Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 223
Filter
1.
Front Immunol ; 12: 696403, 2021.
Article in English | MEDLINE | ID: mdl-34484193

ABSTRACT

Background: Infections are the major cause of morbidity and mortality in patients with primary immunodeficiency disease (PID). Timely and accurate microbiological diagnosis is particularly important in these patients. Metagenomic next-generation sequencing (mNGS) has been used for pathogen detection recently. However, few reports describe the use of mNGS for pathogen identification in patients with PID. Objective: To evaluate the utility of mNGS for detecting pathogens in patients with PID, and to compare it with conventional microbiological tests (CMT). Methods: This single center retrospective study investigated the diagnostic performance of mNGS for pathogens detection in PID patients and compared it with CMT. Sixteen PID patients with suspected infection were enrolled, and medical records were analyzed to extract detailed clinical characteristics such as gene variation, immune status, microbial distribution, time-consuming of mNGS and CMT, treatment, and outcomes. Results: mNGS identified pathogenic microbe in 93.75% samples, compared to 31.25% for culture and 68.75% for conventional methods, and detected an extra 18 pathogenic microorganisms including rare opportunistic pathogens and Mycobacterium tuberculosis. Pathogen identification by mNGS required 48 hours, compared with bacterial culture for 3-7 days and even longer for fungus and Mycobacterium tuberculosis culture. Conclusions: mNGS has marked advantages over conventional methods for pathogenic diagnosis, particularly opportunistic pathogens and mixed infections, in patients with PID. This method might enable clinicians to make more timely and targeted therapeutic decisions, thereby improving the prognosis of these patients.


Subject(s)
Bacterial Infections/diagnosis , High-Throughput Nucleotide Sequencing , Metagenome/genetics , Metagenomics , Mycoses/diagnosis , Opportunistic Infections/diagnosis , Primary Immunodeficiency Diseases/immunology , Adolescent , Bacterial Infections/genetics , Bacterial Infections/immunology , Bacterial Infections/microbiology , Bacteriological Techniques , Child , Child, Preschool , Female , Host-Pathogen Interactions , Humans , Immunocompromised Host , Infant , Male , Metagenome/immunology , Mycoses/genetics , Mycoses/immunology , Mycoses/microbiology , Opportunistic Infections/genetics , Opportunistic Infections/immunology , Opportunistic Infections/microbiology , Predictive Value of Tests , Primary Immunodeficiency Diseases/diagnosis , Primary Immunodeficiency Diseases/genetics , Reproducibility of Results , Retrospective Studies
2.
Front Immunol ; 12: 702378, 2021.
Article in English | MEDLINE | ID: mdl-34276696

ABSTRACT

The field of microbiome research has developed rapidly over the past decades and has become a topic of major interest to basic, preclinical, and clinical research, the pharmaceutical industry as well as the general public. The microbiome is a complex and diverse ecosystem and defined as the collection of all host-associated microorganisms and their genes. It is acquired through vertical transmission and environmental exposure and includes microbes of all kingdoms: bacteria, archaea, prokaryotic and eukaryotic viruses, fungi, protozoa, and the meiofauna. These microorganisms co-evolved with their respective hosts over millions of years, thereby establishing a mutually beneficial, symbiotic relationship on all epithelial barriers. Thus, the microbiome plays a pivotal role in virtually every aspect of mammalian physiology, particularly in the development, homeostasis, and function of the immune system. Consequently, the combination of the host genome and the microbial genome, together referred to as the metagenome, largely drives the mammalian phenotype. So far, the majority of studies have unilaterally focused on the gastrointestinal bacterial microbiota. However, recent work illustrating the impact of viruses, fungi, and protozoa on host immunity urges us towards a holistic view of the mammalian microbiome and the appreciation for its non-bacterial kingdoms. In addition, the importance of microbiota on epithelial barriers other than the gut as well as their systemic effects via microbially-derived biologically active compounds is increasingly recognized. Here, we want to provide a brief but comprehensive overview of the most important findings and the current knowledge on how microbes of all kingdoms and microbial niches shape local and systemic immunity in health and disease.


Subject(s)
Immune System , Metagenome/immunology , Microbiota/immunology , Animals , Humans
3.
Sci Rep ; 10(1): 20618, 2020 11 26.
Article in English | MEDLINE | ID: mdl-33244064

ABSTRACT

Despite being commonly used to collect upper airway epithelial lining fluid, nasal washes are poorly reproducible, not suitable for serial sampling, and limited by a dilution effect. In contrast, nasal filters lack these limitations and are an attractive alternative. To examine whether nasal filters are superior to nasal washes as a sampling method for the characterization of the upper airway microbiome and immune response, we collected paired nasal filters and washes from a group of 40 healthy children and adults. To characterize the upper airway microbiome, we used 16S ribosomal RNA and shotgun metagenomic sequencing. To characterize the immune response, we measured total protein using a BCA assay and 53 immune mediators using multiplex magnetic bead-based assays. We conducted statistical analyses to compare common microbial ecology indices and immune-mediator median fluorescence intensities (MFIs) between sample types. In general, nasal filters were more likely to pass quality control in both children and adults. There were no significant differences in microbiome community richness, α-diversity, or structure between pediatric samples types; however, these were all highly dissimilar between adult sample types. In addition, there were significant differences in the abundance of amplicon sequence variants between sample types in children and adults. In adults, total proteins were significantly higher in nasal filters than nasal washes; consequently, the immune-mediator MFIs were not well detected in nasal washes. Based on better quality control sequencing metrics and higher immunoassay sensitivity, our results suggest that nasal filters are a superior sampling method to characterize the upper airway microbiome and immune response in both children and adults.


Subject(s)
Microbiota/genetics , Microbiota/immunology , Nasal Lavage Fluid/immunology , Nasal Lavage Fluid/microbiology , Nose/immunology , Nose/microbiology , Adult , Child , Female , Humans , Immunity/genetics , Immunity/immunology , Male , Metagenome/genetics , Metagenome/immunology , Nasal Absorption/immunology , Nasal Cavity/immunology , Nasal Cavity/microbiology , RNA, Ribosomal, 16S/genetics , RNA, Ribosomal, 16S/immunology , Specimen Handling/methods
4.
J Crohns Colitis ; 14(11): 1600-1610, 2020 Nov 07.
Article in English | MEDLINE | ID: mdl-32406906

ABSTRACT

BACKGROUND AND AIMS: Dysbiosis of the gut microbiota is a well-known correlate of the pathogenesis of inflammatory bowel disease [IBD]. However, few studies have examined the microbiome in very early-onset [VEO] IBD, which is defined as onset of IBD before 6 years of age. Here we focus on the viral portion of the microbiome-the virome-to assess possible viral associations with disease processes, reasoning that any viruses potentially associated with IBD might grow more robustly in younger subjects, and so be more detectable. METHODS: Virus-like particles [VLPs] were purified from stool samples collected from patients with VEO-IBD [n = 54] and healthy controls [n = 23], and characterized by DNA and RNA sequencing and VLP particle counts. RESULTS: The total number of VLPs was not significantly different between VEO-IBD and healthy controls. For bacterial viruses, the VEO-IBD subjects were found to have a higher ratio of Caudovirales vs to Microviridae compared to healthy controls. An increase in Caudovirales was also associated with immunosuppressive therapy. For viruses infecting human cells, Anelloviridae showed higher prevalence in VEO-IBD compared to healthy controls. Within the VEO-IBD group, higher levels of Anelloviridae DNA were also positively associated with immunosuppressive treatment. To search for new viruses, short sequences enriched in VEO-IBD samples were identified, and some could be validated in an independent cohort, although none was clearly viral; this provides sequence tags to interrogate in future studies. CONCLUSIONS: These data thus document perturbations to normal viral populations associated with VEO-IBD, and provide a biomarker-Anelloviridae DNA levels-potentially useful for reporting the effectiveness of immunosuppression.


Subject(s)
Anelloviridae/isolation & purification , Feces/virology , Immunosuppressive Agents/therapeutic use , Inflammatory Bowel Diseases , Virome/physiology , Age of Onset , Biomarkers, Pharmacological/analysis , Child, Preschool , Correlation of Data , Female , Gastrointestinal Microbiome/physiology , Humans , Inflammatory Bowel Diseases/epidemiology , Inflammatory Bowel Diseases/physiopathology , Inflammatory Bowel Diseases/virology , Male , Metagenome/immunology , Risk Factors , United States/epidemiology , Viruses/classification , Viruses/isolation & purification
5.
Nat Med ; 25(12): 1873-1884, 2019 12.
Article in English | MEDLINE | ID: mdl-31806906

ABSTRACT

Herpes simplex virus-1 (HSV-1) encephalitis (HSE) is typically sporadic. Inborn errors of TLR3- and DBR1-mediated central nervous system cell-intrinsic immunity can account for forebrain and brainstem HSE, respectively. We report five unrelated patients with forebrain HSE, each heterozygous for one of four rare variants of SNORA31, encoding a small nucleolar RNA of the H/ACA class that are predicted to direct the isomerization of uridine residues to pseudouridine in small nuclear RNA and ribosomal RNA. We show that CRISPR/Cas9-introduced bi- and monoallelic SNORA31 deletions render human pluripotent stem cell (hPSC)-derived cortical neurons susceptible to HSV-1. Accordingly, SNORA31-mutated patient hPSC-derived cortical neurons are susceptible to HSV-1, like those from TLR3- or STAT1-deficient patients. Exogenous interferon (IFN)-ß renders SNORA31- and TLR3- but not STAT1-mutated neurons resistant to HSV-1. Finally, transcriptome analysis of SNORA31-mutated neurons revealed normal responses to TLR3 and IFN-α/ß stimulation but abnormal responses to HSV-1. Human SNORA31 thus controls central nervous system neuron-intrinsic immunity to HSV-1 by a distinctive mechanism.


Subject(s)
Encephalitis, Herpes Simplex/genetics , Herpesvirus 1, Human/genetics , Neurons/immunology , RNA, Small Nucleolar/genetics , Adult , Central Nervous System/immunology , Central Nervous System/virology , Child, Preschool , Encephalitis, Herpes Simplex/immunology , Encephalitis, Herpes Simplex/pathology , Encephalitis, Herpes Simplex/virology , Female , Genetic Predisposition to Disease , Herpesvirus 1, Human/immunology , Herpesvirus 1, Human/pathogenicity , Humans , Immunity/genetics , Infant , Male , Metagenome/genetics , Metagenome/immunology , Middle Aged , Neurons/virology , RNA, Small Nucleolar/immunology
6.
Front Immunol ; 10: 1509, 2019.
Article in English | MEDLINE | ID: mdl-31379808

ABSTRACT

The gut microbiome plays an important role in the immune system development, maintenance of normal health status, and in disease progression. In this study, we comparatively examined the fecal microbiomes of Amish (rural) and non-Amish (urban) infants and investigated how they could affect the mucosal immune maturation in germ-free piglets that were inoculated with the two types of infant fecal microbiota (IFM). Differences in microbiome diversity and structure were noted between the two types of fecal microbiotas. The fecal microbiota of the non-Amish (urban) infants had a greater relative abundance of Actinobacteria and Bacteroidetes phyla, while that of the Amish (rural) counterparts was dominated by Firmicutes. Amish infants had greater species richness compared with the non-Amish infants' microbiota. The fecal microbiotas of the Amish and the non-Amish infants were successfully transplanted into germ-free piglets, and the diversity and structure of the microbiota in the transplanted piglets remained similar at phylum level but not at the genus level. Principal coordinates analysis (PCoA) based on Weighted-UniFrac distance revealed distinct microbiota structure in the intestines of the transplanted piglets. Shotgun metagenomic analysis also revealed clear differences in functional diversity of fecal microbiome between Amish and non-Amish donors as well as microbiota transplanted piglets. Specific functional features were enriched in either of the microbiota transplanted piglet groups directly corresponding to the predominance of certain bacterial populations in their gut environment. Some of the colonized bacterial genera were correlated with the frequency of important lymphoid and myeloid immune cells in the ileal submucosa and mesenteric lymph nodes (MLN), both important for mucosal immune maturation. Overall, this study demonstrated that transplantation of diverse IFM into germ-free piglets largely recapitulates the differences in gut microbiota structure between rural (Amish) and urban (non-Amish) infants. Thus, fecal microbiota transplantation to germ-free piglets could be a useful large animal model system for elucidating the impact of gut microbiota on the mucosal immune system development. Future studies can focus on determining the additional advantages of the pig model over the rodent model.


Subject(s)
Feces/microbiology , Gastrointestinal Microbiome/immunology , Microbiota/immunology , Mucous Membrane/immunology , Mucous Membrane/microbiology , Amish , Animals , Fecal Microbiota Transplantation/methods , Firmicutes/immunology , Humans , Infant , Metagenome/immunology , Swine
7.
Front Immunol ; 10: 342, 2019.
Article in English | MEDLINE | ID: mdl-30891034

ABSTRACT

Gut commensal bacteria are known to have a significant role in regulating the innate and adaptive immune homeostasis. Alterations in the intestinal microbial composition have been associated with several disease states, including autoimmune and inflammatory conditions. However, it is not entirely clear how commensal gut microbiota modulate and contribute to the systemic immunity, and whether circulating elements of the host immune system could regulate the microbiome. Thus, we have studied the diversity and abundance of specific taxons in the gut microbiota of inbred GalT-KO mice during 7 months of animal life by metagenetic high-throughput sequencing (16S rRNA gene, variable regions V3-V5). The repertoire of glycan-specific natural antibodies, obtained by printed glycan array technology, was then associated with the microbial diversity for each animal by metagenome-wide association studies (MWAS). Our data show that the orders clostridiales (most abundant), bacteriodales, lactobacillales, and deferribacterales may be associated with the development of the final repertoire of natural anti-glycan antibodies in GalT-KO mice. The main changes in microbiota diversity (month-2 and month-3) were related to important changes in levels and repertoire of natural anti-glycan antibodies in these mice. Additionally, significant positive and negative associations were found between the gut microbiota and the pattern of specific anti-glycan antibodies. Regarding individual features, the gut microbiota and the corresponding repertoire of natural anti-glycan antibodies showed differences among the examined animals. We also found redundancy in different taxa associated with the development of specific anti-glycan antibodies. Differences in microbial diversity did not, therefore, necessarily influence the overall functional output of the gut microbiome of GalT-KO mice. In summary, the repertoire of natural anti-carbohydrate antibodies may be partially determined by the continuous antigenic stimulation produced by the gut bacterial population of each GalT-KO mouse. Small differences in gut microbiota diversity could determine different repertoire and levels of natural anti-glycan antibodies and consequently might induce different immune responses to pathogens or other potential threats.


Subject(s)
Antibodies/immunology , Gastrointestinal Microbiome/immunology , Microbiota/immunology , Polysaccharides/immunology , Animals , Antigens/immunology , Bacteria/immunology , Female , Intestines/immunology , Intestines/microbiology , Male , Metagenome/immunology , Mice , Mice, Knockout , RNA, Ribosomal, 16S/immunology
9.
Nat Med ; 24(12): 1815-1821, 2018 12.
Article in English | MEDLINE | ID: mdl-30397357

ABSTRACT

Human microbiome studies have revealed the intricate interplay of host immunity and bacterial communities to achieve homeostatic balance. Healthy skin microbial communities are dominated by bacteria with low viral representation1-3, mainly bacteriophage. Specific eukaryotic viruses have been implicated in both common and rare skin diseases, but cataloging skin viral communities has been limited. Alterations in host immunity provide an opportunity to expand our understanding of microbial-host interactions. Primary immunodeficient patients manifest with various viral, bacterial, fungal, and parasitic infections, including skin infections4. Dedicator of cytokinesis 8 (DOCK8) deficiency is a rare primary human immunodeficiency characterized by recurrent cutaneous and systemic infections, as well as atopy and cancer susceptibility5. DOCK8, encoding a guanine nucleotide exchange factor highly expressed in lymphocytes, regulates actin cytoskeleton, which is critical for migration through collagen-dense tissues such as skin6. Analyzing deep metagenomic sequencing data from DOCK8-deficient skin samples demonstrated a notable increase in eukaryotic viral representation and diversity compared with healthy volunteers. De novo assembly approaches identified hundreds of novel human papillomavirus genomes, illuminating microbial dark matter. Expansion of the skin virome in DOCK8-deficient patients underscores the importance of immune surveillance in controlling eukaryotic viral colonization and infection.


Subject(s)
Guanine Nucleotide Exchange Factors/genetics , Immunologic Deficiency Syndromes/virology , Skin Diseases/virology , Skin/virology , Adolescent , Bacteriophages/genetics , Child , Female , Genome, Viral/genetics , Guanine Nucleotide Exchange Factors/deficiency , Healthy Volunteers , Host Microbial Interactions/genetics , Host Microbial Interactions/immunology , Humans , Immunity/genetics , Immunologic Deficiency Syndromes/microbiology , Immunologic Deficiency Syndromes/pathology , Lymphocytes/virology , Male , Metagenome/genetics , Metagenome/immunology , Microbiota/genetics , Papillomaviridae/isolation & purification , Papillomaviridae/pathogenicity , Skin/microbiology , Skin Diseases/genetics , Skin Diseases/microbiology , Skin Diseases/pathology
10.
Bioessays ; 39(12)2017 Dec.
Article in English | MEDLINE | ID: mdl-28976007

ABSTRACT

The last decade has seen an explosion of research on the gut microbiota-the trillions of microorganisms that colonize the human gut. It is now clear that interindividual diversity in microbiota composition plays an important role in determining susceptibility to a wide variety of diseases. However, identifying the precise changes in microbiota composition that play causal roles has remained a largely unrealized goal. Here, we propose that functional classifications of microbes based on their interactions with and effects on the host-particularly the host immune system-will illuminate the role of the microbiota in shaping human physiology. We outline the benefits of "functional" classification compared to phylogenetic classifications, and review current efforts at functional classification of the microbiota. Finally, we outline a theoretical framework for classifying host-microbiota interactions. Future advances enabling broader functional classifications of the microbiota promise to revolutionize our understanding of the role of gut microbes in health and disease.


Subject(s)
Dysbiosis/immunology , Gastrointestinal Microbiome/immunology , Gastrointestinal Tract/microbiology , Immunity, Innate , Metagenome/immunology , Dysbiosis/genetics , Dysbiosis/microbiology , Gastrointestinal Tract/immunology , Humans , Immunoglobulin A/genetics , Metabolome/immunology , Microbial Consortia/immunology , Terminology as Topic
11.
Trends Immunol ; 37(10): 647-658, 2016 10.
Article in English | MEDLINE | ID: mdl-27616558

ABSTRACT

Humans are meta-organisms that maintain a diverse population of microorganisms on their barrier surfaces, collectively named the microbiota. Since most pathogens either cross or inhabit barrier surfaces, the microbiota plays a critical and often protective role during infections, both by modulating immune system responses and by mediating colonization resistance. However, the microbiota can also act as a reservoir for opportunistic microorganisms that can 'bloom', significantly complicating diseases of barrier surfaces by contributing to inflammatory immune responses. This review discusses our current understanding of the complex interactions between the host, its microbiota, and pathogenic organisms, focusing in particular on the intestinal mucosa.


Subject(s)
Infections/immunology , Inflammation/immunology , Intestinal Mucosa/immunology , Microbiota/immunology , Opportunistic Infections/immunology , Animals , Host-Pathogen Interactions , Humans , Immunomodulation , Infections/microbiology , Intestinal Mucosa/microbiology , Metagenome/immunology , Opportunistic Infections/microbiology
12.
Ital J Pediatr ; 41: 57, 2015 Aug 14.
Article in English | MEDLINE | ID: mdl-26268374

ABSTRACT

Celiac Disease (CD) is an immune-mediated systemic disorder elicited by gluten and related prolamines in genetically susceptible individuals and it is the result of the interaction between genetic and environmental factors. Among genetic risk factors, the strongest association is with the HLA class II DQ region; nevertheless at least 39 non-HLA loci are associated with CD. Gluten is the main environmental trigger of the disease. In addition, infant feeding and weaning practices as well as timing of gluten introduction in the diet have been suggested to contribute to CD risk. Furthermore a role for infectious agents and microbiota composition in disease development has also been proposed.Aim of this short review is to discuss the current knowledge on both genetic and environmental risk factors for the development of CD; moreover we will provide a brief overview of the possible strategies that could be envisaged to prevent this condition, at least in the population at-risk.


Subject(s)
Celiac Disease/etiology , Celiac Disease/prevention & control , Celiac Disease/immunology , Genetic Predisposition to Disease , Humans , Metagenome/immunology , Microbiota/immunology , Risk Factors
13.
Trends Immunol ; 36(3): 121-3, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25754179

ABSTRACT

The rise of urbanization and an increasingly indoor lifestyle has affected human interactions with our microbiota in unprecedented ways. We discuss how this lifestyle may influence immune development and function, and argue that it is time that we examined ways to manipulate the indoor environment to increase our exposure to a wider phylogeny of microorganisms. An important step is to continue to engage citizen scientists in the efforts to characterize our interactions with the diverse microbial environments that we inhabit.


Subject(s)
Immunity , Metagenome/immunology , Microbiota/immunology , Symbiosis/immunology , Ecological Systems, Closed , Environment , Environment Design/trends , Humans , Information Dissemination , Social Media , Urbanization/trends
14.
Eur J Immunol ; 45(1): 17-31, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25328099

ABSTRACT

Commensal microorganisms colonize barrier surfaces of all multicellular organisms, including those of humans. For more than 500 million years, commensal microorganisms and their hosts have coevolved and adapted to each other. As a result, the commensal microbiota affects many immune and nonimmune functions of their hosts, and de facto the two together comprise one metaorganism. The commensal microbiota communicates with the host via biologically active molecules. Recently, it has been reported that microbial imbalance may play a critical role in the development of multiple diseases, such as cancer, autoimmune conditions, and increased susceptibility to infection. In this review, we focus on the role of the commensal microbiota in the development, progression, and immune evasion of cancer, as well as some modulatory effects on the treatment of cancer. In particular, we discuss the mechanisms of microbiota-mediated regulation of innate and adaptive immune responses to tumors, and the consequences on cancer progression and whether tumors subsequently become resistant or susceptible to different anticancer therapeutic regiments.


Subject(s)
Autoimmune Diseases/microbiology , Carcinogenesis/immunology , Microbiota/immunology , Neoplasms/microbiology , Adaptive Immunity , Animals , Antineoplastic Agents/therapeutic use , Autoimmune Diseases/drug therapy , Autoimmune Diseases/pathology , Biological Evolution , Carcinogenesis/pathology , Humans , Immunity, Innate , Immunomodulation , Inflammation/drug therapy , Inflammation/immunology , Inflammation/microbiology , Inflammation/pathology , Metagenome/immunology , Mice , Neoplasms/drug therapy , Neoplasms/immunology , Neoplasms/pathology , Symbiosis/immunology , Tumor Escape
15.
Pediatr Allergy Immunol ; 25(5): 428-38, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24899389

ABSTRACT

The development of the intestinal microbiota in the first years of life is a dynamic process significantly influenced by early-life nutrition. Pioneer bacteria colonizing the infant intestinal tract and the gradual diversification to a stable climax ecosystem plays a crucial role in establishing host-microbe interactions essential for optimal symbiosis. This colonization process and establishment of symbiosis may profoundly influence health throughout life. Recent developments in microbiologic cultivation-independent methods allow a detailed view of the key players and factors involved in this process and may further elucidate their roles in a healthy gut and immune maturation. Aberrant patterns may lead to identifying key microbial signatures involved in developing immunologic diseases into adulthood, such as asthma and atopic diseases. The central role of early-life nutrition in the developmental human microbiota, immunity, and metabolism offers promising strategies for prevention and treatment of such diseases. This review provides an overview of the development of the intestinal microbiota, its bidirectional relationship with the immune system, and its role in impacting health and disease, with emphasis on allergy, in early life.


Subject(s)
Intestines/microbiology , Metagenome/immunology , Microbiota/immunology , Symbiosis/immunology , Child, Preschool , Humans , Hypersensitivity/immunology , Hypersensitivity/microbiology , Infant , Infant, Newborn
16.
J Innate Immun ; 6(3): 272-83, 2014.
Article in English | MEDLINE | ID: mdl-24525633

ABSTRACT

Severe infection and the patient response constitute sepsis. Here, we review the meta-genome (patient genetics, pathogen communities and host response) and its impact upon the outcome of severe sepsis. Patient genetics, both predisposition for infection and the subsequent response to infection are reviewed. The pathogen is discussed with particular emphasis upon the modern era of microbiome analysis and nucleic acid diagnostics. Finally, we discuss the host clinical and immune responses and present new data to suggest that the immune response is the key to understanding sepsis and improving a death rate of nearly 30%.


Subject(s)
Bacterial Infections/immunology , Metagenome/immunology , Microbiota/immunology , Sepsis/immunology , Acute Disease , Bacterial Infections/diagnosis , Bacterial Infections/genetics , Biodiversity , Genetic Predisposition to Disease , High-Throughput Screening Assays , Humans , Immunity/genetics , Metagenome/genetics , Microbiota/genetics , Pathology, Molecular , Sepsis/diagnosis , Sepsis/genetics
17.
DNA Res ; 21(1): 15-25, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24013298

ABSTRACT

Analysis of microbiota in various biological and environmental samples under a variety of conditions has recently become more practical due to remarkable advances in next-generation sequencing. Changes leading to specific biological states including some of the more complex diseases can now be characterized with relative ease. It is known that gut microbiota is involved in the pathogenesis of inflammatory bowel disease (IBD), mainly Crohn's disease and ulcerative colitis, exhibiting symptoms in the gastrointestinal tract. Recent studies also showed increased frequency of oral manifestations among IBD patients, indicating aberrations in the oral microbiota. Based on these observations, we analyzed the composition of salivary microbiota of 35 IBD patients by 454 pyrosequencing of the bacterial 16S rRNA gene and compared it with that of 24 healthy controls (HCs). The results showed that Bacteroidetes was significantly increased with a concurrent decrease in Proteobacteria in the salivary microbiota of IBD patients. The dominant genera, Streptococcus, Prevotella, Neisseria, Haemophilus, Veillonella, and Gemella, were found to largely contribute to dysbiosis (dysbacteriosis) observed in the salivary microbiota of IBD patients. Analysis of immunological biomarkers in the saliva of IBD patients showed elevated levels of many inflammatory cytokines and immunoglobulin A, and a lower lysozyme level. A strong correlation was shown between lysozyme and IL-1ß levels and the relative abundance of Streptococcus, Prevotella, Haemophilus and Veillonella. Our data demonstrate that dysbiosis of salivary microbiota is associated with inflammatory responses in IBD patients, suggesting that it is possibly linked to dysbiosis of their gut microbiota.


Subject(s)
Dysbiosis/microbiology , Inflammatory Bowel Diseases/microbiology , Microbiota/immunology , Saliva/microbiology , Adult , Biomarkers/analysis , Dysbiosis/immunology , Female , Humans , Inflammatory Bowel Diseases/immunology , Male , Metagenome/immunology , Microbiota/genetics , Middle Aged , RNA, Bacterial/metabolism , RNA, Ribosomal, 16S/metabolism , Saliva/immunology
19.
J Exp Med ; 210(9): 1665-74, 2013 Aug 26.
Article in English | MEDLINE | ID: mdl-23940259

ABSTRACT

We have tracked the fate of immature human B cells at a critical stage in their development when the mature B cell repertoire is shaped. We show that a major subset of bone marrow emigrant immature human B cells, the transitional 2 (T2) B cells, homes to gut-associated lymphoid tissue (GALT) and that most T2 B cells isolated from human GALT are activated. Activation in GALT is a previously unknown potential fate for immature human B cells. The process of maturation from immature transitional B cell through to mature naive B cell includes the removal of autoreactive cells from the developing repertoire, a process which is known to fail in systemic lupus erythematosus (SLE). We observe that immature B cells in SLE are poorly equipped to access the gut and that gut immune compartments are depleted in SLE. Thus, activation of immature B cells in GALT may function as a checkpoint that protects against autoimmunity. In healthy individuals, this pathway may be involved in generating the vast population of IgA plasma cells and also the enigmatic marginal zone B cell subset that is poorly understood in humans.


Subject(s)
B-Lymphocytes/immunology , Gastrointestinal Tract/immunology , Lymphoid Tissue/immunology , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/pathology , Base Sequence , Gastrointestinal Tract/pathology , Humans , Integrin beta Chains/metabolism , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/pathology , Lymphocyte Activation/immunology , Metagenome/immunology , Molecular Sequence Data , Plasma Cells/immunology , Precursor Cells, B-Lymphoid/immunology , Precursor Cells, B-Lymphoid/pathology
20.
Nature ; 500(7461): 232-6, 2013 Aug 08.
Article in English | MEDLINE | ID: mdl-23842501

ABSTRACT

Manipulation of the gut microbiota holds great promise for the treatment of inflammatory and allergic diseases. Although numerous probiotic microorganisms have been identified, there remains a compelling need to discover organisms that elicit more robust therapeutic responses, are compatible with the host, and can affect a specific arm of the host immune system in a well-controlled, physiological manner. Here we use a rational approach to isolate CD4(+)FOXP3(+) regulatory T (Treg)-cell-inducing bacterial strains from the human indigenous microbiota. Starting with a healthy human faecal sample, a sequence of selection steps was applied to obtain mice colonized with human microbiota enriched in Treg-cell-inducing species. From these mice, we isolated and selected 17 strains of bacteria on the basis of their high potency in enhancing Treg cell abundance and inducing important anti-inflammatory molecules--including interleukin-10 (IL-) and inducible T-cell co-stimulator (ICOS)--in Treg cells upon inoculation into germ-free mice. Genome sequencing revealed that the 17 strains fall within clusters IV, XIVa and XVIII of Clostridia, which lack prominent toxins and virulence factors. The 17 strains act as a community to provide bacterial antigens and a TGF-ß-rich environment to help expansion and differentiation of Treg cells. Oral administration of the combination of 17 strains to adult mice attenuated disease in models of colitis and allergic diarrhoea. Use of the isolated strains may allow for tailored therapeutic manipulation of human immune disorders.


Subject(s)
Clostridium/immunology , Metagenome/immunology , T-Lymphocytes, Regulatory/physiology , Adult , Animals , Cell Proliferation , Clostridium/classification , Clostridium/genetics , Colitis/microbiology , Colitis/pathology , Colon/immunology , Colon/microbiology , Disease Models, Animal , Feces/microbiology , Germ-Free Life , Humans , Inducible T-Cell Co-Stimulator Protein/metabolism , Interleukin-10/metabolism , Male , Metagenome/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, SCID , RNA, Ribosomal, 16S/genetics , Rats , Rats, Inbred F344 , T-Lymphocytes, Regulatory/cytology
SELECTION OF CITATIONS
SEARCH DETAIL