751
|
Salcedo R, Cataisson C, Hasan U, Yuspa SH, Trinchieri G. MyD88 and its divergent toll in carcinogenesis. Trends Immunol 2013; 34:379-89. [PMID: 23660392 PMCID: PMC3847901 DOI: 10.1016/j.it.2013.03.008] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2013] [Revised: 03/21/2013] [Accepted: 03/31/2013] [Indexed: 02/07/2023]
Abstract
Toll-like and interleukin-1 (IL-1) family receptors recognize microbial or endogenous ligands and inflammatory mediators, respectively, and with the exception of Toll-like receptor 3 (TLR3), signal via the adaptor molecule myeloid differentiation factor 88 (MyD88). MyD88 is involved in oncogene-induced cell intrinsic inflammation and in cancer-associated extrinsic inflammation, and as such MyD88 contributes to skin, liver, pancreatic, and colon carcinogenesis, as well as sarcomagenesis. MyD88 is also protective, for example in oncogenic virus carcinogenesis or, acting downstream of IL-18R to strengthen mucosal repair, in azoxymethane (AOM)/dextran sulfate sodium (DSS)-induced colon carcinogenesis. Here, we discuss the mechanisms of the divergent effects of MyD88 and the balance of its protumor role in cancer-enhancing inflammation and immunity and its antitumor role in tissue homeostasis, repair, and immunity against the tumor or oncogenic pathogens.
Collapse
Affiliation(s)
- Rosalba Salcedo
- Cancer and Inflammation Program, National Cancer Institute, Frederick, MD 217023, USA
| | | | | | | | | |
Collapse
|
752
|
Börnigen D, Morgan XC, Franzosa EA, Ren B, Xavier RJ, Garrett WS, Huttenhower C. Functional profiling of the gut microbiome in disease-associated inflammation. Genome Med 2013; 5:65. [PMID: 23906180 PMCID: PMC3978847 DOI: 10.1186/gm469] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The microbial residents of the human gut are a major factor in the development and lifelong maintenance of health. The gut microbiota differs to a large degree from person to person and has an important influence on health and disease due to its interaction with the human immune system. Its overall composition and microbial ecology have been implicated in many autoimmune diseases, and it represents a particularly important area for translational research as a new target for diagnostics and therapeutics in complex inflammatory conditions. Determining the biomolecular mechanisms by which altered microbial communities contribute to human disease will be an important outcome of current functional studies of the human microbiome. In this review, we discuss functional profiling of the human microbiome using metagenomic and metatranscriptomic approaches, focusing on the implications for inflammatory conditions such as inflammatory bowel disease and rheumatoid arthritis. Common themes in gut microbial ecology have emerged among these diverse diseases, but they have not yet been linked to targetable mechanisms such as microbial gene and genome composition, pathway and transcript activity, and metabolism. Combining these microbial activities with host gene, transcript and metabolic information will be necessary to understand how and why these complex interacting systems are altered in disease-associated inflammation.
Collapse
Affiliation(s)
- Daniela Börnigen
- Department of Biostatistics, Harvard School of Public Health, Boston, MA 02115, USA ; The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Xochitl C Morgan
- Department of Biostatistics, Harvard School of Public Health, Boston, MA 02115, USA ; The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Eric A Franzosa
- Department of Biostatistics, Harvard School of Public Health, Boston, MA 02115, USA ; The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Boyu Ren
- Department of Biostatistics, Harvard School of Public Health, Boston, MA 02115, USA
| | - Ramnik J Xavier
- The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA ; Gastrointestinal Unit, Massachusetts General Hospital, Boston, MA 02115, USA
| | - Wendy S Garrett
- The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA ; Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, MA 02115, USA ; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA ; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Curtis Huttenhower
- Department of Biostatistics, Harvard School of Public Health, Boston, MA 02115, USA ; The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| |
Collapse
|
753
|
Ursell LK, Van Treuren W, Metcalf JL, Pirrung M, Gewirtz A, Knight R. Replenishing our defensive microbes. Bioessays 2013; 35:810-7. [PMID: 23836415 DOI: 10.1002/bies.201300018] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Large-scale characterization of the human microbiota has largely focused on Western adults, yet these populations may be uncharacteristic because of their diets and lifestyles. In particular, the rise of "Western diseases" may in part stem from reduced exposure to, or even loss of, microbes with which humans have coevolved. Here, we review beneficial microbes associated with pathogen resistance, highlighting the emerging role of complex microbial communities in protecting against disease. We discuss ways in which modern lifestyles and practices may deplete physiologically important microbiota, and explore prospects for reintroducing or encouraging the growth of beneficial microbes to promote the restoration of healthy microbial ecosystems.
Collapse
Affiliation(s)
- Luke K Ursell
- Department of Chemistry and Biochemistry, University of Colorado at Boulder, Boulder, CO, USA
| | | | | | | | | | | |
Collapse
|
754
|
Hu W, Pasare C. Location, location, location: tissue-specific regulation of immune responses. J Leukoc Biol 2013; 94:409-21. [PMID: 23825388 DOI: 10.1189/jlb.0413207] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Discovery of DCs and PRRs has contributed immensely to our understanding of induction of innate and adaptive immune responses. Activation of PRRs leads to secretion of inflammatory cytokines that regulate priming and differentiation of antigen-specific T and B lymphocytes. Pathogens enter the body via different routes, and although the same set of PRRs is likely to be activated, it is becoming clear that the route of immune challenge determines the nature of outcome of adaptive immunity. In addition to the signaling events initiated following innate-immune receptor activation, the cells of the immune system are influenced by the microenvironments in which they reside, and this has a direct impact on the resulting immune response. Specifically, immune responses could be influenced by specialized DCs, specific factors secreted by stromal cells, and also, by commensal microbiota present in certain organs. Following microbial detection, the complex interactions among DCs, stromal cells, and tissue-specific factors influence outcome of immune responses. In this review, we summarize recent findings on the phenotypic heterogeneity of innate and adaptive immune cells and how tissue-specific factors in the systemic and mucosal immune system influence the outcome of adaptive-immune responses.
Collapse
Affiliation(s)
- Wei Hu
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | | |
Collapse
|
755
|
Erturk-Hasdemir D, Kasper DL. Resident commensals shaping immunity. Curr Opin Immunol 2013; 25:450-5. [PMID: 23830047 DOI: 10.1016/j.coi.2013.06.001] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2013] [Revised: 06/05/2013] [Accepted: 06/06/2013] [Indexed: 02/07/2023]
Abstract
All animals coexist with myriad commensal microorganisms in a symbiotic relationship that plays a key role in health and disease. Continuous commensal-host interactions profoundly affect the development and regulation of the host's immune system. The complex interaction of the commensal microbiota with the immune system is a topic of substantial interest. An understanding of these interactions and the mechanisms through which commensal microbes actively shape host immunity may yield new insights into the pathogenesis of many immune-mediated diseases and lead to new prophylactic and therapeutic interventions. This review examines recent advances in this field and their potential implications not just for the colonized tissues but also for the entire immune system.
Collapse
Affiliation(s)
- Deniz Erturk-Hasdemir
- Department of Microbiology and Immunobiology, Harvard Medical School, 77 Avenue Louis Pasteur, Room 1056C, Boston, MA 02115, United States
| | | |
Collapse
|
756
|
Kuo IH, Yoshida T, De Benedetto A, Beck LA. The cutaneous innate immune response in patients with atopic dermatitis. J Allergy Clin Immunol 2013; 131:266-78. [PMID: 23374259 DOI: 10.1016/j.jaci.2012.12.1563] [Citation(s) in RCA: 151] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Accepted: 12/13/2012] [Indexed: 12/28/2022]
Abstract
Orchestrating when and how the cutaneous innate immune system should respond to commensal or pathogenic microbes is a critical function of the epithelium. The cutaneous innate immune system is a key determinant of the physical, chemical, microbial, and immunologic barrier functions of the epidermis. A malfunction in this system can lead to an inadequate host response to a pathogen or a persistent inflammatory state. Atopic dermatitis is the most common inflammatory skin disorder and characterized by abnormalities in both skin barrier structures (stratum corneum and tight junctions), a robust T(H)2 response to environmental antigens, defects in innate immunity, and an altered microbiome. Many of these abnormalities may occur as the consequence of epidermal dysfunction. The epidermis directly interfaces with the environment and, not surprisingly, expresses many pattern recognition receptors that make it a key player in cutaneous innate immune responses to skin infections and injury. This review will discuss the role epidermal innate receptors play in regulation of skin barriers and, where possible, discuss the relevance of these findings for patients with atopic dermatitis.
Collapse
Affiliation(s)
- I-Hsin Kuo
- Department of Dermatology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | | | | | | |
Collapse
|
757
|
Alexeyev OA. Bacterial landscape of human skin: seeing the forest for the trees. Exp Dermatol 2013; 22:443-6. [DOI: 10.1111/exd.12160] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/25/2013] [Indexed: 12/18/2022]
Affiliation(s)
- Oleg A. Alexeyev
- Department of Medical Biosciences/Pathology; Umeå University; Umeå Sweden
| |
Collapse
|
758
|
Signaling via the IL-20 receptor inhibits cutaneous production of IL-1β and IL-17A to promote infection with methicillin-resistant Staphylococcus aureus. Nat Immunol 2013; 14:804-11. [PMID: 23793061 PMCID: PMC3721434 DOI: 10.1038/ni.2637] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2013] [Accepted: 05/02/2013] [Indexed: 12/14/2022]
Abstract
Staphylococcus aureus causes most infections of human skin and soft tissue and is a major infectious cause of mortality. Host defense mechanisms against S. aureus are incompletely understood. Interleukin 19 (IL-19), IL-20 and IL-24 signal through type I and type II IL-20 receptors and are associated with inflammatory skin diseases such as psoriasis and atopic dermatitis. We found here that those cytokines promoted cutaneous infection with S. aureus in mice by downregulating IL-1β- and IL-17A-dependent pathways. We noted similar effects of those cytokines in human keratinocytes after exposure to S. aureus, and antibody blockade of the IL-20 receptor improved outcomes in infected mice. Our findings identify an immunosuppressive role for IL-19, IL-20 and IL-24 during infection that could be therapeutically targeted to alter susceptibility to infection.
Collapse
|
759
|
Smeekens SP, Huttenhower C, Riza A, van de Veerdonk FL, Zeeuwen PLJM, Schalkwijk J, van der Meer JWM, Xavier RJ, Netea MG, Gevers D. Skin microbiome imbalance in patients with STAT1/STAT3 defects impairs innate host defense responses. J Innate Immun 2013; 6:253-62. [PMID: 23796786 DOI: 10.1159/000351912] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Accepted: 05/07/2013] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Chronic mucocutaneous candidiasis (CMC) and hyper-IgE syndrome (HIES) are primary immunodeficiencies mainly caused by mutations in STAT1 and STAT3, respectively. CMC and HIES patients have an increased risk for skin and mucosal infections with fungal pathogens and Staphylococcus aureus. However, it is unknown whether the genetic defects in these patients also affect the skin and mucosal microbiome, which in turn may influence host defense mechanisms. METHODS The skin and oral microbiome of CMC and HIES patients was compared to that of healthy controls at five body sites using 16S rRNA sequencing. The influence of skin colonizers on the immune response was investigated using in vitro experiments. RESULTS The microbiome of CMC and HIES patients contained more Gram-negative bacteria, especially Acinetobacter spp., and less of the normal Corynebacterium spp. compared to healthy controls. Exposure of human primary leukocytes to Acinetobacter suppressed the cytokine response to Candida albicans and S. aureus, while the normal corynebacteria did not suppress cytokine responses. DISCUSSION These results demonstrate that central mediators of immune responses like STAT1 and STAT3 not only directly influence immune responses, but also result in changes in the skin microbiome that in turn can amplify the defective immune response against fungal and microbial pathogens.
Collapse
Affiliation(s)
- Sanne P Smeekens
- Department of Medicine, Radboud University Nijmegen Medical Center, Nijmegen, The Netherlands
| | | | | | | | | | | | | | | | | | | |
Collapse
|
760
|
Yamazaki S, Morita A. Dendritic cells in the periphery control antigen-specific natural and induced regulatory T cells. Front Immunol 2013; 4:151. [PMID: 23801989 PMCID: PMC3689032 DOI: 10.3389/fimmu.2013.00151] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Accepted: 06/04/2013] [Indexed: 12/20/2022] Open
Abstract
Dendritic cells (DCs) are specialized antigen-presenting cells that regulate both immunity and tolerance. DCs in the periphery play a key role in expanding naturally occurring Foxp3+ CD25+ CD4+ regulatory T cells (Natural T-regs) and inducing Foxp3 expression (Induced T-regs) in Foxp3− CD4+ T cells. DCs are phenotypically and functionally heterogeneous, and further classified into several subsets depending on distinct marker expression and their location. Recent findings indicate the presence of specialized DC subsets that act to expand Natural T-regs or induce Foxp3+ T-regs from Foxp3− CD4+ T cells. For example, two major subsets of DCs in lymphoid organs act differentially in inducing Foxp3+ T-regs from Foxp3− cells or expanding Natural T-regs with model-antigen delivery by anti-DC subset monoclonal antibodies in vivo. Furthermore, DCs expressing CD103 in the intestine induce Foxp3+ T-regs from Foxp3− CD4+ T cells with endogenous TGF-β and retinoic acid. In addition, antigen-presenting DCs have a capacity to generate Foxp3+ T-regs in the oral cavity where many antigens and commensals exist, similar to intestine and skin. In skin and skin-draining lymph nodes, at least six DC subsets have been identified, suggesting a complex DC-T-reg network. Here, we will review the specific activity of DCs in expanding Natural T-regs and inducing Foxp3+ T-regs from Foxp3− precursors, and further discuss the critical function of DCs in maintaining tolerance at various locations including skin and oral cavity.
Collapse
Affiliation(s)
- Sayuri Yamazaki
- Department of Geriatric and Environmental Dermatology, Graduate School of Medical Sciences, Nagoya City University , Nagoya , Japan
| | | |
Collapse
|
761
|
The microbiome extends to subepidermal compartments of normal skin. Nat Commun 2013; 4:1431. [PMID: 23385576 PMCID: PMC3655727 DOI: 10.1038/ncomms2441] [Citation(s) in RCA: 308] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2012] [Accepted: 12/19/2012] [Indexed: 12/26/2022] Open
Abstract
Commensal microbes on the skin surface influence the behavior of cells below the epidermis. We hypothesized that bacteria or their products exist below the surface epithelium and thus permit physical interaction between microbes and dermal cells. Here, to test this hypothesis, we employed multiple independent detection techniques for bacteria including qPCR, Gram-staining, immunofluorescence, and in situ hybridization. Bacteria were consistently detectable within the dermis and dermal adipose of normal human skin. Sequencing of DNA from dermis and dermal adipose tissue identified bacterial 16S rRNA reflective of a diverse and partially distinct microbial community in each skin compartment. These results show the microbiota extends within the dermis, therefore enabling physical contact between bacteria and various cells below the basement membrane. These observations show that normal commensal bacterial communities directly communicate with the host in a tissue previously thought to be sterile.
Collapse
|
762
|
Kosiewicz MM, Zirnheld AL, Alard P. Tuning of skin immunity by skin commensal bacteria. Immunotherapy 2013; 5:23-5. [PMID: 23256795 DOI: 10.2217/imt.12.140] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Evaluation of: Naik S, Bouladoux N, Wilhelm C et al. Compartmentalized control of skin immunity by resident commensals. Science 337, 1115-1119 (2012). Most analyses of commensal microbiota have been directed toward the gut microbiota and its role in the development of the intestinal immune system, and in regulating the immune response at sites distant from the gut, including the joints or CNS. However, very little is known about how other niches of commensal microbiota affect local immunity and whether they are influenced by the gut microbiota. The current paper reveals that skin commensals are required for the development of protective immunity against a cutaneous pathogen. This immune response driven by skin commensals occurs independently of the gut microbiota and is mediated by MyD88 and IL-1 signaling that promotes protective effector T-cell responses.
Collapse
Affiliation(s)
- Michele M Kosiewicz
- Department of Microbiology & Immunology, University of Louisville, HSC, Louisville, KY 40202, USA
| | | | | |
Collapse
|
763
|
Ling Z, Liu X, Luo Y, Yuan L, Nelson KE, Wang Y, Xiang C, Li L. Pyrosequencing analysis of the human microbiota of healthy Chinese undergraduates. BMC Genomics 2013; 14:390. [PMID: 23758874 PMCID: PMC3685588 DOI: 10.1186/1471-2164-14-390] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2013] [Accepted: 06/03/2013] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Elucidating the biogeography of bacterial communities on the human body is critical for establishing healthy baselines from which to detect differences associated with disease; however, little is known about the baseline bacterial profiles from various human habitats of healthy Chinese undergraduates. RESULTS Using parallel barcoded 454 pyrosequencing targeting on the 16S rRNA gene V3 region, the bacterial diversity of the nasopharynx, saliva, dominant hands, and feces were investigated from 10 healthy Chinese junior boarding undergraduates at Zhejiang University. The participants were 21-24 years of age with a body mass index (BMI) < 24 kg/m(2). A total of 156,717 high-quality pyrosequencing reads were obtained for evaluating bacterial diversity, which represented 29,887 unique phylotypes. The overall taxonomic distribution of the 16S rRNA gene-based amplicons demonstrated that these 4 habitats of the human body harbored distinct microbiota and could be divided into different clusters according to anatomic site, while the established patterns of bacterial diversity followed the human body habitat (feces, hands, saliva, and nasopharynx). Although significant inter-individual variation was observed, the healthy microbiota still shared a large number of phylotypes in each habitat, but not among the four habitats, indicating that a core microbiome existed in each healthy habitat. The vast majority of sequences from these different habitats were classified into different taxonmies that became the predominant bacteria of the healthy microbiota. CONCLUSIONS We first established the framework of microbial communities from four healthy human habitats of the same participants with similar living environments for the Chinese undergraduates. Our data represent an important step for determining the diversity of Chinese healthy microbiota, and can be used for more large-scale studies that focus on the interactions between healthy and diseases states for young Chinese adults in the same age range.
Collapse
Affiliation(s)
- Zongxin Ling
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, China
| | - Xia Liu
- Department of Intensive Care Unit, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, China
| | - Yueqiu Luo
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, China
| | - Li Yuan
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, China
| | - Karen E Nelson
- J. Craig Venter Institute, Rockville, Maryland, 20850, USA
| | - Yuezhu Wang
- Chinese National Human Genome Center at Shanghai, Shanghai, 201203, China
| | - Charlie Xiang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, China
- J. Craig Venter Institute, Rockville, Maryland, 20850, USA
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, China
| |
Collapse
|
764
|
Mavrommatis B, Young GR, Kassiotis G. Counterpoise between the microbiome, host immune activation and pathology. Curr Opin Immunol 2013; 25:456-62. [PMID: 23743081 DOI: 10.1016/j.coi.2013.05.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Revised: 05/03/2013] [Accepted: 05/08/2013] [Indexed: 12/13/2022]
Abstract
The role of the mammalian intestinal microbiota in health and disease of the host has long been recognized and extensively studied. Largely, these studies have focused on the bacterial component of the microbiota. However, recent technological advances have shed new light on the microbiome at distinct anatomical locations and uncovered the role of additional microbial symbionts, including the virome and endogenous retroelements. Together, they have revealed interactions more intricate than previously recognized. Here, we review recent advances in our knowledge of this collective microbiome and the interactions with the immune system of their host.
Collapse
Affiliation(s)
- Bettina Mavrommatis
- Division of Immunoregulation, MRC National Institute for Medical Research, The Ridgeway, London NW7 1AA, UK
| | | | | |
Collapse
|
765
|
Belkaid Y, Bouladoux N, Hand TW. Effector and memory T cell responses to commensal bacteria. Trends Immunol 2013; 34:299-306. [PMID: 23643444 PMCID: PMC3733441 DOI: 10.1016/j.it.2013.03.003] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Revised: 03/18/2013] [Accepted: 03/18/2013] [Indexed: 02/08/2023]
Abstract
Barrier surfaces are home to a vast population of commensal organisms that together encode millions of proteins; each of them possessing several potential foreign antigens. Regulation of immune responses to this enormous antigenic load represents a tremendous challenge for the immune system. Tissues exposed to commensals have developed elaborate systems of regulation including specialized populations of resident lymphocytes that maintain barrier function and limit potential responses to commensal antigens. However, in settings of infection and inflammation these regulatory mechanisms are compromised and specific effector responses against commensal bacteria can develop. This review discusses the circumstances controlling the fate of commensal specific T cells and how dysregulation of these responses could lead to severe pathological outcomes.
Collapse
Affiliation(s)
- Yasmine Belkaid
- Mucosal Immunology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Disease, NIH, Bethesda 20892, USA.
| | | | | |
Collapse
|
766
|
Grainger JR, Wohlfert EA, Fuss IJ, Bouladoux N, Askenase MH, Legrand F, Koo LY, Brenchley JM, Fraser IDC, Belkaid Y. Inflammatory monocytes regulate pathologic responses to commensals during acute gastrointestinal infection. Nat Med 2013; 19:713-21. [PMID: 23708291 PMCID: PMC3755478 DOI: 10.1038/nm.3189] [Citation(s) in RCA: 213] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2012] [Accepted: 04/09/2013] [Indexed: 12/17/2022]
Abstract
Commensal flora can promote both immunity to pathogens and mucosal inflammation. How commensal driven inflammation is regulated in the context of infection remains poorly understood. Here, we show that during acute mucosal infection, Ly6Chi inflammatory monocytes acquire a tissue specific regulatory phenotype associated with production of the lipid mediator prostaglandin E2 (PGE2). Notably, in response to commensals, Ly6Chi monocytes can directly inhibit neutrophil activation in a PGE2-dependent manner. Further, in the absence of inflammatory monocytes, mice develop severe neutrophil-mediated pathology that can be controlled by PGE2 analog treatment. Complementing these findings, inhibition of PGE2 led to enhanced neutrophil activation and host mortality. These data demonstrate a previously unappreciated dual action of inflammatory monocytes in controlling pathogen expansion while limiting commensal mediated damage to the gut. Collectively, our results place inflammatory monocyte derived PGE2 at the center of a commensal driven regulatory loop required to control host-commensal dialogue during inflammation.
Collapse
Affiliation(s)
- John R Grainger
- Program in Barrier Immunity and Repair, Mucosal Immunology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
767
|
The absence of a microbiota enhances TSLP expression in mice with defective skin barrier but does not affect the severity of their allergic inflammation. J Invest Dermatol 2013; 133:2714-2721. [PMID: 23698100 PMCID: PMC3796202 DOI: 10.1038/jid.2013.228] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2013] [Revised: 04/03/2013] [Accepted: 04/12/2013] [Indexed: 12/20/2022]
Abstract
Evidence is accumulating to suggest that our indigenous microbial communities (microbiota) may play a role in modulating allergic and immune disorders of the skin (Gallo and Nakatsuji, 2011; Macia et al., 2012). To examine the link between the microbiota and atopic dermatitis, we examined a mouse model of defective cutaneous barrier function with an atopic dermatitis-like disease due to loss of Notch signaling. Comparisons of conventionally-raised (CONV-R) and germ-free (GF) mice revealed a similar degree of allergic skin inflammation, systemic atopy, and airway hypersensitivity. GF mutant animals expressed significantly higher levels of thymic stromal lymphopoietin (TSLP), a major proinflammatory cytokine released by skin with defective barrier function, resulting in a more severe B-lymphoproliferative disorder that persisted into adulthood. These findings suggest a role for the microbiota in ameliorating stress signals released by keratinocytes in response to perturbation in cutaneous barrier function.
Collapse
|
768
|
|
769
|
Song SJ, Lauber C, Costello EK, Lozupone CA, Humphrey G, Berg-Lyons D, Caporaso JG, Knights D, Clemente JC, Nakielny S, Gordon JI, Fierer N, Knight R. Cohabiting family members share microbiota with one another and with their dogs. eLife 2013; 2:e00458. [PMID: 23599893 PMCID: PMC3628085 DOI: 10.7554/elife.00458] [Citation(s) in RCA: 743] [Impact Index Per Article: 61.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2012] [Accepted: 03/03/2013] [Indexed: 12/17/2022] Open
Abstract
Human-associated microbial communities vary across individuals: possible contributing factors include (genetic) relatedness, diet, and age. However, our surroundings, including individuals with whom we interact, also likely shape our microbial communities. To quantify this microbial exchange, we surveyed fecal, oral, and skin microbiota from 60 families (spousal units with children, dogs, both, or neither). Household members, particularly couples, shared more of their microbiota than individuals from different households, with stronger effects of co-habitation on skin than oral or fecal microbiota. Dog ownership significantly increased the shared skin microbiota in cohabiting adults, and dog-owning adults shared more ‘skin’ microbiota with their own dogs than with other dogs. Although the degree to which these shared microbes have a true niche on the human body, vs transient detection after direct contact, is unknown, these results suggest that direct and frequent contact with our cohabitants may significantly shape the composition of our microbial communities. DOI:http://dx.doi.org/10.7554/eLife.00458.001 The human body is home to many different microorganisms, with a range of bacteria, fungi and archaea living on the skin, in the intestine and at various other sites in the body. While many of these microorganisms are beneficial to their human hosts, we know very little about most of them. Early research focused primarily on comparing the microorganisms found in healthy individuals with those found in individuals suffering from a particular illness. More recently researchers have become interested in more general issues, such as understanding how these collections of microorganisms, which are also known as the human microbiota or the human microbiome, become established, and exploring the causes of similarities and differences between the microbiota of individuals. We now know that the communities of microorganisms found in the intestines of genetically related people tend to be more similar than those of people who are not related. Moreover, the communities of microorganisms found in the intestines of non-related adults living in the same household are more similar than those of unrelated adults living in different households. We also know that the range of microorganisms found in the intestine changes dramatically between birth and the age of 3 years. However, these studies have focused on the intestine, and little is known about the effect of relatedness, cohabitation and age on the microbiota at other body sites. Song et al. compared the microorganisms found on the skin, on the tongue and in the intestines of 159 people—and 36 dogs—in 60 families. They found that co-habitation resulted in the communities of microorganisms being more similar to each other, with those on the skin being the most similar. This was true for all comparisons, including human pairs, dog pairs and human–dog pairs. This suggests that humans probably acquire many of the microorganisms on their skin through direct contact with their surroundings, and that humans tend to share more microbes with individuals, including their pets, with which they are in frequent contact. Song et al. also discovered that, unlike what happens in the intestine, the microbial communities on the skin and tongue of infants and children were relatively similar to those of adults. Overall, these findings suggest that the communities of microorganisms found in the intestine changes with age in a way that differs significantly from those found on the skin and tongue. DOI:http://dx.doi.org/10.7554/eLife.00458.002
Collapse
Affiliation(s)
- Se Jin Song
- Department of Ecology and Evolutionary Biology , University of Colorado, Boulder , Boulder , United States
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
770
|
Barbarot S. [What's new in pediatric dermatology?]. Ann Dermatol Venereol 2013; 139 Suppl 5:S202-16. [PMID: 23522707 DOI: 10.1016/s0151-9638(12)70135-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
This paper is based on a review of the literature focused on pediatric dermatology, from October 2011 to september 2012. Our objective was to highlight the main advances in fields such as atopic dermatitis, infantile hemangiomas, infectious diseases, inflammatory disorders, and genodermatoses.
Collapse
Affiliation(s)
- S Barbarot
- Clinique dermatologique, Hôtel Dieu, CHU Nantes, Place Alexis Ricordeau, 44000 Nantes, France.
| |
Collapse
|
771
|
Hubo M, Trinschek B, Kryczanowsky F, Tuettenberg A, Steinbrink K, Jonuleit H. Costimulatory molecules on immunogenic versus tolerogenic human dendritic cells. Front Immunol 2013; 4:82. [PMID: 23565116 PMCID: PMC3615188 DOI: 10.3389/fimmu.2013.00082] [Citation(s) in RCA: 167] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2012] [Accepted: 03/20/2013] [Indexed: 12/12/2022] Open
Abstract
Dendritic cells (DC) are sentinels of immunity, essential for homeostasis of T cell-dependent immune responses. Both functions of DC, initiation of antigen-specific T cell immunity and maintenance of tissue-specific tolerance originate from distinct stages of differentiation, immunogenic versus tolerogenic. Dependent on local micro milieu and inflammatory stimuli, tissue resident immature DC with functional plasticity differentiate into tolerogenic or immunogenic DC with stable phenotypes. They efficiently link innate and adaptive immunity and are ideally positioned to modify T cell-mediated immune responses. Since the T cell stimulatory properties of DC are significantly influenced by their expression of signal II ligands, it is critical to understand the impact of distinct costimulatory pathways on DC function. This review gives an overview of functional different human DC subsets with unique profiles of costimulatory molecules and outlines how different costimulatory pathways together with the immunosuppressive cytokine IL-10 bias immunogenic versus tolerogenic DC functions. Furthermore, we exemplarily describe protocols for the generation of two well-defined monocyte-derived DC subsets for their clinical use, immunogenic versus tolerogenic.
Collapse
Affiliation(s)
- Mario Hubo
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University Mainz Mainz, Germany
| | | | | | | | | | | |
Collapse
|
772
|
Misery L. [What's new in dermatological research?]. Ann Dermatol Venereol 2013; 139 Suppl 5:S188-93. [PMID: 23522705 DOI: 10.1016/s0151-9638(12)70133-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Dermatological research is more and more productive and its level higher and higher. Choosing the most significant articles is difficult. Mast cell plays a role in the initiation of inflammation and therefore in poorer healing. Keratinocytes derive from stem cells and progenitors, which are independent. They can be activated directly by heat through sensory proteins at their surface. The cutaneous nervous system has an organization similar to that of the most complex sensory organs. In psoriasis, denervation induces a significant plaque regression. The cerebral integration of skin appearance modulates the skin reactivity to histamine. Pruritus is linked to specific receptors in the skin, which give specific projections into the brain and are histamine-dependent or not. Atopic dermatitis may be linked to the nonspecific activation of Th2 immune system, particularly to abnormalities of the skin barrier. Skin bacteria, but not intestinal, modulate the formation of skin immunity. Raf kinases are well known in melanoma and play an important role in physiological conditions: they are not essential to the initial development of the melanocyte lineage but to maintain it. In culture, melanocytes can be dedifferentiated in melanoblasts. Sunburns are consecutive to the activation of TLR3 by UVB. ANRIL gene is involved in the polymorphism of neurofibromatosis 1 and gene RAD51B is linked to the risk of male breast cancer. MCV infection is linked to sites with sialic acid. Aging objectified by telomere shortening is accelerated by stress.
Collapse
Affiliation(s)
- L Misery
- Service de Dermatologie, CHU de Brest, 2, avenue Foch, 29200 Brest, France.
| |
Collapse
|
773
|
Affiliation(s)
- Nicolas Bouladoux
- Mucosal Immunology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 4 Center Drive, Bethesda, MD 20892, États-Unis.
| | | | | | | |
Collapse
|
774
|
Khosravi A, Mazmanian SK. Disruption of the gut microbiome as a risk factor for microbial infections. Curr Opin Microbiol 2013; 16:221-7. [PMID: 23597788 PMCID: PMC5695238 DOI: 10.1016/j.mib.2013.03.009] [Citation(s) in RCA: 149] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2013] [Revised: 03/22/2013] [Accepted: 03/22/2013] [Indexed: 12/22/2022]
Abstract
The discovery that microorganisms can be etiologic agents of disease has driven clinical, research and public health efforts to reduce exposure to bacteria. However, despite extensive campaigns to eradicate pathogens (via antibiotics, vaccinations, hygiene, sanitation, etc.), the incidence and/or severity of multiple immune-mediated diseases including, paradoxically, infectious disease have increased in recent decades. We now appreciate that most microbes in our environment are not pathogenic, and that many human-associated bacteria are symbiotic or beneficial. Notably, recent examples have emerged revealing that the microbiome augments immune system function. This review will focus on how commensal-derived signals enhance various aspects of the host response against pathogens. We suggest that modern lifestyle advances may be depleting specific microbes that enhance immunity against pathogens. Validation of the notion that absence of beneficial microbes is a risk factor for infectious disease may have broad implications for future medical practices.
Collapse
Affiliation(s)
- Arya Khosravi
- Division of Biology, California Institute of Technology, Pasadena, California, 91125
| | - Sarkis K. Mazmanian
- Division of Biology, California Institute of Technology, Pasadena, California, 91125
| |
Collapse
|
775
|
Gonzalez-Lombana C, Gimblet C, Bacellar O, Oliveira WW, Passos S, Carvalho LP, Goldschmidt M, Carvalho EM, Scott P. IL-17 mediates immunopathology in the absence of IL-10 following Leishmania major infection. PLoS Pathog 2013; 9:e1003243. [PMID: 23555256 PMCID: PMC3605236 DOI: 10.1371/journal.ppat.1003243] [Citation(s) in RCA: 123] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Accepted: 01/29/2013] [Indexed: 12/31/2022] Open
Abstract
Leishmaniasis, resulting from infection with the protozoan parasite Leishmania, consists of a wide spectrum of clinical manifestations, from healing cutaneous lesions to fatal visceral infections. A particularly severe form of cutaneous leishmaniasis, termed mucosal leishmaniasis, exhibits decreased IL-10 levels and an exaggerated inflammatory response that perpetuates the disease. Using a mouse model of leishmaniasis, we investigated what cytokines contribute to increased pathology when IL-10-mediated regulation is absent. Leishmania major infected C57BL/6 mice lacking IL-10 regulation developed larger lesions than controls, but fewer parasites. Both IFN-γ and IL-17 levels were substantially elevated in mice lacking the capacity to respond to IL-10. IFN-γ promoted an increased infiltration of monocytes, while IL-17 contributed to an increase in neutrophils. Surprisingly, however, we found that IFN-γ did not contribute to increased pathology, but instead regulated the IL-17 response. Thus, blocking IFN-γ led to a significant increase in IL-17, neutrophils and disease. Similarly, the production of IL-17 by cells from leishmaniasis patients was also regulated by IL-10 and IFN-γ. Additional studies found that the IL-1 receptor was required for both the IL-17 response and increased pathology. Therefore, we propose that regulating IL-17, possibly by downregulating IL-1β, may be a useful approach for controlling immunopathology in leishmaniasis. Leishmaniasis is a tropical disease transmitted by sand flies that causes visceral and cutaneous lesions. In humans, the most severe form of cutaneous leishmaniasis is the mucosal form, causing disfiguring lesions in the nasal and oral mucosa. Why these patients develop severe disease is not clear. It is known, however, that the severe disease is not due to an overwhelming number of parasites, but rather appears to be due to an uncontrolled inflammatory response that includes elevated production of IFN-γ and IL-17. Here, we used a murine model of leishmaniasis to identify the factors involved in this pathology, and found that mice infected with Leishmania major developed severe lesions in the absence of IL-10 or IL-10 signaling, and similar to patients, contained high levels of IFN-γ and IL-17. While both of these cytokines have the potential to induce pathology, we found that IL-17 was responsible for the severe pathology seen in the absence of IL-10 regulation, and furthermore that IL-17 levels were higher and pathology greater in the absence of IFN-γ. Thus, our study suggests that IL-17, but not the IFN-γ, is a strong candidate to be targeted in strategies to control the severe immunopathology observed in mucosal leishmaniasis patients.
Collapse
Affiliation(s)
- Claudia Gonzalez-Lombana
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Ciara Gimblet
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Olivia Bacellar
- Instituto Nacional de Ciência e Tecnologia de Doenças Tropicais-INCT-DT(CNPq/MCT), Serviço de Imunologia, Hospital Universitario Prof. Edgard Santos, Universidade Federal da Bahia Salvador, Bahia, Brasil
| | - Walker W. Oliveira
- Instituto Nacional de Ciência e Tecnologia de Doenças Tropicais-INCT-DT(CNPq/MCT), Serviço de Imunologia, Hospital Universitario Prof. Edgard Santos, Universidade Federal da Bahia Salvador, Bahia, Brasil
| | - Sara Passos
- Instituto Nacional de Ciência e Tecnologia de Doenças Tropicais-INCT-DT(CNPq/MCT), Serviço de Imunologia, Hospital Universitario Prof. Edgard Santos, Universidade Federal da Bahia Salvador, Bahia, Brasil
| | - Lucas P. Carvalho
- Instituto Nacional de Ciência e Tecnologia de Doenças Tropicais-INCT-DT(CNPq/MCT), Serviço de Imunologia, Hospital Universitario Prof. Edgard Santos, Universidade Federal da Bahia Salvador, Bahia, Brasil
| | - Michael Goldschmidt
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Edgar M. Carvalho
- Instituto Nacional de Ciência e Tecnologia de Doenças Tropicais-INCT-DT(CNPq/MCT), Serviço de Imunologia, Hospital Universitario Prof. Edgard Santos, Universidade Federal da Bahia Salvador, Bahia, Brasil
| | - Phillip Scott
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
776
|
Chen YE, Tsao H. The skin microbiome: current perspectives and future challenges. J Am Acad Dermatol 2013; 69:143-55. [PMID: 23489584 DOI: 10.1016/j.jaad.2013.01.016] [Citation(s) in RCA: 117] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2012] [Revised: 12/19/2012] [Accepted: 01/06/2013] [Indexed: 02/08/2023]
Abstract
Complex communities of bacteria, fungi, and viruses thrive on our skin. The composition of these communities depends on skin characteristics, such as sebaceous gland concentration, moisture content, and temperature, as well as on host genetics and exogenous environmental factors. Recent metagenomic studies have uncovered a surprising diversity within these ecosystems and have fostered a new view of commensal organisms as playing a much larger role in immune modulation and epithelial health than previously expected. Understanding microbe-host interactions and discovering the factors that drive microbial colonization will help us understand the pathogenesis of skin diseases and develop new promicrobial and antimicrobial therapeutics.
Collapse
Affiliation(s)
- Yiyin Erin Chen
- Medical Scientist Training Program, Health Sciences and Technology, Harvard Medical School, Boston, Massachusetts, USA
| | | |
Collapse
|
777
|
Abstract
Commensal bacteria are necessary for the development and maintenance of a healthy immune system. Harnessing the ability of microbiota to affect host immunity is considered an important therapeutic strategy for many mucosal and nonmucosal immune-related conditions, such as inflammatory bowel diseases (IBDs), celiac disease, metabolic syndrome, diabetes, and microbial infections. In addition to well-established immunostimulatory effects of the microbiota, the presence of individual mutualistic commensal bacteria with immunomodulatory effects has been described. These organisms are permanent members of the commensal microbiota and affect host immune homeostasis in specific ways. Identification of individual examples of such immunomodulatory commensals and understanding their mechanisms of interaction with the host will be invaluable in designing therapeutic strategies to reverse intestinal dysbiosis and recover immunological homeostasis.
Collapse
Affiliation(s)
- Ivaylo I Ivanov
- Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY 10032, USA.
| | | |
Collapse
|
778
|
Jain R, Weninger W. Shedding light on cutaneous innate immune responses: the intravital microscopy approach. Immunol Cell Biol 2013; 91:263-70. [PMID: 23459295 DOI: 10.1038/icb.2012.76] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The skin is under constant assault by environmental factors and microbes. Innate immune cells in epidermis and dermis regulate immune responses against pathogens while maintaining tolerance against commensal bacteria and autoantigens. The introduction of intravital imaging approaches, in particular multiphoton microscopy, has enabled studying the cellular and molecular regulation of cutaneous immunity in real time within intact skin. Here, we discuss recent advances in our understanding of innate immune cell behaviour in the skin, as unravelled by intravital microscopy, with emphasis on the function of myeloid cells, including dendritic cells, neutrophils and monocytes.
Collapse
Affiliation(s)
- Rohit Jain
- Immune Imaging Program, The Centenary Institute, Newtown, New South Wales, Australia
| | | |
Collapse
|
779
|
Russell SL, Gold MJ, Willing BP, Thorson L, McNagny KM, Finlay BB. Perinatal antibiotic treatment affects murine microbiota, immune responses and allergic asthma. Gut Microbes 2013; 4:158-64. [PMID: 23333861 PMCID: PMC3595077 DOI: 10.4161/gmic.23567] [Citation(s) in RCA: 197] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
There is convincing evidence from recent human and animal studies that suggests the intestinal microbiota plays an important role in regulating immune responses associated with the development of allergic asthma, particularly during early infancy. Although identifying the mechanistic link between host-microbe interactions in the gut and lung mucosal tissues has proved challenging, several very recent studies are now providing significant insights. We have shown that administering vancomycin to mice early in life shifts resident gut flora and enhances future susceptibility to allergic asthma. This effect was not observed in mice given another antibiotic, streptomycin, nor when either antibiotic was administered to adult mice. In this addendum, we further analyze the link between early life administration of vancomycin and future susceptibility to asthma and describe how specific immune cell populations, which have been implicated in other asthma-related microbiota studies, are affected. We propose that shifts in gut microbiota exacerbate asthma-related immune responses when they occur shortly after birth and before weaning (perinatal period), and suggest that these effects may be mediated, at least in the case of vancomycin, by elevated serum IgE and reduced regulatory T cell populations.
Collapse
Affiliation(s)
- Shannon L. Russell
- Department of Microbiology and Immunology; University of British Columbia; Vancouver, BC Canada,Michael Smith Laboratories; University of British Columbia; Vancouver, BC Canada
| | - Matthew J. Gold
- The Biomedical Research Center; University of British Columbia; Vancouver, BC Canada
| | - Benjamin P. Willing
- Michael Smith Laboratories; University of British Columbia; Vancouver, BC Canada
| | - Lisa Thorson
- Michael Smith Laboratories; University of British Columbia; Vancouver, BC Canada
| | - Kelly M. McNagny
- The Biomedical Research Center; University of British Columbia; Vancouver, BC Canada,Correspondence to: Kelly M. McNagny, and Brett B. Finlay,
| | - Brett B. Finlay
- Department of Microbiology and Immunology; University of British Columbia; Vancouver, BC Canada,Michael Smith Laboratories; University of British Columbia; Vancouver, BC Canada,Correspondence to: Kelly M. McNagny, and Brett B. Finlay,
| |
Collapse
|
780
|
Hartley MA, Kohl K, Ronet C, Fasel N. The therapeutic potential of immune cross-talk in leishmaniasis. Clin Microbiol Infect 2013; 19:119-30. [DOI: 10.1111/1469-0691.12095] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2012] [Revised: 10/31/2012] [Accepted: 11/01/2012] [Indexed: 11/30/2022]
|
781
|
Abt MC, Artis D. The dynamic influence of commensal bacteria on the immune response to pathogens. Curr Opin Microbiol 2013; 16:4-9. [PMID: 23332724 DOI: 10.1016/j.mib.2012.12.002] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2012] [Accepted: 12/20/2012] [Indexed: 12/17/2022]
Abstract
Alterations in the composition of commensal bacterial communities are associated with enhanced susceptibility to multiple inflammatory, allergic, metabolic and infectious diseases in humans. In the context of infection, commensal bacteria-derived signals can influence the host immune response to invasive pathogens by acting as an adjuvant to boost the immune response to infection or by providing tonic stimulation to induce basal expression of factors required for host defense. Conversely, some pathogens have evolved mechanisms that can utilize commensal bacteria to establish a replicative advantage within the host. Thus, examining the dynamic relationship that exists between the mammalian host, commensal bacteria and invasive pathogens can provide insights into the etiology of pathogenesis from an infection.
Collapse
Affiliation(s)
- Michael C Abt
- Department of Microbiology and Institute for Immunology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | |
Collapse
|
782
|
Mueller SN. Effector T-cell responses in non-lymphoid tissues: insights from in vivo imaging. Immunol Cell Biol 2013; 91:290-6. [PMID: 23295362 DOI: 10.1038/icb.2012.75] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
T-cell responses are initiated within secondary lymphoid organs, and effector T-cells are released into the circulation where they home to inflamed tissues and mediate protective immune responses. Within non-lymphoid tissues, the types of cellular interactions and the dynamics that lead to clearance of infections are still relatively poorly understood. Here I review how imaging of effector T-cells within tissues has contributed to our understanding of immune responses, and examine some of the remaining questions that may benefit from in vivo imaging to reveal the intricacies of how immune cells function. A detailed understanding of the dynamics of T-cell responses within non-lymphoid tissues is important for the rational design of targeted therapies that influence key steps in disease progression.
Collapse
Affiliation(s)
- Scott N Mueller
- Department of Microbiology and Immunology, The University of Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|
783
|
|
784
|
|
785
|
Yamazaki S, Maruyama A, Okada K, Matsumoto M, Morita A, Seya T. Dendritic cells from oral cavity induce Foxp3(+) regulatory T cells upon antigen stimulation. PLoS One 2012; 7:e51665. [PMID: 23272135 PMCID: PMC3525649 DOI: 10.1371/journal.pone.0051665] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2012] [Accepted: 11/06/2012] [Indexed: 02/06/2023] Open
Abstract
Evidence is accumulating that dendritic cells (DCs) from the intestines have the capacity to induce Foxp3(+)CD4(+) regulatory T cells (T-regs) and regulate immunity versus tolerance in the intestines. However, the contribution of DCs to controlling immunity versus tolerance in the oral cavity has not been addressed. Here, we report that DCs from the oral cavity induce Foxp3(+) T-regs as well as DCs from intestine. We found that oral-cavity-draining cervical lymph nodes contained higher frequencies of Foxp3(+) T-regs and ROR-γt(+) CD4(+)T cells than other lymph nodes. The high frequency of Foxp3(+) T-regs in the oral-cavity-draining cervical lymph nodes was not dependent on the Toll like receptor (TLR) adaptor molecules, Myd88 and TICAM-1 (TRIF). In contrast, the high frequency of ROR-γt(+) CD4(+)T cells relies on Myd88 and TICAM-1. In vitro data showed that CD11c(+) DCs from oral-cavity-draining cervical lymph nodes have the capacity to induce Foxp3(+) T-regs in the presence of antigen. These data suggest that, as well as in the intestinal environment, antigen-presenting DCs may play a vital role in maintaining tolerance by inducing Foxp3(+) T-regs in the oral cavity.
Collapse
Affiliation(s)
- Sayuri Yamazaki
- Department of Microbiology and Immunology, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
- Department of Geriatric and Environmental Dermatology, Nagoya City University, Graduate School of Medical Sciences, Nagoya, Japan
- * E-mail: (TS); (SY)
| | - Akira Maruyama
- Department of Microbiology and Immunology, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Kohei Okada
- Department of Microbiology and Immunology, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Misako Matsumoto
- Department of Microbiology and Immunology, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Akimichi Morita
- Department of Geriatric and Environmental Dermatology, Nagoya City University, Graduate School of Medical Sciences, Nagoya, Japan
| | - Tsukasa Seya
- Department of Microbiology and Immunology, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
- * E-mail: (TS); (SY)
| |
Collapse
|
786
|
Issa F, Robb RJ, Wood KJ. The where and when of T cell regulation in transplantation. Trends Immunol 2012; 34:107-13. [PMID: 23228885 DOI: 10.1016/j.it.2012.11.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2012] [Revised: 11/08/2012] [Accepted: 11/13/2012] [Indexed: 01/03/2023]
Abstract
Multiple cell types contribute to the peripheral regulation of T cell alloresponses in haematopoieitc cell transplantation (HCT) and solid organ transplantation (SOT). Of these, regulatory T cells (Tregs) are the principal players and have shown the greatest success in the therapeutic control of detrimental immune responses. Investigations into the induction, location, and mechanism of suppression utilised by Tregs to control alloreactive responses are ongoing. The activation and homing characteristics of Tregs are important to their regulatory capabilities, with activation and homing occurring in the same time and space as conventional T cells. This review discusses these characteristics and recent advances in the field as we move closer to the ultimate goal of utilising Tregs as treatment for allograft rejection and graft-versus-host disease (GvHD).
Collapse
Affiliation(s)
- Fadi Issa
- Transplantation Research Immunology Group, Nuffield Department of Surgical Sciences, University of Oxford, UK
| | | | | |
Collapse
|
787
|
Nylén S, Eidsmo L. Tissue damage and immunity in cutaneous leishmaniasis. Parasite Immunol 2012; 34:551-61. [DOI: 10.1111/pim.12007] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2012] [Accepted: 09/17/2012] [Indexed: 12/21/2022]
Affiliation(s)
- S. Nylén
- Department of Microbiology; Tumor and Cell Biology; Karolinska Institutet; Stockholm; Sweden
| | - L. Eidsmo
- Molecular Dermatology; Department of Medicine Solna; Karolinska Institutet; Stockholm; Sweden
| |
Collapse
|
788
|
Yazdi AS. [Skin bacteria direct the immune response]. Hautarzt 2012; 63:985-7. [PMID: 23160452 DOI: 10.1007/s00105-012-2490-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Affiliation(s)
- A S Yazdi
- Universitäts-Hautklinik, Liebermeisterstr. 25, 72076, Tübingen, Deutschland.
| |
Collapse
|
789
|
Otto M. Coagulase-negative staphylococci as reservoirs of genes facilitating MRSA infection: Staphylococcal commensal species such as Staphylococcus epidermidis are being recognized as important sources of genes promoting MRSA colonization and virulence. Bioessays 2012; 35:4-11. [PMID: 23165978 DOI: 10.1002/bies.201200112] [Citation(s) in RCA: 157] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Recent research has suggested that Staphylococcus epidermidis is a reservoir of genes that, after horizontal transfer, facilitate the potential of Staphylococcus aureus to colonize, survive during infection, or resist antibiotic treatment, traits that are notably manifest in methicillin-resistant S. aureus (MRSA). S. aureus is a dangerous human pathogen and notorious for acquiring antibiotic resistance. MRSA in particular is one of the most frequent causes of morbidity and death in hospitalized patients. S. aureus is an extremely versatile pathogen with a multitude of mechanisms to cause disease and circumvent immune defenses. In contrast, most other staphylococci, such as S. epidermidis, are commonly benign commensals and only occasionally cause disease. Recent findings highlight the key importance of efforts to better understand how genes of staphylococci other than S. aureus contribute to survival in the human host, how they are transferred to S. aureus, and why this exchange appears to be uni-directional.
Collapse
Affiliation(s)
- Michael Otto
- Pathogen Molecular Genetics Section, Laboratory of Human Bacterial Pathogenesis, National Institute of Allergy and Infectious Diseases, The National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
790
|
Zeeuwen PLJM, Boekhorst J, van den Bogaard EH, de Koning HD, van de Kerkhof PMC, Saulnier DM, van Swam II, van Hijum SAFT, Kleerebezem M, Schalkwijk J, Timmerman HM. Microbiome dynamics of human epidermis following skin barrier disruption. Genome Biol 2012; 13:R101. [PMID: 23153041 PMCID: PMC3580493 DOI: 10.1186/gb-2012-13-11-r101] [Citation(s) in RCA: 174] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2012] [Revised: 10/01/2012] [Accepted: 11/15/2012] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Recent advances in sequencing technologies have enabled metagenomic analyses of many human body sites. Several studies have catalogued the composition of bacterial communities of the surface of human skin, mostly under static conditions in healthy volunteers. Skin injury will disturb the cutaneous homeostasis of the host tissue and its commensal microbiota, but the dynamics of this process have not been studied before. Here we analyzed the microbiota of the surface layer and the deeper layers of the stratum corneum of normal skin, and we investigated the dynamics of recolonization of skin microbiota following skin barrier disruption by tape stripping as a model of superficial injury. RESULTS We observed gender differences in microbiota composition and showed that bacteria are not uniformly distributed in the stratum corneum. Phylogenetic distance analysis was employed to follow microbiota development during recolonization of injured skin. Surprisingly, the developing neo-microbiome at day 14 was more similar to that of the deeper stratum corneum layers than to the initial surface microbiome. In addition, we also observed variation in the host response towards superficial injury as assessed by the induction of antimicrobial protein expression in epidermal keratinocytes. CONCLUSIONS We suggest that the microbiome of the deeper layers, rather than that of the superficial skin layer, may be regarded as the host indigenous microbiome. Characterization of the skin microbiome under dynamic conditions, and the ensuing response of the microbial community and host tissue, will shed further light on the complex interaction between resident bacteria and epidermis.
Collapse
Affiliation(s)
- Patrick LJM Zeeuwen
- Nijmegen Centre for Molecular Life Sciences (NCMLS), Radboud University Nijmegen Medical Centre, PO BOX 9101, 6500 HB Nijmegen, The Netherlands
- Nijmegen Institute for Infection, Inflammation and Immunity (N4i), Radboud University Nijmegen Medical Centre, PO BOX 9101, 6500 HB Nijmegen, The Netherlands
| | - Jos Boekhorst
- Nijmegen Centre for Molecular Life Sciences (NCMLS), Radboud University Nijmegen Medical Centre, PO BOX 9101, 6500 HB Nijmegen, The Netherlands
- Centre for Molecular and Biomolecular Informatics (CMBI), Radboud University Nijmegen Medical Centre, PO BOX 9101, 6500 HB Nijmegen, The Netherlands
- NIZO Food Research B.V., Kernhemseweg 2, 6718 ZB, Ede, The Netherlands
| | - Ellen H van den Bogaard
- Nijmegen Centre for Molecular Life Sciences (NCMLS), Radboud University Nijmegen Medical Centre, PO BOX 9101, 6500 HB Nijmegen, The Netherlands
- Nijmegen Institute for Infection, Inflammation and Immunity (N4i), Radboud University Nijmegen Medical Centre, PO BOX 9101, 6500 HB Nijmegen, The Netherlands
| | - Heleen D de Koning
- Nijmegen Centre for Molecular Life Sciences (NCMLS), Radboud University Nijmegen Medical Centre, PO BOX 9101, 6500 HB Nijmegen, The Netherlands
- Nijmegen Institute for Infection, Inflammation and Immunity (N4i), Radboud University Nijmegen Medical Centre, PO BOX 9101, 6500 HB Nijmegen, The Netherlands
| | - Peter MC van de Kerkhof
- Nijmegen Institute for Infection, Inflammation and Immunity (N4i), Radboud University Nijmegen Medical Centre, PO BOX 9101, 6500 HB Nijmegen, The Netherlands
| | | | - Iris I van Swam
- NIZO Food Research B.V., Kernhemseweg 2, 6718 ZB, Ede, The Netherlands
| | - Sacha AFT van Hijum
- Nijmegen Centre for Molecular Life Sciences (NCMLS), Radboud University Nijmegen Medical Centre, PO BOX 9101, 6500 HB Nijmegen, The Netherlands
- Centre for Molecular and Biomolecular Informatics (CMBI), Radboud University Nijmegen Medical Centre, PO BOX 9101, 6500 HB Nijmegen, The Netherlands
- NIZO Food Research B.V., Kernhemseweg 2, 6718 ZB, Ede, The Netherlands
| | - Michiel Kleerebezem
- NIZO Food Research B.V., Kernhemseweg 2, 6718 ZB, Ede, The Netherlands
- Wageningen University, Host-Microbe Interactomics Group, De Elst 1, 6708 WD, Wageningen, The Netherlands
| | - Joost Schalkwijk
- Nijmegen Centre for Molecular Life Sciences (NCMLS), Radboud University Nijmegen Medical Centre, PO BOX 9101, 6500 HB Nijmegen, The Netherlands
- Nijmegen Institute for Infection, Inflammation and Immunity (N4i), Radboud University Nijmegen Medical Centre, PO BOX 9101, 6500 HB Nijmegen, The Netherlands
| | - Harro M Timmerman
- NIZO Food Research B.V., Kernhemseweg 2, 6718 ZB, Ede, The Netherlands
| |
Collapse
|
791
|
Hulcr J, Latimer AM, Henley JB, Rountree NR, Fierer N, Lucky A, Lowman MD, Dunn RR. A jungle in there: bacteria in belly buttons are highly diverse, but predictable. PLoS One 2012; 7:e47712. [PMID: 23144827 PMCID: PMC3492386 DOI: 10.1371/journal.pone.0047712] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2012] [Accepted: 09/13/2012] [Indexed: 11/18/2022] Open
Abstract
The belly button is one of the habitats closest to us, and yet it remains relatively unexplored. We analyzed bacteria and arachaea from the belly buttons of humans from two different populations sampled within a nation-wide citizen science project. We examined bacterial and archaeal phylotypes present and their diversity using multiplex pyrosequencing of 16S rDNA libraries. We then tested the oligarchy hypothesis borrowed from tropical macroecology, namely that the frequency of phylotypes in one sample of humans predicts its frequency in another independent sample. We also tested the predictions that frequent phylotypes (the oligarchs) tend to be common when present, and tend to be more phylogenetically clustered than rare phylotypes. Once rarefied to four hundred reads per sample, bacterial communities from belly buttons proved to be at least as diverse as communities known from other skin studies (on average 67 bacterial phylotypes per belly button). However, the belly button communities were strongly dominated by a few taxa: only 6 phylotypes occurred on >80% humans. While these frequent bacterial phylotypes (the archaea were all rare) are a tiny part of the total diversity of bacteria in human navels (<0.3% of phylotypes), they constitute a major portion of individual reads (∼1/3), and are predictable among independent samples of humans, in terms of both the occurrence and evolutionary relatedness (more closely related than randomly drawn equal sets of phylotypes). Thus, the hypothesis that “oligarchs” dominate diverse assemblages appears to be supported by human-associated bacteria. Although it remains difficult to predict which species of bacteria might be found on a particular human, predicting which species are most frequent (or rare) seems more straightforward, at least for those species living in belly buttons.
Collapse
Affiliation(s)
- Jiri Hulcr
- School of Forest Resources and Conservation, University of Florida, Gainesville, Florida, United States of America
- Department of Biology and Keck Center for Behavioral Biology, North Carolina State University, Raleigh, North Carolina, United States of America
| | - Andrew M. Latimer
- Department of Plant Sciences, University of California Davis, California, United States of America
| | - Jessica B. Henley
- Cooperative Institute for Research in Environmental Sciences, University of Colorado, Boulder, Colorado, United States of America
| | - Nina R. Rountree
- Department of Biology and Keck Center for Behavioral Biology, North Carolina State University, Raleigh, North Carolina, United States of America
| | - Noah Fierer
- Cooperative Institute for Research in Environmental Sciences, University of Colorado, Boulder, Colorado, United States of America
- Department of Ecology and Evolutionary Biology, University of Colorado, Boulder, Colorado, United States of America
| | - Andrea Lucky
- Department of Biology and Keck Center for Behavioral Biology, North Carolina State University, Raleigh, North Carolina, United States of America
- Department of Entomology, University of Florida, Gainesville, Florida, United States of America
| | - Margaret D. Lowman
- School of Forest Resources and Conservation, University of Florida, Gainesville, Florida, United States of America
- North Carolina Museum of Natural Sciences, Raleigh, North Carolina, United States of America
| | - Robert R. Dunn
- Department of Biology and Keck Center for Behavioral Biology, North Carolina State University, Raleigh, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
792
|
|
793
|
Local government. Nat Rev Immunol 2012; 12:615. [DOI: 10.1038/nri3289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
794
|
Hwang JS, Im CR, Im SH. Immune disorders and its correlation with gut microbiome. Immune Netw 2012; 12:129-38. [PMID: 23091436 PMCID: PMC3467411 DOI: 10.4110/in.2012.12.4.129] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2012] [Revised: 07/19/2012] [Accepted: 07/27/2012] [Indexed: 01/05/2023] Open
Abstract
Allergic disorders such as atopic dermatitis and asthma are common hyper-immune disorders in industrialized countries. Along with genetic association, environmental factors and gut microbiota have been suggested as major triggering factors for the development of atopic dermatitis. Numerous studies support the association of hygiene hypothesis in allergic immune disorders that a lack of early childhood exposure to diverse microorganism increases susceptibility to allergic diseases. Among the symbiotic microorganisms (e.g. gut flora or probiotics), probiotics confer health benefits through multiple action mechanisms including modification of immune response in gut associated lymphoid tissue (GALT). Although many human clinical trials and mouse studies demonstrated the beneficial effects of probiotics in diverse immune disorders, this effect is strain specific and needs to apply specific probiotics for specific allergic diseases. Herein, we briefly review the diverse functions and regulation mechanisms of probiotics in diverse disorders.
Collapse
Affiliation(s)
- Ji-Sun Hwang
- School of Life Sciences and Immune Synapse Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju 500-712, Korea
| | | | | |
Collapse
|
795
|
The skin’s secret surveillance system. Nature 2012. [DOI: 10.1038/nature.2012.11075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|