551
|
Salgado VR, Queiroz ATLD, Sanabani SS, Oliveira CID, Carvalho EM, Costa JML, Barral-Netto M, Barral A. The microbiological signature of human cutaneous leishmaniasis lesions exhibits restricted bacterial diversity compared to healthy skin. Mem Inst Oswaldo Cruz 2016; 111:241-51. [PMID: 27074253 PMCID: PMC4830113 DOI: 10.1590/0074-02760150436] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 03/01/2016] [Indexed: 02/07/2023] Open
Abstract
Localised cutaneous leishmaniasis (LCL) is the most common form of cutaneous
leishmaniasis characterised by single or multiple painless chronic ulcers, which
commonly presents with secondary bacterial infection. Previous culture-based studies
have found staphylococci, streptococci, and opportunistic pathogenic bacteria in LCL
lesions, but there have been no comparisons to normal skin. In addition, this
approach has strong bias for determining bacterial composition. The present study
tested the hypothesis that bacterial communities in LCL lesions differ from those
found on healthy skin (HS). Using a high throughput amplicon sequencing approach,
which allows for better populational evaluation due to greater depth coverage and the
Quantitative Insights Into Microbial Ecology pipeline, we compared the
microbiological signature of LCL lesions with that of contralateral HS from the same
individuals.Streptococcus,
Staphylococcus,Fusobacterium and other strict or
facultative anaerobic bacteria composed the LCL microbiome. Aerobic and facultative
anaerobic bacteria found in HS, including environmental bacteria, were significantly
decreased in LCL lesions (p < 0.01). This paper presents the first comprehensive
microbiome identification from LCL lesions with next generation sequence methodology
and shows a marked reduction of bacterial diversity in the lesions.
Collapse
Affiliation(s)
- Vanessa R Salgado
- Centro de Pesquisas Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, BA, Brasil
| | | | - Sabri S Sanabani
- Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil
| | | | - Edgar M Carvalho
- Centro de Pesquisas Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, BA, Brasil
| | - Jackson M L Costa
- Centro de Pesquisas Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, BA, Brasil
| | - Manoel Barral-Netto
- Centro de Pesquisas Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, BA, Brasil
| | - Aldina Barral
- Centro de Pesquisas Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, BA, Brasil
| |
Collapse
|
552
|
McIntyre MK, Peacock TJ, Akers KS, Burmeister DM. Initial Characterization of the Pig Skin Bacteriome and Its Effect on In Vitro Models of Wound Healing. PLoS One 2016; 11:e0166176. [PMID: 27824921 PMCID: PMC5100914 DOI: 10.1371/journal.pone.0166176] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 10/24/2016] [Indexed: 02/01/2023] Open
Abstract
Elucidating the roles and composition of the human skin microbiome has revealed a delicate interplay between resident microbes and wound healing. Evolutionarily speaking, normal cutaneous flora likely has been selected for because it potentiates or, at minimum, does not impede wound healing. While pigs are the gold standard model for wound healing studies, the porcine skin microbiome has not been studied in detail. Herein, we performed 16S rDNA sequencing to characterize the pig skin bacteriome at several anatomical locations. Additionally, we used bacterial conditioned-media with in vitro techniques to examine the paracrine effects of bacterial-derived proteins on human keratinocytes (NHEK) and fibroblasts (NHDF). We found that at the phyla level, the pig skin bacteriome is similar to that of humans and largely consists of Firmicutes (55.6%), Bacteroidetes (20.8%), Actinobacteria (13.3%), and Proteobacteria (5.1%) however species-level differences between anatomical locations exist. Studies of bacterial supernatant revealed location-dependent effects on NHDF migration and NHEK apoptosis and growth factor release. These results expand the limited knowledge of the cutaneous bacteriome of healthy swine, and suggest that naturally occurring bacterial flora affects wound healing differentially depending on anatomical location. Ultimately, the pig might be considered the best surrogate for not only wound healing studies but also the cutaneous microbiome. This would not only facilitate investigations into the microbiome’s role in recovery from injury, but also provide microbial targets for enhancing or accelerating wound healing.
Collapse
Affiliation(s)
- Matthew K. McIntyre
- Damage Control Resuscitation, United States Army Institute of Surgical Research, Fort Sam Houston, Texas, United States of America
| | - Trent J. Peacock
- Dental Trauma Research Detachment, United States Army Institute of Surgical Research, Fort Sam Houston, Texas, United States of America
| | - Kevin S. Akers
- Extremity Trauma and Regenerative Medicine, United States Army Institute of Surgical Research, Fort Sam Houston, Texas, United States of America
| | - David M. Burmeister
- Damage Control Resuscitation, United States Army Institute of Surgical Research, Fort Sam Houston, Texas, United States of America
- * E-mail:
| |
Collapse
|
553
|
Gonçalves SM, Lagrou K, Duarte-Oliveira C, Maertens JA, Cunha C, Carvalho A. The microbiome-metabolome crosstalk in the pathogenesis of respiratory fungal diseases. Virulence 2016; 8:673-684. [PMID: 27820674 DOI: 10.1080/21505594.2016.1257458] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Filamentous fungi of the genus Aspergillus are responsible for several superficial and invasive infections and allergic syndromes. The risk of infection and its clinical outcome vary significantly even among patients with similar predisposing clinical factors and pathogen exposure. There is increasing evidence that the individual microbiome supervises the outcome of the host-fungus interaction by influencing mechanisms of immune regulation, inflammation, metabolism, and other physiological processes. Microbiome-mediated mechanisms of resistance allow therefore the control of fungal colonization, preventing the onset of overt disease, particularly in patients with underlying immune dysfunction. Here, we review this emerging area of research and discuss the contribution of the microbiota (and its dysbiosis), including its immunoregulatory properties and relationship with the metabolic activity of commensals, to respiratory fungal diseases. Finally, we highlight possible strategies aimed at decoding the microbiome-metabolome dialog and at its exploitation toward personalized medical interventions in patients at high risk of infection.
Collapse
Affiliation(s)
- Samuel M Gonçalves
- a Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho , Braga , Portugal.,b ICVS/3B's - PT Government Associate Laboratory , Guimarães , Portugal
| | - Katrien Lagrou
- c Department of Microbiology and Immunology , KU Leuven - University of Leuven , Leuven , Belgium.,d Department of Laboratory Medicine and National Reference Center for Medical Mycology , University Hospitals Leuven , Leuven , Belgium
| | - Cláudio Duarte-Oliveira
- a Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho , Braga , Portugal.,b ICVS/3B's - PT Government Associate Laboratory , Guimarães , Portugal
| | - Johan A Maertens
- e Department of Hematology , University Hospitals Leuven , Leuven , Belgium
| | - Cristina Cunha
- a Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho , Braga , Portugal.,b ICVS/3B's - PT Government Associate Laboratory , Guimarães , Portugal
| | - Agostinho Carvalho
- a Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho , Braga , Portugal.,b ICVS/3B's - PT Government Associate Laboratory , Guimarães , Portugal
| |
Collapse
|
554
|
Nakatsuji T, Chen TH, Two AM, Chun KA, Narala S, Geha RS, Hata TR, Gallo RL. Staphylococcus aureus Exploits Epidermal Barrier Defects in Atopic Dermatitis to Trigger Cytokine Expression. J Invest Dermatol 2016; 136:2192-2200. [PMID: 27381887 PMCID: PMC5103312 DOI: 10.1016/j.jid.2016.05.127] [Citation(s) in RCA: 248] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Revised: 05/09/2016] [Accepted: 05/22/2016] [Indexed: 01/24/2023]
Abstract
Patients with atopic dermatitis (AD) have an abnormal skin barrier and are frequently colonized by S. aureus. In this study we investigated if S. aureus penetrates the epidermal barrier of subjects with AD and sought to understand the mechanism and functional significance of this entry. S. aureus was observed to be more abundant in the dermis of lesional skin from AD patients. Bacterial entry past the epidermis was observed in cultured human skin equivalents and in mice but was found to be increased in the skin of cathelicidin knockout and ovalbumin-sensitized filaggrin mutant mice. S. aureus penetration through the epidermis was dependent on bacterial viability and protease activity, because killed bacteria and a protease-null mutant strain of S. aureus were unable to penetrate. Entry of S. aureus directly correlated with increased expression of IL-4, IL-13, IL-22, thymic stromal lymphopoietin, and other cytokines associated with AD and with decreased expression of cathelicidin. These data illustrate how abnormalities of the epidermal barrier in AD can alter the balance of S. aureus entry into the dermis and provide an explanation for how such dermal dysbiosis results in increased inflammatory cytokines and exacerbation of disease.
Collapse
Affiliation(s)
- Teruaki Nakatsuji
- Department of Dermatology, University of California-San Diego, California, USA
| | - Tiffany H Chen
- Department of Dermatology, University of California-San Diego, California, USA
| | - Aimee M Two
- Department of Dermatology, University of California-San Diego, California, USA
| | - Kimberly A Chun
- Department of Dermatology, University of California-San Diego, California, USA
| | - Saisindhu Narala
- Department of Dermatology, University of California-San Diego, California, USA
| | - Raif S Geha
- Division of Immunology, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Tissa R Hata
- Department of Dermatology, University of California-San Diego, California, USA
| | - Richard L Gallo
- Department of Dermatology, University of California-San Diego, California, USA.
| |
Collapse
|
555
|
Pellegatta T, Saler M, Bonfanti V, Nicoletti G, Faga A. Novel perspectives on the role of the human microbiota in regenerative medicine and surgery. Biomed Rep 2016; 5:519-524. [PMID: 27882211 PMCID: PMC5103662 DOI: 10.3892/br.2016.778] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 09/21/2016] [Indexed: 12/19/2022] Open
Abstract
Plastic surgery is transitioning from a fine craftsmanship to a regenerative science. In wound healing, the role of microorganisms is no longer considered to be just counteracting, but also promoting. Furthermore, host-microbe interactions are essential for numerous aspects of normal mammalian physiology, from metabolic activity to immune homeostasis. Each area of the human body hosts a unique microbial community, and the composition of microbiota is dependent on the host, age and the anatomical area, and it changes according to the characteristics of the microenvironment. Every squared centimeter of skin contains ~1 billion bacteria. The majority of microorganisms of the skin are commensal or temporary passing members. Skin flora mechanisms interacting or influencing the human physical skin barrier are not well defined. Resident skin bacteria provide the first line of defence against potentially dangerous pathogens and produce small molecules that influence their microbial neighbours. Furthermore, the microbiota activates and assists innate immunity and influences adaptive immunity. Various types of immune and non-immune cells contribute to wound healing. The proliferative phase of wound healing is inversely proportional to the extent of the post-traumatic inflammatory reaction. Topical bacterial lipopolysaccharide application markedly affects wound healing by accelerating the resolution of inflammation, increasing macrophage infiltration, enhancing collagen synthesis and altering the secretion of mediators involved in skin regeneration. Various studies have investigated the biological contents of thermal spring waters, and their anti-inflammatory and immune protective roles. In addition, the regenerative properties of thermal spring waters were analysed in an experimental animal wound model. The areas treated with thermal water healed faster than the areas treated with conventional dressings, and exhibited a collagen and elastic fiber network comparable with the normal skin. Thus, the microbial environment may be considered as a potential tool in regenerative medicine and surgery.
Collapse
Affiliation(s)
- Tommaso Pellegatta
- Department of Clinical-Surgical, Diagnostic and Pediatric Sciences, Plastic Surgery Unit, University of Pavia, Pavia, I-27100 Lombardy, Italy
| | - Marco Saler
- Department of Clinical-Surgical, Diagnostic and Pediatric Sciences, Plastic Surgery Unit, University of Pavia, Pavia, I-27100 Lombardy, Italy
| | - Viola Bonfanti
- Department of Clinical-Surgical, Diagnostic and Pediatric Sciences, Plastic Surgery Unit, University of Pavia, Pavia, I-27100 Lombardy, Italy
| | - Giovanni Nicoletti
- Department of Clinical-Surgical, Diagnostic and Pediatric Sciences, Plastic Surgery Unit, University of Pavia, Pavia, I-27100 Lombardy, Italy
- Plastic and Reconstructive Surgery Unit, Salvatore Maugeri Research and Care Institute, Pavia, I-27100 Lombardy, Italy
| | - Angela Faga
- Department of Clinical-Surgical, Diagnostic and Pediatric Sciences, Plastic Surgery Unit, University of Pavia, Pavia, I-27100 Lombardy, Italy
- Plastic and Reconstructive Surgery Unit, Salvatore Maugeri Research and Care Institute, Pavia, I-27100 Lombardy, Italy
| |
Collapse
|
556
|
Brunner PM, Silverberg JI, Guttman-Yassky E, Paller AS, Kabashima K, Amagai M, Luger TA, Deleuran M, Werfel T, Eyerich K, Stingl G. Increasing Comorbidities Suggest that Atopic Dermatitis Is a Systemic Disorder. J Invest Dermatol 2016; 137:18-25. [PMID: 27771048 DOI: 10.1016/j.jid.2016.08.022] [Citation(s) in RCA: 280] [Impact Index Per Article: 31.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 08/04/2016] [Accepted: 08/19/2016] [Indexed: 12/12/2022]
Abstract
Atopic dermatitis comorbidities extend well beyond the march to allergic conditions (food allergy, asthma, allergic rhinitis, allergic conjunctivitis, and eosinophilic esophagitis), suggesting both cutaneous and systemic immune activation. In reviewing atopic dermatitis comorbidities, Councilors of the International Eczema Council found a strong pattern of immune activation in peripheral blood and the propensity to both skin and systemic infections. Associations with cardiovascular, neuropsychiatric, and malignant diseases were increasingly reported, but confirmation of their link with atopic dermatitis requires longitudinal studies. Given the possibility of atopic dermatitis-related systemic immune activation, future investigations of new interventions should concurrently examine the impact on these comorbidities.
Collapse
Affiliation(s)
- Patrick M Brunner
- The Laboratory for Investigative Dermatology, The Rockefeller University, New York, New York, USA
| | - Jonathan I Silverberg
- Department of Dermatology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Emma Guttman-Yassky
- Department of Dermatology and the Laboratory for Inflammatory Skin Diseases, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Amy S Paller
- Department of Dermatology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA.
| | - Kenji Kabashima
- Department of Dermatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Masayuki Amagai
- Department of Dermatology, Keio University School of Medicine, Tokyo, Japan
| | - Thomas A Luger
- Department of Dermatology, University of Münster, Münster, Germany
| | - Mette Deleuran
- Department of Dermato-Venereology, Aarhus University Hospital, Aarhus C, Denmark
| | - Thomas Werfel
- Division of Immunodermatology and Allergy Research, Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany
| | - Kilian Eyerich
- Department of Dermatology and Allergy, Technical University Munich, Munich, Germany
| | - Georg Stingl
- Division of Immunology, Allergy and Infectious Diseases, Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | | |
Collapse
|
557
|
Wang Z, Mascarenhas N, Eckmann L, Miyamoto Y, Sun X, Kawakami T, Di Nardo A. Skin microbiome promotes mast cell maturation by triggering stem cell factor production in keratinocytes. J Allergy Clin Immunol 2016; 139:1205-1216.e6. [PMID: 27746235 DOI: 10.1016/j.jaci.2016.09.019] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 08/22/2016] [Accepted: 09/23/2016] [Indexed: 12/25/2022]
Abstract
BACKGROUND Mast cell (MC) progenitors leave the bone marrow, enter the circulation, and settle in the skin and other tissues. Their maturation in tissues is influenced by the surrounding microenvironment. OBJECTIVE We tested the hypothesis that environmental factors play a role in MC maturation in the skin. METHODS MCs were numerically, phenotypically, and functionally compared between germ-free (GF), specific pathogen-free, and GF mice reconstituted with microbiota. The maturity of MCs was then correlated with skin levels of stem cell factor (SCF), a critical MC differentiation factor, and lipoteichoic acid (LTA), a Toll-like receptor 2 ligand. MCs were also evaluated in mice with keratinocyte-specific deletion of Scf. RESULTS We found that GF mice express abnormally low amounts of SCF, a critical MC differentiation factor, and contain MCs that are largely undifferentiated. Reconstituting the GF microbiota reverted this MC phenotype to normal, indicating that the phenotype is related to ongoing interactions of the microbiota and skin. Consistent with the immaturity of GF MCs, degranulation-provoking compound 48/80 induced less edema in the skin of GF mice than in conventional mice. Our results show that the skin microbiome drives SCF production in keratinocytes, which triggers the differentiation of dermal MCs. Because the skin microbiome is a rich source of LTA, a Toll-like receptor 2 ligand, we mimicked the GF microbiome's effect on MCs by applying LTA to the skin of GF mice. We also demonstrated that MC migration within the skin depends exclusively on keratinocyte-produced SCF. CONCLUSION This study has revealed a novel mechanism by which the skin microbiota signals the recruitment and maturation of MCs within the dermis through SCF production by LTA-stimulated keratinocytes.
Collapse
Affiliation(s)
- Zhenping Wang
- Department of Dermatology, School of Medicine, University of California, San Diego, La Jolla, Calif
| | - Nicholas Mascarenhas
- Department of Dermatology, School of Medicine, University of California, San Diego, La Jolla, Calif
| | - Lars Eckmann
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, Calif
| | - Yukiko Miyamoto
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, Calif
| | - Xiaojun Sun
- Department of Dermatology, School of Medicine, University of California, San Diego, La Jolla, Calif
| | - Toshiaki Kawakami
- Division of Cell Biology, La Jolla Institute for Allergy and Immunology, La Jolla, Calif
| | - Anna Di Nardo
- Department of Dermatology, School of Medicine, University of California, San Diego, La Jolla, Calif.
| |
Collapse
|
558
|
Deines P, Bosch TCG. Transitioning from Microbiome Composition to Microbial Community Interactions: The Potential of the Metaorganism Hydra as an Experimental Model. Front Microbiol 2016; 7:1610. [PMID: 27790207 PMCID: PMC5061769 DOI: 10.3389/fmicb.2016.01610] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 09/26/2016] [Indexed: 01/08/2023] Open
Abstract
Animals are home to complex microbial communities, which are shaped through interactions within the community, interactions with the host, and through environmental factors. The advent of high-throughput sequencing methods has led to novel insights in changing patterns of community composition and structure. However, deciphering the different types of interactions among community members, with their hosts and their interplay with their environment is still a challenge of major proportion. The emerging fields of synthetic microbial ecology and community systems biology have the potential to decrypt these complex relationships. Studying host-associated microbiota across multiple spatial and temporal scales will bridge the gap between individual microorganism studies and large-scale whole community surveys. Here, we discuss the unique potential of Hydra as an emerging experimental model in microbiome research. Through in vivo, in vitro, and in silico approaches the interaction structure of host-associated microbial communities and the effects of the host on the microbiota and its interactions can be disentangled. Research in the model system Hydra can unify disciplines from molecular genetics to ecology, opening up the opportunity to discover fundamental rules that govern microbiome community stability.
Collapse
Affiliation(s)
- Peter Deines
- Zoological Institute and Interdisciplinary Research Center, Kiel Life Science, Christian-Albrechts-Universität zu Kiel Kiel, Germany
| | - Thomas C G Bosch
- Zoological Institute and Interdisciplinary Research Center, Kiel Life Science, Christian-Albrechts-Universität zu Kiel Kiel, Germany
| |
Collapse
|
559
|
Alegre ML, Lakkis FG, Morelli AE. Antigen Presentation in Transplantation. Trends Immunol 2016; 37:831-843. [PMID: 27743777 DOI: 10.1016/j.it.2016.09.003] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 09/12/2016] [Accepted: 09/15/2016] [Indexed: 02/07/2023]
Abstract
Transplantation of solid organs between genetically distinct individuals leads, in the absence of immunosuppression, to T cell-dependent transplant rejection. Activation of graft-reactive T cells relies on the presentation of transplant-derived antigens (intact donor MHC molecules or processed peptides on host MHC molecules) by mature dendritic cells (DCs). This review focuses on novel insights regarding the steps for maturation and differentiation of DCs that are necessary for productive presentation of transplant antigens to host T cells. These steps include the licensing of DCs by the microbiota, their activation and maturation following recognition of allogeneic non-self, and their capture of donor cell exosomes to amplify the presentation of transplant antigens.
Collapse
Affiliation(s)
- Maria-Luisa Alegre
- Department of Medicine, University of Chicago, 924 East 57th Street, JFK-R312, Chicago, IL 60637, USA.
| | - Fadi G Lakkis
- Thomas E. Starzl Transplantation Institute and Departments of Surgery and Immunology, University of Pittsburgh School of Medicine, Biomedical Science Tower, 200 Lothrop Street, Pittsburgh, PA 15261, USA; Thomas E. Starzl Transplantation Institute and Department of Medicine, University of Pittsburgh School of Medicine, Biomedical Science Tower, 200 Lothrop Street, Pittsburgh, PA 15261, USA
| | - Adrian E Morelli
- Thomas E. Starzl Transplantation Institute and Departments of Surgery and Immunology, University of Pittsburgh School of Medicine, Biomedical Science Tower, 200 Lothrop Street, Pittsburgh, PA 15261, USA
| |
Collapse
|
560
|
Halliday A, Bates PA, Chance ML, Taylor MJ. Toll-like receptor 2 (TLR2) plays a role in controlling cutaneous leishmaniasis in vivo, but does not require activation by parasite lipophosphoglycan. Parasit Vectors 2016; 9:532. [PMID: 27716391 PMCID: PMC5053327 DOI: 10.1186/s13071-016-1807-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 09/21/2016] [Indexed: 01/27/2023] Open
Abstract
Background Leishmaniasis is a neglected tropical disease affecting millions of individuals worldwide. Despite several studies reporting involvement of the innate immune receptor Toll-like receptor 2 (TLR2) in the recognition of surface glycolipids from Leishmania parasites in vitro, the role of TLR2 and its co-receptors during cutaneous leishmaniasis infection in vivo is unknown. Methods To explore the role of TLR2 and its co-receptors in cutaneous leishmaniasis, mice deficient in either TLR2, 4, 1 or 6, or wild-type (WT) controls, were infected with either Leishmania major promastigotes, L. mexicana promastigotes, L. mexicana amastigotes, or LPG1−/−L. mexicana promastigotes. For each infection, lesion sizes were monitored and parasite burden was assessed at various time points. To assess immune responses, draining lymph node (DLN) cells were re-stimulated with parasite antigens and the production of cytokines and parasite-specific antibody isotypes in blood was determined by ELISA. Results Mice deficient in TLR2 and TLR4 presented with larger lesions and higher parasite burdens than WT controls. Mice lacking TLR2 co-receptors TLR1 or TLR6 did not show exacerbated infection, suggesting that TLR2 does not require either co-receptor in the recognition of Leishmania infection. Furthermore, it appears that lipophosphoglycan (LPG) is not the major mediator of TLR2 activation during infection with L. mexicana, as parasites lacking LPG (axenic amastigotes and LPG1−/− promastigotes) also resulted in exacerbated disease in TLR2−/− mice. Infected TLR2−/− mice show a skewed Th2 immune response to Leishmania parasites, as demonstrated by elevated IL-4, IL-13 and IL-10 production by DLN cells from L. mexicana infected mice in response to antigen. Furthermore, L. major infected TLR2−/− mice have elevated antigen-specific IgG1 antibodies. Conclusions TLR2 deficiency leads to exacerbation of disease and parasite burden through promotion of Th2 immunity. TLR2 activation in vivo occurs independently of parasite LPG, suggesting other parasite ligands are involved in TLR2 recognition of Leishmania. Electronic supplementary material The online version of this article (doi:10.1186/s13071-016-1807-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Alice Halliday
- Department of Parasitology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, UK
| | - Paul A Bates
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Furness Building, Lancaster University, Bailrigg, Lancaster, LA1 4YG, UK
| | - Michael L Chance
- Department of Parasitology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, UK
| | - Mark J Taylor
- Department of Parasitology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, UK.
| |
Collapse
|
561
|
Marrs T, Flohr C. The role of skin and gut microbiota in the development of atopic eczema. Br J Dermatol 2016; 175 Suppl 2:13-18. [PMID: 27667310 DOI: 10.1111/bjd.14907] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/25/2016] [Indexed: 12/19/2022]
Abstract
Conventional culture-based studies have suggested that a reduction in microbial exposure in early life predisposes to atopic eczema and allergies. However, molecular microbiological methods have shown that conventional culture fails to grow around 80% of the bacterial flora. More recent work reviewed in this paper has employed next generation sequencing to study the influence of the gut and skin microbiota, both with regard to the risk of developing atopic eczema but also the role of pathogenic and commensal bacteria in established disease. Birth cohorts investigating the gastrointestinal tract reported reduced faecal microbiota diversity among those who later developed atopic eczema, using gel electrophoresis, real-time PCR or 16S ribosomal RNA gene pyrosequencing. However, the inverse association with reduced faecal bacterial diversity was not confirmed in cross-sectional studies among patients with established atopic eczema. Only two studies investigated the cutaneous microbiota in a longitudinal study design and both were unable to provide evidence that Staphylococcus aureus colonisation precedes the development of atopic eczema. Next generation sequencing has confirmed the cross-sectional association between atopic eczema and S. aureus colonisation. The two studies that used this approach have also shown that disease flares are associated with a significant fall in skin microbiota diversity and an increase in the relative abundance of both S. aureus and epidermidis. Interestingly, S. aureus elimination does not appear to be the main reason why atopic eczema improves after a flare and antimicrobial and anti-inflammatory therapy enhances bacterial diversity. Further, well-phenotyped birth cohorts that take key confounders, such as antibiotic exposure, into account are required.
Collapse
Affiliation(s)
- T Marrs
- Division of Asthma, Allergy and Lung Biology, Department of Paediatric Allergy, King's College London, London, U.K.,Children's Allergies Department, Guy's and St Thomas' NHS Foundation Trust, St Thomas' Hospital, Westminster Bridge Road, Lambeth, SE1 7EH, U.K
| | - C Flohr
- Unit for Population-Based Dermatology Research, St John's Institute of Dermatology, Division of Genetics and Molecular Medicine, King's College London and Guy's and St Thomas' Hospital NHS Foundation Trust, London, U.K.
| |
Collapse
|
562
|
Christensen SM, Dillon LAL, Carvalho LP, Passos S, Novais FO, Hughitt VK, Beiting DP, Carvalho EM, Scott P, El-Sayed NM, Mosser DM. Meta-transcriptome Profiling of the Human-Leishmania braziliensis Cutaneous Lesion. PLoS Negl Trop Dis 2016; 10:e0004992. [PMID: 27631090 PMCID: PMC5025153 DOI: 10.1371/journal.pntd.0004992] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Accepted: 08/19/2016] [Indexed: 11/24/2022] Open
Abstract
Host and parasite gene expression in skin biopsies from Leishmania braziliensis-infected patients were simultaneously analyzed using high throughput RNA-sequencing. Biopsies were taken from 8 patients with early cutaneous leishmaniasis and 17 patients with late cutaneous leishmaniasis. Although parasite DNA was found in all patient lesions at the time of biopsy, the patients could be stratified into two groups: one lacking detectable parasite transcripts (PTNeg) in lesions, and another in which parasite transcripts were readily detected (PTPos). These groups exhibited substantial differences in host responses to infection. PTPos biopsies contained an unexpected increase in B lymphocyte-specific and immunoglobulin transcripts in the lesions, and an upregulation of immune inhibitory molecules. Biopsies without detectable parasite transcripts showed decreased evidence for B cell activation, but increased expression of antimicrobial genes and genes encoding skin barrier functions. The composition and abundance of L. braziliensis transcripts in PTPos lesions were surprisingly conserved among all six patients, with minimal meaningful differences between lesions from patients with early and late cutaneous leishmaniasis. The most abundant parasite transcripts expressed in lesions were distinct from transcripts expressed in vitro in human macrophage cultures infected with L. amazonensis or L. major. Therefore in vitro gene expression in macrophage monolayers may not be a strong predictor of gene expression in lesions. Some of the most highly expressed in vivo transcripts encoded amastin-like proteins, hypothetical genes, putative parasite virulence factors, as well as histones and tubulin. In summary, RNA sequencing allowed us to simultaneously analyze human and L. braziliensis transcriptomes in lesions of infected patients, and identify unexpected differences in host immune responses which correlated with active transcription of parasite genes. Leishmania spp are intracellular protozoan parasites that replicate primarily within host tissue macrophages. In this paper we simultaneously query host and parasite gene expression in human cutaneous L. braziliensis lesions. We observe an unexpectedly prominent role for B cells and immunoglobulins in lesions in which actively transcribing parasites reside. We also observe that parasite gene expression is surprisingly conserved among L. braziliensis lesions, and the genes that are expressed in lesions are not those that have been previously associated with parasite growth in vitro. This analysis of parasite and host gene expression in lesions may lead to the identification of new parasite virulence factors and may identify host responses that promote parasite persistence in lesions.
Collapse
Affiliation(s)
- Stephen M. Christensen
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland, United States of America
| | - Laura A. L. Dillon
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland, United States of America
- Center for Bioinformatics and Computational Biology, University of Maryland, College Park, Maryland, United States of America
| | | | - Sara Passos
- Universidade Federal da Bahia Salvador, Bahia, Brazil
| | - Fernanda O. Novais
- Department of Pathobiology, School of Veterinary Medicine, The University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - V. Keith Hughitt
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland, United States of America
- Center for Bioinformatics and Computational Biology, University of Maryland, College Park, Maryland, United States of America
| | - Daniel P. Beiting
- Department of Pathobiology, School of Veterinary Medicine, The University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | | | - Phillip Scott
- Department of Pathobiology, School of Veterinary Medicine, The University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Najib M. El-Sayed
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland, United States of America
- Center for Bioinformatics and Computational Biology, University of Maryland, College Park, Maryland, United States of America
| | - David M. Mosser
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland, United States of America
- * E-mail:
| |
Collapse
|
563
|
Hand TW. The Role of the Microbiota in Shaping Infectious Immunity. Trends Immunol 2016; 37:647-658. [PMID: 27616558 DOI: 10.1016/j.it.2016.08.007] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 08/09/2016] [Accepted: 08/11/2016] [Indexed: 02/07/2023]
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.
Collapse
Affiliation(s)
- Timothy W Hand
- Richard King Mellon Institute for Pediatric Research, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh, Pittsburgh, PA 15224, USA.
| |
Collapse
|
564
|
Kennedy EA, Connolly J, Hourihane JO, Fallon PG, McLean WHI, Murray D, Jo JH, Segre JA, Kong HH, Irvine AD. Skin microbiome before development of atopic dermatitis: Early colonization with commensal staphylococci at 2 months is associated with a lower risk of atopic dermatitis at 1 year. J Allergy Clin Immunol 2016; 139:166-172. [PMID: 27609659 PMCID: PMC5207796 DOI: 10.1016/j.jaci.2016.07.029] [Citation(s) in RCA: 238] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Revised: 07/20/2016] [Accepted: 07/27/2016] [Indexed: 12/19/2022]
Abstract
BACKGROUND Disease flares of established atopic dermatitis (AD) are generally associated with a low-diversity skin microbiota and Staphylococcus aureus dominance. The temporal transition of the skin microbiome between early infancy and the dysbiosis of established AD is unknown. METHODS We randomly selected 50 children from the Cork Babies After SCOPE: Evaluating the Longitudinal Impact Using Neurological and Nutritional Endpoints (BASELINE) longitudinal birth cohort for microbiome sampling at 3 points in the first 6 months of life at 4 skin sites relevant to AD: the antecubital and popliteal fossae, nasal tip, and cheek. We identified 10 infants with AD and compared them with 10 randomly selected control infants with no AD. We performed bacterial 16S ribosomal RNA sequencing and analysis directly from clinical samples. RESULTS Bacterial community structures and diversity shifted over time, suggesting that age strongly affects the skin microbiome in infants. Unlike established AD, these patients with infantile AD did not have noticeably dysbiotic communities before or with disease and were not colonized by S aureus. In comparing patients and control subjects, infants who had affected skin at month 12 had statistically significant differences in bacterial communities on the antecubital fossa at month 2 compared with infants who were unaffected at month 12. In particular, commensal staphylococci were significantly less abundant in infants affected at month 12, suggesting that this genus might protect against the later development of AD. CONCLUSIONS This study suggests that 12-month-old infants with AD were not colonized with S aureus before having AD. Additional studies are needed to confirm whether colonization with commensal staphylococci modulates skin immunity and attenuates development of AD.
Collapse
Affiliation(s)
- Elizabeth A Kennedy
- Dermatology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Md
| | - Jennifer Connolly
- Paediatrics and Child Health, University College, Cork, and the Irish Centre for Fetal and Neonatal Translational (INFANT) Research, Cork University Maternity Hospital, Cork, Ireland
| | - Jonathan O'B Hourihane
- Paediatrics and Child Health, University College, Cork, and the Irish Centre for Fetal and Neonatal Translational (INFANT) Research, Cork University Maternity Hospital, Cork, Ireland
| | - Padraic G Fallon
- Clinical Medicine, Trinity College Dublin, Dublin, Ireland; National Children's Research Centre, Our Lady's Children's Hospital Crumlin, Dublin, Ireland
| | - W H Irwin McLean
- Dermatology and Genetic Medicine, University of Dundee, Dundee, United Kingdom
| | - Deirdre Murray
- Paediatrics and Child Health, University College, Cork, and the Irish Centre for Fetal and Neonatal Translational (INFANT) Research, Cork University Maternity Hospital, Cork, Ireland
| | - Jay-Hyun Jo
- Dermatology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Md
| | - Julia A Segre
- Translational and Functional Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Md
| | - Heidi H Kong
- Dermatology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Md.
| | - Alan D Irvine
- Clinical Medicine, Trinity College Dublin, Dublin, Ireland; National Children's Research Centre, Our Lady's Children's Hospital Crumlin, Dublin, Ireland; Paediatric Dermatology, Our Lady's Children's Hospital Crumlin, Dublin, Ireland.
| |
Collapse
|
565
|
Lowry CA, Smith DG, Siebler PH, Schmidt D, Stamper CE, Hassell JE, Yamashita PS, Fox JH, Reber SO, Brenner LA, Hoisington AJ, Postolache TT, Kinney KA, Marciani D, Hernandez M, Hemmings SMJ, Malan-Muller S, Wright KP, Knight R, Raison CL, Rook GAW. The Microbiota, Immunoregulation, and Mental Health: Implications for Public Health. Curr Environ Health Rep 2016; 3:270-86. [PMID: 27436048 PMCID: PMC5763918 DOI: 10.1007/s40572-016-0100-5] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The hygiene or "Old Friends" hypothesis proposes that the epidemic of inflammatory disease in modern urban societies stems at least in part from reduced exposure to microbes that normally prime mammalian immunoregulatory circuits and suppress inappropriate inflammation. Such diseases include but are not limited to allergies and asthma; we and others have proposed that the markedly reduced exposure to these Old Friends in modern urban societies may also increase vulnerability to neurodevelopmental disorders and stress-related psychiatric disorders, such as anxiety and affective disorders, where data are emerging in support of inflammation as a risk factor. Here, we review recent advances in our understanding of the potential for Old Friends, including environmental microbial inputs, to modify risk for inflammatory disease, with a focus on neurodevelopmental and psychiatric conditions. We highlight potential mechanisms, involving bacterially derived metabolites, bacterial antigens, and helminthic antigens, through which these inputs promote immunoregulation. Though findings are encouraging, significant human subjects' research is required to evaluate the potential impact of Old Friends, including environmental microbial inputs, on biological signatures and clinically meaningful mental health prevention and intervention outcomes.
Collapse
Affiliation(s)
- Christopher A Lowry
- Department of Integrative Physiology and Center for Neuroscience, University of Colorado Boulder, 1725 Pleasant Street, Boulder, CO, 80309-0354, USA.
| | - David G Smith
- Department of Integrative Physiology and Center for Neuroscience, University of Colorado Boulder, 1725 Pleasant Street, Boulder, CO, 80309-0354, USA
| | - Philip H Siebler
- Department of Integrative Physiology and Center for Neuroscience, University of Colorado Boulder, 1725 Pleasant Street, Boulder, CO, 80309-0354, USA
| | - Dominic Schmidt
- Department of Integrative Physiology and Center for Neuroscience, University of Colorado Boulder, 1725 Pleasant Street, Boulder, CO, 80309-0354, USA
| | - Christopher E Stamper
- Department of Integrative Physiology and Center for Neuroscience, University of Colorado Boulder, 1725 Pleasant Street, Boulder, CO, 80309-0354, USA
| | - James E Hassell
- Department of Integrative Physiology and Center for Neuroscience, University of Colorado Boulder, 1725 Pleasant Street, Boulder, CO, 80309-0354, USA
| | - Paula S Yamashita
- Department of Integrative Physiology and Center for Neuroscience, University of Colorado Boulder, 1725 Pleasant Street, Boulder, CO, 80309-0354, USA
| | - James H Fox
- Department of Integrative Physiology and Center for Neuroscience, University of Colorado Boulder, 1725 Pleasant Street, Boulder, CO, 80309-0354, USA
| | - Stefan O Reber
- Laboratory for Molecular Psychosomatics, Clinic for Psychosomatic Medicine and Psychotherapy, University of Ulm, D-89081, Ulm, Germany
| | - Lisa A Brenner
- Departments of Psychiatry, Physical Medicine & Rehabilitation, University of Colorado, Anschutz School of Medicine, Aurora, CO, 80045, USA
- Rocky Mountain Mental Illness Research Education and Clinical Center (MIRECC), Veterans Integrated Service Network (VISN) 19, Denver, CO, 80220, USA
| | - Andrew J Hoisington
- Department of Civil and Environmental Engineering, United States Air Force Academy, Colorado Springs, CO, 80840, USA
| | - Teodor T Postolache
- University of Maryland School of Medicine, Baltimore, MD, USA
- Rocky Mountain MIRECC, Denver, CO, 80220, USA
- VISN 5 MIRECC, Baltimore, MD, 21201, USA
| | - Kerry A Kinney
- Civil, Architectural and Environmental Engineering, University of Texas Austin, Austin, TX, 78712, USA
| | | | - Mark Hernandez
- Department of Civil, Environmental and Architectural Engineering, University of Colorado Boulder, Boulder, CO, 80309, USA
| | - Sian M J Hemmings
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, Cape Town, 7505, South Africa
| | - Stefanie Malan-Muller
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, Cape Town, 7505, South Africa
| | - Kenneth P Wright
- Department of Integrative Physiology and Center for Neuroscience, University of Colorado Boulder, 1725 Pleasant Street, Boulder, CO, 80309-0354, USA
| | - Rob Knight
- Departments of Pediatrics and Computer Science and Engineering, and Center for Microbiome Innovation, University of California San Diego, La Jolla, CA, 92093, USA
| | - Charles L Raison
- School of Human Ecology and School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Graham A W Rook
- Center for Clinical Microbiology, UCL (University College London), WC1E 6BT, London, UK
| |
Collapse
|
566
|
Abstract
Hypomorphic Rag mutations in humans cause Omenn Syndrome (OS) a severe immunodeficiency associated with autoimmune-like manifestations mediated by oligoclonal activated T and B cells. The clinical and immunological spectrum of OS presentation is extremely broad. However, the role played by environmental triggers in the disease pathogenesis remains largely unknown. We have recently shown in a murine model that gut microbiota has a substantial role in determining the distinctive immune dysregulation of OS. Here, we describe how dysbiosis and loss of T cell tolerance to commensals influence the expression of autoimmunity at the barrier site and beyond, and the disease hallmark hyper-IgE. We discuss how commensal antigens and gut-derived pathogenic T cells could potentially modulate skin immunity to determine cutaneous degenerations in OS. These mechanisms may have broader implications for a deeper understanding of the role of gut microbes in influencing barriers integrity and host physiology.
Collapse
Affiliation(s)
- Rosita Rigoni
- Milan Unit, Istituto di Ricerca Genetica e
Biomedica, Consiglio Nazionale delle Ricerche, Milan,
Italy,Humanitas Clinical and Research
Center, Rozzano, Milan, Italy
| | - Fabio Grassi
- Istituto Nazionale Genetica Molecolare,
Department of Medical Biotechnology and Translational Medicine, University of
Milan, Milan, Italy,Institute for Research in
Biomedicine, Bellinzona, Switzerland
| | - Anna Villa
- Milan Unit, Istituto di Ricerca Genetica e
Biomedica, Consiglio Nazionale delle Ricerche, Milan,
Italy,Telethon Institute for Gene Therapy, Division
of Regenerative Medicine, Stem Cells and Gene Therapy, Istituto di Ricovero e Cura a
Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan,
Italy
| | - Barbara Cassani
- Milan Unit, Istituto di Ricerca Genetica e
Biomedica, Consiglio Nazionale delle Ricerche, Milan,
Italy,Humanitas Clinical and Research
Center, Rozzano, Milan, Italy,CONTACT Barbara Cassani Humanitas Clinical and Research Center, via Manzoni
56, 20089 Rozzano (Mi), Italy
| |
Collapse
|
567
|
O'Neill CA, Monteleone G, McLaughlin JT, Paus R. The gut-skin axis in health and disease: A paradigm with therapeutic implications. Bioessays 2016; 38:1167-1176. [PMID: 27554239 DOI: 10.1002/bies.201600008] [Citation(s) in RCA: 250] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
As crucial interface organs gut and skin have much in common. Therefore it is unsurprising that several gut pathologies have skin co-morbidities. Nevertheless, the reason for this remains ill explored, and neither mainstream gastroenterology nor dermatology research have systematically investigated the 'gut-skin axis'. Here, in reviewing the field, we propose several mechanistic levels on which gut and skin may interact under physiological and pathological circumstances. We focus on the gut microbiota, with its huge metabolic capacity, and the role of dietary components as potential principle actors along the gut-skin axis. We suggest that metabolites from either the diet or the microbiota are skin accessible. After defining open key questions around the nature of these metabolites, how they are sensed, and which cutaneous changes they can induce, we propose that understanding of these pathways will lead to novel therapeutic strategies based on targeting one organ to improve the health of the other.
Collapse
Affiliation(s)
- Catherine A O'Neill
- Dermatology Research Centre, Institute of Inflammation and Repair, University of Manchester and Manchester Academic Health Sciences Centre, Manchester, UK.
| | - Giovanni Monteleone
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - John T McLaughlin
- Gastrointestinal Research Centre, Institute of Inflammation and Repair, University of Manchester and Manchester Academic Health Sciences Centre, Manchester, UK
| | - Ralf Paus
- Dermatology Research Centre, Institute of Inflammation and Repair, University of Manchester and Manchester Academic Health Sciences Centre, Manchester, UK.,Department of Dermatology, University of Münster, Münster, Germany
| |
Collapse
|
568
|
Kitaoka M, Wakabayashi R, Kamiya N, Goto M. Solid-in-oil nanodispersions for transdermal drug delivery systems. Biotechnol J 2016; 11:1375-1385. [PMID: 27529824 PMCID: PMC5132072 DOI: 10.1002/biot.201600081] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Revised: 07/10/2016] [Accepted: 07/11/2016] [Indexed: 12/15/2022]
Abstract
Transdermal administration of drugs has advantages over conventional oral administration or administration using injection equipment. The route of administration reduces the opportunity for drug evacuation before systemic circulation, and enables long-lasting drug administration at a modest body concentration. In addition, the skin is an attractive route for vaccination, because there are many immune cells in the skin. Recently, solid-in-oil nanodisperison (S/O) technique has demonstrated to deliver cosmetic and pharmaceutical bioactives efficiently through the skin. S/O nanodispersions are nanosized drug carriers designed to overcome the skin barrier. This review discusses the rationale for preparation of efficient and stable S/O nanodispersions, as well as application examples in cosmetic and pharmaceutical materials including vaccines. Drug administration using a patch is user-friendly, and may improve patient compliance. The technique is a potent transcutaneous immunization method without needles.
Collapse
Affiliation(s)
- Momoko Kitaoka
- Graduate School of Engineering, Kyushu University, Fukuoka, Japan
| | - Rie Wakabayashi
- Graduate School of Engineering, Kyushu University, Fukuoka, Japan.,Center for Transdermal Drug Delivery, Kyushu University, Fukuoka, Japan
| | - Noriho Kamiya
- Graduate School of Engineering, Kyushu University, Fukuoka, Japan.,Center for Transdermal Drug Delivery, Kyushu University, Fukuoka, Japan.,Center for Future Chemistry, Kyushu University, Fukuoka, Japan
| | - Masahiro Goto
- Graduate School of Engineering, Kyushu University, Fukuoka, Japan.,Center for Transdermal Drug Delivery, Kyushu University, Fukuoka, Japan.,Center for Future Chemistry, Kyushu University, Fukuoka, Japan
| |
Collapse
|
569
|
Patra V, Byrne SN, Wolf P. The Skin Microbiome: Is It Affected by UV-induced Immune Suppression? Front Microbiol 2016; 7:1235. [PMID: 27559331 PMCID: PMC4979252 DOI: 10.3389/fmicb.2016.01235] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 07/25/2016] [Indexed: 12/12/2022] Open
Abstract
Human skin apart from functioning as a physical barricade to stop the entry of pathogens, also hosts innumerable commensal organisms. The skin cells and the immune system constantly interact with microbes, to maintain cutaneous homeostasis, despite the challenges offered by various environmental factors. A major environmental factor affecting the skin is ultraviolet radiation (UV-R) from sunlight. UV-R is well known to modulate the immune system, which can be both beneficial and deleterious. By targeting the cells and molecules within skin, UV-R can trigger the production and release of antimicrobial peptides, affect the innate immune system and ultimately suppress the adaptive cellular immune response. This can contribute to skin carcinogenesis and the promotion of infectious agents such as herpes simplex virus and possibly others. On the other hand, a UV-established immunosuppressive environment may protect against the induction of immunologically mediated skin diseases including some of photodermatoses such as polymorphic light eruption. In this article, we share our perspective about the possibility that UV-induced immune suppression may alter the landscape of the skin’s microbiome and its components. Alternatively, or in concert with this, direct UV-induced DNA and membrane damage to the microbiome may result in pathogen associated molecular patterns (PAMPs) that interfere with UV-induced immune suppression.
Collapse
Affiliation(s)
- VijayKumar Patra
- Research Unit for Photodermatology, Department of Dermatology, Medical University of GrazGraz, Austria; Center for Medical Research, Medical University of GrazGraz, Austria
| | - Scott N Byrne
- Cellular Photoimmunology Group, Infectious Diseases and Immunology, Sydney Medical School, The Charles Perkins Center Hub at The University of Sydney, Sydney NSW, Australia
| | - Peter Wolf
- Research Unit for Photodermatology, Department of Dermatology, Medical University of Graz Graz, Austria
| |
Collapse
|
570
|
Honda K, Littman DR. The microbiota in adaptive immune homeostasis and disease. Nature 2016; 535:75-84. [PMID: 27383982 DOI: 10.1038/nature18848] [Citation(s) in RCA: 1249] [Impact Index Per Article: 138.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2016] [Accepted: 04/25/2016] [Indexed: 12/12/2022]
Abstract
In the mucosa, the immune system's T cells and B cells have position-specific phenotypes and functions that are influenced by the microbiota. These cells play pivotal parts in the maintenance of immune homeostasis by suppressing responses to harmless antigens and by enforcing the integrity of the barrier functions of the gut mucosa. Imbalances in the gut microbiota, known as dysbiosis, can trigger several immune disorders through the activity of T cells that are both near to and distant from the site of their induction. Elucidation of the mechanisms that distinguish between homeostatic and pathogenic microbiota-host interactions could identify therapeutic targets for preventing or modulating inflammatory diseases and for boosting the efficacy of cancer immunotherapy.
Collapse
Affiliation(s)
- Kenya Honda
- Department of Microbiology and Immunology, Keio University School of Medicine, Shinjuku, Tokyo 160-8582, Japan.,RIKEN Center for Integrative Medical Sciences, Tsurumi, Yokohama, Kanagawa 230-0045, Japan.,AMED-CREST, Chiyoda, Tokyo 100-0004, Japan
| | - Dan R Littman
- The Helen L. and Martin S. Kimmel Center for Biology and Medicine at the Skirball Institute of Biomolecular Medicine, New York University School of Medicine, New York, New York 10016, USA.,The Howard Hughes Medical Institute, New York University School of Medicine, New York, New York 10016, USA
| |
Collapse
|
571
|
Wang Y, Zhang L, Yu J, Huang S, Wang Z, Chun KA, Lee TL, Chen YT, Gallo RL, Huang CM. A Co-Drug of Butyric Acid Derived from Fermentation Metabolites of the Human Skin Microbiome Stimulates Adipogenic Differentiation of Adipose-Derived Stem Cells: Implications in Tissue Augmentation. J Invest Dermatol 2016; 137:46-56. [PMID: 27498050 DOI: 10.1016/j.jid.2016.07.030] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Revised: 06/28/2016] [Accepted: 07/12/2016] [Indexed: 12/25/2022]
Abstract
We show that Staphylococcus epidermidis, a commensal bacterium in the human skin microbiome, produces short-chain fatty acids by glycerol fermentation that can induce adipogenesis. Although the antimicrobial and anti-inflammatory activities of short-chain fatty acids have been previously well characterized, little is known about the contribution of short-chain fatty acids to the adipogenic differentiation of adipose-derived stem cells (ADSCs). We show that ADSCs differentiated into adipocytes and accumulated lipids in the cytoplasm when cultured with butyric acid, a principal short-chain fatty acid in the fermentation metabolites of S. epidermidis. Additionally, a co-drug, butyric acid 2-(2-butyryloxyethoxy) ethyl ester (BA-DEG-BA), released active butyric acid when it was intradermally injected into mouse ears and induced ADSC differentiation, characterized by an increased expression of cytoplasmic lipids and perilipin A. The BA-DEG-BA-induced adipogenic differentiation was mediated via peroxisome proliferator-activated receptor gamma. Furthermore, intradermal injection of ADSCs along with BA-DEG-BA into mouse ears markedly enhanced the adipogenic differentiation of ADSCs, leading to dermal augmentation. Our study introduces BA-DEG-BA as an enhancer of ADSC adipogenesis and suggests an integral interaction between the human skin microbiome and ADSCs.
Collapse
Affiliation(s)
- Yanhan Wang
- Department of Dermatology, School of Medicine, University of California, San Diego, California, USA
| | - Lingjuan Zhang
- Department of Dermatology, School of Medicine, University of California, San Diego, California, USA
| | - Jinghua Yu
- Sanford-Burnham Institute for Medical Research, La Jolla, California, USA
| | - Stephen Huang
- Surface Bioadvances Incorporated, San Diego, California, USA
| | - Zhenping Wang
- Department of Dermatology, School of Medicine, University of California, San Diego, California, USA
| | - Kimberly Ann Chun
- Department of Dermatology, School of Medicine, University of California, San Diego, California, USA
| | - Tammy Ling Lee
- Department of Dermatology, School of Medicine, University of California, San Diego, California, USA
| | - Ying-Tung Chen
- Nerd SkinCare Incorporated, San Francisco, California, USA
| | - Richard L Gallo
- Department of Dermatology, School of Medicine, University of California, San Diego, California, USA
| | - Chun-Ming Huang
- Department of Dermatology, School of Medicine, University of California, San Diego, California, USA; Moores Cancer Center, University of California, San Diego, California, USA.
| |
Collapse
|
572
|
Microbiota, regulatory T cell subsets, and allergic disorders. ACTA ACUST UNITED AC 2016; 25:114-123. [PMID: 27656354 PMCID: PMC5016534 DOI: 10.1007/s40629-016-0118-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2015] [Accepted: 01/26/2016] [Indexed: 02/07/2023]
|
573
|
Ohnmacht C. Microbiota, regulatory T cell subsets, and allergic disorders. ALLERGO JOURNAL 2016. [DOI: 10.1007/s15007-016-1137-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
574
|
Sabino-Pinto J, Bletz MC, Islam MM, Shimizu N, Bhuju S, Geffers R, Jarek M, Kurabayashi A, Vences M. Composition of the Cutaneous Bacterial Community in Japanese Amphibians: Effects of Captivity, Host Species, and Body Region. MICROBIAL ECOLOGY 2016; 72:460-9. [PMID: 27278778 DOI: 10.1007/s00248-016-0797-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2016] [Accepted: 05/25/2016] [Indexed: 05/09/2023]
Abstract
The cutaneous microbiota plays a significant role in the biology of their vertebrate hosts, and its composition is known to be influenced both by host and environment, with captive conditions often altering alpha diversity. Here, we compare the cutaneous bacterial communities of 61 amphibians (both wild and captive) from Hiroshima, Japan, using high-throughput amplicon sequencing of a segment of the 16S rRNA gene. The majority of these samples came from a captive breeding facility at Hiroshima University where specimens from six species are maintained under highly standardized conditions for several generations. This allowed to identify host effects on the bacterial communities under near identical environmental conditions in captivity. We found the structure of the cutaneous bacterial community significantly differing between wild and captive individuals of newts, Cynops pyrrhogaster, with a higher alpha diversity found in the wild individuals. Community structure also showed distinct patterns when comparing different species of amphibians kept under highly similar conditions, revealing an intrinsic host effect. Bacterial communities of dorsal vs. ventral skin surfaces did not significantly differ in most species, but a trend of higher alpha diversity on the ventral surface was found in Oriental fire-bellied toads, Bombina orientalis. This study confirms the cutaneous microbiota of amphibians as a highly dynamic system influenced by a complex interplay of numerous factors.
Collapse
Affiliation(s)
- Joana Sabino-Pinto
- Zoological Institute, Braunschweig University of Technology, Braunschweig, Germany.
| | | | - Mohammed Mafizul Islam
- Institute for Amphibian Biology, Graduate School of Science, Hiroshima University, Higashi-Hiroshima, Japan
| | - Norio Shimizu
- Hiroshima University Museums, Hiroshima University, Higashi-Hiroshima, Japan
| | - Sabin Bhuju
- Department of Genome Analytics, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Robert Geffers
- Department of Genome Analytics, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Michael Jarek
- Department of Genome Analytics, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Atsushi Kurabayashi
- Institute for Amphibian Biology, Graduate School of Science, Hiroshima University, Higashi-Hiroshima, Japan
| | - Miguel Vences
- Zoological Institute, Braunschweig University of Technology, Braunschweig, Germany
| |
Collapse
|
575
|
Abstract
Cutaneous leishmaniasis is a major public health problem and causes a range of diseases from self-healing infections to chronic disfiguring disease. Currently, there is no vaccine for leishmaniasis, and drug therapy is often ineffective. Since the discovery of CD4(+) T helper 1 (TH1) cells and TH2 cells 30 years ago, studies of cutaneous leishmaniasis in mice have answered basic immunological questions concerning the development and maintenance of CD4(+) T cell subsets. However, new strategies for controlling the human disease have not been forthcoming. Nevertheless, advances in our knowledge of the cells that participate in protection against Leishmania infection and the cells that mediate increased pathology have highlighted new approaches for vaccine development and immunotherapy. In this Review, we discuss the early events associated with infection, the CD4(+) T cells that mediate protective immunity and the pathological role that CD8(+) T cells can have in cutaneous leishmaniasis.
Collapse
|
576
|
Nuriel-Ohayon M, Neuman H, Koren O. Microbial Changes during Pregnancy, Birth, and Infancy. Front Microbiol 2016; 7:1031. [PMID: 27471494 PMCID: PMC4943946 DOI: 10.3389/fmicb.2016.01031] [Citation(s) in RCA: 374] [Impact Index Per Article: 41.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 06/17/2016] [Indexed: 12/19/2022] Open
Abstract
Several healthy developmental processes such as pregnancy, fetal development, and infant development include a multitude of physiological changes: weight gain, hormonal, and metabolic changes, as well as immune changes. In this review, we present an additional important factor which both influences and is affected by these physiological processes-the microbiome. We summarize the known changes in microbiota composition at a variety of body sites including gut, vagina, oral cavity, and placenta, throughout pregnancy, fetal development, and early childhood. There is still a lot to be discovered; yet several pieces of research point to the healthy desired microbial changes. Future research is likely to unravel precise roles and mechanisms of the microbiota in gestation; perhaps linking the metabolic, hormonal, and immune changes together. Although some research has started to link microbial dysbiosis and specific microbial populations with unhealthy pregnancy complications, it is important to first understand the context of the natural healthy microbial changes occurring. Until recently the placenta and developing fetus were considered to be germ free, containing no apparent microbiome. We present multiple study results showing distinct microbiota compositions in the placenta and meconium, alluding to early microbial colonization. These results may change dogmas and our overall understanding of the importance and roles of microbiota from the beginning of life. We further review the main factors shaping the infant microbiome-modes of delivery, feeding, weaning, and exposure to antibiotics. Taken together, we are starting to build a broader understanding of healthy vs. abnormal microbial alterations throughout major developmental time-points.
Collapse
Affiliation(s)
| | - Hadar Neuman
- Faculty of Medicine, Bar-Ilan University Safed, Israel
| | - Omry Koren
- Faculty of Medicine, Bar-Ilan University Safed, Israel
| |
Collapse
|
577
|
Grice EA. The intersection of microbiome and host at the skin interface: genomic- and metagenomic-based insights. Genome Res 2016; 25:1514-20. [PMID: 26430162 PMCID: PMC4579337 DOI: 10.1101/gr.191320.115] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The past two decades have been marked by a surge in research to understand the microbial communities that live in association with the human body, in part stimulated by affordable, high-throughput DNA sequencing technology. In the context of the skin, this Perspective focuses on the current state of genomic- and metagenomic-based host–microbe research and future challenges and opportunities to move the field forward. These include elucidating nonbacterial components of the skin microbiome (i.e., viruses); systematic studies to address common perturbations to the skin microbiome (e.g., antimicrobial drugs, topical cosmetic/hygienic products); improved approaches for identifying potential microbial triggers for skin diseases, including species- and strain-level resolution; and improved, clinically relevant models for studying the functional and mechanistic roles of the skin microbiome. In the next 20 years, we can realistically expect that our knowledge of the skin microbiome will inform the clinical management and treatment of skin disorders through diagnostic tests to stratify patient subsets and predict best treatment modality and outcomes and through treatment strategies such as targeted manipulation or reconstitution of microbial communities.
Collapse
Affiliation(s)
- Elizabeth A Grice
- Department of Dermatology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19014, USA
| |
Collapse
|
578
|
McAleer JP, Nguyen NLH, Chen K, Kumar P, Ricks DM, Binnie M, Armentrout RA, Pociask DA, Hein A, Yu A, Vikram A, Bibby K, Umesaki Y, Rivera A, Sheppard D, Ouyang W, Hooper LV, Kolls JK. Pulmonary Th17 Antifungal Immunity Is Regulated by the Gut Microbiome. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2016; 197:97-107. [PMID: 27217583 PMCID: PMC4912941 DOI: 10.4049/jimmunol.1502566] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 04/25/2016] [Indexed: 12/18/2022]
Abstract
Commensal microbiota are critical for the development of local immune responses. In this article, we show that gut microbiota can regulate CD4 T cell polarization during pulmonary fungal infections. Vancomycin drinking water significantly decreased lung Th17 cell numbers during acute infection, demonstrating that Gram-positive commensals contribute to systemic inflammation. We next tested a role for RegIIIγ, an IL-22-inducible antimicrobial protein with specificity for Gram-positive bacteria. Following infection, increased accumulation of Th17 cells in the lungs of RegIIIγ(-/-) and Il22(-/-) mice was associated with intestinal segmented filamentous bacteria (SFB) colonization. Although gastrointestinal delivery of rRegIIIγ decreased lung inflammatory gene expression and protected Il22(-/-) mice from weight loss during infection, it had no direct effect on SFB colonization, fungal clearance, or lung Th17 immunity. We further show that vancomycin only decreased lung IL-17 production in mice colonized with SFB. To determine the link between gut microbiota and lung immunity, serum-transfer experiments revealed that IL-1R ligands increase the accumulation of lung Th17 cells. These data suggest that intestinal microbiota, including SFB, can regulate pulmonary adaptive immune responses.
Collapse
Affiliation(s)
- Jeremy P McAleer
- Richard King Mellon Foundation Institute for Pediatric Research, Children's Hospital of Pittsburgh of University of Pittsburgh Medical Center, Pittsburgh, PA 15224
| | - Nikki L H Nguyen
- Richard King Mellon Foundation Institute for Pediatric Research, Children's Hospital of Pittsburgh of University of Pittsburgh Medical Center, Pittsburgh, PA 15224; Department of Genetics, Louisiana State University Health Sciences Center, New Orleans, LA 70112
| | - Kong Chen
- Richard King Mellon Foundation Institute for Pediatric Research, Children's Hospital of Pittsburgh of University of Pittsburgh Medical Center, Pittsburgh, PA 15224
| | - Pawan Kumar
- Richard King Mellon Foundation Institute for Pediatric Research, Children's Hospital of Pittsburgh of University of Pittsburgh Medical Center, Pittsburgh, PA 15224
| | - David M Ricks
- Richard King Mellon Foundation Institute for Pediatric Research, Children's Hospital of Pittsburgh of University of Pittsburgh Medical Center, Pittsburgh, PA 15224; Department of Genetics, Louisiana State University Health Sciences Center, New Orleans, LA 70112
| | - Matthew Binnie
- Division of Respirology, Department of Medicine, University of Toronto, Ontario M5B 1W8, Canada
| | - Rachel A Armentrout
- Richard King Mellon Foundation Institute for Pediatric Research, Children's Hospital of Pittsburgh of University of Pittsburgh Medical Center, Pittsburgh, PA 15224
| | - Derek A Pociask
- Richard King Mellon Foundation Institute for Pediatric Research, Children's Hospital of Pittsburgh of University of Pittsburgh Medical Center, Pittsburgh, PA 15224
| | - Aaron Hein
- Richard King Mellon Foundation Institute for Pediatric Research, Children's Hospital of Pittsburgh of University of Pittsburgh Medical Center, Pittsburgh, PA 15224
| | - Amy Yu
- Department of Genetics, Louisiana State University Health Sciences Center, New Orleans, LA 70112
| | - Amit Vikram
- Department of Civil and Environmental Engineering, University of Pittsburgh, Pittsburgh, PA 15261
| | - Kyle Bibby
- Department of Civil and Environmental Engineering, University of Pittsburgh, Pittsburgh, PA 15261; Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA 15260
| | - Yoshinori Umesaki
- Yakult Central Institute for Microbiological Research, Kunitachi-shi, Tokyo 186-8650, Japan
| | - Amariliz Rivera
- Department of Pediatrics, Center for Immunity and Inflammation, New Jersey Medical School, Newark, NJ 07101
| | - Dean Sheppard
- Lung Biology Center, University of California, San Francisco, San Francisco, CA 94143
| | - Wenjun Ouyang
- Department of Immunology, Genentech, South San Francisco, CA 94080; and
| | - Lora V Hooper
- Department of Immunology, Howard Hughes Medical Institute, The University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Jay K Kolls
- Richard King Mellon Foundation Institute for Pediatric Research, Children's Hospital of Pittsburgh of University of Pittsburgh Medical Center, Pittsburgh, PA 15224;
| |
Collapse
|
579
|
Li J, Leyva-Castillo JM, Hener P, Eisenmann A, Zaafouri S, Jonca N, Serre G, Birling MC, Li M. Counterregulation between thymic stromal lymphopoietin– and IL-23–driven immune axes shapes skin inflammation in mice with epidermal barrier defects. J Allergy Clin Immunol 2016; 138:150-161.e13. [DOI: 10.1016/j.jaci.2016.01.013] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Revised: 01/11/2016] [Accepted: 01/22/2016] [Indexed: 12/11/2022]
|
580
|
Gurung P, Kanneganti TD. Autoinflammatory Skin Disorders: The Inflammasomme in Focus. Trends Mol Med 2016; 22:545-564. [PMID: 27267764 PMCID: PMC4925313 DOI: 10.1016/j.molmed.2016.05.003] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 05/10/2016] [Indexed: 12/24/2022]
Abstract
Autoinflammatory skin disorders are a group of heterogeneous diseases that include diseases such as cryopyrin-associated periodic syndrome (CAPS) and familial Mediterranean fever (FMF). Therapeutic strategies targeting IL-1 cytokines have proved helpful in ameliorating some of these diseases. While inflammasomes are the major regulators of IL-1 cytokines, inflammasome-independent complexes can also process IL-1 cytokines. Herein, we focus on recent advances in our understanding of how IL-1 cytokines, stemming from inflammasome-dependent and -independent pathways are involved in the regulation of skin conditions. Importantly, we discuss several mouse models of skin inflammation generated to help elucidate the basic cellular and molecular effects and modulation of IL-1 in the skin. Such models offer perspectives on how these signaling pathways could be targeted to improve therapeutic approaches in the treatment of these rare and debilitating inflammatory skin disorders.
Collapse
Affiliation(s)
- Prajwal Gurung
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | | |
Collapse
|
581
|
Borges TJ, O’Malley JT, Wo L, Murakami N, Smith B, Azzi J, Tripathi S, Lane JD, Bueno EM, Clark RA, Tullius SG, Chandraker A, Lian CG, Murphy GF, Strom TB, Pomahac B, Najafian N, Riella LV. Codominant Role of Interferon-γ- and Interleukin-17-Producing T Cells During Rejection in Full Facial Transplant Recipients. Am J Transplant 2016; 16:2158-71. [PMID: 26749226 PMCID: PMC4979599 DOI: 10.1111/ajt.13705] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Revised: 12/23/2015] [Accepted: 12/27/2015] [Indexed: 01/25/2023]
Abstract
Facial transplantation is a life-changing procedure for patients with severe composite facial defects. However, skin is the most immunogenic of all transplants, and better understanding of the immunological processes after facial transplantation is of paramount importance. Here, we describe six patients who underwent full facial transplantation at our institution, with a mean follow-up of 2.7 years. Seum, peripheral blood mononuclear cells, and skin biopsy specimens were collected prospectively, and a detailed characterization of their immune response (51 time points) was performed, defining 47 immune cell subsets, 24 serum cytokines, anti-HLA antibodies, and donor alloreactivity on each sample, producing 4269 data points. In a nonrejecting state, patients had a predominant T helper 2 cell phenotype in the blood. All patients developed at least one episode of acute cellular rejection, which was characterized by increases in interferon-γ/interleukin-17-producing cells in peripheral blood and in the allograft's skin. Serum monocyte chemotactic protein-1 level was significantly increased during rejection compared with prerejection time points. None of the patients developed de novo donor-specific antibodies, despite a fourfold expansion in T follicular helper cells at 1 year posttransplantation. In sum, facial transplantation is frequently complicated by a codominant interferon-γ/interleukin-17-mediated acute cellular rejection process. Despite that, medium-term outcomes are promising with no evidence of de novo donor-specific antibody development.
Collapse
Affiliation(s)
- T. J. Borges
- Schuster Transplantation Research Center, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA
| | - J. T. O’Malley
- Department of Dermatology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - L. Wo
- Division of Plastic Surgery, Department of Surgery, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA
| | - N. Murakami
- Schuster Transplantation Research Center, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA
| | - B. Smith
- Schuster Transplantation Research Center, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA
| | - J. Azzi
- Schuster Transplantation Research Center, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA
| | - S. Tripathi
- Schuster Transplantation Research Center, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA
| | - J. D. Lane
- Division of Plastic Surgery, Department of Surgery, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA
| | - E. M. Bueno
- Division of Plastic Surgery, Department of Surgery, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA
| | - R. A. Clark
- Department of Dermatology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - S. G. Tullius
- Division of Transplant Surgery, Department of Surgery, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA
| | - A. Chandraker
- Schuster Transplantation Research Center, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA
| | - C. G. Lian
- Program in Dermatopathology, Department of Pathology, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA
| | - G. F. Murphy
- Program in Dermatopathology, Department of Pathology, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA
| | - T. B. Strom
- Transplant Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - B. Pomahac
- Division of Plastic Surgery, Department of Surgery, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA
| | - N. Najafian
- Schuster Transplantation Research Center, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA,Department of Nephrology, Cleveland Clinic Florida, Weston, FL
| | - L. V. Riella
- Schuster Transplantation Research Center, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA,Corresponding author: Leonardo V. Riella,
| |
Collapse
|
582
|
Abstract
The increase in allergic airways disease has been linked to modern urbanization and lifestyle. Recent evidence suggests that the associated reduction in microbial exposure, reduction in dietary fibre intake and increased antibiotic use may cause early dysbiosis in infancy, which predisposes to immune dysregulation and allergic airways disease later in life. This implies that there may be a window of opportunity for primary prevention strategies aimed to protect or restore the microbiome early in life and thereby decrease the risk of developing allergic airways disease. Alternatively, strategies that correct dysbiosis may aid in the treatment of established allergic airways disease.
Collapse
|
583
|
|
584
|
Skin CD4(+) memory T cells exhibit combined cluster-mediated retention and equilibration with the circulation. Nat Commun 2016; 7:11514. [PMID: 27160938 PMCID: PMC4866325 DOI: 10.1038/ncomms11514] [Citation(s) in RCA: 144] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 04/04/2016] [Indexed: 12/20/2022] Open
Abstract
Although memory T cells within barrier tissues can persist as permanent residents, at least some exchange with blood. The extent to which this occurs is unclear. Here we show that memory CD4+ T cells in mouse skin are in equilibrium with the circulation at steady state. These cells are dispersed throughout the inter-follicular regions of the dermis and form clusters with antigen presenting cells around hair follicles. After infection or administration of a contact sensitizing agent, there is a sustained increase in skin CD4+ T-cell content, which is confined to the clusters, with a concomitant CCL5-dependent increase in CD4+ T-cell recruitment. Skin CCL5 is derived from CD11b+ cells and CD8+ T cells, with the elimination of the latter decreasing CD4+ T-cell numbers. These results reveal a complex pattern of tissue-retention and equilibration for CD4+ memory T cells in skin, which is altered by infection and inflammation history. Memory T cells are vital responders to skin inflammation, but cell localization and dynamics of exchange with the bloodstream are not clear. Here the authors use parabiosis and intravital microscopy to show that CD4+ memory T cells equilibrate with the circulation and cluster around hair follicles in response to CCL5-dependent responses to viral infection or contact sensitization.
Collapse
|
585
|
Direct and Indirect Horizontal Transmission of the Antifungal Probiotic Bacterium Janthinobacterium lividum on Green Frog (Lithobates clamitans) Tadpoles. Appl Environ Microbiol 2016; 82:2457-2466. [PMID: 26873311 DOI: 10.1128/aem.04147-15] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Accepted: 02/09/2016] [Indexed: 01/11/2023] Open
Abstract
Amphibian populations worldwide are being threatened by the disease chytridiomycosis, which is caused by Batrachochytrium dendrobatidis To mitigate the effects of B. dendrobatidis, bioaugmentation of antifungal bacteria has been shown to be a promising strategy. One way to implement bioaugmentation is through indirect horizontal transmission, defined as the transfer of bacteria from a host to the environment and to another host. In addition, direct horizontal transmission among individuals can facilitate the spread of a probiotic in a population. In this study, we tested whether the antifungal bacterium Janthinobacterium lividum could be horizontally transferred, directly or indirectly, in a laboratory experiment using Lithobates clamitans tadpoles. We evaluated the ability of J. lividumto colonize the tadpoles' skin and to persist through time using culture-dependent and culture-independent techniques. We also tested whether the addition of J. lividum affected the skin community in L. clamitans tadpoles. We found that transmission occurred rapidly by direct and indirect horizontal transmission, but indirect transmission that included a potential substrate was more effective. Even though J. lividum colonized the skin, its relative abundance on the tadpole skin decreased over time. The inoculation of J. lividum did not significantly alter the skin bacterial diversity of L. clamitans tadpoles, which was dominated by Pseudomonas Our results show that indirect horizontal transmission can be an effective bioaugmentation method. Future research is needed to determine the best conditions, including the presence of substrates, under which a probiotic can persist on the skin so that bioaugmentation becomes a successful strategy to mitigate chytridiomycosis.
Collapse
|
586
|
Skin microbiota-associated inflammation precedes autoantibody induced tissue damage in experimental epidermolysis bullosa acquisita. J Autoimmun 2016; 68:14-22. [DOI: 10.1016/j.jaut.2015.08.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2014] [Revised: 08/09/2015] [Accepted: 08/14/2015] [Indexed: 11/22/2022]
|
587
|
Cole LE, Zhang J, Kesselly A, Anosova NG, Lam H, Kleanthous H, Yethon JA. Limitations of Murine Models for Assessment of Antibody-Mediated Therapies or Vaccine Candidates against Staphylococcus epidermidis Bloodstream Infection. Infect Immun 2016; 84:1143-1149. [PMID: 26857577 PMCID: PMC4807487 DOI: 10.1128/iai.01472-15] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 01/29/2016] [Indexed: 02/02/2023] Open
Abstract
Staphylococcus epidermidis is normally a commensal colonizer of human skin and mucus membranes, but, due to its ability to form biofilms on indwelling medical devices, it has emerged as a leading cause of nosocomial infections. Bacteremia or bloodstream infection is a frequent and costly complication resulting from biofilm fouling of medical devices. Our goal was to develop a murine model of S. epidermidis infection to identify potential vaccine targets for the prevention of S. epidermidis bacteremia. However, assessing the contribution of adaptive immunity to protection against S. epidermidis challenge was complicated by a highly efficacious innate immune response in mice. Naive mice rapidly cleared S. epidermidis infections from blood and solid organs, even when the animals were immunocompromised. Cyclophosphamide-mediated leukopenia reduced the size of the bacterial challenge dose required to cause lethality but did not impair clearance after a nonlethal challenge. Nonspecific innate immune stimulation, such as treatment with a Toll-like receptor 4 (TLR4) agonist, enhanced bacterial clearance. TLR2 signaling was confirmed to accelerate the clearance of S. epidermidis bacteremia, but TLR2(-/-)mice could still resolve a bloodstream infection. Furthermore, TLR2 signaling played no role in the clearance of bacteria from the spleen. In conclusion, these data suggest that S. epidermidis bloodstream infection is cleared in a highly efficient manner that is mediated by both TLR2-dependent and -independent innate immune mechanisms. The inability to establish a persistent infection in mice, even in immunocompromised animals, rendered these murine models unsuitable for meaningful assessment of antibody-mediated therapies or vaccine candidates.
Collapse
Affiliation(s)
- Leah E Cole
- Sanofi Pasteur, Cambridge, Massachusetts, USA
| | | | | | | | - Hubert Lam
- Sanofi Pasteur, Cambridge, Massachusetts, USA
| | | | | |
Collapse
|
588
|
Greer R, Dong X, Morgun A, Shulzhenko N. Investigating a holobiont: Microbiota perturbations and transkingdom networks. Gut Microbes 2016; 7:126-35. [PMID: 26979110 PMCID: PMC4856449 DOI: 10.1080/19490976.2015.1128625] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
The scientific community has recently come to appreciate that, rather than existing as independent organisms, multicellular hosts and their microbiota comprise a complex evolving superorganism or metaorganism, termed a holobiont. This point of view leads to a re-evaluation of our understanding of different physiological processes and diseases. In this paper we focus on experimental and computational approaches which, when combined in one study, allowed us to dissect mechanisms (traditionally named host-microbiota interactions) regulating holobiont physiology. Specifically, we discuss several approaches for microbiota perturbation, such as use of antibiotics and germ-free animals, including advantages and potential caveats of their usage. We briefly review computational approaches to characterize the microbiota and, more importantly, methods to infer specific components of microbiota (such as microbes or their genes) affecting host functions. One such approach called transkingdom network analysis has been recently developed and applied in our study. (1) Finally, we also discuss common methods used to validate the computational predictions of host-microbiota interactions using in vitro and in vivo experimental systems.
Collapse
Affiliation(s)
- Renee Greer
- College of Veterinary Medicine, Oregon State University, Corvallis, OR, USA
| | - Xiaoxi Dong
- College of Pharmacy, Oregon State University, Corvallis, OR, USA
| | - Andrey Morgun
- College of Pharmacy, Oregon State University, Corvallis, OR, USA
| | | |
Collapse
|
589
|
Scharschmidt TC, Vasquez KS, Truong HA, Gearty SV, Pauli ML, Nosbaum A, Gratz IK, Otto M, Moon JJ, Liese J, Abbas AK, Fischbach MA, Rosenblum MD. A Wave of Regulatory T Cells into Neonatal Skin Mediates Tolerance to Commensal Microbes. Immunity 2016; 43:1011-21. [PMID: 26588783 DOI: 10.1016/j.immuni.2015.10.016] [Citation(s) in RCA: 390] [Impact Index Per Article: 43.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Revised: 08/06/2015] [Accepted: 09/14/2015] [Indexed: 02/06/2023]
Abstract
The skin is a site of constant dialog between the immune system and commensal bacteria. However, the molecular mechanisms that allow us to tolerate the presence of skin commensals without eliciting destructive inflammation are unknown. Using a model system to study the antigen-specific response to S. epidermidis, we demonstrated that skin colonization during a defined period of neonatal life was required for establishing immune tolerance to commensal microbes. This crucial window was characterized by an abrupt influx of highly activated regulatory T (Treg) cells into neonatal skin. Selective inhibition of this Treg cell wave completely abrogated tolerance. Thus, the host-commensal relationship in the skin relied on a unique Treg cell population that mediated tolerance to bacterial antigens during a defined developmental window. This suggests that the cutaneous microbiome composition in neonatal life is crucial in shaping adaptive immune responses to commensals, and disrupting these interactions might have enduring health implications.
Collapse
Affiliation(s)
- Tiffany C Scharschmidt
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Kimberly S Vasquez
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Hong-An Truong
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Sofia V Gearty
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Mariela L Pauli
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Audrey Nosbaum
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Iris K Gratz
- Department of Molecular Biology, University of Salzburg, 5020 Salzburg, Austria
| | - Michael Otto
- National Institute of Allergy and Infectious Disease, NIH, Bethesda, MD 20892, USA
| | - James J Moon
- Center for Immunology and Inflammatory Diseases, Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA
| | - Jan Liese
- Institute for Medical Microbiology and Hygiene, University Hospital Tübingen, 72076 Tübingen, Germany
| | - Abul K Abbas
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Michael A Fischbach
- Department of Bioengineering and Therapeutic Sciences and California Institute for Quantitative Biosciences, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Michael D Rosenblum
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
590
|
Zhang N, He QS. Commensal Microbiome Promotes Resistance to Local and Systemic Infections. Chin Med J (Engl) 2016; 128:2250-5. [PMID: 26265621 PMCID: PMC4717980 DOI: 10.4103/0366-6999.162502] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Objective: In this review, to illustrate the resistance mechanism for pathogen insult, we discussed the role of the intestinal microbiome in promoting resistance to local gastrointestinal tract infections and to respiratory tract infections. Data Sources: The review was based on data obtained from the published research articles. Study Selection: A total of 49 original articles were selected in accordance with our main objective to illustrate the resistance mechanism(s) by which commensal microbiota can contribute to host defense against local and systemic infections. Results: Diverse microorganisms colonize human environmentally exposed surfaces such as skin, respiratory tract, and gastrointestinal tract. Co-evolution has resulted in these microbes with extensive and diverse impacts on multiple aspects of host biological functions. During the last decade, high-throughput sequencing technology developed has been applied to study commensal microbiota and their impact on host biological functions. By using pathogen recognition receptors pathway and nucleotide binding oligomerization domain-like receptors pathway, the commensal microbiome promotes resistance to local and systemic infections, respectively. To protect against the local infections, the microbiome functions contain the following: The competing for sites of colonization, direct production of inhibition molecules or depletion of nutrients needed for pathogens, and priming immune defenses against pathogen insult. At the same time, with the purpose to maintain homeostasis, the commensal bacteria can program systemic signals toward not only local tissue but also distal tissue to modify their function for infections accordingly. Conclusions: Commensal bacteria play an essential role in protecting against infections, shaping and regulating immune responses, and maintaining host immune homeostasis.
Collapse
Affiliation(s)
| | - Qiu-Shui He
- Department of Medical Microbiology and Research Centre of Microbiome, Capital Medical University, Beijing 100069, China; Department of Medical Microbiology and Immunology, University of Turku, Turku, Finland,
| |
Collapse
|
591
|
Abstract
Atopic dermatitis (AD) is a common skin disease that affects a large proportion of the population worldwide. The incidence of AD has increased over the last several decades along with AD's burden on the physical and psychological health of the patient and family. However, current advances in understanding the mechanisms behind the pathophysiology of AD are leading to a hopeful outlook for the future. Staphylococcus aureus (S. aureus) colonization on AD skin has been directly correlated to disease severity but the functions of other members of the skin bacterial community may be equally important. Applying knowledge gained from understanding the role of the skin microbiome in maintaining normal skin immune function, and addressing the detrimental consequences of microbial dysbiosis in driving inflammation, is a promising direction for development of new treatments. This review discusses current preclinical and clinical research focused on determining how the skin microbiome may influence the development of AD.
Collapse
|
592
|
Oh JE, Kim BC, Chang DH, Kwon M, Lee SY, Kang D, Kim JY, Hwang I, Yu JW, Nakae S, Lee HK. Dysbiosis-induced IL-33 contributes to impaired antiviral immunity in the genital mucosa. Proc Natl Acad Sci U S A 2016; 113:E762-71. [PMID: 26811463 PMCID: PMC4760794 DOI: 10.1073/pnas.1518589113] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Commensal microbiota are well known to play an important role in antiviral immunity by providing immune inductive signals; however, the consequence of dysbiosis on antiviral immunity remains unclear. We demonstrate that dysbiosis caused by oral antibiotic treatment directly impairs antiviral immunity following viral infection of the vaginal mucosa. Antibiotic-treated mice succumbed to mucosal herpes simplex virus type 2 infection more rapidly than water-fed mice, and also showed delayed viral clearance at the site of infection. However, innate immune responses, including type I IFN and proinflammatory cytokine production at infection sites, as well as induction of virus-specific CD4 and CD8 T-cell responses in draining lymph nodes, were not impaired in antibiotic-treated mice. By screening the factors controlling antiviral immunity, we found that IL-33, an alarmin released in response to tissue damage, was secreted from vaginal epithelium after the depletion of commensal microbiota. This cytokine suppresses local antiviral immunity by blocking the migration of effector T cells to the vaginal tissue, thereby inhibiting the production of IFN-γ, a critical cytokine for antiviral defense, at local infection sites. These findings provide insight into the mechanisms of homeostasis maintained by commensal bacteria, and reveal a deleterious consequence of dysbiosis in antiviral immune defense.
Collapse
Affiliation(s)
- Ji Eun Oh
- Laboratory of Host Defenses, Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Byoung-Chan Kim
- Korean Collection for Type Cultures, Biological Resource Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea
| | - Dong-Ho Chang
- Korean Collection for Type Cultures, Biological Resource Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea
| | - Meehyang Kwon
- Center for Bioanalysis, Division of Metrology for Quality of Life, Korea Research Institute of Standards and Science, Daejeon 34113, Republic of Korea
| | - Sun Young Lee
- Center for Bioanalysis, Division of Metrology for Quality of Life, Korea Research Institute of Standards and Science, Daejeon 34113, Republic of Korea
| | - Dukjin Kang
- Center for Bioanalysis, Division of Metrology for Quality of Life, Korea Research Institute of Standards and Science, Daejeon 34113, Republic of Korea
| | - Jin Young Kim
- Division of Mass Spectrometry Research, Korea Basic Science Institute, Ochang 28119, Republic of Korea
| | - Inhwa Hwang
- Department of Microbiology, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Je-Wook Yu
- Department of Microbiology, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Susumu Nakae
- Laboratory of Systems Biology, Center for Experimental Medicine and Systems Biology, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan; Precursory Research for Embryonic Science and Technology, Japan Science and Technology Agency, Saitama 332-0012, Japan
| | - Heung Kyu Lee
- Laboratory of Host Defenses, Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea;
| |
Collapse
|
593
|
Abstract
Plasmodium infections result in clinical presentations that range from asymptomatic to severe malaria, resulting in ∼1 million deaths annually. Despite this toll on humanity, the factors that determine disease severity remain poorly understood. Here, we show that the gut microbiota of mice influences the pathogenesis of malaria. Genetically similar mice from different commercial vendors, which exhibited differences in their gut bacterial community, had significant differences in parasite burden and mortality after infection with multiple Plasmodium species. Germfree mice that received cecal content transplants from "resistant" or "susceptible" mice had low and high parasite burdens, respectively, demonstrating the gut microbiota shaped the severity of malaria. Among differences in the gut flora were increased abundances of Lactobacillus and Bifidobacterium in resistant mice. Susceptible mice treated with antibiotics followed by yogurt made from these bacterial genera displayed a decreased parasite burden. Consistent with differences in parasite burden, resistant mice exhibited an elevated humoral immune response compared with susceptible mice. Collectively, these results identify the composition of the gut microbiota as a previously unidentified risk factor for severe malaria and modulation of the gut microbiota (e.g., probiotics) as a potential treatment to decrease parasite burden.
Collapse
|
594
|
Skabytska Y, Kaesler S, Volz T, Biedermann T. Wie das angeborene Immunsystem die Immunität trainiert: neue Erkenntnisse über die atopische Dermatitis und kutane Bakterien. J Dtsch Dermatol Ges 2016. [DOI: 10.1111/ddg.100_12843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Yuliya Skabytska
- Klinik und Poliklinik für Dermatologie und Allergologie; TUM Fakultät für Medizin; Technische Universität München; Deutschland
- Universitäts-Hautklinik; Eberhard- Karls-Universität Tübingen; Deutschland
| | - Susanne Kaesler
- Klinik und Poliklinik für Dermatologie und Allergologie; TUM Fakultät für Medizin; Technische Universität München; Deutschland
- Universitäts-Hautklinik; Eberhard- Karls-Universität Tübingen; Deutschland
| | - Thomas Volz
- Klinik und Poliklinik für Dermatologie und Allergologie; TUM Fakultät für Medizin; Technische Universität München; Deutschland
| | - Tilo Biedermann
- Klinik und Poliklinik für Dermatologie und Allergologie; TUM Fakultät für Medizin; Technische Universität München; Deutschland
| |
Collapse
|
595
|
Skabytska Y, Kaesler S, Volz T, Biedermann T. How the innate immune system trains immunity: lessons from studying atopic dermatitis and cutaneous bacteria. J Dtsch Dermatol Ges 2016; 14:153-6. [PMID: 26788792 DOI: 10.1111/ddg.12843] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The skin is the largest organ at the interface between environment and host. It plays a major protective role against pathogens as physical barrier, as site of first recognition, and as orchestrator of consecutive immune responses. In this process, immunological crosstalk between skin-resident and immune cells is required, and fixed innate immune responses were previously believed to orchestrate adaptive immunity of B and T lymphocytes. Today, we understand that diverse qualities of immune responses to different microbes need to be regulated by also varying responses at the level of first microbe recognition through receptors of the innate immune system. Only fine-tuning of the innate immune system allows for the orchestration of immune responses to the microbiota in the absence of inflammation as well as to pathogens in the context of protective responses including inflammation. Understanding how innate immunity precisely adapts is also important for diseases such as atopic dermatitis (AD) with chronic inflammation. In this review, we present data on how the innate immune system actually fine-tunes its responses with special focus on the immunological consequences of cutaneous innate immune sensing through TLR2. These new insights are highly relevant for understanding microbiota-associated state of health, immune defense, and the pathogenesis underlying chronic cutaneous inflammation as seen in AD.
Collapse
Affiliation(s)
- Yuliya Skabytska
- Department of Dermatology and Allergology, TUM School of Medicine, Technische Universität München, Germany.,Department of Dermatology, Eberhard-Karls-University Tübingen, Germany
| | - Susanne Kaesler
- Department of Dermatology and Allergology, TUM School of Medicine, Technische Universität München, Germany.,Department of Dermatology, Eberhard-Karls-University Tübingen, Germany
| | - Thomas Volz
- Department of Dermatology and Allergology, TUM School of Medicine, Technische Universität München, Germany
| | - Tilo Biedermann
- Department of Dermatology and Allergology, TUM School of Medicine, Technische Universität München, Germany
| |
Collapse
|
596
|
Microbial community profiling shows dysbiosis in the lesional skin of Vitiligo subjects. Sci Rep 2016; 6:18761. [PMID: 26758568 PMCID: PMC4725359 DOI: 10.1038/srep18761] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 11/26/2015] [Indexed: 12/18/2022] Open
Abstract
Healthy human skin harbours a diverse array of microbes that comprise the skin microbiome. Commensal bacteria constitute an important component of resident microbiome and are intricately linked to skin health. Recent studies describe an association between altered skin microbial community and epidemiology of diseases, like psoriasis, atopic dermatitis etc. In this study, we compare the differences in bacterial community of lesional and non-lesional skin of vitiligo subjects. Our study reveals dysbiosis in the diversity of microbial community structure in lesional skin of vitiligo subjects. Although individual specific signature is dominant over the vitiligo-specific microbiota, a clear decrease in taxonomic richness and evenness can be noted in lesional patches. Investigation of community specific correlation networks reveals distinctive pattern of interactions between resident bacterial populations of the two sites (lesional and non-lesional). While Actinobacterial species constitute the central regulatory nodes (w.r.t. degree of interaction) in non-lesional skin, species belonging to Firmicutes dominate on lesional sites. We propose that the changes in taxonomic characteristics of vitiligo lesions, as revealed by our study, could play a crucial role in altering the maintenance and severity of disease. Future studies would elucidate mechanistic relevance of these microbial dynamics that can provide new avenues for therapeutic interventions.
Collapse
|
597
|
Kashem SW, Riedl MS, Yao C, Honda CN, Vulchanova L, Kaplan DH. Nociceptive Sensory Fibers Drive Interleukin-23 Production from CD301b+ Dermal Dendritic Cells and Drive Protective Cutaneous Immunity. Immunity 2016; 43:515-26. [PMID: 26377898 DOI: 10.1016/j.immuni.2015.08.016] [Citation(s) in RCA: 334] [Impact Index Per Article: 37.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Revised: 06/30/2015] [Accepted: 07/28/2015] [Indexed: 12/24/2022]
Abstract
Innate resistance to Candida albicans in mucosal tissues requires the production of interleukin-17A (IL-17A) by tissue-resident cells early during infection, but the mechanism of cytokine production has not been precisely defined. In the skin, we found that dermal γδ T cells were the dominant source of IL-17A during C. albicans infection and were required for pathogen resistance. Induction of IL-17A from dermal γδ T cells and resistance to C. albicans required IL-23 production from CD301b(+) dermal dendritic cells (dDCs). In addition, we found that sensory neurons were directly activated by C. albicans. Ablation of sensory neurons increased susceptibility to C. albicans infection, which could be rescued by exogenous addition of the neuropeptide CGRP. These data define a model in which nociceptive pathways in the skin drive production of IL-23 by CD301b(+) dDCs resulting in IL-17A production from γδ T cells and resistance to cutaneous candidiasis.
Collapse
MESH Headings
- Animals
- Candida albicans/immunology
- Candida albicans/physiology
- Candidiasis/genetics
- Candidiasis/immunology
- Candidiasis/microbiology
- Cells, Cultured
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Dermis/cytology
- Flow Cytometry
- Host-Pathogen Interactions/immunology
- Immunity/genetics
- Immunity/immunology
- Interleukin-17/genetics
- Interleukin-17/immunology
- Interleukin-17/metabolism
- Interleukin-23/genetics
- Interleukin-23/immunology
- Interleukin-23/metabolism
- Lectins, C-Type/immunology
- Lectins, C-Type/metabolism
- Mice, Inbred Strains
- Mice, Knockout
- Mice, Transgenic
- Oligonucleotide Array Sequence Analysis
- Receptors, Antigen, T-Cell, gamma-delta/genetics
- Receptors, Antigen, T-Cell, gamma-delta/immunology
- Receptors, Antigen, T-Cell, gamma-delta/metabolism
- Receptors, Calcitonin Gene-Related Peptide/genetics
- Receptors, Calcitonin Gene-Related Peptide/immunology
- Receptors, Calcitonin Gene-Related Peptide/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Sensory Receptor Cells/immunology
- Sensory Receptor Cells/metabolism
- Skin/immunology
- Skin/metabolism
- Skin/microbiology
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Transcriptome/genetics
- Transcriptome/immunology
Collapse
Affiliation(s)
- Sakeen W Kashem
- Department of Dermatology, Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Maureen S Riedl
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - Chen Yao
- Department of Dermatology, Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Christopher N Honda
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - Lucy Vulchanova
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - Daniel H Kaplan
- Department of Dermatology, Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
598
|
Sargent JL, Löhr CV, Diggs HE. Scratching Responses to Epidermal Injury in C57BL/6, DBA/2, BALB/c, and CD1 Mice. Comp Med 2016; 66:208-215. [PMID: 27298245 PMCID: PMC4907529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 09/28/2015] [Accepted: 10/29/2015] [Indexed: 06/06/2023]
Abstract
Whereas early investigations into ulcerative dermatitis (UD) focused on the possibility of a primary dermatopathology, several recent studies have advocated scratching behavior as a primary driver for UD. The aim of this study was to assess whether B6 mice exhibit excessive scratching under resting conditions or when provoked by epidermal barrier disruption. We hypothesized that B6 mice would exhibit more spontaneous scratching behavior and that B6 mice would be more pruritic after mild epidermal barrier injury compared with the other strains and stock tested. The behavior of the retired breeder female C57BL/6J, DBA/2J, BALB/cByJ, and Crl:CD1 mice was videotaped for 60 min. Behavior filming occurred at 17:15 and at 07:00 the next morning prior to (baseline) and after tape-stripping to initiate epidermal barrier disruption. Scratching duration was recorded as brief (less than 3 s) or prolonged (3 s or longer), on the basis of observations during a pilot study. In contrast to the hypothesis, B6 mice did not scratch significantly more frequently, have more long-duration scratching events, nor have a higher median scratching duration of prolonged scratching as compared with the other types of mice tested. In fact, B6 mice showed the lowest average scratching frequency and duration under both conditions. B6 mice demonstrated increased scratching behavior after epidermal barrier disruption, but the increased scratching did not surpass the rate or duration of scratching in the other types of mice tested. These findings do not support the idea that a strain-related tendency toward exaggerated scratching behavior under resting or epidermal barrier disruption conditions predisposes B6 mice to UD.
Collapse
Affiliation(s)
- Jennifer L Sargent
- Laboratory Animal Resources Center and Department of Biomedical Sciences, Oregon State University, Corvallis, Oregon, USA
| | - Christiane V Löhr
- Department of Biomedical Sciences, and Oregon Veterinary Diagnostic Laboratory, Oregon State University, Corvallis, Oregon, USA
| | - Helen E Diggs
- Laboratory Animal Resources Center and Department of Biomedical Sciences, Oregon State University, Corvallis, Oregon, USA
| |
Collapse
|
599
|
|
600
|
Egert M, Simmering R. The Microbiota of the Human Skin. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 902:61-81. [PMID: 27161351 DOI: 10.1007/978-3-319-31248-4_5] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The aim of this chapter is to sum up important progress in the field of human skin microbiota research that was achieved over the last years.The human skin is one of the largest and most versatile organs of the human body. Owing to its function as a protective interface between the largely sterile interior of the human body and the highly microbially contaminated outer environment, it is densely colonized with a diverse and active microbiota. This skin microbiota is of high importance for human health and well-being. It is implicated in several severe skin diseases and plays a major role in wound infections. Many less severe, but negatively perceived cosmetic skin phenomena are linked with skin microbes, too. In addition, skin microorganisms, in particular on the human hands, are crucial for the field of hygiene research. Notably, apart from being only a potential source of disease and contamination, the skin microbiota also contributes to the protective functions of the human skin in many ways. Finally, the analysis of structure and function of the human skin microbiota is interesting from a basic, evolutionary perspective on human microbe interactions.Key questions in the field of skin microbiota research deal with (a) a deeper understanding of the structure (species inventory) and function (physiology) of the healthy human skin microbiota in space and time, (b) the distinction of resident and transient skin microbiota members,
Collapse
Affiliation(s)
- Markus Egert
- Faculty of Medical and Life Sciences, Institute of Precision Medicine, Microbiology and Hygiene Group, Furtwangen University, Jakob-Kienzle-Str. 17, 78054, Villingen-Schwenningen, Germany.
| | - Rainer Simmering
- Corporate Scientific Services, Henkel AG & Co. KGaA, Henkelstr. 67, 40589, Düsseldorf, Germany
| |
Collapse
|