1
|
Spencer BL, Nguyen DT, Marroquin SM, Gapin L, O’Brien RL, Doran KS. Characterization of the Cellular Immune Response to Group B Streptococcal Vaginal Colonization. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.29.635275. [PMID: 39975125 PMCID: PMC11838357 DOI: 10.1101/2025.01.29.635275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Introduction Group B Streptococcus (GBS) asymptomatic colonizes the female genital tract (FGT) but can contribute to adverse pregnancy outcomes including pre-term birth, chorioamnionitis, and neonatal infection. We previously observed that GBS elicits FGT cytokine responses, including IL-17, during murine vaginal colonization; yet the anti-GBS cellular immune response during colonization remained unknown. We hypothesized that GBS may induce cellular immunity, resulting in FGT clearance. Methods Herein, we utilize depleting antibodies and knockout mice and performed flow cytometry to investigate cellular immunes responses during GBS colonization. Results We found that neutrophils (effectors of the IL-17 response) are important for GBS mucosal control as neutrophil depletion promoted increased GBS burdens in FGT tissues. Flow cytometric analysis of immune populations in the vagina, cervix, and uterus revealed, however, that GBS colonization did not induce a marked increase in FGT CD45+ immune cells. We also found that that Vγ6+ γδ T cells comprise a primary source of FGT IL-17. Finally, using knockout mice, we observed that IL-17-producing γδ T cells are important for the control of GBS in the FGT during murine colonization. Conclusions Taken together, this work characterizes FGT cellular immunity and suggests that GBS colonization does not elicit a significant immune response, which may be a bacterial directed adaptive outcome. However, certain FGT immune cells, such as neutrophils and ɣδ T cells, contribute to host defense and control of GBS colonization.
Collapse
Affiliation(s)
- Brady L. Spencer
- University of Colorado-Anschutz, Department of Immunology & Microbiology, Aurora, CO, USA
| | - Dustin T. Nguyen
- University of Colorado-Anschutz, Department of Immunology & Microbiology, Aurora, CO, USA
| | - Stephanie M. Marroquin
- University of Colorado-Anschutz, Department of Immunology & Microbiology, Aurora, CO, USA
| | - Laurent Gapin
- University of Colorado-Anschutz, Department of Immunology & Microbiology, Aurora, CO, USA
| | - Rebecca L. O’Brien
- National Jewish Health, Department of Biomedical Research, Denver, CO, USA
| | - Kelly S. Doran
- University of Colorado-Anschutz, Department of Immunology & Microbiology, Aurora, CO, USA
| |
Collapse
|
2
|
Guo W, Lian X, Li H, Jiang L, Chen Y, Shen P, Yu W. Characteristics of Immunocytes and Cytokines in Patients with Bloodstream Infections Caused by Carbapenem-Resistant Klebsiella pneumoniae in China. Infect Drug Resist 2024; 17:719-725. [PMID: 38410794 PMCID: PMC10896100 DOI: 10.2147/idr.s431207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 02/02/2024] [Indexed: 02/28/2024] Open
Abstract
Objective To evaluate the characteristics of immunocytes and cytokines associated with bloodstream infections (BSIs) caused by carbapenem-resistant Klebsiella pneumoniae (CRKP). Methods Patients with BSIs K. pneumoniae (BSIs-Kpn) were enrolled in our hospital between 2015 and 2022. Whole blood and serum samples were collected on the first day after diagnosis. Immunocytes and cytokines profiles were assessed using multicolor flow cytometry and multiplex immunoassays, respectively. The test cytokines included interferon-gamma (IFN-γ), tumor necrosis factor-alpha (TNF-α), interleukin (IL)-2, IL-4, IL-6, IL-10, and IL-17A. Results A total of 313 patients had BSIs-Kpn, including 145 with CRKP, 43 with extended-spectrum β-lactamases (ESBL) producing Kpn (ESBL-Kpn) and 125 with non-CRKP or non-ESBL-Kpn (susceptible Kpn, S-Kpn). Absolute number of leukomonocyte (CD45+) in CRKP, ESBL-Kpn and S-Kpn were 280.0 (138.0-523.0) cells/μL, 354.5 (150.3-737.3) cells/μL, and 637.0 (245.0-996.5) cells/μL, respectively. Compared with S-Kpn group, the absolute numbers of leukomonocyte (including T lymphocytes, B lymphocytes and natural killer cells) in patients with CRKP were significantly lower than that in patients with S-Kpn (P < 0.01). The levels of cytokines IL-2 and IL-17A were significantly higher in patients with S-Kpn than in those patients with CRKP (P<0.05). The area under receiver operating curve (AUC) of IL-2, IL-4, and IL-17A for S-Kpn was 0.576, 0.513, and 0.561, respectively, whereas that for the combination of these three cytokines with immunocytes was 0.804. Conclusion Patients with BSIs-CRKP had lower leukomonocyte counts. High levels of IL-2 and IL-17A combined with immunocyte subpopulations showed relatively high diagnostic value for BSIs-S-Kpn from BSIs-CRKP.
Collapse
Affiliation(s)
- Wenhui Guo
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People’s Republic of China
| | - Xiang Lian
- Department of Infectious Diseases, The Affiliated Xiangshan Hospital of Wenzhou Medical University; Xiangshan First People’s Hospital Medical and Health Group; Ningbo Fourth Hospital, Ningbo, People’s Republic of China
| | - Hong Li
- Department of Infectious Diseases, The Affiliated Xiangshan Hospital of Wenzhou Medical University; Xiangshan First People’s Hospital Medical and Health Group; Ningbo Fourth Hospital, Ningbo, People’s Republic of China
| | - Lushun Jiang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People’s Republic of China
| | - Yingsha Chen
- Department of Infectious Diseases, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People’s Republic of China
| | - Ping Shen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People’s Republic of China
| | - Wei Yu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People’s Republic of China
| |
Collapse
|
3
|
Hirata K, Yamada Y, Hamamoto Y, Tsunoda K, Muramatsu H, Horie S, Sukawa Y, Naganuma M, Nakagawa T, Kanai T. Prospective feasibility study of indigo naturalis ointment for chemotherapy-induced oral mucositis. BMJ Support Palliat Care 2024; 13:e1001-e1007. [PMID: 34649840 PMCID: PMC10850637 DOI: 10.1136/bmjspcare-2021-003199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 09/28/2021] [Indexed: 11/04/2022]
Abstract
OBJECTIVES Indigo naturalis, a herbal medicine effective against ulcerative colitis, exhibits anti-inflammatory effects and induces interleukin-22-mediated antimicrobial peptide production. Anti-inflammatory activity and the prevention of secondary infection are essential for the management of chemotherapy-induced oral mucositis (CIOM); therefore, we developed an indigo naturalis ointment to be administered topically for CIOM and evaluated its feasibility. METHODS We performed a single-centre, open-label, prospective feasibility study from March 2017 to December 2018. The key eligibility criteria for the subjects were as follows: (1) receiving chemotherapy for a malignant tumour; (2) grade 1 or 2 CIOM and (3) receiving continuous oral care. The treatment protocol comprised topical indigo naturalis ointment application three times a day for 7 days. The primary endpoint assessed was feasibility. The secondary endpoints assessed were the changes in oral findings, oral cavity pain and safety. RESULTS Nineteen patients with CIOM were enrolled. The average feasibility (the proportion of prescribed applications that were carried out) observed in this study was 94.7%±8.9% (95% CI 90.5% to 99.0%), which was higher than the expected feasibility. The revised oral assessment guide scores of the mucous membrane domain and total scores were significantly improved. All patients reported a reduction in oral cavity pain, with a median pain resolution duration of 6 days. No serious adverse events were observed. CONCLUSIONS The indigo naturalis ointment was feasible, and showed the potential for efficacy and safety. Larger randomised controlled trials are needed to further assess the efficacy and safety of indigo naturalis compared with a placebo. TRIAL REGISTRATION NUMBER UMIN000024271.
Collapse
Affiliation(s)
- Kenro Hirata
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Yuka Yamada
- Department of Dentistry and Oral Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Yasuo Hamamoto
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Kazuyuki Tsunoda
- Department of Dentistry and Oral Surgery, Keio University School of Medicine, Tokyo, Japan
| | | | - Sara Horie
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Yasutaka Sukawa
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Makoto Naganuma
- The Third Department of Internal Medicine, Division of Gastroenterology and Hepatology, Kansai Medical University, Osaka, Japan
| | - Taneaki Nakagawa
- Department of Dentistry and Oral Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Takanori Kanai
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
4
|
Role of wound microbiome, strategies of microbiota delivery system and clinical management. Adv Drug Deliv Rev 2023; 192:114671. [PMID: 36538989 DOI: 10.1016/j.addr.2022.114671] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 11/23/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022]
Abstract
Delayed wound healing is one of the most global public health threats affecting nearly 100 million people each year, particularly the chronic wounds. Many confounding factors such as aging, diabetic disease, medication, peripheral neuropathy, immunocompromises or arterial and venous insufficiency hyperglycaemia are considered to inhibit wound healing. Therapeutic approaches for slow wound healing include anti-infection, debridement and the use of various wound dressings. However, the current clinical outcomes are still unsatisfied. In this review, we discuss the role of skin and wound commensal microbiota in the different healing stages, including inflammation, cell proliferation, re-epithelialization and remodelling phase, followed by multiple immune cell responses to commensal microbiota. Current clinical management in treating surgical wounds and chronic wounds was also reviewed together with potential controlled delivery systems which may be utilized in the future for the topical administration of probiotics and microbiomes. This review aims to introduce advances, novel strategies, and pioneer ideas in regulating the wound microbiome and the design of controlled delivery systems.
Collapse
|
5
|
Ouyang J, Yan J, Zhou X, Isnard S, Harypursat V, Cui H, Routy JP, Chen Y. Relevance of biomarkers indicating gut damage and microbial translocation in people living with HIV. Front Immunol 2023; 14:1173956. [PMID: 37153621 PMCID: PMC10160480 DOI: 10.3389/fimmu.2023.1173956] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Accepted: 04/10/2023] [Indexed: 05/10/2023] Open
Abstract
The intestinal barrier has the daunting task of allowing nutrient absorption while limiting the entry of microbial products into the systemic circulation. HIV infection disrupts the intestinal barrier and increases intestinal permeability, leading to microbial product translocation. Convergent evidence has shown that gut damage and an enhanced level of microbial translocation contribute to the enhanced immune activation, the risk of non-AIDS comorbidity, and mortality in people living with HIV (PLWH). Gut biopsy procedures are invasive, and are not appropriate or feasible in large populations, even though they are the gold standard for intestinal barrier investigation. Thus, validated biomarkers that measure the degree of intestinal barrier damage and microbial translocation are needed in PLWH. Hematological biomarkers represent an objective indication of specific medical conditions and/or their severity, and should be able to be measured accurately and reproducibly via easily available and standardized blood tests. Several plasma biomarkers of intestinal damage, i.e., intestinal fatty acid-binding protein (I-FABP), zonulin, and regenerating islet-derived protein-3α (REG3α), and biomarkers of microbial translocation, such as lipopolysaccharide (LPS) and (1,3)-β-D-Glucan (BDG) have been used as markers of risk for developing non-AIDS comorbidities in cross sectional analyses and clinical trials, including those aiming at repair of gut damage. In this review, we critically discuss the value of different biomarkers for the estimation of gut permeability levels, paving the way towards developing validated diagnostic and therapeutic strategies to repair gut epithelial damage and to improve overall disease outcomes in PLWH.
Collapse
Affiliation(s)
- Jing Ouyang
- Department of Infectious Diseases, Chongqing Public Health Medical Center, Chongqing, China
- Clinical Research Center, Chongqing Public Health Medical Center, Chongqing, China
| | - Jiangyu Yan
- Department of Infectious Diseases, Chongqing Public Health Medical Center, Chongqing, China
- Clinical Research Center, Chongqing Public Health Medical Center, Chongqing, China
| | - Xin Zhou
- Clinical Research Center, Chongqing Public Health Medical Center, Chongqing, China
- Cancer Center, Medical Research Institute, Southwest University, Chongqing, China
| | - Stéphane Isnard
- Infectious Diseases and Immunity in Global Health Program, Research Institute, McGill University Health Centre, Montréal, QC, Canada
- Chronic Viral Illness Service, McGill University Health Centre, Montréal, QC, Canada
- Canadian HIV Trials Network, Canadian Institutes for Health Research, Vancouver, BC, Canada
| | - Vijay Harypursat
- Department of Infectious Diseases, Chongqing Public Health Medical Center, Chongqing, China
- Clinical Research Center, Chongqing Public Health Medical Center, Chongqing, China
| | - Hongjuan Cui
- Cancer Center, Medical Research Institute, Southwest University, Chongqing, China
| | - Jean-Pierre Routy
- Infectious Diseases and Immunity in Global Health Program, Research Institute, McGill University Health Centre, Montréal, QC, Canada
- Chronic Viral Illness Service, McGill University Health Centre, Montréal, QC, Canada
- Division of Hematology, McGill University Health Centre, Montréal, QC, Canada
- *Correspondence: Jean-Pierre Routy, ; Yaokai Chen,
| | - Yaokai Chen
- Department of Infectious Diseases, Chongqing Public Health Medical Center, Chongqing, China
- *Correspondence: Jean-Pierre Routy, ; Yaokai Chen,
| |
Collapse
|
6
|
Borges ÁH, Follmann F, Dietrich J. Chlamydia trachomatis vaccine development - a view on the current challenges and how to move forward. Expert Rev Vaccines 2022; 21:1555-1567. [PMID: 36004386 DOI: 10.1080/14760584.2022.2117694] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Chlamydia trachomatis is the most common sexually transmitted bacterial pathogen in the world. A licensed vaccine is not yet available, but the first vaccines have entered clinical trials. AREAS COVERED : We describe the progress that has been made in our understanding of the type of immunity that a protective vaccine should induce, and the challenges that vaccine developers face. We also focus on the clinical development of a chlamydia vaccine. The first chlamydia vaccine candidate has now been tested in a clinical phase-I trial, and another phase-I trial is currently running. We discuss what it will take to continue this development and what future trial setups could look like. EXPERT OPINION The chlamydia field is coming of age and the first phase I clinical trial of a C. trachomatis vaccine has been successfully completed. We expect and hope that this will motivate various stakeholders to support further development of chlamydia vaccines in humans.
Collapse
Affiliation(s)
- Álvaro H Borges
- Statens Serum Institut, Department of Infectious Diseases Immunology, Kobenhavn, 2300 Denmark
| | | | - Jes Dietrich
- Statens Serum Institut, Department of Infectious Diseases Immunology, Kobenhavn, 2300 Denmark
| |
Collapse
|
7
|
Mkhikian H, Hayama KL, Khachikyan K, Li C, Zhou RW, Pawling J, Klaus S, Tran PQN, Ly KM, Gong AD, Saryan H, Hai JL, Grigoryan D, Lee PL, Newton BL, Raffatellu M, Dennis JW, Demetriou M. Age-associated impairment of T cell immunity is linked to sex-dimorphic elevation of N-glycan branching. NATURE AGING 2022; 2:231-242. [PMID: 35528547 PMCID: PMC9075523 DOI: 10.1038/s43587-022-00187-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 02/02/2022] [Indexed: 11/08/2022]
Abstract
Impaired T cell immunity with aging increases mortality from infectious disease. The branching of Asparagine-linked glycans is a critical negative regulator of T cell immunity. Here we show that branching increases with age in females more than males, in naïve more than memory T cells, and in CD4+ more than CD8+ T cells. Female sex hormones and thymic output of naïve T cells (TN) decrease with age, however neither thymectomy nor ovariectomy altered branching. Interleukin-7 (IL-7) signaling was increased in old female more than male mouse TN cells, and triggered increased branching. N-acetylglucosamine, a rate-limiting metabolite for branching, increased with age in humans and synergized with IL-7 to raise branching. Reversing elevated branching rejuvenated T cell function and reduced severity of Salmonella infection in old female mice. These data suggest sex-dimorphic antagonistic pleiotropy, where IL-7 initially benefits immunity through TN maintenance but inhibits TN function by raising branching synergistically with age-dependent increases in N-acetylglucosamine.
Collapse
Affiliation(s)
- Haik Mkhikian
- Department of Pathology and Laboratory Medicine, University of California, Irvine, Irvine, CA, USA
| | - Ken L Hayama
- Department of Microbiology and Molecular Genetics, University of California, Irvine, Irvine, CA, USA
| | - Khachik Khachikyan
- Department of Neurology, University of California, Irvine, Irvine, CA, USA
| | - Carey Li
- Department of Neurology, University of California, Irvine, Irvine, CA, USA
| | - Raymond W Zhou
- Department of Neurology, University of California, Irvine, Irvine, CA, USA
| | - Judy Pawling
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Suzi Klaus
- Department of Microbiology and Molecular Genetics, University of California, Irvine, Irvine, CA, USA
| | - Phuong Q N Tran
- Department of Neurology, University of California, Irvine, Irvine, CA, USA
| | - Kim M Ly
- Department of Neurology, University of California, Irvine, Irvine, CA, USA
| | - Andrew D Gong
- Department of Neurology, University of California, Irvine, Irvine, CA, USA
| | - Hayk Saryan
- Department of Neurology, University of California, Irvine, Irvine, CA, USA
| | - Jasper L Hai
- Department of Neurology, University of California, Irvine, Irvine, CA, USA
| | - David Grigoryan
- Department of Neurology, University of California, Irvine, Irvine, CA, USA
| | - Philip L Lee
- Department of Neurology, University of California, Irvine, Irvine, CA, USA
| | - Barbara L Newton
- Department of Neurology, University of California, Irvine, Irvine, CA, USA
| | - Manuela Raffatellu
- Department of Microbiology and Molecular Genetics, University of California, Irvine, Irvine, CA, USA
- Division of Host-Microbe Systems and Therapeutics, Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA
- Center for Mucosal Immunology, Allergy, and Vaccines, Chiba University-UC San Diego, La Jolla, CA, USA
| | - James W Dennis
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Michael Demetriou
- Department of Microbiology and Molecular Genetics, University of California, Irvine, Irvine, CA, USA.
- Department of Neurology, University of California, Irvine, Irvine, CA, USA.
| |
Collapse
|
8
|
Wang J, Zhang W, Chu X, Wang S, Wang Y, Ji H. Deoxyshikonin-Induced Gene Expression Profile in Porcine Epithelial Cells. Front Vet Sci 2022; 8:711721. [PMID: 35097037 PMCID: PMC8792893 DOI: 10.3389/fvets.2021.711721] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 12/20/2021] [Indexed: 11/30/2022] Open
Affiliation(s)
- Jing Wang
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
- Sino-US Joint Laboratory of Animal Science, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
- *Correspondence: Jing Wang
| | - Wei Zhang
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
- Sino-US Joint Laboratory of Animal Science, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
| | - Xu Chu
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
- College of Agriculture and Animal Husbandry, Qinghai University, Xining, China
| | - Sixin Wang
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
| | - Yamin Wang
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
| | - Haifeng Ji
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
- Sino-US Joint Laboratory of Animal Science, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
| |
Collapse
|
9
|
Schmidt S, Sassu EL, Vatzia E, Pierron A, Lagler J, Mair KH, Stadler M, Knecht C, Spergser J, Dolezal M, Springer S, Theuß T, Fachinger V, Ladinig A, Saalmüller A, Gerner W. Vaccination and Infection of Swine With Salmonella Typhimurium Induces a Systemic and Local Multifunctional CD4 + T-Cell Response. Front Immunol 2021; 11:603089. [PMID: 33584671 PMCID: PMC7874209 DOI: 10.3389/fimmu.2020.603089] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Accepted: 11/25/2020] [Indexed: 01/14/2023] Open
Abstract
The gram-negative facultative intracellular bacteria Salmonella Typhimurium (STM) often leads to subclinical infections in pigs, but can also cause severe enterocolitis in this species. Due to its high zoonotic potential, the pathogen is likewise dangerous for humans. Vaccination with a live attenuated STM strain (Salmoporc) is regarded as an effective method to control STM infections in affected pig herds. However, information on the cellular immune response of swine against STM is still scarce. In this study, we investigated the T-cell immune response in pigs that were vaccinated twice with Salmoporc followed by a challenge infection with a virulent STM strain. Blood- and organ-derived lymphocytes (spleen, tonsils, jejunal and ileocolic lymph nodes, jejunum, ileum) were stimulated in vitro with heat-inactivated STM. Subsequently, CD4+ T cells present in these cell preparations were analyzed for the production of IFN-γ, TNF-α, and IL-17A by flow cytometry and Boolean gating. Highest frequencies of STM-specific cytokine-producing CD4+ T cells were found in lamina propria lymphocytes of jejunum and ileum. Significant differences of the relative abundance of cytokine-producing phenotypes between control group and vaccinated + infected animals were detected in most organs, but dominated in gut and lymph node-residing CD4+ T cells. IL-17A producing CD4+ T cells dominated in gut and gut-draining lymph nodes, whereas IFN-γ/TNF-α co-producing CD4+ T cells were present in all locations. Additionally, the majority of cytokine-producing CD4+ T cells had a CD8α+CD27- phenotype, indicative of a late effector or effector memory stage of differentiation. In summary, we show that Salmonella-specific multifunctional CD4+ T cells exist in vaccinated and infected pigs, dominate in the gut and most likely contribute to protective immunity against STM in the pig.
Collapse
Affiliation(s)
- Selma Schmidt
- Institute of Immunology, Department of Pathobiology, University of Veterinary Medicine, Vienna, Austria
| | - Elena L Sassu
- University Clinic for Swine, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine, Vienna, Austria
| | - Eleni Vatzia
- Institute of Immunology, Department of Pathobiology, University of Veterinary Medicine, Vienna, Austria
| | - Alix Pierron
- Institute of Immunology, Department of Pathobiology, University of Veterinary Medicine, Vienna, Austria
| | - Julia Lagler
- Institute of Immunology, Department of Pathobiology, University of Veterinary Medicine, Vienna, Austria.,Clinic for Poultry and Fish Medicine, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine, Vienna, Austria
| | - Kerstin H Mair
- Institute of Immunology, Department of Pathobiology, University of Veterinary Medicine, Vienna, Austria
| | - Maria Stadler
- Institute of Immunology, Department of Pathobiology, University of Veterinary Medicine, Vienna, Austria
| | - Christian Knecht
- University Clinic for Swine, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine, Vienna, Austria
| | - Joachim Spergser
- Institute of Microbiology, Department of Pathobiology, University of Veterinary Medicine, Vienna, Austria
| | - Marlies Dolezal
- Platform for Bioinformatics and Biostatistics, Department of Biomedical Sciences, University of Veterinary Medicine, Vienna, Austria
| | | | - Tobias Theuß
- Ceva Innovation Center GmbH, Dessau-Roßlau, Germany
| | | | - Andrea Ladinig
- University Clinic for Swine, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine, Vienna, Austria
| | - Armin Saalmüller
- Institute of Immunology, Department of Pathobiology, University of Veterinary Medicine, Vienna, Austria
| | - Wilhelm Gerner
- Institute of Immunology, Department of Pathobiology, University of Veterinary Medicine, Vienna, Austria
| |
Collapse
|
10
|
Zhang H, Pan S, Zhang K, Michiels J, Zeng Q, Ding X, Wang J, Peng H, Bai J, Xuan Y, Su Z, Bai S. Impact of Dietary Manganese on Intestinal Barrier and Inflammatory Response in Broilers Challenged with Salmonella Typhimurium. Microorganisms 2020; 8:microorganisms8050757. [PMID: 32443502 PMCID: PMC7285304 DOI: 10.3390/microorganisms8050757] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 05/10/2020] [Accepted: 05/16/2020] [Indexed: 11/16/2022] Open
Abstract
Growing concern for public health and food safety has prompted a special interest in developing nutritional strategies for removing waterborne and foodborne pathogens, including Salmonella. Strong links between manganese (Mn) and intestinal barrier or immune function hint that dietary Mn supplementation is likely to be a promising approach to limit the loads of pathogens in broilers. Here, we provide evidence that Salmonella Typhimurium (S. Typhimurium, 4 × 108 CFUs) challenge-induced intestinal injury along with systemic Mn redistribution in broilers. Further examining of the effect of dietary Mn treatments (a basal diet plus additional 0, 40, or 100 mg Mn/kg for corresponding to Mn-deficient, control, or Mn-surfeit diet, respectively) on intestinal barrier and inflammation status of broilers infected with S. Typhimurium revealed that birds fed the control and Mn-surfeit diets exhibited improved intestinal tight junctions and microbiota composition. Even without Salmonella infection, dietary Mn deficiency alone increased intestinal permeability by impairing intestinal tight junctions. In addition, when fed the control and Mn-surfeit diets, birds showed decreased Salmonella burdens in cecal content and spleen, with a concomitant increase in inflammatory cytokine levels in spleen. Furthermore, the dietary Mn-supplementation-mediated induction of cytokine production was probably associated with the nuclear factor kappa-B (NF-κB)/hydrogen peroxide (H2O2) pathway, as judged by the enhanced manganese superoxide dismutase activity and the increased H2O2 level in mitochondria, together with the increased mRNA level of NF-κB in spleen. Ingenuity-pathway analysis indicated that acute-phase response pathways, T helper type 1 pathway, and dendritic cell maturation were significantly activated by the dietary Mn supplementation. Our data suggest that dietary Mn supplementation could enhance intestinal barrier and splenic inflammatory response to fight against Salmonella infection in broilers.
Collapse
Affiliation(s)
- Huaiyong Zhang
- Key Laboratory of Animal Disease-Resistant Nutrition, Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (H.Z.); (S.P.); (K.Z.); (Q.Z.); (X.D.); (J.W.); (H.P.); (J.B.); (Y.X.); (Z.S.)
| | - Shuqin Pan
- Key Laboratory of Animal Disease-Resistant Nutrition, Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (H.Z.); (S.P.); (K.Z.); (Q.Z.); (X.D.); (J.W.); (H.P.); (J.B.); (Y.X.); (Z.S.)
| | - Keying Zhang
- Key Laboratory of Animal Disease-Resistant Nutrition, Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (H.Z.); (S.P.); (K.Z.); (Q.Z.); (X.D.); (J.W.); (H.P.); (J.B.); (Y.X.); (Z.S.)
| | - Joris Michiels
- Laboratory for Animal Nutrition and Animal Product Quality, Department of Animal Sciences and Aquatic Ecology, Ghent University, 9000 Ghent, Belgium;
| | - Qiufeng Zeng
- Key Laboratory of Animal Disease-Resistant Nutrition, Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (H.Z.); (S.P.); (K.Z.); (Q.Z.); (X.D.); (J.W.); (H.P.); (J.B.); (Y.X.); (Z.S.)
| | - Xuemei Ding
- Key Laboratory of Animal Disease-Resistant Nutrition, Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (H.Z.); (S.P.); (K.Z.); (Q.Z.); (X.D.); (J.W.); (H.P.); (J.B.); (Y.X.); (Z.S.)
| | - Jianping Wang
- Key Laboratory of Animal Disease-Resistant Nutrition, Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (H.Z.); (S.P.); (K.Z.); (Q.Z.); (X.D.); (J.W.); (H.P.); (J.B.); (Y.X.); (Z.S.)
| | - Huanwei Peng
- Key Laboratory of Animal Disease-Resistant Nutrition, Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (H.Z.); (S.P.); (K.Z.); (Q.Z.); (X.D.); (J.W.); (H.P.); (J.B.); (Y.X.); (Z.S.)
| | - Jie Bai
- Key Laboratory of Animal Disease-Resistant Nutrition, Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (H.Z.); (S.P.); (K.Z.); (Q.Z.); (X.D.); (J.W.); (H.P.); (J.B.); (Y.X.); (Z.S.)
| | - Yue Xuan
- Key Laboratory of Animal Disease-Resistant Nutrition, Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (H.Z.); (S.P.); (K.Z.); (Q.Z.); (X.D.); (J.W.); (H.P.); (J.B.); (Y.X.); (Z.S.)
| | - Zhuowei Su
- Key Laboratory of Animal Disease-Resistant Nutrition, Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (H.Z.); (S.P.); (K.Z.); (Q.Z.); (X.D.); (J.W.); (H.P.); (J.B.); (Y.X.); (Z.S.)
| | - Shiping Bai
- Key Laboratory of Animal Disease-Resistant Nutrition, Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (H.Z.); (S.P.); (K.Z.); (Q.Z.); (X.D.); (J.W.); (H.P.); (J.B.); (Y.X.); (Z.S.)
- Correspondence: ; Tel.: +86-028-86290922
| |
Collapse
|
11
|
Nguyen NDNT, Olsen AW, Lorenzen E, Andersen P, Hvid M, Follmann F, Dietrich J. Parenteral vaccination protects against transcervical infection with Chlamydia trachomatis and generate tissue-resident T cells post-challenge. NPJ Vaccines 2020; 5:7. [PMID: 31993218 PMCID: PMC6978417 DOI: 10.1038/s41541-020-0157-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 12/20/2019] [Indexed: 11/21/2022] Open
Abstract
The optimal protective immunity against Chlamydia trachomatis (C.t.) is still not fully resolved. One of the unresolved issues concerns the importance of resident immunity, since a recent study showed that optimal protection against a transcervical (TC) infection required genital tissue-resident memory T cells. An important question in the Chlamydia field is therefore if a parenteral vaccine strategy, inducing only circulating immunity primed at a nonmucosal site, should be pursued by Chlamydia vaccine developers. To address this question we studied the protective efficacy of a parenteral Chlamydia vaccine, formulated in the Th1/Th17 T cell-inducing adjuvant CAF01. We found that a parenteral vaccination induced significant protection against a TC infection and against development of chronic pathology. Protection correlated with rapid recruitment of Th1/Th17 T cells to the genital tract (GT), which efficiently prevented infection-driven generation of low quality Th1 or Th17 T cells, and instead maintained a pool of high quality multifunctional Th1/Th17 T cells in the GT throughout the infection. After clearance of the infection, a pool of these cells settled in the GT as tissue-resident Th1 and Th17 cells expressing CD69 but not CD103, CD49d, or CCR7, where they responded rapidly to a reinfection. These results show that a nonmucosal parenteral strategy inducing Th1 and Th17 T cells mediates protection against both infection with C.t. as well as development of chronic pathology, and lead to post-challenge protective tissue-resident memory immunity in the genital tract.
Collapse
Affiliation(s)
| | - Anja W Olsen
- 1Statens Serum Institut, Department for Infectious Disease Immunology, Copenhagen, Denmark
| | - Emma Lorenzen
- 1Statens Serum Institut, Department for Infectious Disease Immunology, Copenhagen, Denmark
| | - Peter Andersen
- 1Statens Serum Institut, Department for Infectious Disease Immunology, Copenhagen, Denmark
| | - Malene Hvid
- 2Department of Biomedicine and Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Frank Follmann
- 1Statens Serum Institut, Department for Infectious Disease Immunology, Copenhagen, Denmark
| | - Jes Dietrich
- 1Statens Serum Institut, Department for Infectious Disease Immunology, Copenhagen, Denmark
| |
Collapse
|
12
|
Abstract
Helicobacter pylori is a Gram-negative bacterium that infects the gastric epithelia of its human host. Everyone who is colonized with these pathogenic bacteria can develop gastric inflammation, termed gastritis. Additionally, a small proportion of colonized people develop more adverse outcomes, including gastric ulcer disease, gastric adenocarcinoma, or gastric mucosa-associated lymphoid tissue lymphoma. The development of these adverse outcomes is dependent on the establishment of a chronic inflammatory response. The development and control of this chronic inflammatory response are significantly impacted by CD4+ T helper cell activity. Noteworthy, T helper 17 (Th17) cells, a proinflammatory subset of CD4+ T cells, produce several proinflammatory cytokines that activate innate immune cell antimicrobial activity, drive a pathogenic immune response, regulate B cell responses, and participate in wound healing. Therefore, this review was written to take an intricate look at the involvement of Th17 cells and their affiliated cytokines (interleukin-17A [IL-17A], IL-17F, IL-21, IL-22, and IL-26) in regulating the immune response to H. pylori colonization and carcinogenesis.
Collapse
|
13
|
Ceccarelli G, Statzu M, Santinelli L, Pinacchio C, Bitossi C, Cavallari EN, Vullo V, Scagnolari C, d'Ettorre G. Challenges in the management of HIV infection: update on the role of probiotic supplementation as a possible complementary therapeutic strategy for cART treated people living with HIV/AIDS. Expert Opin Biol Ther 2019; 19:949-965. [PMID: 31260331 DOI: 10.1080/14712598.2019.1638907] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Introduction: Recent insights show that gut-mucosal immunity and intestinal microbiota play a key role in the pathogenesis of HIV infection. Alterations in the composition of intestinal flora (dysbiosis) could be associated with an impaired intestinal epithelium barrier activity and an impaired mucosal immunity function, significantly contributing to microbial translocation which is considered a major driver of chronic immune activation. Areas covered: This article provides an overview on the novel trends in probiotic therapy application. A particular emphasis is addressed to the importance of probiotics as a novel strategy to attenuate or prevent gastrointestinal involvement and to improve gut-mucosal immunity in HIV-infected subjects. Therefore, opportunities, limits and methodological criticalities of supplementation with probiotic therapy are considered and analyzed. Expert opinion: Use of probiotics is emerging as a novel strategy to manage dysbiosis and gut-mucosal impairment, to reduce immune activation and to limit a number of non-AIDS-related disorders. However, despite the growing use of probiotic therapy, mechanisms by which oral bacteria intake exhibits its effects are strain-related and disease-specific, hence clinicians need to take these two factors into consideration when suggesting probiotic supplementation to HIV-infected patients.
Collapse
Affiliation(s)
- Giancarlo Ceccarelli
- a Department of Public Health and Infectious Diseases, Sapienza University of Rome , Rome , Italy
| | - Maura Statzu
- b Laboratory of Virology, Department of Molecular Medicine, affiliated to Istituto Pasteur Italia - Cenci Bolognetti Foundation, Sapienza University of Rome , Rome , Italy
| | - Letizia Santinelli
- b Laboratory of Virology, Department of Molecular Medicine, affiliated to Istituto Pasteur Italia - Cenci Bolognetti Foundation, Sapienza University of Rome , Rome , Italy
| | - Claudia Pinacchio
- a Department of Public Health and Infectious Diseases, Sapienza University of Rome , Rome , Italy
| | - Camilla Bitossi
- b Laboratory of Virology, Department of Molecular Medicine, affiliated to Istituto Pasteur Italia - Cenci Bolognetti Foundation, Sapienza University of Rome , Rome , Italy
| | - Eugenio Nelson Cavallari
- a Department of Public Health and Infectious Diseases, Sapienza University of Rome , Rome , Italy
| | - Vincenzo Vullo
- a Department of Public Health and Infectious Diseases, Sapienza University of Rome , Rome , Italy
| | - Carolina Scagnolari
- b Laboratory of Virology, Department of Molecular Medicine, affiliated to Istituto Pasteur Italia - Cenci Bolognetti Foundation, Sapienza University of Rome , Rome , Italy
| | - GabrieIla d'Ettorre
- a Department of Public Health and Infectious Diseases, Sapienza University of Rome , Rome , Italy
| |
Collapse
|
14
|
Kondo S, Araki T, Okita Y, Yamamoto A, Hamada Y, Katsurahara M, Horiki N, Nakamura M, Shimoyama T, Yamamoto T, Takei Y, Kusunoki M. Colitis with wall thickening and edematous changes during oral administration of the powdered form of Qing-dai in patients with ulcerative colitis: a report of two cases. Clin J Gastroenterol 2018; 11:268-272. [DOI: 10.1007/s12328-018-0851-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Accepted: 03/09/2018] [Indexed: 12/28/2022]
|
15
|
Pan S, Zhang K, Ding X, Wang J, Peng H, Zeng Q, Xuan Y, Su Z, Wu B, Bai S. Effect of High Dietary Manganese on the Immune Responses of Broilers Following Oral Salmonella typhimurium Inoculation. Biol Trace Elem Res 2018; 181:347-360. [PMID: 28555440 DOI: 10.1007/s12011-017-1060-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 05/22/2017] [Indexed: 12/17/2022]
Abstract
Manganese (Mn) is an essential nutrient for both host and pathogen. Recent studies have demonstrated the nutritional immunity of Mn against Salmonella infection in mammals. To investigate the effect of high dietary Mn on immune responses of broilers following Salmonella challenge, 144 1-day-old male broilers were fed a basal diet (containing 20.04 mg Mn/kg) plus an additional 40 (the control group) or 400 mg Mn/kg (the H-Mn group) for 7 days. The 72 broilers in each group were then orally inoculated with 5 × 107 CFUs of Salmonella typhimurium (ATCC#14028) or phosphate-buffered saline. Peripheral blood, spleens, cecal tonsils, and bursa of Fabricius were collected from Salmonella-inoculated and Salmonella-noninoculated broilers (n = 6) at 2 days post inoculation (2 DPI) and 7 days post inoculation (7 DPI). Peripheral blood lymphocyte subpopulations were determined by flow cytometry. The messenger RNA (mRNA) abundance of genes was determined by quantitative real-time polymerase chain reaction. Salmonella counts were higher (P < 0.05) in the H-Mn group than that in the control group at 2 DPI in the cecal contents of Salmonella-inoculated broilers. High dietary Mn increased CD3+CD4+ and CD3+CD8+ percentages in the peripheral blood of Salmonella-inoculated broilers at 2 DPI. Salmonella inoculation increased interleukin (IL)-6 mRNA expression in spleens and bursa of Fabricius at 2 DPI and increased IL-1β and IL-6 mRNA expression in cecal tonsils at 7 DPI in the H-Mn group. These changes were not observed in the control group. High dietary Mn increased interferon-γ (IFN-γ) in spleens and decreased IFN-γ and IL-12 mRNA expression in cecal tonsils of Salmonella-inoculated broilers at 2 DPI. High dietary Mn decreased IL-17 mRNA expression in the bursa of Fabricius at 7 DPI, but increased this expression in cecal tonsils at 2 and 7 DPI in Salmonella-inoculated broilers. These results suggested that dietary Mn level affected T helper (Th) 1-cytokine reaction in spleens and cecal tonsils, and Th17-mediated immunity in cecal tonsils and the bursa of Fabricius of broilers when challenged with Salmonella.
Collapse
Affiliation(s)
- Shuqin Pan
- Institute of Animal Nutrition, Feed Engineering Research Centre of Sichuan Province, Sichuan Agricultural University, Huimin Road 211#, Wenjiang District, Chengdu, Sichuan, 611130, China
| | - Keying Zhang
- Institute of Animal Nutrition, Feed Engineering Research Centre of Sichuan Province, Sichuan Agricultural University, Huimin Road 211#, Wenjiang District, Chengdu, Sichuan, 611130, China
| | - Xuemei Ding
- Institute of Animal Nutrition, Feed Engineering Research Centre of Sichuan Province, Sichuan Agricultural University, Huimin Road 211#, Wenjiang District, Chengdu, Sichuan, 611130, China
| | - Jianping Wang
- Institute of Animal Nutrition, Feed Engineering Research Centre of Sichuan Province, Sichuan Agricultural University, Huimin Road 211#, Wenjiang District, Chengdu, Sichuan, 611130, China
| | - Huanwei Peng
- Institute of Animal Nutrition, Feed Engineering Research Centre of Sichuan Province, Sichuan Agricultural University, Huimin Road 211#, Wenjiang District, Chengdu, Sichuan, 611130, China
| | - Qiufeng Zeng
- Institute of Animal Nutrition, Feed Engineering Research Centre of Sichuan Province, Sichuan Agricultural University, Huimin Road 211#, Wenjiang District, Chengdu, Sichuan, 611130, China
| | - Yue Xuan
- Institute of Animal Nutrition, Feed Engineering Research Centre of Sichuan Province, Sichuan Agricultural University, Huimin Road 211#, Wenjiang District, Chengdu, Sichuan, 611130, China
| | - Zuowei Su
- Institute of Animal Nutrition, Feed Engineering Research Centre of Sichuan Province, Sichuan Agricultural University, Huimin Road 211#, Wenjiang District, Chengdu, Sichuan, 611130, China
| | - Bing Wu
- Chinese Chelota Group, Liangshui Industrial Estate, Jinyu District, Guanghan, Sihuan, 618300, China
| | - Shiping Bai
- Institute of Animal Nutrition, Feed Engineering Research Centre of Sichuan Province, Sichuan Agricultural University, Huimin Road 211#, Wenjiang District, Chengdu, Sichuan, 611130, China.
| |
Collapse
|
16
|
Guo Q, Betts C, Pennock N, Mitchell E, Schedin P. Mammary Gland Involution Provides a Unique Model to Study the TGF-β Cancer Paradox. J Clin Med 2017; 6:jcm6010010. [PMID: 28098775 PMCID: PMC5294963 DOI: 10.3390/jcm6010010] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2016] [Revised: 12/21/2016] [Accepted: 12/27/2016] [Indexed: 12/12/2022] Open
Abstract
Transforming Growth Factor-β (TGF-β) signaling in cancer has been termed the “TGF-β paradox”, acting as both a tumor suppresser and promoter. The complexity of TGF-β signaling within the tumor is context dependent, and greatly impacted by cellular crosstalk between TGF-β responsive cells in the microenvironment including adjacent epithelial, endothelial, mesenchymal, and hematopoietic cells. Here we utilize normal, weaning-induced mammary gland involution as a tissue microenvironment model to study the complexity of TGF-β function. This article reviews facets of mammary gland involution that are TGF-β regulated, namely mammary epithelial cell death, immune activation, and extracellular matrix remodeling. We outline how distinct cellular responses and crosstalk between cell types during physiologically normal mammary gland involution contribute to simultaneous tumor suppressive and promotional microenvironments. We also highlight alternatives to direct TGF-β blocking anti-cancer therapies with an emphasis on eliciting concerted microenvironmental-mediated tumor suppression.
Collapse
Affiliation(s)
- Qiuchen Guo
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University, Portland, OR 97239, USA.
| | - Courtney Betts
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University, Portland, OR 97239, USA.
| | - Nathan Pennock
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University, Portland, OR 97239, USA.
| | - Elizabeth Mitchell
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University, Portland, OR 97239, USA.
| | - Pepper Schedin
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University, Portland, OR 97239, USA.
- Young Women's Breast Cancer Translational Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA.
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97239, USA.
| |
Collapse
|
17
|
Serious Non-AIDS Events: Therapeutic Targets of Immune Activation and Chronic Inflammation in HIV Infection. Drugs 2016; 76:533-49. [PMID: 26915027 DOI: 10.1007/s40265-016-0546-7] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
In the antiretroviral therapy (ART) era, serious non-AIDS events (SNAEs) have become the major causes of morbidity and mortality in HIV-infected persons. Early ART initiation has the strongest evidence for reducing SNAEs and mortality. Biomarkers of immune activation, inflammation and coagulopathy do not fully normalize despite virologic suppression and persistent immune activation is an important contributor to SNAEs. A number of strategies aimed to reduce persistent immune activation including ART intensification to reduce residual viremia; treatment of co-infections to reduce chronic antigen stimulation; the use of anti-inflammatory agents, reducing microbial translocation as well as interventions to improve immune recovery through cytokine administration and reducing lymphoid tissue fibrosis, have been investigated. To date, there is little conclusive evidence on which strategies beyond treatment of hepatitis B and C co-infections and reducing cardiovascular risk factors will result in clinical benefits in patients already on ART with viral suppression. The use of statins seems to show early promise and larger clinical trials are underway to confirm their efficacy. At this stage, clinical care of HIV-infected patients should therefore focus on early diagnosis and prompt ART initiation, treatment of active co-infections and the aggressive management of co-morbidities until further data are available.
Collapse
|
18
|
Naganuma M, Mizuno S, Nanki K, Sugimoto S, Kanai T. Recent trends and future directions for the medical treatment of ulcerative colitis. Clin J Gastroenterol 2016; 9:329-336. [DOI: 10.1007/s12328-016-0686-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 09/22/2016] [Indexed: 02/07/2023]
|
19
|
Beneficial Effects of Sodium Phenylbutyrate Administration during Infection with Salmonella enterica Serovar Typhimurium. Infect Immun 2016; 84:2639-52. [PMID: 27382022 PMCID: PMC4995890 DOI: 10.1128/iai.00132-16] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 06/24/2016] [Indexed: 02/07/2023] Open
Abstract
Sodium phenylbutyrate (PBA) is a derivative of the short-chain fatty acid butyrate and is approved for treatment of urea cycle disorders and progressive familial intrahepatic cholestasis type 2. Previously known functions include histone deacetylase inhibitor, endoplasmic reticulum stress inhibitor, ammonia sink, and chemical chaperone. Here, we show that PBA has a previously undiscovered protective role in host mucosal defense during infection. Administration of PBA to Taconic mice resulted in the increase of intestinal Lactobacillales and segmented filamentous bacteria (SFB), as well as an increase of interleukin 17 (IL-17) production by intestinal cells. This effect was not observed in Jackson Laboratory mice, which are not colonized with SFB. Because previous studies showed that IL-17 plays a protective role during infection with mucosal pathogens, we hypothesized that Taconic mice treated with PBA would be more resistant to infection with Salmonella enterica serovar Typhimurium (S Typhimurium). By using the streptomycin-treated mouse model, we found that Taconic mice treated with PBA exhibited significantly lower S Typhimurium intestinal colonization and dissemination to the reticuloendothelial system, as well as lower levels of inflammation. The lower levels of S Typhimurium gut colonization and intestinal inflammation were not observed in Jackson Laboratory mice. Although PBA had no direct effect on bacterial replication, its administration reduced S Typhimurium epithelial cell invasion and lowered the induction of the proinflammatory cytokine IL-23 in macrophage-like cells. These effects likely contributed to the better outcome of infection in PBA-treated mice. Overall, our results suggest that PBA induces changes in the microbiota and in the mucosal immune response that can be beneficial to the host during infection with S Typhimurium and possibly other enteric pathogens.
Collapse
|
20
|
Dixon BREA, Radin JN, Piazuelo MB, Contreras DC, Algood HMS. IL-17a and IL-22 Induce Expression of Antimicrobials in Gastrointestinal Epithelial Cells and May Contribute to Epithelial Cell Defense against Helicobacter pylori. PLoS One 2016; 11:e0148514. [PMID: 26867135 PMCID: PMC4750979 DOI: 10.1371/journal.pone.0148514] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 01/19/2016] [Indexed: 12/11/2022] Open
Abstract
Helicobacter pylori colonization of the human stomach can lead to adverse clinical outcomes including gastritis, peptic ulcers, or gastric cancer. Current data suggest that in addition to bacterial virulence factors, the magnitude and types of immune responses influence the outcome of colonization. Specifically, CD4+ T cell responses impact the pathology elicited in response to H. pylori. Because gastritis is believed to be the initiating host response to more detrimental pathological outcomes, there has been a significant interest in pro-inflammatory T cell cytokines, including the cytokines produced by T helper 17 cells. Th17 cells produce IL-17A, IL-17F, IL-21 and IL-22. While these cytokines have been linked to inflammation, IL-17A and IL-22 are also associated with anti-microbial responses and control of bacterial colonization. The goal of this research was to determine the role of IL-22 in activation of antimicrobial responses in models of H. pylori infection using human gastric epithelial cell lines and the mouse model of H. pylori infection. Our data indicate that IL-17A and IL-22 work synergistically to induce antimicrobials and chemokines such as IL-8, components of calprotectin (CP), lipocalin (LCN) and some β-defensins in both human and primary mouse gastric epithelial cells (GEC) and gastroids. Moreover, IL-22 and IL-17A-activated GECs were capable of inhibiting growth of H. pylori in vitro. While antimicrobials were activated by IL-17A and IL-22 in vitro, using a mouse model of H. pylori infection, the data herein indicate that IL-22 deficiency alone does not render mice more susceptible to infection, change their antimicrobial gene transcription, or significantly change their inflammatory response.
Collapse
Affiliation(s)
- Beverly R. E. A. Dixon
- Department of Medicine, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Jana N. Radin
- Department of Medicine, Vanderbilt University, Nashville, Tennessee, United States of America
| | - M. Blanca Piazuelo
- Department of Medicine, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Diana C. Contreras
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Holly M. Scott Algood
- Veterans Affairs Tennessee Valley Healthcare Services, Nashville, Tennessee, United States of America
- Department of Medicine, Vanderbilt University, Nashville, Tennessee, United States of America
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, Tennessee, United States of America
| |
Collapse
|
21
|
Pichavant M, Sharan R, Le Rouzic O, Olivier C, Hennegrave F, Rémy G, Pérez-Cruz M, Koné B, Gosset P, Just N, Gosset P. IL-22 Defect During Streptococcus pneumoniae Infection Triggers Exacerbation of Chronic Obstructive Pulmonary Disease. EBioMedicine 2015; 2:1686-96. [PMID: 26870795 PMCID: PMC4740310 DOI: 10.1016/j.ebiom.2015.09.040] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Revised: 09/22/2015] [Accepted: 09/23/2015] [Indexed: 12/22/2022] Open
Abstract
Progression of chronic obstructive pulmonary disease (COPD) is linked to episodes of exacerbations caused by bacterial infections due to Streptococcus pneumoniae. Our objective was to identify during COPD, factors of susceptibility to bacterial infections among cytokine network and their role in COPD exacerbations. S. pneumoniae was used to sub-lethally challenge mice chronically exposed to air or cigarette smoke (CS) and to stimulate peripheral blood mononuclear cells (PBMC) from non-smokers, smokers and COPD patients. The immune response and the cytokine production were evaluated. Delayed clearance of the bacteria and stronger lung inflammation observed in infected CS-exposed mice were associated with an altered production of IL-17 and IL-22 by innate immune cells. This defect was related to a reduced production of IL-1β and IL-23 by antigen presenting cells. Importantly, supplementation with recombinant IL-22 restored bacterial clearance in CS-exposed mice and limited lung alteration. In contrast with non-smokers, blood NK and NKT cells from COPD patients failed to increase IL-17 and IL-22 levels in response to S. pneumoniae, in association with a defect in IL-1β and IL-23 secretion. This study identified IL-17 and IL-22 as susceptibility factors in COPD exacerbation. Therefore targeting such cytokines could represent a potent strategy to control COPD exacerbation.
Collapse
Key Words
- AM, alveolar macrophages
- APC, antigen presenting cells
- BAL, broncho-alveolar lavage
- Bacterial infection
- CFU, colony forming unit
- COPD, chronic obstructive pulmonary disease
- CS, cigarette smoke
- Chronic obstructive pulmonary disease
- DC, dendritic cells
- IL-22
- Innate immunity
- NK, natural killer cells
- NKT, natural killer T cells
- PBMC, peripheral blood mononuclear cells
- Sp, Streptococcus pneumoniae
Collapse
Affiliation(s)
- Muriel Pichavant
- Institut Pasteur de Lille, Centre d'Infection et d'Immunité de Lille, F-59019 Lille, France
- Université Lille Nord de France, F-59000 Lille, France
- Centre National de la Recherche Scientifique, UMR 8204, F-59021 Lille, France
- Institut National de la Santé et de la Recherche Médicale, U1019, F-59019 Lille, France
- Institut Fédératif de Recherche 142, F-59019 Lille, France
| | - Riti Sharan
- Institut Pasteur de Lille, Centre d'Infection et d'Immunité de Lille, F-59019 Lille, France
- Université Lille Nord de France, F-59000 Lille, France
- Centre National de la Recherche Scientifique, UMR 8204, F-59021 Lille, France
- Institut National de la Santé et de la Recherche Médicale, U1019, F-59019 Lille, France
- Institut Fédératif de Recherche 142, F-59019 Lille, France
| | - Olivier Le Rouzic
- Institut Pasteur de Lille, Centre d'Infection et d'Immunité de Lille, F-59019 Lille, France
- Université Lille Nord de France, F-59000 Lille, France
- Centre National de la Recherche Scientifique, UMR 8204, F-59021 Lille, France
- Institut National de la Santé et de la Recherche Médicale, U1019, F-59019 Lille, France
- Institut Fédératif de Recherche 142, F-59019 Lille, France
- Service de Pneumologie, Hôpital Calmette, CHRU, Lille, France
| | - Cécile Olivier
- Institut Pasteur de Lille, Centre d'Infection et d'Immunité de Lille, F-59019 Lille, France
- Université Lille Nord de France, F-59000 Lille, France
- Centre National de la Recherche Scientifique, UMR 8204, F-59021 Lille, France
- Institut National de la Santé et de la Recherche Médicale, U1019, F-59019 Lille, France
- Institut Fédératif de Recherche 142, F-59019 Lille, France
- Service de Pneumologie, Hôpital Calmette, CHRU, Lille, France
| | - Florence Hennegrave
- Institut Pasteur de Lille, Centre d'Infection et d'Immunité de Lille, F-59019 Lille, France
- Université Lille Nord de France, F-59000 Lille, France
- Centre National de la Recherche Scientifique, UMR 8204, F-59021 Lille, France
- Institut National de la Santé et de la Recherche Médicale, U1019, F-59019 Lille, France
- Institut Fédératif de Recherche 142, F-59019 Lille, France
- Service de Pneumologie, Hôpital Calmette, CHRU, Lille, France
| | - Gaëlle Rémy
- Institut Pasteur de Lille, Centre d'Infection et d'Immunité de Lille, F-59019 Lille, France
- Université Lille Nord de France, F-59000 Lille, France
- Centre National de la Recherche Scientifique, UMR 8204, F-59021 Lille, France
- Institut National de la Santé et de la Recherche Médicale, U1019, F-59019 Lille, France
- Institut Fédératif de Recherche 142, F-59019 Lille, France
| | - Magdiel Pérez-Cruz
- Institut Pasteur de Lille, Centre d'Infection et d'Immunité de Lille, F-59019 Lille, France
- Université Lille Nord de France, F-59000 Lille, France
- Centre National de la Recherche Scientifique, UMR 8204, F-59021 Lille, France
- Institut National de la Santé et de la Recherche Médicale, U1019, F-59019 Lille, France
- Institut Fédératif de Recherche 142, F-59019 Lille, France
| | - Bachirou Koné
- Institut Pasteur de Lille, Centre d'Infection et d'Immunité de Lille, F-59019 Lille, France
- Université Lille Nord de France, F-59000 Lille, France
- Centre National de la Recherche Scientifique, UMR 8204, F-59021 Lille, France
- Institut National de la Santé et de la Recherche Médicale, U1019, F-59019 Lille, France
- Institut Fédératif de Recherche 142, F-59019 Lille, France
| | - Pierre Gosset
- Service d'Anatomo-Pathologie, Hôpital Saint Vincent de Paul, Lille, France
| | - Nicolas Just
- Service de Pneumologie, Hôpital Victor Provo, Roubaix, France
| | - Philippe Gosset
- Institut Pasteur de Lille, Centre d'Infection et d'Immunité de Lille, F-59019 Lille, France
- Université Lille Nord de France, F-59000 Lille, France
- Centre National de la Recherche Scientifique, UMR 8204, F-59021 Lille, France
- Institut National de la Santé et de la Recherche Médicale, U1019, F-59019 Lille, France
- Institut Fédératif de Recherche 142, F-59019 Lille, France
| |
Collapse
|
22
|
Behnsen J, Perez-Lopez A, Nuccio SP, Raffatellu M. Exploiting host immunity: the Salmonella paradigm. Trends Immunol 2015; 36:112-20. [PMID: 25582038 DOI: 10.1016/j.it.2014.12.003] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Revised: 12/12/2014] [Accepted: 12/12/2014] [Indexed: 01/08/2023]
Abstract
Pathogens have evolved clever strategies to evade and in some cases exploit the attacks of an activated immune system. Salmonella enterica is one such pathogen, exploiting multiple aspects of host defense to promote its replication in the host. Here we review recent findings on the mechanisms by which Salmonella establishes systemic and chronic infection, including strategies involving manipulation of innate immune signaling and inflammatory forms of cell death, as well as immune evasion by establishing residency in M2 macrophages. We also examine recent evidence showing that the oxidative environment and the high levels of antimicrobial proteins produced in response to localized Salmonella gastrointestinal infection enable the pathogen to successfully outcompete the resident gut microbiota.
Collapse
Affiliation(s)
- Judith Behnsen
- Department of Microbiology and Molecular Genetics, University of California, Irvine School of Medicine, Irvine, CA 92697-4025, USA; Institute for Immunology, University of California, Irvine School of Medicine, Irvine, CA 92697-4025, USA
| | - Araceli Perez-Lopez
- Department of Microbiology and Molecular Genetics, University of California, Irvine School of Medicine, Irvine, CA 92697-4025, USA; Institute for Immunology, University of California, Irvine School of Medicine, Irvine, CA 92697-4025, USA
| | - Sean-Paul Nuccio
- Department of Microbiology and Molecular Genetics, University of California, Irvine School of Medicine, Irvine, CA 92697-4025, USA
| | - Manuela Raffatellu
- Department of Microbiology and Molecular Genetics, University of California, Irvine School of Medicine, Irvine, CA 92697-4025, USA; Institute for Immunology, University of California, Irvine School of Medicine, Irvine, CA 92697-4025, USA.
| |
Collapse
|
23
|
Schuetz A, Deleage C, Sereti I, Rerknimitr R, Phanuphak N, Phuang-Ngern Y, Estes JD, Sandler NG, Sukhumvittaya S, Marovich M, Jongrakthaitae S, Akapirat S, Fletscher JLK, Kroon E, Dewar R, Trichavaroj R, Chomchey N, Douek DC, O′Connell RJ, Ngauy V, Robb ML, Phanuphak P, Michael NL, Excler JL, Kim JH, de Souza MS, Ananworanich J. Initiation of ART during early acute HIV infection preserves mucosal Th17 function and reverses HIV-related immune activation. PLoS Pathog 2014; 10:e1004543. [PMID: 25503054 PMCID: PMC4263756 DOI: 10.1371/journal.ppat.1004543] [Citation(s) in RCA: 209] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Accepted: 10/28/2014] [Indexed: 02/07/2023] Open
Abstract
Mucosal Th17 cells play an important role in maintaining gut epithelium integrity and thus prevent microbial translocation. Chronic HIV infection is characterized by mucosal Th17 cell depletion, microbial translocation and subsequent immune-activation, which remain elevated despite antiretroviral therapy (ART) correlating with increased mortality. However, when Th17 depletion occurs following HIV infection is unknown. We analyzed mucosal Th17 cells in 42 acute HIV infection (AHI) subjects (Fiebig (F) stage I-V) with a median duration of infection of 16 days and the short-term impact of early initiation of ART. Th17 cells were defined as IL-17+ CD4+ T cells and their function was assessed by the co-expression of IL-22, IL-2 and IFNγ. While intact during FI/II, depletion of mucosal Th17 cell numbers and function was observed during FIII correlating with local and systemic markers of immune-activation. ART initiated at FI/II prevented loss of Th17 cell numbers and function, while initiation at FIII restored Th17 cell numbers but not their polyfunctionality. Furthermore, early initiation of ART in FI/II fully reversed the initially observed mucosal and systemic immune-activation. In contrast, patients treated later during AHI maintained elevated mucosal and systemic CD8+ T-cell activation post initiation of ART. These data support a loss of Th17 cells at early stages of acute HIV infection, and highlight that studies of ART initiation during early AHI should be further explored to assess the underlying mechanism of mucosal Th17 function preservation.
Collapse
Affiliation(s)
- Alexandra Schuetz
- Department of Retrovirology, Armed Forces Research Institute of Medical Sciences – United States Component, Bangkok, Thailand
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, United States of America
| | - Claire Deleage
- AIDS and Cancer Virus Program, Leidos Biomedical Research Inc., Frederick National Laboratory of Cancer Research, Frederick, Maryland, United States of America
| | - Irini Sereti
- Clinical and Molecular Retrovirology Section/Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Rungsun Rerknimitr
- Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Nittaya Phanuphak
- SEARCH, Bangkok, Thailand
- The Thai Red Cross AIDS Research Centre, Bangkok, Thailand
| | - Yuwadee Phuang-Ngern
- Department of Retrovirology, Armed Forces Research Institute of Medical Sciences – United States Component, Bangkok, Thailand
| | - Jacob D. Estes
- AIDS and Cancer Virus Program, Leidos Biomedical Research Inc., Frederick National Laboratory of Cancer Research, Frederick, Maryland, United States of America
| | - Netanya G. Sandler
- Human Immunology Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Suchada Sukhumvittaya
- Department of Retrovirology, Armed Forces Research Institute of Medical Sciences – United States Component, Bangkok, Thailand
| | - Mary Marovich
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| | - Surat Jongrakthaitae
- Department of Retrovirology, Armed Forces Research Institute of Medical Sciences – United States Component, Bangkok, Thailand
| | - Siriwat Akapirat
- Department of Retrovirology, Armed Forces Research Institute of Medical Sciences – United States Component, Bangkok, Thailand
| | | | - Eugene Kroon
- Department of Retrovirology, Armed Forces Research Institute of Medical Sciences – United States Component, Bangkok, Thailand
- SEARCH, Bangkok, Thailand
| | - Robin Dewar
- Virus Isolation and Serology Laboratory Applied and Developmental Research Directorate Science Applications International Corporation, Frederick, Inc. National Cancer Institute, Frederick Cancer Research and Development Center, Frederick, Maryland, United States of America
| | - Rapee Trichavaroj
- Department of Retrovirology, Armed Forces Research Institute of Medical Sciences – United States Component, Bangkok, Thailand
| | - Nitiya Chomchey
- SEARCH, Bangkok, Thailand
- The Thai Red Cross AIDS Research Centre, Bangkok, Thailand
| | - Daniel C. Douek
- Human Immunology Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Robert J. O′Connell
- Department of Retrovirology, Armed Forces Research Institute of Medical Sciences – United States Component, Bangkok, Thailand
| | - Viseth Ngauy
- Department of Retrovirology, Armed Forces Research Institute of Medical Sciences – United States Component, Bangkok, Thailand
| | - Merlin L. Robb
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, United States of America
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| | - Praphan Phanuphak
- Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- SEARCH, Bangkok, Thailand
- The Thai Red Cross AIDS Research Centre, Bangkok, Thailand
| | - Nelson L. Michael
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| | - Jean-Louis Excler
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, United States of America
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| | - Jerome H. Kim
- SEARCH, Bangkok, Thailand
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| | - Mark S. de Souza
- Department of Retrovirology, Armed Forces Research Institute of Medical Sciences – United States Component, Bangkok, Thailand
- SEARCH, Bangkok, Thailand
| | - Jintanat Ananworanich
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, United States of America
- SEARCH, Bangkok, Thailand
- The Thai Red Cross AIDS Research Centre, Bangkok, Thailand
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| | | |
Collapse
|
24
|
Le PT, Pearce MM, Zhang S, Campbell EM, Fok CS, Mueller ER, Brincat CA, Wolfe AJ, Brubaker L. IL22 regulates human urothelial cell sensory and innate functions through modulation of the acetylcholine response, immunoregulatory cytokines and antimicrobial peptides: assessment of an in vitro model. PLoS One 2014; 9:e111375. [PMID: 25354343 PMCID: PMC4213028 DOI: 10.1371/journal.pone.0111375] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Accepted: 09/26/2014] [Indexed: 11/21/2022] Open
Abstract
Human urinary disorders are generally studied in rodent models due to limitations of functional in vitro culture models of primary human urothelial cells (HUCs). Current HUC culture models are often derived from immortalized cancer cell lines, which likely have functional characteristics differ from healthy human urothelium. Here, we described a simple explant culture technique to generate HUCs and assessed their in vitro functions. Using transmission electron microscopy, we assessed morphology and heterogeneity of the generated HUCs and characterized their intercellular membrane structural proteins relative to ex vivo urothelium tissue. We demonstrated that our cultured HUCs are free of fibroblasts. They are also heterogeneous, containing cells characteristic of both immature basal cells and mature superficial urothelial cells. The cultured HUCs expressed muscarinic receptors (MR1 and MR2), carnitine acetyltransferase (CarAT), immunoregulatory cytokines IL7, IL15, and IL23, as well as the chemokine CCL20. HUCs also expressed epithelial cell-specific molecules essential for forming intercellular structures that maintain the functional capacity to form the physiological barrier of the human bladder urothelium. A subset of HUCs, identified by the high expression of CD44, expressed the Toll-like receptor 4 (TLR4) along with its co-receptor CD14. We demonstrated that HUCs express, at the mRNA level, both forms of the IL22 receptor, the membrane-associated (IL22RA1) and the secreted soluble (IL22RA2) forms; in turn, IL22 inhibited expression of MR1 and induced expression of CarAT and two antimicrobial peptides (S100A9 and lipocalin-2). While the cellular sources of IL22 have yet to be identified, the HUC cytokine and chemokine profiles support the concept that IL22-producing cells are present in the human bladder mucosa tissue and that IL22 plays a regulatory role in HUC functions. Thus, the described explant technique is clearly capable of generating functional HUCs suitable for the study of human urinary tract disorders, including interactions between urothelium and IL22-producing cells.
Collapse
Affiliation(s)
- Phong T. Le
- Department of Microbiology and Immunology, Stritch School of Medicine, Loyola University Chicago, Maywood, Illinois, United States of America
| | - Meghan M. Pearce
- Department of Microbiology and Immunology, Stritch School of Medicine, Loyola University Chicago, Maywood, Illinois, United States of America
| | - Shubin Zhang
- Department of Microbiology and Immunology, Stritch School of Medicine, Loyola University Chicago, Maywood, Illinois, United States of America
| | - Edward M. Campbell
- Department of Microbiology and Immunology, Stritch School of Medicine, Loyola University Chicago, Maywood, Illinois, United States of America
| | - Cynthia S. Fok
- University of Minnesota, Department of Urology, Minneapolis, Minnesota, United States of America
| | - Elizabeth R. Mueller
- Department of Obstetrics and Gynecology, Stritch School of Medicine, Loyola University Chicago, Maywood, Illinois, United States of America
| | - Cynthia A. Brincat
- Department of Obstetrics and Gynecology, Stritch School of Medicine, Loyola University Chicago, Maywood, Illinois, United States of America
| | - Alan J. Wolfe
- Department of Microbiology and Immunology, Stritch School of Medicine, Loyola University Chicago, Maywood, Illinois, United States of America
| | - Linda Brubaker
- Department of Obstetrics and Gynecology, Stritch School of Medicine, Loyola University Chicago, Maywood, Illinois, United States of America
| |
Collapse
|
25
|
Behnsen J, Jellbauer S, Wong CP, Edwards RA, George MD, Ouyang W, Raffatellu M. The cytokine IL-22 promotes pathogen colonization by suppressing related commensal bacteria. Immunity 2014; 40:262-73. [PMID: 24508234 PMCID: PMC3964146 DOI: 10.1016/j.immuni.2014.01.003] [Citation(s) in RCA: 230] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Accepted: 11/27/2013] [Indexed: 12/16/2022]
Abstract
Interleukin-22 (IL-22) is highly induced in response to infections with a variety of pathogens, and its main functions are considered to be tissue repair and host defense at mucosal surfaces. Here we showed that IL-22 has a unique role during infection in that its expression suppressed the intestinal microbiota and enhanced the colonization of a pathogen. IL-22 induced the expression of antimicrobial proteins, including lipocalin-2 and calprotectin, which sequester essential metal ions from microbes. Because Salmonella enterica ser. Typhimurium can overcome metal ion starvation mediated by lipocalin-2 and calprotectin via alternative pathways, IL-22 boosted its colonization of the inflamed intestine by suppressing commensal Enterobacteriaceae, which are susceptible to the antimicrobial proteins. Thus, IL-22 tipped the balance between pathogenic and commensal bacteria in favor of a pathogen. Taken together, IL-22 induction can be exploited by pathogens to suppress the growth of their closest competitors, thereby enhancing pathogen colonization of mucosal surfaces.
Collapse
Affiliation(s)
- Judith Behnsen
- Department of Microbiology, University of California, Irvine, Irvine, CA 92697, USA; Institute for Immunology, University of California, Irvine, Irvine, CA 92697, USA
| | - Stefan Jellbauer
- Department of Microbiology, University of California, Irvine, Irvine, CA 92697, USA; Institute for Immunology, University of California, Irvine, Irvine, CA 92697, USA
| | - Christina P Wong
- Department of Microbiology, University of California, Irvine, Irvine, CA 92697, USA; Institute for Immunology, University of California, Irvine, Irvine, CA 92697, USA
| | - Robert A Edwards
- Institute for Immunology, University of California, Irvine, Irvine, CA 92697, USA; Department of Pathology and Laboratory Medicine, University of California, Irvine, Irvine, CA 92697, USA
| | - Michael D George
- Department of Medical Microbiology and Immunology, School of Medicine, University of California, Davis, CA 95616, USA
| | - Wenjun Ouyang
- Department of Immunology, Genentech, South San Francisco, CA 94080, USA
| | - Manuela Raffatellu
- Department of Microbiology, University of California, Irvine, Irvine, CA 92697, USA; Institute for Immunology, University of California, Irvine, Irvine, CA 92697, USA.
| |
Collapse
|
26
|
Sinha B, Rubens M. Systemic immune activation in HIV and potential therapeutic options. Immunopharmacol Immunotoxicol 2014; 36:89-95. [PMID: 24552614 DOI: 10.3109/08923973.2014.890217] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
CONTEXT Advancement in HIV treatment has evolved over the last two decades with the discovery of new drugs and approaches. Studies have demonstrated that HIV-infected individuals have elevated immune activation even during effective antiretroviral therapy. Persistently elevated immune activation has been one of the main obstacles against developing an effective approach for curing HIV. OBJECTIVE This review examines the mechanism of microbial translocation in HIV-infected individuals and currently investigated potential therapeutic approaches. METHODS We searched PubMed and Medline for peer-reviwed articles and recent HIV/AIDS conference abstracts and papers. Narrative review method was used since the objectives of the study were mechanism of microbial translocation and mechanism of action of multiple drugs against it. RESULTS Microbial translocation occurs as a result of the disruption of epithelial barrier and immunological dysfunction within the intestinal tract due to defective tight junctions, loss of TH17 type CD4(+) T cells, impaired liver architecture, and depletion of intestinal myelomonocytic cells. Potent and effective way to intervene microbial translocation is to target the mechanism of actions involved in microbial translocation by restoration of beneficial microbiata with supplemental probiotics/prebiotics, increased clearance of microbial products from systemic circulation with targeted antibodies and restoration of intestinal integrity with antibiotics. CONCLUSIONS Number of promising drug molecules against microbial translocation are currently under various stages of trials and the results of these trials will hopefully contribute significantly toward effective therapeutic intervention. However, studies also need to explore the effect of combination drugs to abrogate microbial translocation.
Collapse
|
27
|
Interleukin-17-induced protein lipocalin 2 is dispensable for immunity to oral candidiasis. Infect Immun 2013; 82:1030-5. [PMID: 24343647 DOI: 10.1128/iai.01389-13] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Oropharyngeal candidiasis (OPC; thrush) is an opportunistic fungal infection caused by the commensal microbe Candida albicans. Immunity to OPC is strongly dependent on CD4+ T cells, particularly those of the Th17 subset. Interleukin-17 (IL-17) deficiency in mice or humans leads to chronic mucocutaneous candidiasis, but the specific downstream mechanisms of IL-17-mediated host defense remain unclear. Lipocalin 2 (Lcn2; 24p3; neutrophil gelatinase-associated lipocalin [NGAL]) is an antimicrobial host defense factor produced in response to inflammatory cytokines, particularly IL-17. Lcn2 plays a key role in preventing iron acquisition by bacteria that use catecholate-type siderophores, and lipocalin 2(-/-) mice are highly susceptible to infection by Escherichia coli and Klebsiella pneumoniae. The role of Lcn2 in mediating immunity to fungi is poorly defined. Accordingly, in this study, we evaluated the role of Lcn2 in immunity to oral infection with C. albicans. Lcn2 is strongly upregulated following oral infection with C. albicans, and its expression is almost entirely abrogated in mice with defective IL-17 signaling (IL-17RA(-/-) or Act1(-/-) mice). However, Lcn2(-/-) mice were completely resistant to OPC, comparably to wild-type (WT) mice. Moreover, Lcn2 deficiency mediated protection from OPC induced by steroid immunosuppression. Therefore, despite its potent regulation during C. albicans infection, Lcn2 is not required for immunity to mucosal candidiasis.
Collapse
|
28
|
Inflammatory Responses to Salmonella Infections Are Serotype-Specific. INTERNATIONAL JOURNAL OF BACTERIOLOGY 2013; 2013:168179. [PMID: 26904722 PMCID: PMC4745457 DOI: 10.1155/2013/168179] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Revised: 07/15/2013] [Accepted: 08/13/2013] [Indexed: 12/16/2022]
Abstract
The main purpose of this study was to investigate the profile of inflammatory response in patients with acute salmonellosis caused by two serotypes of Salmonella enterica, S. Enteritidis and S. Typhimurium, as well as in convalescent patients with previous acute disease caused by S. Enteritidis. Patients with acute disease showed significantly elevated levels of IL-1β, IL-17, IL-10, and calprotectin compared to healthy control subjects. In convalescent patients, these markers were also significantly elevated, with the exception of IL-1β. Multivariate statistical analyses with the use of these variables produced models with a good predictive accuracy resulting in excellent separation of the diseased and healthy cohorts studied. Overall, the results suggest that the profile of inflammatory response in this disease is determined, to a significant degree, by the serotype of Salmonella, and the profile of certain cytokines and calprotectin remains abnormal for a number of months following the acute disease stage.
Collapse
|
29
|
Liu KL, Yu RJ, Wu J, Lin XC. Significance of interleukin 17-A expression in colorectal cancer and adenoma. Shijie Huaren Xiaohua Zazhi 2013; 21:2522-2529. [DOI: 10.11569/wcjd.v21.i25.2522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the expression and role of interleukin-17 (IL-17) in colorectal cancer and adenoma.
METHODS: Tissue samples taken from patients with colorectal cancer or adenoma who were treated in our gastrointestinal endoscopy unit between 2009 and 2011 were used in this retrospective study. The expression of IL-17A in the above tissue samples was detected by immunohistochemistry. The relationship between IL-17A expression and clinicopathological characteristics and β-catenin localization was analyzed.
RESULTS: The positive rate of IL-17A expression was significantly higher in colorectal cancer, colorectal adenoma and ulcerative colitis than in normal colorectal tissue (54.2%, 45.8%, 50.0% vs 0.0%, all P < 0.01). IL-17A expression was not associated with tumor location, degree of differentiation, Duke's stage, lymph node metastasis, MVD or Ki-67 expression in colorectal cancer (all P > 0.05). IL-17A expression was also not associated with histological type, MVD, concomitant colorectal cancer, location, size, color, number or shape of adenomas (all P > 0.05) in colorectal adenoma. The expression of IL-17A was higher in adenomas with high-grade dysplasia than in those with mild to moderate dysplasia, but the difference was not significant (29.62 ± 26.77 vs 16.98 ± 10.62, P > 0.05). The expression of IL-17A was significantly higher in adenomas with nuclear expression of β-catenin than in those with cytoplasmic or membrane expression of β-catenin (29.11 ± 6.16 vs 16.95 ± 7.99, 15.10 ± 11.46, P = 0.038, 0.021).
CONCLUSION: IL-17A expression is elevated in colorectal cancer and adenomas and may mediate nuclear translocation of β-catenin.
Collapse
|
30
|
Behnsen J, Raffatellu M. Keeping the peace: aryl hydrocarbon receptor signaling modulates the mucosal microbiota. Immunity 2013; 39:206-7. [PMID: 23973218 DOI: 10.1016/j.immuni.2013.08.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
In this issue of Immunity, Zelante et al. (2013) and Qiu et al. (2013) provide mechanistic insights into functional interactions between commensal microbes and innate lymphoid cells via the aryl hydrocarbon receptor.
Collapse
Affiliation(s)
- Judith Behnsen
- Department of Microbiology and Molecular Genetics, School of Medicine, University of California, Irvine, Irvine, CA 92697-4025, USA
| | | |
Collapse
|
31
|
Zhang H, Li H, Li Y, Zou Y, Dong X, Song W, Jia C, Li S, Xi H, Liu D, Wang Y. IL-17 plays a central role in initiating experimental Candida albicans infection in mouse corneas. Eur J Immunol 2013; 43:2671-82. [PMID: 23843112 DOI: 10.1002/eji.201242891] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2012] [Revised: 06/05/2013] [Accepted: 07/05/2013] [Indexed: 12/13/2022]
Abstract
The pathogenesis of fungal infection in the cornea remains largely unclear. To understand how the immune system influences the progression of fungal infection in corneas, we inoculated immunocompetent BALB/c mice, neutrophil- or CD4⁺ T-cell-depleted BALB/c mice, and nude mice with Candida albicans. We found that only immunocompetent BALB/c mice developed typical Candida keratitis (CaK), while the other mouse strains lacked obvious clinical manifestations. Furthermore, CaK development was blocked in immunocompetent mice treated with anti-IL-17A or anti-IL-23p19 to neutralize IL-17 activity. However, no significant effects were observed when Treg cells, γδ T cells, or IFN-γ were immunodepleted. Upon infection, the corneas of BALB/c mice were infiltrated with IL-17-producing leukocytes, including neutrophils and, to a lesser degree, CD4⁺ T cells. In contrast, leukocyte recruitment to corneas was significantly diminished in nude mice. Indeed, nude mice produced much less chemokines (e.g. CXCL1, CXCL2, CXCL10, CXCL12, CCL2, and IL-6) in response to inoculation. Remarkably, addition of CXCL2 during inoculation restored CaK induction in nude mice. In contrast to its therapeutic effect on CaK, neutralization of IL-17 exacerbated Candida-induced dermatitis in skin. We conclude that IL-17, mainly produced by neutrophils and CD4⁺ T cells in the corneas, is essential in the pathogenesis of CaK.
Collapse
Affiliation(s)
- Hongbo Zhang
- Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong Academy of Medical Sciences, Qingdao, P. R. China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Taooka Y, Ohe M, Chen L, Sutani A, Higashi Y, Isobe T. Increased expression levels of integrin α9β1 and CD11b on circulating neutrophils and elevated serum IL-17A in elderly aspiration pneumonia. Respiration 2012; 86:367-75. [PMID: 23257917 DOI: 10.1159/000345390] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2012] [Accepted: 10/22/2012] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Repeated aspiration pneumonia is a serious problem in the elderly. In aspiration pneumonia, neutrophils play an important role in acute lung injury, while CD18-independent neutrophil transmigration pathways have also been reported in acid-aspiration pneumonia animal models. However, the involvement of IL-17A and β1 integrin still remains unclear. The β1 integrin subfamily integrin α9β1 has been shown to be expressed on human neutrophils and to mediate adhesion to extracellular matrix proteins including the vascular cell adhesion molecule-1. OBJECTIVES To elucidate the possible involvement of β1 integrin subfamily and IL-17A in aspiration pneumonia. METHODS We analyzed the expression levels of CD11b, CD18 and integrin α9β1 in circulating neutrophils and serum concentration of IL-17A, IL-22 and IL-23 in elderly aspiration pneumonia patients (n = 32, 14 males and 18 females, 78.8 ± 3.9 years old) at 2 time points (on the day of admission before starting antibiotics and the day after finishing antibiotics) and compared the results with those of a control group (n = 30, 13 males and 17 females, 76.1 ± 3.4 years old). RESULTS Recombinant IL-17A stimulated integrin α9β1 and CD11b expression levels in healthy human neutrophils in vitro. The expression levels of integrin α9β1 and CD11b in circulating neutrophils were significantly higher in pneumonia patients compared with the controls. In addition, serum IL-17A concentration was significantly increased in pneumonia patients. Integrin α9β1 levels positively correlated with serum IL-17A and CD18 expression levels. CONCLUSIONS These findings suggest a potential role of integrin α9β1 expressed in neutrophils and elevated serum IL-17A in extravasation of neutrophils in cases of aspiration pneumonia.
Collapse
Affiliation(s)
- Yasuyuki Taooka
- Division of Medical Oncology and Respiratory Medicine, Department of Internal Medicine, Faculty of Medicine, Shimane University, Izumo, Japan
| | | | | | | | | | | |
Collapse
|
33
|
Nyagode BA, Watkins WJ, Kinloch RD, Morgan ET. Selective modulation of hepatic cytochrome P450 and flavin monooxygenase 3 expression during citrobacter rodentium infection in severe combined immune-deficient mice. Drug Metab Dispos 2012; 40:1894-9. [PMID: 22745333 PMCID: PMC3463824 DOI: 10.1124/dmd.112.046557] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2012] [Accepted: 06/27/2012] [Indexed: 12/13/2022] Open
Abstract
The profile of selective modulation of hepatic cytochrome P450 (P450) gene expression caused by infection with the murine intestinal pathogen Citrobacter rodentium has been well characterized in multiple genetic backgrounds; yet, the mechanisms underlying this modulation are still not entirely understood. Although several studies have addressed the roles of cytokines from the innate immune system, the influence of the adaptive immune system is not known. To address this deficiency, we used mice harboring the severe combined immune deficiency (SCID) spontaneous mutation, which lack mature T and B lymphocytes and are unable to mount an acquired immune response. Female C57BL/6 (B6) and SCID mice were infected orally with C. rodentium and assessed for bacterial colonization/translocation and P450 and flavin monooxygenase-3 (Fmo3) expression levels after 7 days. SCID mice showed similar patterns of colonic bacterial colonization and a similar degree of colonic mucosal hypertrophy compared with infected B6 mice, but SCID mice displayed 6-fold greater bacterial translocation to the liver. In the SCID mice, Cyp4a10 and Cyp2b9 down-regulations were partially and fully blocked, respectively, whereas the regulation of other P450s and Fmo3 was similar in both strains. In the C3H genetic background, the SCID mutation also blocked the down-regulation of Cyp3a11, Cyp3a25, Cyp2d22, and Cyp2c29. The results clearly dissociate bacterial translocation to the liver from hepatic drug-metabolizing enzyme regulation and suggest a possible role of T cells, T-cell cytokines, or other proteins regulated by such cytokines in the selective regulation of a limited subset of hepatic P450 enzymes during C. rodentium infection.
Collapse
Affiliation(s)
- Beatrice A Nyagode
- Department of Pharmacology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | | | | | | |
Collapse
|
34
|
Kaakoush NO, Day AS, Huinao KD, Leach ST, Lemberg DA, Dowd SE, Mitchell HM. Microbial dysbiosis in pediatric patients with Crohn's disease. J Clin Microbiol 2012; 50:3258-3266. [PMID: 22837318 PMCID: PMC3457451 DOI: 10.1128/jcm.01396-12] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2012] [Accepted: 07/17/2012] [Indexed: 12/13/2022] Open
Abstract
Microbial dysbiosis has been suggested to be involved in the pathogenesis of Crohn's disease (CD); however, many studies of gut microbial communities have been confounded by environmental and patient-related factors. In this study, the microbial flora of fecal samples from 19 children newly diagnosed with CD and 21 age-matched controls were analyzed using high-throughput sequencing to determine differences in the microbial composition between CD patients and controls. Analysis of the microbial composition of specific bacterial groups revealed that Firmicutes percentages were significantly lower in CD patients than in controls and that this was due largely to changes in the class Clostridia. Bacteroidetes and Proteobacteria percentages were higher and significantly higher in CD patients than in controls, respectively. Both the detection frequencies of Bacteroidetes and Firmicutes correlated (positively and negatively, respectively) with the calculated pediatric Crohn's disease activity index scores of patients. Upon further analysis, differences in the microbial compositions of patients with mild disease and moderate to severe disease were identified. Our findings indicate that a combination of different bacterial species or a dynamic interplay between individual species is important for disease and is consistent with the dysbiosis hypothesis of CD.
Collapse
Affiliation(s)
- Nadeem O. Kaakoush
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, Australia
| | - Andrew S. Day
- School of Women's and Children's Health, The University of New South Wales, Sydney, Australia
- Department of Gastroenterology, Sydney Children's Hospital, Sydney, Australia
- Department of Paediatrics, University of Otago, Christchurch, New Zealand
| | - Karina D. Huinao
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, Australia
| | - Steven T. Leach
- School of Women's and Children's Health, The University of New South Wales, Sydney, Australia
| | - Daniel A. Lemberg
- Department of Gastroenterology, Sydney Children's Hospital, Sydney, Australia
| | - Scot E. Dowd
- Department of Biology, Texas Tech University, Lubbock, Texas, USA
| | - Hazel M. Mitchell
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, Australia
| |
Collapse
|
35
|
Sandler NG, Douek DC. Microbial translocation in HIV infection: causes, consequences and treatment opportunities. Nat Rev Microbiol 2012; 10:655-66. [PMID: 22886237 DOI: 10.1038/nrmicro2848] [Citation(s) in RCA: 343] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Systemic immune activation is increased in HIV-infected individuals, even in the setting of virus suppression with antiretroviral therapy. Although numerous factors may contribute, microbial products have recently emerged as potential drivers of this immune activation. In this Review, we describe the intestinal damage that occurs in HIV infection, the evidence for translocation of microbial products into the systemic circulation and the pathways by which these products activate the immune system. We also discuss novel therapies that disrupt the translocation of microbial products and the downstream effects of microbial translocation.
Collapse
Affiliation(s)
- Netanya G Sandler
- Human Immunology Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | |
Collapse
|
36
|
Abstract
Mucosal surfaces are a major portal of entry for many human pathogens that are the cause of infectious diseases worldwide. Vaccines capable of eliciting mucosal immune responses can fortify defenses at mucosal front lines and protect against infection. However, most licensed vaccines are administered parenterally and fail to elicit protective mucosal immunity. Immunization by mucosal routes may be more effective at inducing protective immunity against mucosal pathogens at their sites of entry. Recent advances in our understanding of mucosal immunity and identification of correlates of protective immunity against specific mucosal pathogens have renewed interest in the development of mucosal vaccines. Efforts have focused on efficient delivery of vaccine antigens to mucosal sites that facilitate uptake by local antigen-presenting cells to generate protective mucosal immune responses. Discovery of safe and effective mucosal adjuvants are also being sought to enhance the magnitude and quality of the protective immune response.
Collapse
Affiliation(s)
- Kim A Woodrow
- Department of Bioengineering, University of Washington, Seattle, Washington 98195, USA.
| | | | | |
Collapse
|
37
|
Ruby T, McLaughlin L, Gopinath S, Monack D. Salmonella's long-term relationship with its host. FEMS Microbiol Rev 2012; 36:600-15. [PMID: 22335190 DOI: 10.1111/j.1574-6976.2012.00332.x] [Citation(s) in RCA: 103] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2011] [Revised: 01/30/2012] [Accepted: 02/07/2012] [Indexed: 12/23/2022] Open
Abstract
Host-adapted strains of Salmonella enterica cause systemic infections and have the ability to persist systemically for long periods of time and pose significant public-health problems. Multidrug-resistant S. enterica serovar Typhi (S. Typhi) and nontyphoidal Salmonella (NTS) are on the increase and are often associated with HIV infection. Chronically infected hosts are often asymptomatic and transmit disease to naïve hosts via fecal shedding of bacteria, thereby serving as a critical reservoir for disease. Salmonella utilizes multiple ways to evade and modulate host innate and adaptive immune responses in order to persist in the presence of a robust immune response. Survival in macrophages and modulation of immune cells migration allow Salmonella to evade various immune responses. The ability of Salmonella to persist depends on a balance between immune responses that lead to the clearance of the pathogen and avoidance of damage to host tissues.
Collapse
Affiliation(s)
- Thomas Ruby
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, USA
| | | | | | | |
Collapse
|
38
|
Fujimoto Y, Pradipta AR, Inohara N, Fukase K. Peptidoglycan as Nod1 ligand; fragment structures in the environment, chemical synthesis, and their innate immunostimulation. Nat Prod Rep 2012; 29:568-79. [PMID: 22370813 DOI: 10.1039/c2np00091a] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Covering: up to 2011. This review focuses on the recent revealing of the immunostimulatory bacterial cell wall peptidoglycan (PGN) fragments as Nod1 ligands, especially a newly developed chemical synthesis of the partial structures, fragment structures in the environment and bacterial supernatant, and the immunostimulatory activities of the Nod1 ligands.
Collapse
Affiliation(s)
- Yukari Fujimoto
- Department of Chemistry, Graduate School of Science, Osaka University, Machikaneyama 1-1, Toyonaka, Osaka 560-0043, Japan.
| | | | | | | |
Collapse
|
39
|
Bharathan M, Mullarky IK. Targeting mucosal immunity in the battle to develop a mastitis vaccine. J Mammary Gland Biol Neoplasia 2011; 16:409-19. [PMID: 21968537 DOI: 10.1007/s10911-011-9233-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2011] [Accepted: 09/14/2011] [Indexed: 01/31/2023] Open
Abstract
The mucosal immune system encounters antigens that enhance and suppress immune function, and serves as a selective barrier against invading pathogens. The mammary gland not only encounters antigens but also produces a nutrient evolved to protect and enhance mucosal development in the neonate. Efforts to manipulate antibody concentrations in milk to prevent mastitis, an infection of the mammary gland, have been hampered both by complexity and variation in target pathogens and limited knowledge of cellular immunity in the gland. Successful vaccination strategies must overcome the natural processes that regulate types and concentrations of milk antibodies for neonatal development, and enhance cellular immunity. Furthermore, the need to overcome dampening of immunity caused by non-pathogenic encounters to successfully prevent establishment of infection is an additional obstacle in vaccine development at mucosal sites. A significant mastitis pathogen, Staphylococcus aureus, not only resides as a normal flora on a multitude of species, but also causes clinical disease with limited treatment options. Using the bovine model of S. aureus mastitis, researchers can decipher the role of antigen selection and presentation by mammary dendritic cells, enhance development of central and effector memory function, and subsequently target specific memory cells to the mammary gland for successful vaccine development. This brief review provides an overview of adaptive immunity, previous vaccine efforts, current immunological findings relevant to enhancing immune memory, and research technologies that show promise in directing future vaccine efforts to enhance mammary gland immunity and prevent mastitis.
Collapse
Affiliation(s)
- Mini Bharathan
- Immunology, Human Therapeutic Division, Intrexon Corporation, Germantown, MD, USA
| | | |
Collapse
|
40
|
Bansal AS, Henriquez F, Sumar N, Patel S. T helper cell subsets in arthritis and the benefits of immunomodulation by 1,25(OH)2 vitamin D. Rheumatol Int 2011; 32:845-52. [DOI: 10.1007/s00296-011-2077-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2011] [Accepted: 08/21/2011] [Indexed: 12/16/2022]
|
41
|
Abstract
Salmonella enterica is a genetically broad species harboring isolates that display considerable antigenic heterogeneity and significant differences in virulence potential. Salmonella generally exhibit an invasive potential and they can survive for extended periods within cells of the immune system. They cause acute or chronic infections that can be local (e.g. gastroenteritis) or systemic (e.g. typhoid). In vivo Salmonella infections are complex with multiple arms of the immune system being engaged. Both humoral and cellular responses can be detected and characterized, but full protective immunity is not always induced, even following natural infection. The murine model has proven to be a fertile ground for exploring immune mechanisms and observations in the mouse have often, although not always, correlated with those in other infectable species, including humans. Host genetic studies have identified a number of mammalian genes that are central to controlling infection, operating both in innate and acquired immune pathways. Vaccines, both oral and parenteral, are available or under development, and these have been used with some success to explore immunity in both model systems and clinically in humans.
Collapse
Affiliation(s)
- Gordon Dougan
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Cambridge, UK.
| | | | | | | |
Collapse
|
42
|
Early MyD88-dependent induction of interleukin-17A expression during Salmonella colitis. Infect Immun 2011; 79:3131-40. [PMID: 21576324 DOI: 10.1128/iai.00018-11] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The development of T helper 17 (T(H)17) cells is a well-established adaptive mechanism for the production of interleukin-17A (IL-17A), a cytokine involved in neutrophil recruitment. However, pathways contributing to mucosal expression of IL-17A during the initial phase of a bacterial infection have received less attention. Here we used the mouse colitis model of Salmonella enterica serotype Typhimurium infection to investigate the contribution of myeloid differentiation primary response protein 88 (MyD88) to inflammation and mucosal IL-17A expression. Expression of IL-23 in the cecal mucosa during S. Typhimurium colitis was dependent on the presence of MyD88. Furthermore, initial expression of IL-17A at 24 h after S. Typhimurium infection was dependent on MyD88 and the receptor for IL-1β. IL-23 and IL-1β synergized in inducing expression of IL-17A in splenic T cells in vitro. In the intestinal mucosa, IL-17A was produced by three distinct T cell populations, including δγ T cells, T(H)17 cells, and CD4(-)CD8(-) T cells. The absence of IL-1β signaling or IL-17 signaling reduced CXC chemokine expression but did not alter the overall severity of pathological lesions in the cecal mucosa. In contrast, cecal pathology and neutrophil recruitment were markedly reduced in Myd88-deficient mice during the initial phases of S. Typhimurium infection. Collectively, these data demonstrate that MyD88-dependent mechanisms, including an initial expression of IL-17A, are important for orchestrating early inflammatory responses during S. Typhimurium colitis.
Collapse
|
43
|
Perry S, Hussain R, Parsonnet J. The impact of mucosal infections on acquisition and progression of tuberculosis. Mucosal Immunol 2011; 4:246-51. [PMID: 21412228 PMCID: PMC5480373 DOI: 10.1038/mi.2011.11] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
More than one-third of the world's population, or over 2 billion people, are infected with Mycobacterium tuberculosis, the causative pathogen of tuberculosis in humans. Why only 10% of those infected develop active disease while the remainder harbor latent infection remains one of the greatest scientific and public health mysteries. Bacterial persistence is characterized by a dynamic state of immunological tolerance between pathogen and host. The critical role of CD4(+) T cells in defense against intracellular pathogens became evident during epidemiological studies of HIV-1 infection, which showed a clear inverse relationship between CD4(+) T-cell count in peripheral blood and increased risk of infection with M. tuberculosis, pneumocystis and Toxoplasma gondii. There is also growing evidence of a common mucosal immune system, whereby immune cells activated at one mucosal site may disseminate to remote effector sites. In this commentary, we review emerging evidence from human studies that the outcome of M. tuberculosis infection is influenced by concurrent mucosal infections, using Helicobacter pylori and geohelminths as examples. Understanding how the complexity of microbial exposures influences host immunity may have important implications for vaccine development and therapeutic interventions.
Collapse
Affiliation(s)
- S Perry
- Division of Infectious Diseases and Geographic Medicine, Stanford University, Stanford, CA, USA
| | - R Hussain
- Department of Molecular Biology, Aga Khan University, Karachi, Pakistan
| | - J Parsonnet
- Division of Infectious Diseases and Geographic Medicine, Stanford University, Stanford, CA, USA
| |
Collapse
|
44
|
Dlubek D, Turlej E, Sedzimirska M, Lange J, Lange A. Interleukin-17-producing cells increase among CD4+ lymphocytes before overt manifestation of acute graft-versus-host disease. Transplant Proc 2011; 42:3277-9. [PMID: 20970671 DOI: 10.1016/j.transproceed.2010.07.026] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Interleukin-17A is a hallmark of a subset of CD4+ lymphocytes called T(H)17. Allogeneic hematopoietic stem cell transplantation (HSCT) induces an immune response that facilitates graft acceptance, but if clinically apparent as acute graft-versus-host disease (aGvHD), it may adversely affect transplantation outcomes. TH17 cells are involved in the inflammatory processes associated with several diseases, including inflammatory bowel disease (IBD) as a prototype. In this study we investigated the presence of IL-17-producing cells among peripheral blood mononuclear cells (PBMC) of patients after HSCT. The 48 patients of median age 45 years (range, 1.0-64 years), experienced hematologic malignancies (n=45) or nonmalignant disorders (n=3), treated with matched unrelated (n=24) or sibling (n=24) transplants. We examined IL-17-producing cells in alloreactive reactions after HSCT. PBMC were stimulated with BD Leukocyte Activation Cocktail (Ionomycin, Brefeldin A, and phorbol myristic acetate (PMA)) in the presence of BD GolgiStop. After stimulation the cells were labeled with anti-CD4 and intracellular anti-IL-17A monoclonal antibodies. IL-17+ cell proportions were analyzed in the CD4+ lymphocyte gate. We observed that patients at the time of hematologic reconstitution had higher proportions of IL-17-producing cells than healthy control subjects (0.73±0.13 vs 0.19±0.06%; P=.019). Fourteen patients displayed the first symptoms of aGvHD at the time of hematologic reconstitution, when they showed lower proportions of IL-17+ cells among CD4+ lymphocytes than their counterparts lacking aGvHD at a similar time after transplantation (0.29±0.09 vs 0.73±0.13%; P=.024). Eight patients developed aGvHD after hematologic reconstitution (median, 34 days). All of these patients displayed lower proportions of IL-17-producing CD4+ cells on the day of aGvHD compared with their initial measurements preceding this complication (0.34±0.14 vs 1.07±0.37%; P=.01).
Collapse
Affiliation(s)
- D Dlubek
- Institute of Immunology and Experimental Therapy, Polish Academy of Science, Wroclaw, Poland
| | | | | | | | | |
Collapse
|
45
|
Abstract
While raised cellular immunity mediated by T helper (Th) 1 type cells may be harmful for the developing embryo/foetus, it is likely that Th2 type immunity may be helpful. The role of natural killer (NK) cells is presently underestimated, although they are clearly important in angiogenesis and the coordinated invasion of the decidua by the trophoblast. Deficient T regulatory cell (Treg) function is evident in women with recurrent miscarriage particularly when this occurs in early pregnancy. The role of the pro-inflammatory Th17 cells is presently unclear. However, early evidence suggests that excessive Th17 activity may promote miscarriage and preterm delivery. This may relate to the ability of these cells to produce those cytokines that encourage Th1 and NK cell activity. As such recurrent miscarriage may be caused not only by chromosomal abnormalities, autoimmunity and uterine abnormalities but also by subclinical uterine infection and inflammation which by stimulating interleukin 6 favours Th17 development over Tregs. This review examines the role of these different cells in early pregnancy and suggests a schema that may join the dots of the immunological puzzle called pregnancy. Finally, suggestions are made as to how inappropriate immunity in recurrent miscarriage may be down-regulated using currently available therapies.
Collapse
|
46
|
Jha R, Srivastava P, Salhan S, Finckh A, Gabay C, Mittal A, Bas S. Spontaneous secretion of interleukin-17 and -22 by human cervical cells in Chlamydia trachomatis infection. Microbes Infect 2011; 13:167-78. [PMID: 21034849 DOI: 10.1016/j.micinf.2010.10.012] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2009] [Revised: 10/12/2010] [Accepted: 10/14/2010] [Indexed: 01/10/2023]
Abstract
To investigate whether IL-17A (IL-17) and IL-22 are produced in response to Chlamydia trachomatis infection, the cervical washes of 27 women with C. trachomatis infection and 17 C. trachomatis negative controls were collected. The levels of cytokines were determined in the cervical wash and in the supernatant of cervical and systemic cell cultures upon C. trachomatis antigen stimulation. C. trachomatis infection appeared to activate local IL-17 and IL-22 production more efficiently than IFN-γ production. In the cervical wash of infected women, median concentrations of IL-17 and -22 were 5- and 3-fold higher, respectively, than in negative controls. The spontaneous intracellular expression of these cytokines was analysed by flow cytometry in blood and cervical cells and 26% of cervical mononuclear cells from infected women were shown to produce IL-22 and 12% to coproduce IL-17 and IL-22. In addition, it was demonstrated that 20-25% of IL-22 producing and IL-17-IL-22 coproducing cervical CD4+ T cells expressed the mucosal homing receptor CCR6. These results suggest that CCR6 is involved in the migration of these cells to the cervix and that IL-17 and IL-22 might play a role in the immune response at the site of C. trachomatis infection.
Collapse
Affiliation(s)
- Rajneesh Jha
- Institute of Pathology-ICMR, Safdarjung Hospital Campus, New Delhi, India
| | | | | | | | | | | | | |
Collapse
|
47
|
Georgel P, Radosavljevic M, Macquin C, Bahram S. The non-conventional MHC class I MR1 molecule controls infection by Klebsiella pneumoniae in mice. Mol Immunol 2010; 48:769-75. [PMID: 21190736 DOI: 10.1016/j.molimm.2010.12.002] [Citation(s) in RCA: 169] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2010] [Revised: 12/04/2010] [Accepted: 12/06/2010] [Indexed: 01/10/2023]
Abstract
As opposed to the well established role of MHC-linked, polymorphic, class I (MHC-I) genes in adaptive immunity, a universal role for non-conventional MHC-I is unknown, thus requiring a case-by-case study. The MHC unlinked, monomorphic, but β₂microglobulin (β₂m)-associated "MHC class I related" MR1 molecule interacts with a semi-invariant TCR. The pathophysiology of this interaction or more importantly of this peculiar MHC-I remains mostly unknown. Recently it was shown that β₂m deficient mice were more susceptible to infection by Klebsiella pneumoniae, a widely spread Gram-negative bacteria that causes diverse and often severe ailments in man. Here we demonstrate, using both an in vivo imaging system and survival tests, the increased susceptibility to K. pneumoniae (but not to several other Gram negative bacteria) of MR1 deficient mice. This is accompanied by a consequent change in body temperature and systemic cytokine profile. Hence MR1 controls K. pneumoniae infection in vivo.
Collapse
Affiliation(s)
- Philippe Georgel
- Laboratoire d'Immunogénétique Moléculaire Humaine, Centre de Recherche d'Immunologie et d'Hématologie, Faculté de Médecine, Université de Strasbourg, 4 rue Kirschleger, 67085 Strasbourg Cedex, France
| | | | | | | |
Collapse
|
48
|
A role for syndecan-1 and claudin-2 in microbial translocation during HIV-1 infection. J Acquir Immune Defic Syndr 2010; 55:306-15. [PMID: 20700059 DOI: 10.1097/qai.0b013e3181ecfeca] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Microbial translocation from the gastrointestinal tract has been implicated in chronic activation of the immune system during progressive HIV-1 infection by ill-defined mechanisms. We recently identified a gene encoding syndecan-1 (SYN1) in microarray studies of HIV-1 infection in lymphatic tissues and show here that increased expression of SYN1 in the gut of HIV-1-infected individuals is associated with increased microbial translocation. We further show that: (1) microbial access to SYN1 in the intestinal epithelium could be mediated by compromised barrier function through the upregulation of claudin-2; (2) increases in SYN1 and microbial translocation are associated with systemic immune activation; and (3) SYN1 expression and microbial translocation are inversely correlated with peripheral blood CD4 T-cell counts. We thus propose a new mechanism in which claudin-2 and SYN1 work in concert to enhance microbial translocation across the intestinal epithelial barrier to contribute to chronic immune activation and CD4 T-cell depletion.
Collapse
|
49
|
The role of the IL-12 cytokine family in directing T-cell responses in oral candidosis. Clin Dev Immunol 2010; 2011:697340. [PMID: 20981280 PMCID: PMC2963117 DOI: 10.1155/2011/697340] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2010] [Accepted: 09/03/2010] [Indexed: 12/24/2022]
Abstract
Candida albicans is an opportunistic fungal pathogen that normally exists as a harmless commensal in humans. In instances where host debilitation occurs, Candida can cause a range of clinical infections, and whilst these are primarily superficial, effecting mucosal membranes, systemic infections can develop in severely immunocompromised individuals. The mechanism of host immunity during commensal carriage of C. albicans has been intensively studied. In this paper, we present the most recent information concerning host recognition of C. albicans leading to cytokine production and the subsequent T-cell responses generated in response to C. albicans. Particular focus is given to the role of the IL-12 cytokine family including IL-12, IL-23, IL-27, and IL-35, in host immunity to Candida. CD4+ T-cells are considered crucial in the regulation of immunity and inflammation. In this regard, the role of Th1/2, helper cells, together with the recently identified Th17 and Treg cells in candidosis will be discussed. Understanding the detailed mechanisms that underlie host immunity to Candida not only will be of benefit in terms of the infections caused by this organism but could also be exploited in the development of therapeutic interventions for other diseases.
Collapse
|
50
|
Conti HR, Gaffen SL. Host responses to Candida albicans: Th17 cells and mucosal candidiasis. Microbes Infect 2010; 12:518-27. [PMID: 20381638 PMCID: PMC2892252 DOI: 10.1016/j.micinf.2010.03.013] [Citation(s) in RCA: 114] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2010] [Accepted: 03/24/2010] [Indexed: 12/14/2022]
Abstract
Candida albicans causes mucosal and disseminated candidiasis, which represent serious problems for the rapidly expanding immunocompromised population. Until recently, Th1-mediated immunity was thought to confer the primary protection, particularly for oral candidiasis. However, emerging data indicate that the newly-defined Th17 compartment appears to play the predominant role in mucosal candidiasis.
Collapse
Affiliation(s)
| | - Sarah L. Gaffen
- University at Buffalo, SUNY, Dept. of Oral Biology, Buffalo NY
- University of Pittsburgh, Department of Medicine, Division of Rheumatology and Clinical Immunology, Pittsburgh PA
| |
Collapse
|