines/immunology , Humans , Immunity, Innate/immunology , Immunity, Mucosal/immunology , Microbial Interactions/immunology , Microbiota/immunology , Periodontal Diseases/microbiology
14.
J Clin Periodontol ; 39(10): 913-22, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22860513

ABSTRACT

BACKGROUND: Human beta-defensins (hBDs) and the C-C chemokine ligand 20 (CCL20) produced by gingival epithelial cells (GECs) and fibroblasts (HGFs) are antimicrobial peptides (AMPs) that play an important role in innate immunity. The aim of this study was to determine the differential immune response of GECs and HGFs to the oral commensal Streptococcus gordonii (SG) and the pathogen Porphyromonas gingivalis (PG). MATERIAL AND METHODS: In addition to the analysis of gingival biopsies, primary GECs and HGFs were exposed to SG and/or PG, and expression of various AMPs and pro-inflammatory mediators was studied by real-time PCR and ELISA. RESULTS: Gene expression of AMPs was detected in gingival connective tissue. Both SG and PG induced the mRNA-expression of hBD-2 and hBD-3 in GECs as well as HGFs after 24 h (p < 0.05). In HGFs, the commensal bacterium SG stimulated the mRNAs of hBD-3 and CCL20 after 24 h (p < 0.05), while not in GECs. In GECs, the inductive effect of PG on the mRNA-expression of hBD-2 was amplified when cells were first exposed to commensal SG (for 1 h) prior to stimulation with PG (SG-PG; p < 0.05). CONCLUSION: Our data indicate that cell-bacteria interactions and/or bacteria-bacteria cross-talk may have an impact on AMP-regulation in gingiva.


Subject(s)
Chronic Periodontitis/metabolism , Epithelial Cells/metabolism , Fibroblasts/metabolism , Porphyromonas gingivalis/immunology , beta-Defensins/metabolism , Adult , Case-Control Studies , Chemokine CCL20/metabolism , Chronic Periodontitis/immunology , Chronic Periodontitis/microbiology , Chronic Periodontitis/pathology , Epithelial Cells/immunology , Fibroblasts/immunology , Gingiva/cytology , Gingiva/immunology , Gingiva/microbiology , Humans , Microbial Interactions/immunology , Middle Aged , RNA, Messenger/analysis , Reference Values , Streptococcus gordonii/immunology , beta-Defensins/genetics
15.
Gastroenterol Hepatol ; 34(3): 147-54, 2011 Mar.
Article in Spanish | MEDLINE | ID: mdl-21377761

ABSTRACT

The intestine hosts a complex ecosystem of microbial communities. Experimental data suggests that the microbiota has metabolic functions that contribute to nutrient and energy recovery from non-digestible substrates. Moreover, microbial colonization is essential for the normal development of the immune system and therefore seems to influence homeostasis between environmental antigen load and immune response. In genetically-susceptible individuals, an imbalance may give rise to diseases of immune dysregulation, including chronic inflammatory bowel diseases, in which there is an exaggerated immune response to harmless microbial antigens. Despite the availability of new molecular technologies, the normal composition of the human intestinal microbiota remains unknown. In the next few years, the results of international projects designed to determine the precise impact of the microbiota in various physiological and pathological processes will hopefully lead to major advances.


Subject(s)
Inflammatory Bowel Diseases/microbiology , Intestines/microbiology , Metagenome/physiology , Microbial Consortia/physiology , Microbial Interactions/physiology , Animals , Antigens, Bacterial/immunology , Genetic Predisposition to Disease , Germ-Free Life , Homeostasis/immunology , Humans , Immune Tolerance , Immunity, Mucosal/immunology , Inflammatory Bowel Diseases/etiology , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/therapy , Intestines/immunology , Metagenome/genetics , Metagenome/immunology , Microbial Consortia/immunology , Microbial Interactions/immunology , RNA, Ribosomal, 16S/genetics , Species Specificity , Symbiosis , T-Lymphocytes, Regulatory/immunology
16.
Front Immunol ; 12: 819560, 2021.
Article in English | MEDLINE | ID: mdl-35140712

ABSTRACT

Filarial helminths infect approximately 120 million people worldwide initiating a type 2 immune response in the host. Influenza A viruses stimulate a virulent type 1 pro-inflammatory immune response that in some individuals can cause uncontrolled immunopathology and fatality. Although coinfection with filariasis and influenza is a common occurrence, the impact of filarial infection on respiratory viral infection is unknown. The aim of this study was to determine the impact of pre-existing filarial infection on concurrent infection with influenza A virus. A murine model of co-infection was established using the filarial helminth Litomosoides sigmodontis and the H1N1 (A/WSN/33) influenza A virus (IAV). Co-infection was performed at 3 different stages of L. sigmodontis infection (larval, juvenile adult, and patency), and the impact of co-infection was determined by IAV induced weight loss and clinical signs, quantification of viral titres, and helminth counts. Significant alterations of IAV pathogenesis, dependent upon stage of infection, was observed on co-infection with L. sigmodontis. Larval stage L. sigmodontis infection alleviated clinical signs of IAV co-infection, whilst more established juvenile adult infection also significantly delayed weight loss. Viral titres remained unaltered at either infection stage. In contrast, patent L. sigmdodontis infection led to a reversal of age-related resistance to IAV infection, significantly increasing weight loss and clinical signs of infection as well as increasing IAV titre. These data demonstrate that the progression of influenza infection can be ameliorated or worsened by pre-existing filarial infection, with the outcome dependent upon the stage of filarial infection.


Subject(s)
Coinfection , Filariasis/immunology , Filariasis/parasitology , Filarioidea , Influenza A virus , Microbial Interactions , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Animals , Cell Line , Disease Models, Animal , Disease Susceptibility , Female , Mice , Microbial Interactions/immunology
17.
Curr Opin Immunol ; 72: 324-330, 2021 10.
Article in English | MEDLINE | ID: mdl-34537476

ABSTRACT

The skin represents the largest area for direct contact between microbes and host immunocytes and is a site for constant communication between the host and this diverse and essential microbial community. Coagulase-negative staphylococci are an abundant bacterial genus on the human skin and are regulated through various mechanisms that include the epidermal barrier environment and innate and adaptive immune systems within the epidermis and dermis. In turn, some species and strains of these bacteria produce beneficial products that augment host immunity by exerting specifically targeted antimicrobial, anti-inflammatory, or anti-neoplastic activity while also promoting broad innate and adaptive immune responses. The use of selected skin commensals as a therapeutic has shown promise in recent human clinical trials. This emerging concept of bacteriotherapy is defining mechanisms of action and validating the dependence on the microbiome for maintenance of immune homeostasis.


Subject(s)
Host Microbial Interactions/immunology , Microbiota/immunology , Skin Physiological Phenomena , Skin/immunology , Skin/microbiology , Animals , Biomarkers , Disease Management , Disease Susceptibility , Dysbiosis , Humans , Immunomodulation , Microbial Interactions/immunology
18.
Front Immunol ; 12: 828887, 2021.
Article in English | MEDLINE | ID: mdl-35154087

ABSTRACT

Objective: Probiotics can modulate immune responses to resist influenza infection. This study aims to evaluate the anti-viral efficacy of B. dorei. Methods: C57BL/6J mice were infected with influenza virus together with treatment of PBS vehicle, B. dorei, or oseltamivir respectively. Anti-influenza potency of B. dorei and the underlying mechanism were determined by measuring survival rate, lung viral load and pathology, gene expression and production of cytokines and chemokines, and analysis of gut microbiota. Results: Administration of B. dorei increased (by 30%) the survival of influenza-infected mice, and improved their weight loss, lung pathology, lung index, and colon length compared to the vehicle control group. B. dorei treatment reduced (by 61%) the viral load of lung tissue and increased expression of type 1 interferon more rapidly at day 3 postinfection. At day 7 postinfection, B. dorei-treated mice showed lower local (lung) and systemic (serum) levels of interferon and several proinflammatory cytokines or chemokines (IL-1ß, IL-6, TNF-α, IL-10, MCP-1 and IP-10) with a efficacy comparable to oseltamivi treatment. B. dorei treatment also altered gut microbiota as indicated by increased levels of Bacteroides, Prevotella, and Lactobacillus and decreased levels of Escherichia, Shigella, and Parabacteroides. Conclusion: B. dorei has anti-influenza effect. Its working mechanisms involve promoting earlier interferon expression and down-regulating both local and systemic inflammatory response. B. dorei changes the composition of gut microbiota, which may also contribute to its beneficial effects.


Subject(s)
Bacteroides/immunology , Host-Pathogen Interactions/immunology , Influenza A virus/physiology , Microbial Interactions/immunology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Animals , Biomarkers , Brain-Gut Axis/immunology , Cytokines/blood , Cytokines/metabolism , Disease Models, Animal , Female , Gastrointestinal Microbiome/immunology , Immunomodulation , Interferons/metabolism , Metagenome , Metagenomics/methods , Mice , Orthomyxoviridae Infections/metabolism , Orthomyxoviridae Infections/pathology , Probiotics , Quercetin/metabolism , Viral Load
19.
Cell Host Microbe ; 29(3): 334-346, 2021 03 10.
Article in English | MEDLINE | ID: mdl-33705705

ABSTRACT

Immunoglobulin A (IgA) is the most abundant antibody at mucosal surfaces and has been the subject of many investigations involving microbiota research in the last decade. Although the classic functions of IgA include neutralization of harmful toxins, more recent investigations have highlighted an important role for IgA in regulating the composition and function of the commensal microbiota. Multiple reviews have comprehensively covered the literature that describes recent, novel mechanisms of action of IgA and development of the IgA response within the intestine. Here we focus on how the interaction between IgA and the microbiota promotes homeostasis with the host to prevent disease.


Subject(s)
Gastrointestinal Microbiome/immunology , Homeostasis , Host Microbial Interactions/immunology , Host Microbial Interactions/physiology , Animals , Bacteria/metabolism , Colorectal Neoplasms/immunology , Colorectal Neoplasms/microbiology , Gastrointestinal Microbiome/genetics , Gastrointestinal Microbiome/physiology , Gene Expression , Host Microbial Interactions/genetics , Humans , Immunoglobulin A/immunology , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/microbiology , Intestinal Mucosa/immunology , Intestines/microbiology , Metabolic Diseases/microbiology , Microbial Interactions/immunology , Microbial Interactions/physiology , Species Specificity , Symbiosis
20.
J Clin Periodontol ; 37(4): 313-23, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20447254

ABSTRACT

AIM: To examine relationships between subgingival biofilm composition and levels of gingival crevicular fluid (GCF) cytokines in periodontal health and generalized aggressive periodontitis (GAP). MATERIALS AND METHODS: Periodontal parameters were measured in 25 periodontally healthy and 31 GAP subjects. Subgingival plaque and GCF samples were obtained from 14 sites from each subject. Forty subgingival taxa were quantified using checkerboard DNA-DNA hybridization and the concentrations of eight GCF cytokines were measured using Luminex. Cluster analysis was used to define sites with similar subgingival microbiotas in each clinical group. Significance of differences in clinical, microbiological and immunological parameters among clusters was determined using the Kruskal-Wallis test. RESULTS: GAP subjects had statistically significantly higher GCF levels of interleukin-1beta (IL-1beta) (p<0.001), granulocyte-macrophage colony-stimulating factor (GM-CSF) (p<0.01) and IL-1beta/IL-10 ratio (p<0.001) and higher proportions of Red and Orange complex species than periodontally healthy subjects. There were no statistically significant differences in the mean proportion of cytokines among clusters in the periodontally healthy subjects, while the ratio IL-1beta/IL-10 (p<0.05) differed significantly among clusters in the aggressive periodontitis group. CONCLUSIONS: Different subgingival biofilm profiles are associated with distinct patterns of GCF cytokine expression. Aggressive periodontitis subjects were characterized by a higher IL-1beta/IL-10 ratio than periodontally healthy subjects, suggesting an imbalance between pro- and anti-inflammatory cytokines in aggressive periodontitis.


Subject(s)
Aggressive Periodontitis/immunology , Dental Plaque/microbiology , Gingival Crevicular Fluid/immunology , Interleukin-10/metabolism , Interleukin-1beta/metabolism , Microbial Interactions/immunology , Adult , Aggressive Periodontitis/metabolism , Aggressive Periodontitis/microbiology , Bacteria/classification , Bacteria/genetics , Biofilms , Biomarkers/analysis , Case-Control Studies , Cluster Analysis , DNA, Bacterial/analysis , Dental Plaque/immunology , Female , Gingival Crevicular Fluid/metabolism , Gingival Crevicular Fluid/microbiology , Granulocyte-Macrophage Colony-Stimulating Factor/analysis , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Interleukin-10/analysis , Interleukin-1beta/analysis , Male , Reference Values , Subgingival Curettage
SELECTION OF CITATIONS
SEARCH DETAIL