1
|
Mouttoulingam N, Taleb S. Exploring tryptophan metabolism in cardiometabolic diseases. Trends Endocrinol Metab 2024:S1043-2760(24)00317-5. [PMID: 39694729 DOI: 10.1016/j.tem.2024.11.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 11/18/2024] [Accepted: 11/25/2024] [Indexed: 12/20/2024]
Abstract
Tryptophan (Trp) metabolism is linked to health and disease, with indoleamine 2,3-dioxygenase 1 (IDO) being a key enzyme in its breakdown outside the liver. This process produces metabolites that influence metabolic and inflammatory responses. A distinctive feature of the gut is its involvement in three major Trp catabolic pathways: the IDO-driven kynurenine pathway, bacteria-produced indoles, and serotonin. Dysregulation of these pathways is associated with gastrointestinal and chronic inflammatory diseases. Understanding these mechanisms could reveal how gut function affects overall systemic health and disease susceptibility. Here, we review current insights into Trp metabolism, its impact on host physiology and cardiometabolic diseases, and its role in the gut-periphery connection, highlighting its relevance for therapeutic innovation.
Collapse
Affiliation(s)
| | - Soraya Taleb
- Université Paris Cité, Inserm, PARCC, F-75015 Paris, France.
| |
Collapse
|
2
|
Hu Y, Zhang R, Li J, Wang H, Wang M, Ren Q, Fang Y, Tian L. Association Between Gut and Nasal Microbiota and Allergic Rhinitis: A Systematic Review. J Asthma Allergy 2024; 17:633-651. [PMID: 39006241 PMCID: PMC11246088 DOI: 10.2147/jaa.s472632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 07/01/2024] [Indexed: 07/16/2024] Open
Abstract
Allergic rhinitis is a chronic non-infectious inflammation of the nasal mucosa mediated by specific IgE. Recently, the human microbiome has drawn broad interest as a potential new target for treating this condition. This paper succinctly summarizes the main findings of 17 eligible studies published by February 2024, involving 1044 allergic rhinitis patients and 954 healthy controls from 5 countries. These studies examine differences in the human microbiome across important mucosal interfaces, including the nasal and intestinal areas, between patients and controls. Overall, findings suggest variations in the gut microbiota between allergic rhinitis patients and healthy individuals, although the specific bacterial taxa that significantly changed were not always consistent across studies. Due to the limited scope of existing research and patient coverage, the relationship between the nasal microbiome and allergic rhinitis remains inconclusive. The article discusses the potential immune-regulating role of the gut microbiome in allergic rhinitis. Further well-designed clinical trials with large-scale recruitment of allergic rhinitis patients are encouraged.
Collapse
Affiliation(s)
- Yucheng Hu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, People’s Republic of China
| | - Rong Zhang
- Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, People’s Republic of China
| | - Junjie Li
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, People’s Republic of China
| | - Huan Wang
- Chengdu university of Traditional Chinese Medicine, Chengdu, Sichuan, People’s Republic of China
| | - Meiya Wang
- Chengdu university of Traditional Chinese Medicine, Chengdu, Sichuan, People’s Republic of China
| | - Qiuyi Ren
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, People’s Republic of China
| | - Yueqi Fang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, People’s Republic of China
| | - Li Tian
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, People’s Republic of China
| |
Collapse
|
3
|
Nasb M, Li F, Dayoub L, Wu T, Wei M, Chen N. Bridging the gap: Integrating exercise mimicry into chronic disease management through suppressing chronic inflammation. J Adv Res 2024:S2090-1232(24)00176-0. [PMID: 38704088 DOI: 10.1016/j.jare.2024.04.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 03/25/2024] [Accepted: 04/30/2024] [Indexed: 05/06/2024] Open
Abstract
BACKGROUND Chronic inflammation is a common hallmark of many chronic diseases. Although exercise holds paramount importance in preventing and managing chronic diseases, adherence to exercise programs can be challenging for some patients. Consequently, there is a pressing need to explore alternative strategies to emulate the anti-inflammatory effects of exercise for chronic diseases. AIM OF REVIEW This review explores the emerging role of green tea bioactive components as potential mitigators of chronic inflammation, offering insights into their capacity to mimic the beneficial effects of exercise. We propose that bioactive components in green tea are promising agents for suppressing chronic inflammation, suggesting their unique capability to replicate the health benefits of exercise. KEY SCIENTIFIC CONCEPTS OF REVIEW This review focuses on several key concepts, including chronic inflammation and its role in chronic diseases, the anti-inflammatory effects of regular exercise, and bioactive components in green tea responsible for its health benefits. It elaborates on scientific evidence supporting the anti-inflammatory properties of green tea bioactive components, such as epigallocatechin gallate (EGCG), and theorizes how these bioactive components might replicate the effects of exercise at a molecular level. Through a comprehensive analysis of current research, this review proposes a novel perspective on the application of green tea as a potential intervention strategy to suppress chronic inflammation, thereby extending the benefits akin to those achieved through exercise.
Collapse
Affiliation(s)
- Mohammad Nasb
- Tianjiu Research and Development Center for Exercise Nutrition and Foods, Hubei Key Laboratory of Exercise Training and Monitoring, College of Sports Medicine, Wuhan Sports University, Wuhan 430079, China
| | - Fengxing Li
- Tianjiu Research and Development Center for Exercise Nutrition and Foods, Hubei Key Laboratory of Exercise Training and Monitoring, College of Sports Medicine, Wuhan Sports University, Wuhan 430079, China
| | - Lamis Dayoub
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Tong Wu
- Tianjiu Research and Development Center for Exercise Nutrition and Foods, Hubei Key Laboratory of Exercise Training and Monitoring, College of Sports Medicine, Wuhan Sports University, Wuhan 430079, China
| | - Minhui Wei
- Tianjiu Research and Development Center for Exercise Nutrition and Foods, Hubei Key Laboratory of Exercise Training and Monitoring, College of Sports Medicine, Wuhan Sports University, Wuhan 430079, China
| | - Ning Chen
- Tianjiu Research and Development Center for Exercise Nutrition and Foods, Hubei Key Laboratory of Exercise Training and Monitoring, College of Sports Medicine, Wuhan Sports University, Wuhan 430079, China.
| |
Collapse
|
4
|
Huang Y, Chen L, Liu F, Xiong X, Ouyang Y, Deng Y. Tryptophan, an important link in regulating the complex network of skin immunology response in atopic dermatitis. Front Immunol 2024; 14:1300378. [PMID: 38318507 PMCID: PMC10839033 DOI: 10.3389/fimmu.2023.1300378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 12/31/2023] [Indexed: 02/07/2024] Open
Abstract
Atopic dermatitis (AD) is a common chronic relapsing inflammatory skin disease, of which the pathogenesis is a complex interplay between genetics and environment. Although the exact mechanisms of the disease pathogenesis remain unclear, the immune dysregulation primarily involving the Th2 inflammatory pathway and accompanied with an imbalance of multiple immune cells is considered as one of the critical etiologies of AD. Tryptophan metabolism has long been firmly established as a key regulator of immune cells and then affect the occurrence and development of many immune and inflammatory diseases. But the relationship between tryptophan metabolism and the pathogenesis of AD has not been profoundly discussed throughout the literatures. Therefore, this review is conducted to discuss the relationship between tryptophan metabolism and the complex network of skin inflammatory response in AD, which is important to elucidate its complex pathophysiological mechanisms, and then lead to the development of new therapeutic strategies and drugs for the treatment of this frequently relapsing disease.
Collapse
Affiliation(s)
- Yaxin Huang
- Department of Dermatology & Sexually Transmitted Disease (STD), the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Lingna Chen
- Department of Dermatology & Sexually Transmitted Disease (STD), the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Fuming Liu
- Department of Dermatology & Sexually Transmitted Disease (STD), the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Xia Xiong
- Department of Dermatology & Sexually Transmitted Disease (STD), the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Yongliang Ouyang
- Department of Dermatology & Sexually Transmitted Disease (STD), Chengdu First People’s Hospital, Chengdu, Sichuan, China
- Health Management Center, Luzhou People’s Hospital, Luzhou, China
| | - Yongqiong Deng
- Department of Dermatology & Sexually Transmitted Disease (STD), the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Department of Dermatology & Sexually Transmitted Disease (STD), Chengdu First People’s Hospital, Chengdu, Sichuan, China
| |
Collapse
|
5
|
Murakami Y, Imamura Y, Kasahara Y, Yoshida C, Momono Y, Fang K, Sakai D, Konishi Y, Nishiyama T. Maternal Inflammation with Elevated Kynurenine Metabolites Is Related to the Risk of Abnormal Brain Development and Behavioral Changes in Autism Spectrum Disorder. Cells 2023; 12:1087. [PMID: 37048160 PMCID: PMC10093447 DOI: 10.3390/cells12071087] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/27/2023] [Accepted: 03/31/2023] [Indexed: 04/14/2023] Open
Abstract
Several studies show that genetic and environmental factors contribute to the onset and progression of neurodevelopmental disorders. Maternal immune activation (MIA) during gestation is considered one of the major environmental factors driving this process. The kynurenine pathway (KP) is a major route of the essential amino acid L-tryptophan (Trp) catabolism in mammalian cells. Activation of the KP following neuro-inflammation can generate various endogenous neuroactive metabolites that may impact brain functions and behaviors. Additionally, neurotoxic metabolites and excitotoxicity cause long-term changes in the trophic support, glutamatergic system, and synaptic function following KP activation. Therefore, investigating the role of KP metabolites during neurodevelopment will likely promote further understanding of additional pathophysiology of neurodevelopmental disorders, including autism spectrum disorder (ASD). In this review, we describe the changes in KP metabolism in the brain during pregnancy and represent how maternal inflammation and genetic factors influence the KP during development. We overview the patients with ASD clinical data and animal models designed to verify the role of perinatal KP elevation in long-lasting biochemical, neuropathological, and behavioral deficits later in life. Our review will help shed light on new therapeutic strategies and interventions targeting the KP for neurodevelopmental disorders.
Collapse
Affiliation(s)
- Yuki Murakami
- Department of Hygiene and Public Health, Kansai Medical University, Hirakata 573-1010, Japan
| | - Yukio Imamura
- Department of Architecture and Architectual Systems Engineering, Graduate School of Engineering, Kyoto University, Kyoto 615-8530, Japan
- Department of Traumatology and Acute Critical Medicine, Graduate School of Medicine/Faculty of Medicine, Osaka University, Suita 565-0871, Japan
| | - Yoshiyuki Kasahara
- Department of Maternal and Fetal Therapeutics, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Chihiro Yoshida
- Department of Maternal and Fetal Therapeutics, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Yuta Momono
- Department of Maternal and Fetal Therapeutics, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Ke Fang
- Department of Hygiene and Public Health, Kansai Medical University, Hirakata 573-1010, Japan
| | - Daisuke Sakai
- Department of Biology, Kanazawa Medical University, Kanazawa 920-0293, Japan
| | - Yukuo Konishi
- Center for Baby Science, Doshisha University, Kyotanabe 619-0225, Japan
- Healthcare and Medical Data Multi-Level Integration Platform Group, RIKEN Medical Sciences Innovation Hub Program, Yokohama 230-0045, Japan
| | - Toshimasa Nishiyama
- Department of Hygiene and Public Health, Kansai Medical University, Hirakata 573-1010, Japan
| |
Collapse
|
6
|
Iraji D, Oftedal BE, Wolff ASB. Th17 Cells: Orchestrators of Mucosal Inflammation and Potential Therapeutic Targets. Crit Rev Immunol 2023; 43:25-52. [PMID: 37831521 DOI: 10.1615/critrevimmunol.2023050360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2023]
Abstract
T helper 17 (Th17) cells represent a specialized subgroup of effector CD4+ T cells known for their role in provoking neutrophil-driven tissue inflammation, particularly within mucosal tissues. Although they are pivotal for defending the host against extracellular bacteria and fungi, they have also been associated with development of various T cell-mediated inflammatory conditions, autoimmune diseases, and even cancer. Notably, Th17 cells exhibit a dual nature, with different Th17 cell subtypes showcasing distinct effector functions and varying capacities to incite autoimmune tissue inflammation. Furthermore, Th17 cells exhibit significant plasticity, which carries important functional implications, both in terms of their expression of cytokines typically associated with other effector T cell subsets and in their interactions with regulatory CD4+ T cells. The intricate balance of Th17 cytokines can also be a double-edged sword in inflammation, autoimmunity, and cancer. Within this article, we delve into the mechanisms that govern the differentiation, function, and adaptability of Th17 cells. We culminate with an exploration of therapeutic potentials in harnessing the power of Th17 cells and their cytokines. Targeted interventions to modulate Th17 responses are emerging as promising strategies for autoimmunity, inflammation, and cancer treatment. By precisely fine-tuning Th17-related pathways, we may unlock new avenues for personalized therapeutic approaches, aiming to restore immune balance, alleviate the challenges of these disorders, and ultimately enhance the quality of life for individuals affected by them.
Collapse
Affiliation(s)
- Dorsa Iraji
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Bergithe E Oftedal
- Department of Clinical Science, University of Bergen, Bergen, Norway; Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Anette S B Wolff
- Department of Clinical Science, University of Bergen, Bergen, Norway; Department of Medicine, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
7
|
Hu W, Huang L, Zhou Z, Yin L, Tang J. Diallyl Disulfide (DADS) Ameliorates Intestinal Candida albicans Infection by Modulating the Gut microbiota and Metabolites and Providing Intestinal Protection in Mice. Front Cell Infect Microbiol 2022; 11:743454. [PMID: 35071031 PMCID: PMC8777027 DOI: 10.3389/fcimb.2021.743454] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Accepted: 12/13/2021] [Indexed: 12/12/2022] Open
Abstract
Diallyl disulfide (DADS), a garlic extract also known as allicin, has been reported to have numerous biological activities, including anticancer, antifungal, and inflammation-inhibiting activities, among others. Although many studies have assessed whether DADS can treat Candida albicans infection in vitro, its in vivo function and the underlying mechanism are still not clear. Accumulated evidence has implicated the gut microbiota as an important factor in the colonization and invasion of C. albicans. Thus, this study aimed to identify the mechanism by which DADS ameliorates dextran sulfate (DSS)-induced intestinal C. albicans infection based on the systematic analysis of the gut microbiota and metabolomics in mice. Here, we determined the body weight, survival, colon length, histological score, and inflammatory cytokine levels in the serum and intestines of experimental mice. Fecal samples were collected for gut microbiota and metabolite analysis by 16S rRNA gene sequencing and LC-MS metabolomics, respectively. DADS significantly alleviated DSS-induced intestinal C. albicans infection and altered the gut microbial community structure and metabolic profile in the mice. The abundances of some pathogenic bacteria, such as Proteobacteria, Escherichia-Shigella, and Streptococcus, were notably decreased after treatment with DADS. In contrast, SCFA-producing bacteria, namely, Ruminiclostridium, Oscillibacter, and Ruminococcaceae_UCG-013, greatly increased in number. The perturbance of metabolites in infectious mice was improved by DADS, with increases in secondary bile acids, arachidonic acid, indoles and their derivatives, which were highly related to the multiple differentially altered metabolic pathways, namely, bile secretion, arachidonic acid metabolism, and tryptophan metabolism. This study indicated that DADS could modulate gut microbiota and metabolites and protect the gut barrier to alleviate DSS-induced intestinal C. albicans infection in mice. Moreover, this work might also provide novel insight into the treatment of C. albicans infection using DADS.
Collapse
Affiliation(s)
| | | | | | | | - Jianguo Tang
- Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People’s Hospital, Fudan University, Shanghai, China
| |
Collapse
|
8
|
Dos Santos AR, Fraga-Silva TF, de Fátima Almeida-Donanzam D, Dos Santos RF, Finato AC, Soares CT, Lara VS, Almeida NLM, Andrade MI, de Arruda OS, de Arruda MSP, Venturini J. IFN-γ Mediated Signaling Improves Fungal Clearance in Experimental Pulmonary Mucormycosis. Mycopathologia 2021; 187:15-30. [PMID: 34716549 PMCID: PMC8555725 DOI: 10.1007/s11046-021-00598-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 10/13/2021] [Indexed: 12/28/2022]
Abstract
We established three immunocompetent murine models of pulmonary mucormycosis to determine the involvement of the adaptive immune response in host resistance in pulmonary mucormycosis, a rapidly fatal disease caused mainly by Rhizopus spp. Immunocompetent inbred (C57BL/6, BALB/c) and outbred (Swiss) strains of mice were inoculated with R. oryzae via the intratracheal route. The inoculation resulted in a disseminated infection that spread to the brain, spleen, kidney, and liver. After 7 and 30 days of R. oryzae infection, BALB/c mice showed the lowest fungal load and highest production of IFN-γ and IL-2 by splenocytes. Swiss mice showed a higher fungal load 30 days p.i. and was associated with a weak development of the Th-1 profile. To confirm our findings, R. oryzae-infected IFN-γ−/− mice were evaluated after 60 days, where the mice still showed viable fungi in the lungs. This study showed, for the first time, that pulmonary mucormycosis in three widely used mouse strains resulted in an acute fungal dissemination without immunosuppression whose outcome varies according to the genetic background of the mice. We also identified the partial role of IFN-γ in the efficient elimination of R. oryzae during pulmonary infection.
Collapse
Affiliation(s)
- Amanda Ribeiro Dos Santos
- Faculdade de Ciências, Universidade Estadual Paulista (Unesp), Bauru, SP, 17033-360, Brazil.,Faculdade de Medicina, Universidade Federal de Mato Grosso do Sul (UFMS), Cidade Universitária, Unit 9, Campo Grande, MS, 79070-900, Brazil
| | - Thais Fernanda Fraga-Silva
- Departamento de Bioquimica e Imunologia, Universidade de São Paulo, Escola de Medicina de Ribeirão Preto, São Paulo, SP, 14049-900, Brazil
| | - Débora de Fátima Almeida-Donanzam
- Faculdade de Ciências, Universidade Estadual Paulista (Unesp), Bauru, SP, 17033-360, Brazil.,Faculdade de Medicina, Universidade Federal de Mato Grosso do Sul (UFMS), Cidade Universitária, Unit 9, Campo Grande, MS, 79070-900, Brazil
| | | | - Angela Carolina Finato
- Faculdade de Ciências, Universidade Estadual Paulista (Unesp), Bauru, SP, 17033-360, Brazil
| | | | - Vanessa Soares Lara
- Faculdade de Odontologia de Bauru (FOB), Universidade de São Paulo (USP), Bauru, SP, 17012-901, Brazil
| | | | | | | | | | - James Venturini
- Faculdade de Ciências, Universidade Estadual Paulista (Unesp), Bauru, SP, 17033-360, Brazil. .,Faculdade de Medicina, Universidade Federal de Mato Grosso do Sul (UFMS), Cidade Universitária, Unit 9, Campo Grande, MS, 79070-900, Brazil.
| |
Collapse
|
9
|
Kanova M, Kohout P. Tryptophan: A Unique Role in the Critically Ill. Int J Mol Sci 2021; 22:ijms222111714. [PMID: 34769144 PMCID: PMC8583765 DOI: 10.3390/ijms222111714] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 10/21/2021] [Accepted: 10/26/2021] [Indexed: 02/06/2023] Open
Abstract
Tryptophan is an essential amino acid whose metabolites play key roles in diverse physiological processes. Due to low reserves in the body, especially under various catabolic conditions, tryptophan deficiency manifests itself rapidly, and both the serotonin and kynurenine pathways of metabolism are clinically significant in critically ill patients. In this review, we highlight these pathways as sources of serotonin and melatonin, which then regulate neurotransmission, influence circadian rhythm, cognitive functions, and the development of delirium. Kynurenines serve important signaling functions in inter-organ communication and modulate endogenous inflammation. Increased plasma kynurenine levels and kynurenine-tryptophan ratios are early indicators for the development of sepsis. They also influence the regulation of skeletal muscle mass and thereby the development of polyneuromyopathy in critically ill patients. The modulation of tryptophan metabolism could help prevent and treat age-related disease with low grade chronic inflammation as well as post intensive care syndrome in all its varied manifestations: cognitive decline (including delirium or dementia), physical impairment (catabolism, protein breakdown, loss of muscle mass and tone), and mental impairment (depression, anxiety or post-traumatic stress disorder).
Collapse
Affiliation(s)
- Marcela Kanova
- Department of Anaesthesiology and Intensive Care Medicine, University Hospital Ostrava, 708 52 Ostrava, Czech Republic
- Institute of Physiology and Pathophysiology, Faculty of Medicine, University of Ostrava, 703 00 Ostrava, Czech Republic
- Correspondence: or (M.K.); (P.K.); Tel.: +420-597-372-702 (M.K.); +420-261-083-802 (P.K.)
| | - Pavel Kohout
- Department of Internal Medicine, 3rd Faculty of Medicine, Charles University Prague and Teaching Thomayer Hospital, 140 59 Prague, Czech Republic
- Correspondence: or (M.K.); (P.K.); Tel.: +420-597-372-702 (M.K.); +420-261-083-802 (P.K.)
| |
Collapse
|
10
|
The network interplay of interferon and Toll-like receptor signaling pathways in the anti-Candida immune response. Sci Rep 2021; 11:20281. [PMID: 34645905 PMCID: PMC8514550 DOI: 10.1038/s41598-021-99838-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 09/30/2021] [Indexed: 01/22/2023] Open
Abstract
Fungal infections represent a major global health problem affecting over a billion people that kills more than 1.5 million annually. In this study, we employed an integrative approach to reveal the landscape of the human immune responses to Candida spp. through meta-analysis of microarray, bulk, and single-cell RNA sequencing (scRNA-seq) data for the blood transcriptome. We identified across these different studies a consistent interconnected network interplay of signaling molecules involved in both Toll-like receptor (TLR) and interferon (IFN) signaling cascades that is activated in response to different Candida species (C. albicans, C. auris, C. glabrata, C. parapsilosis, and C. tropicalis). Among these molecules are several types I IFN, indicating an overlap with antiviral immune responses. scRNA-seq data confirmed that genes commonly identified by the three transcriptomic methods show cell type-specific expression patterns in various innate and adaptive immune cells. These findings shed new light on the anti-Candida immune response, providing putative molecular pathways for therapeutic intervention.
Collapse
|
11
|
Murakami Y, Imamura Y, Kasahara Y, Yoshida C, Momono Y, Fang K, Nishiyama T, Sakai D, Konishi Y. The Effects of Maternal Interleukin-17A on Social Behavior, Cognitive Function, and Depression-Like Behavior in Mice with Altered Kynurenine Metabolites. Int J Tryptophan Res 2021; 14:11786469211026639. [PMID: 34262289 PMCID: PMC8243115 DOI: 10.1177/11786469211026639] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 05/31/2021] [Indexed: 01/03/2023] Open
Abstract
Viral infection and chronic maternal inflammation during pregnancy are correlated
with a higher prevalence of autism spectrum disorder (ASD). However, the
pathoetiology of ASD is not fully understood; moreover, the key molecules that
can cross the placenta following maternal inflammation and contribute to the
development of ASD have not been identified. Recently, the pro-inflammatory
cytokine, interleukin-17A (IL-17A) was identified as a potential mediator of
these effects. To investigate the impact of maternal IL-17A on offspring,
C57BL/6J dams were injected with IL-17A-expressing plasmids via
the tail vein on embryonic day 12.5 (E12.5), and maternal IL-17A was expressed
continuously throughout pregnancy. By adulthood, IL-17A-injected offspring
exhibited behavioral abnormalities, including social and cognitive defects.
Additionally, maternal IL-17A promoted metabolism of the essential amino acid
tryptophan, which produces several neuroactive compounds and may affect fetal
neurodevelopment. We observed significantly increased levels of kynurenine in
maternal serum and fetal plasma. Thus, we investigated the effects of high
maternal concentration of kynurenine on offspring by continuously administering
mouse dams with kynurenine from E12.5 during gestation. Obviously, maternal
kynurenine administration rapidly increased kynurenine levels in the fetal
plasma and brain, pointing to the ability of kynurenine to cross the placenta
and change the KP metabolites which are affected as neuroactive compounds in the
fetal brain. Notably, the offspring of kynurenine-injected mice exhibited
behavioral abnormalities similar to those observed in offspring of
IL-17A-conditioned mice. Several tryptophan metabolites were significantly
altered in the prefrontal cortex of the IL-17A-conditioned and
kynurenine-injected adult mice, but not in the hippocampus. Even though we
cannot exclude the possibility or other molecules being related to ASD
pathogenesis and the presence of a much lower degree of pathway activation, our
results suggest that increased kynurenine following maternal inflammation may be
a key factor in changing the balance of KP metabolites in fetal brain during
neuronal development and represents a therapeutic target for
inflammation-induced ASD-like phenotypes.
Collapse
Affiliation(s)
- Yuki Murakami
- Department of Hygiene and Public Health, Kansai Medical University, Osaka, Japan
| | - Yukio Imamura
- Organization for Research Initiatives and Development, Doshisha University, Kyoto, Japan.,Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yoshiyuki Kasahara
- Department of Maternal and Fetal Therapeutics, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Chihiro Yoshida
- Department of Maternal and Fetal Therapeutics, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Yuta Momono
- Department of Maternal and Fetal Therapeutics, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Ke Fang
- Department of Hygiene and Public Health, Kansai Medical University, Osaka, Japan
| | - Toshimasa Nishiyama
- Department of Hygiene and Public Health, Kansai Medical University, Osaka, Japan
| | - Daisuke Sakai
- Department of Biology, Kanazawa Medical University, Ishikawa, Japan
| | - Yukuo Konishi
- Center for Baby Science, Doshisha University, Kyoto, Japan.,Healthcare and Medical Data Multi-level Integration Platform Group, RIKEN Medical Sciences Innovation Hub Program, Kanagawa, Japan
| |
Collapse
|
12
|
Whiting-Fawcett F, Field KA, Puechmaille SJ, Blomberg AS, Lilley TM. Heterothermy and antifungal responses in bats. Curr Opin Microbiol 2021; 62:61-67. [PMID: 34098511 DOI: 10.1016/j.mib.2021.05.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/21/2021] [Accepted: 05/10/2021] [Indexed: 11/28/2022]
Abstract
Hibernation, a period where bats have suppressed immunity and low body temperatures, provides the psychrophilic fungus Pseudogymnoascus destructans the opportunity to colonise bat skin, leading to severe disease in susceptible species. Innate immunity, which requires less energy and may remain more active during torpor, can control infections with local inflammation in some bat species that are resistant to infection. If infection is not controlled before emergence from hibernation, ineffective adaptive immune mechanisms are activated, including incomplete Th1, ineffective Th2, and variable Th17 responses. The Th17 and neutrophil responses, normally beneficial antifungal mechanisms, appear to be sources of immunopathology for susceptible bat species, because they are hyperactivated after return to homeothermy. Non-susceptible species show both well-balanced and suppressed immune responses both during and after hibernation.
Collapse
Affiliation(s)
- Flora Whiting-Fawcett
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | | | | | | | - Thomas M Lilley
- Finnish Museum of Natural History, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
13
|
de Araújo EF, Loures FV, Preite NW, Feriotti C, Galdino NA, Costa TA, Calich VLG. AhR Ligands Modulate the Differentiation of Innate Lymphoid Cells and T Helper Cell Subsets That Control the Severity of a Pulmonary Fungal Infection. Front Immunol 2021; 12:630938. [PMID: 33936043 PMCID: PMC8085362 DOI: 10.3389/fimmu.2021.630938] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 03/30/2021] [Indexed: 11/13/2022] Open
Abstract
In agreement with other fungal infections, immunoprotection in pulmonary paracoccidioidomycosis (PCM) is mediated by Th1/Th17 cells whereas disease progression by prevalent Th2/Th9 immunity. Treg cells play a dual role, suppressing immunity but also controlling excessive tissue inflammation. Our recent studies have demonstrated that the enzyme indoleamine 2,3 dioxygenase (IDO) and the transcription factor aryl hydrocarbon receptor (AhR) play an important role in the immunoregulation of PCM. To further evaluate the immunomodulatory activity of AhR in this fungal infection, Paracoccidioides brasiliensis infected mice were treated with two different AhR agonists, L-Kynurenin (L-Kyn) or 6-formylindole [3,2-b] carbazole (FICZ), and one AhR specific antagonist (CH223191). The disease severity and immune response of treated and untreated mice were assessed 96 hours and 2 weeks after infection. Some similar effects on host response were shared by FICZ and L-Kyn, such as the reduced fungal loads, decreased numbers of CD11c+ lung myeloid cells expressing activation markers (IA, CD40, CD80, CD86), and early increased expression of IDO and AhR. In contrast, the AhR antagonist CH223191 induced increased fungal loads, increased number of pulmonary CD11c+ leukocytes expressing activation markers, and a reduction in AhR and IDO production. While FICZ treatment promoted large increases in ILC3, L-Kyn and CH223191 significantly reduced this cell population. Each of these AhR ligands induced a characteristic adaptive immunity. The large expansion of FICZ-induced myeloid, lymphoid, and plasmacytoid dendritic cells (DCs) led to the increased expansion of all CD4+ T cell subpopulations (Th1, Th2, Th17, Th22, and Treg), but with a clear predominance of Th17 and Th22 subsets. On the other hand, L-Kyn, that preferentially activated plasmacytoid DCs, reduced Th1/Th22 development but caused a robust expansion of Treg cells. The AhR antagonist CH223191 induced a preferential expansion of myeloid DCs, reduced the number of Th1, Th22, and Treg cells, but increased Th17 differentiation. In conclusion, the present study showed that the pathogen loads and the immune response in pulmonary PCM can be modulated by AhR ligands. However, further studies are needed to define the possible use of these compounds as adjuvant therapy for this fungal infection.
Collapse
Affiliation(s)
- Eliseu F de Araújo
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Flávio V Loures
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Nycolas W Preite
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Cláudia Feriotti
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Nayane Al Galdino
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Tânia A Costa
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Vera L G Calich
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
14
|
Amerikanou C, Dimitropoulou E, Gioxari A, Papada E, Tanaini A, Fotakis C, Zoumpoulakis P, Kaliora AC. Linking the IL-17A immune response with NMR-based faecal metabolic profile in IBD patients treated with Mastiha. Biomed Pharmacother 2021; 138:111535. [PMID: 34311533 DOI: 10.1016/j.biopha.2021.111535] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 03/14/2021] [Accepted: 03/21/2021] [Indexed: 12/28/2022] Open
Abstract
Dysregulation of intestinal immune response plays a critical role in the pathogenesis of Inflammatory Bowel Disease (IBD). Mastiha's anti-inflammatory properties are well established. Our aim was to investigate Mastiha's regulatory effect on IL-17A serum levels in IBD patients. Alterations of the faecal metabolome as a functional readout of microbial activity were explored. A randomized, double-blind, placebo-controlled, parallel-group design was applied for a total of 3 months in active and 6 months in inactive IBD patients. Serum IL-17A increased significantly in Mastiha group (p = 0.006), and the mean change differed significantly between Mastiha and placebo (p = 0.003) even after adjusting for age, sex and BMI (p = 0.001) in inactive patients. In inactive UC patients IL-17A decreased significantly only in placebo (p = 0.033). No significant differences were detected in active disease. Faecal metabolomics indicated that intervention with Mastiha influenced considerably the metabolic profile of IBD patients in remission exhibiting, in between others, increased levels of glycine and tryptophan. Glycine has been proposed to have a therapeutic effect against IBD, while tryptophan derivatives are involved in immunoregalutory mechanisms, such as the Th17 cells differentiation. Thus, it is quite possible that the immunoregulatory role of Mastiha in quiescent IBD involves the regulation of Th17 cells function and differentiation.
Collapse
Affiliation(s)
- Charalampia Amerikanou
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University, Athens, Greece
| | - Eirini Dimitropoulou
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University, Athens, Greece
| | - Aristea Gioxari
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University, Athens, Greece
| | - Efstathia Papada
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University, Athens, Greece
| | - Anthi Tanaini
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University, Athens, Greece
| | - Charalambos Fotakis
- Institute of Chemical Biology, National Hellenic Research Foundation, Athens, Greece
| | - Panagiotis Zoumpoulakis
- Institute of Chemical Biology, National Hellenic Research Foundation, Athens, Greece; Department of Food Science and Technology, University of West Attica, Athens, Greece.
| | - Andriana C Kaliora
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University, Athens, Greece.
| |
Collapse
|
15
|
Melhem NJ, Chajadine M, Gomez I, Howangyin KY, Bouvet M, Knosp C, Sun Y, Rouanet M, Laurans L, Cazorla O, Lemitre M, Vilar J, Mallat Z, Tedgui A, Ait-Oufella H, Hulot JS, Callebert J, Launay JM, Fauconnier J, Silvestre JS, Taleb S. Endothelial Cell Indoleamine 2, 3-Dioxygenase 1 Alters Cardiac Function After Myocardial Infarction Through Kynurenine. Circulation 2020; 143:566-580. [PMID: 33272024 DOI: 10.1161/circulationaha.120.050301] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
BACKGROUND Ischemic cardiovascular diseases, particularly acute myocardial infarction (MI), is one of the leading causes of mortality worldwide. Indoleamine 2, 3-dioxygenase 1 (IDO) catalyzes 1 rate-limiting step of L-tryptophan metabolism, and emerges as an important regulator of many pathological conditions. We hypothesized that IDO could play a key role to locally regulate cardiac homeostasis after MI. METHODS Cardiac repair was analyzed in mice harboring specific endothelial or smooth muscle cells or cardiomyocyte or myeloid cell deficiency of IDO and challenged with acute myocardial infarction. RESULTS We show that kynurenine generation through IDO is markedly induced after MI in mice. Total genetic deletion or pharmacological inhibition of IDO limits cardiac injury and cardiac dysfunction after MI. Distinct loss of function of IDO in smooth muscle cells, inflammatory cells, or cardiomyocytes does not affect cardiac function and remodeling in infarcted mice. In sharp contrast, mice harboring endothelial cell-specific deletion of IDO show an improvement of cardiac function as well as cardiomyocyte contractility and reduction in adverse ventricular remodeling. In vivo kynurenine supplementation in IDO-deficient mice abrogates the protective effects of IDO deletion. Kynurenine precipitates cardiomyocyte apoptosis through reactive oxygen species production in an aryl hydrocarbon receptor-dependent mechanism. CONCLUSIONS These data suggest that IDO could constitute a new therapeutic target during acute MI.
Collapse
Affiliation(s)
- Nada Joe Melhem
- Université de Paris, Paris-Centre de Recherche Cardiovasculaire (PARCC), Institut National de la Santé et de la Recherche Médicale, France (N.-J.M., M.C., I.G., K.-Y.H., M.B., C.K., Y.S., M.R., L.L., M.L., J.V., Z.M., A.T., H.A.-O., J.-S.H., J.-S.S., S.T.)
| | - Mouna Chajadine
- Université de Paris, Paris-Centre de Recherche Cardiovasculaire (PARCC), Institut National de la Santé et de la Recherche Médicale, France (N.-J.M., M.C., I.G., K.-Y.H., M.B., C.K., Y.S., M.R., L.L., M.L., J.V., Z.M., A.T., H.A.-O., J.-S.H., J.-S.S., S.T.)
| | - Ingrid Gomez
- Université de Paris, Paris-Centre de Recherche Cardiovasculaire (PARCC), Institut National de la Santé et de la Recherche Médicale, France (N.-J.M., M.C., I.G., K.-Y.H., M.B., C.K., Y.S., M.R., L.L., M.L., J.V., Z.M., A.T., H.A.-O., J.-S.H., J.-S.S., S.T.)
| | - Kiave-Yune Howangyin
- Université de Paris, Paris-Centre de Recherche Cardiovasculaire (PARCC), Institut National de la Santé et de la Recherche Médicale, France (N.-J.M., M.C., I.G., K.-Y.H., M.B., C.K., Y.S., M.R., L.L., M.L., J.V., Z.M., A.T., H.A.-O., J.-S.H., J.-S.S., S.T.)
| | - Marion Bouvet
- Université de Paris, Paris-Centre de Recherche Cardiovasculaire (PARCC), Institut National de la Santé et de la Recherche Médicale, France (N.-J.M., M.C., I.G., K.-Y.H., M.B., C.K., Y.S., M.R., L.L., M.L., J.V., Z.M., A.T., H.A.-O., J.-S.H., J.-S.S., S.T.)
| | - Camille Knosp
- Université de Paris, Paris-Centre de Recherche Cardiovasculaire (PARCC), Institut National de la Santé et de la Recherche Médicale, France (N.-J.M., M.C., I.G., K.-Y.H., M.B., C.K., Y.S., M.R., L.L., M.L., J.V., Z.M., A.T., H.A.-O., J.-S.H., J.-S.S., S.T.)
| | - Yanyi Sun
- Université de Paris, Paris-Centre de Recherche Cardiovasculaire (PARCC), Institut National de la Santé et de la Recherche Médicale, France (N.-J.M., M.C., I.G., K.-Y.H., M.B., C.K., Y.S., M.R., L.L., M.L., J.V., Z.M., A.T., H.A.-O., J.-S.H., J.-S.S., S.T.)
| | - Marie Rouanet
- Université de Paris, Paris-Centre de Recherche Cardiovasculaire (PARCC), Institut National de la Santé et de la Recherche Médicale, France (N.-J.M., M.C., I.G., K.-Y.H., M.B., C.K., Y.S., M.R., L.L., M.L., J.V., Z.M., A.T., H.A.-O., J.-S.H., J.-S.S., S.T.)
| | - Ludivine Laurans
- Université de Paris, Paris-Centre de Recherche Cardiovasculaire (PARCC), Institut National de la Santé et de la Recherche Médicale, France (N.-J.M., M.C., I.G., K.-Y.H., M.B., C.K., Y.S., M.R., L.L., M.L., J.V., Z.M., A.T., H.A.-O., J.-S.H., J.-S.S., S.T.)
| | - Olivier Cazorla
- PHYSIOLOGIE ET MÉDECINE EXPÉRIMENTALE DU COEUR ET DES MUSCLES (PHYMEDEXP), Institut national de la santé et de la Recherche Médicale (INSERM), Centre National de la Recherche Scientifique (CNRS), Université de Montpellier, Centre Hospitalier Régional Universitaire (CHRU) Montpellier, France (O.C., J.F.)
| | - Mathilde Lemitre
- Université de Paris, Paris-Centre de Recherche Cardiovasculaire (PARCC), Institut National de la Santé et de la Recherche Médicale, France (N.-J.M., M.C., I.G., K.-Y.H., M.B., C.K., Y.S., M.R., L.L., M.L., J.V., Z.M., A.T., H.A.-O., J.-S.H., J.-S.S., S.T.)
| | - José Vilar
- Université de Paris, Paris-Centre de Recherche Cardiovasculaire (PARCC), Institut National de la Santé et de la Recherche Médicale, France (N.-J.M., M.C., I.G., K.-Y.H., M.B., C.K., Y.S., M.R., L.L., M.L., J.V., Z.M., A.T., H.A.-O., J.-S.H., J.-S.S., S.T.)
| | - Ziad Mallat
- Université de Paris, Paris-Centre de Recherche Cardiovasculaire (PARCC), Institut National de la Santé et de la Recherche Médicale, France (N.-J.M., M.C., I.G., K.-Y.H., M.B., C.K., Y.S., M.R., L.L., M.L., J.V., Z.M., A.T., H.A.-O., J.-S.H., J.-S.S., S.T.).,Division of Cardiovascular Medicine, University of Cambridge, Addenbrooke's Hospital, United Kingdom (Z.M.)
| | - Alain Tedgui
- Université de Paris, Paris-Centre de Recherche Cardiovasculaire (PARCC), Institut National de la Santé et de la Recherche Médicale, France (N.-J.M., M.C., I.G., K.-Y.H., M.B., C.K., Y.S., M.R., L.L., M.L., J.V., Z.M., A.T., H.A.-O., J.-S.H., J.-S.S., S.T.)
| | - Hafid Ait-Oufella
- Université de Paris, Paris-Centre de Recherche Cardiovasculaire (PARCC), Institut National de la Santé et de la Recherche Médicale, France (N.-J.M., M.C., I.G., K.-Y.H., M.B., C.K., Y.S., M.R., L.L., M.L., J.V., Z.M., A.T., H.A.-O., J.-S.H., J.-S.S., S.T.)
| | - Jean-Sébastien Hulot
- Université de Paris, Paris-Centre de Recherche Cardiovasculaire (PARCC), Institut National de la Santé et de la Recherche Médicale, France (N.-J.M., M.C., I.G., K.-Y.H., M.B., C.K., Y.S., M.R., L.L., M.L., J.V., Z.M., A.T., H.A.-O., J.-S.H., J.-S.S., S.T.)
| | - Jacques Callebert
- Service de Biochimie, Assistance Publique Hôpitaux de Paris, and Institut National de la Santé et de la Recherche Médicale UMR942, Hôpital Lariboisière, France (J.C., J.-M.L.)
| | - Jean-Marie Launay
- Service de Biochimie, Assistance Publique Hôpitaux de Paris, and Institut National de la Santé et de la Recherche Médicale UMR942, Hôpital Lariboisière, France (J.C., J.-M.L.)
| | - Jeremy Fauconnier
- PHYSIOLOGIE ET MÉDECINE EXPÉRIMENTALE DU COEUR ET DES MUSCLES (PHYMEDEXP), Institut national de la santé et de la Recherche Médicale (INSERM), Centre National de la Recherche Scientifique (CNRS), Université de Montpellier, Centre Hospitalier Régional Universitaire (CHRU) Montpellier, France (O.C., J.F.)
| | - Jean-Sébastien Silvestre
- Université de Paris, Paris-Centre de Recherche Cardiovasculaire (PARCC), Institut National de la Santé et de la Recherche Médicale, France (N.-J.M., M.C., I.G., K.-Y.H., M.B., C.K., Y.S., M.R., L.L., M.L., J.V., Z.M., A.T., H.A.-O., J.-S.H., J.-S.S., S.T.)
| | - Soraya Taleb
- Université de Paris, Paris-Centre de Recherche Cardiovasculaire (PARCC), Institut National de la Santé et de la Recherche Médicale, France (N.-J.M., M.C., I.G., K.-Y.H., M.B., C.K., Y.S., M.R., L.L., M.L., J.V., Z.M., A.T., H.A.-O., J.-S.H., J.-S.S., S.T.)
| |
Collapse
|
16
|
Aaron L, Torsten M. Candida albicans in celiac disease: A wolf in sheep's clothing. Autoimmun Rev 2020; 19:102621. [PMID: 32693029 DOI: 10.1016/j.autrev.2020.102621] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 03/20/2020] [Indexed: 12/16/2022]
Abstract
Candida albicans is a commensal fungus with a potential pathogenicity and celiac disease is an autoimmune condition. Both share multiple pathophysiological junctions, including serological markers against cell-wall proteins of Candida, anti-gliadin antibodies are positive in both entities, gluten and a candidal virulence factor share sequence similarity and the autoantigen of celiac disease, the tissue transglutaminase, is pivotal in Candida albicans commensalism and hostile behavior and its covalently cross linked products are stable and resistant to breakdown in the two entities. Those autoimmune/infectious cross roads are the basis for the hypothesis that Candida albicans is an additional environmental factor for celiac disease autoimmunogenesis.
Collapse
|
17
|
Pulmonary paracoccidioidomycosis in AhR deficient hosts is severe and associated with defective Treg and Th22 responses. Sci Rep 2020; 10:11312. [PMID: 32647342 PMCID: PMC7347857 DOI: 10.1038/s41598-020-68322-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 06/11/2020] [Indexed: 12/20/2022] Open
Abstract
AhR is a ligand-activated transcription factor that plays an important role in the innate and adaptive immune responses. In infection models, it has been associated with host responses that promote or inhibit disease progression. In pulmonary paracoccidioidomycosis, a primary fungal infection endemic in Latin America, immune protection is mediated by Th1/Th17 cells and disease severity with predominant Th2/Th9/Treg responses. Because of its important role at epithelial barriers, we evaluate the role of AhR in the outcome of a pulmonary model of paracoccidioidomycosis. AhR−/− mice show increased fungal burdens, enhanced tissue pathology and mortality. During the infection, AhR−/− mice have more pulmonary myeloid cells with activated phenotype and reduced numbers expressing indoleamine 2,3 dioxygenase 1. AhR-deficient lungs have altered production of cytokines and reduced numbers of innate lymphoid cells (NK, ILC3 and NCR IL-22). The lungs of AhR−/− mice showed increased presence Th17 cells concomitant with reduced numbers of Th1, Th22 and Foxp3+ Treg cells. Furthermore, treatment of infected WT mice with an AhR-specific antagonist (CH223191) reproduced the main findings obtained in AhR−/− mice. Collectively our data demonstrate that in pulmonary paracoccidioidomycosis AhR controls fungal burden and excessive tissue inflammation and is a possible target for antifungal therapy.
Collapse
|
18
|
Huang YS, Ogbechi J, Clanchy FI, Williams RO, Stone TW. IDO and Kynurenine Metabolites in Peripheral and CNS Disorders. Front Immunol 2020; 11:388. [PMID: 32194572 PMCID: PMC7066259 DOI: 10.3389/fimmu.2020.00388] [Citation(s) in RCA: 113] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 02/18/2020] [Indexed: 12/12/2022] Open
Abstract
The importance of the kynurenine pathway in normal immune system function has led to an appreciation of its possible contribution to autoimmune disorders such as rheumatoid arthritis. Indoleamine-2,3-dioxygenase (IDO) activity exerts a protective function, limiting the severity of experimental arthritis, whereas deletion or inhibition exacerbates the symptoms. Other chronic disorder with an inflammatory component, such as atherosclerosis, are also suppressed by IDO activity. It is suggested that this overall anti-inflammatory activity is mediated by a change in the relative production or activity of Th17 and regulatory T cell populations. Kynurenines may play an anti-inflammatory role also in CNS disorders such as Huntington's disease, Alzheimer's disease and multiple sclerosis, in which signs of inflammation and neurodegeneration are involved. The possibility is discussed that in Huntington's disease kynurenines interact with other anti-inflammatory molecules such as Human Lymphocyte Antigen-G which may be relevant in other disorders. Kynurenine involvement may account for the protection afforded to animals with cerebral malaria and trypanosomiasis when they are treated with an inhibitor of kynurenine-3-monoxygenase (KMO). There is some evidence that changes in IL-10 may contribute to this protection and the relationship between kynurenines and IL-10 in arthritis and other inflammatory conditions should be explored. In addition, metabolites of kynurenine downstream of KMO, such as anthranilic acid and 3-hydroxy-anthranilic acid can influence inflammation, and the ratio of these compounds is a valuable biomarker of inflammatory status although the underlying molecular mechanisms of the changes require clarification. Hence it is essential that more effort be expended to identify their sites of action as potential targets for drug development. Finally, we discuss increasing awareness of the epigenetic regulation of IDO, for example by DNA methylation, a phenomenon which may explain differences between individuals in their susceptibility to arthritis and other inflammatory disorders.
Collapse
Affiliation(s)
- Yi-Shu Huang
- The Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, United Kingdom
| | - Joy Ogbechi
- The Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, United Kingdom
| | - Felix I Clanchy
- The Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, United Kingdom
| | - Richard O Williams
- The Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, United Kingdom
| | - Trevor W Stone
- The Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
19
|
Hu Q, Jin L, Zeng J, Wang J, Zhong S, Fan W, Liao W. Tryptophan metabolite-regulated Treg responses contribute to attenuation of airway inflammation during specific immunotherapy in a mouse asthma model. Hum Vaccin Immunother 2020; 16:1891-1899. [PMID: 31951781 DOI: 10.1080/21645515.2019.1698900] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
In allergen-specific immunotherapy for asthma, antigens attached to dendritic cells increase the tryptophan metabolism in these cells and alter the Th17/Treg balance in the airways. Tryptophan metabolism has long been suggested to be relevant in the pathophysiology of allergic disorders, including asthma. Our study investigated whether tryptophan metabolites are responsible for the changes in Th17/Treg balance and decreases in airway hyperreactivity and inflammation seen during allergen-specific immunotherapy in an asthma model. Ovalbumin was injected intraperitoneally into mice to establish an asthma model, and then high dose ovalbumin allergen-specific immunotherapy was administered to induce immune tolerance. Airway hyperreactivity and serum ovalbumin-specific immunoglobulin E were measured to assess whether the animal model was successfully established. We then examined the influence of inhibition of tryptophan metabolism and the addition of tryptophan metabolites on allergen-specific immunotherapy-induced changes in the Th17/Treg balance and decreases in airway inflammation and inflammatory cytokines. Production of tryptophan metabolites was partly responsible for the allergen-specific immunotherapy-induced increase in Tregs, decrease in airway inflammation, and decrease in inflammatory cytokines. Ovalbumin-specific immunoglobulin E and airway hyperreactivity were not affected. In the context of asthma, an increase in tryptophan metabolites is one of the mechanisms by which allergen-specific immunotherapy achieves immune tolerance.
Collapse
Affiliation(s)
- Qi Hu
- Department of Pediatrics, The First Hospital Affiliated to Army Medical University , Chongqing, China
| | - Ling Jin
- Department of Pediatrics, The First Hospital Affiliated to Army Medical University , Chongqing, China
| | - Jing Zeng
- Department of Pediatrics, The First Hospital Affiliated to Army Medical University , Chongqing, China
| | - Jinyu Wang
- Department of the Laboratory of Neurosurgery, The First Hospital Affiliated to Army Medical University , Chongqing, China
| | - Shimin Zhong
- Department of Pediatrics, The First Hospital Affiliated to Army Medical University , Chongqing, China
| | - Wenting Fan
- Department of Pediatrics, The First Hospital Affiliated to Army Medical University , Chongqing, China
| | - Wei Liao
- Department of Pediatrics, The First Hospital Affiliated to Army Medical University , Chongqing, China
| |
Collapse
|
20
|
Lee SB, Han YR, Jeon HJ, Jun CH, Kim SK, Chin J, Lee SJ, Jeong M, Lee JE, Lee CH, Cho SJ, Kim DS, Jeon YH. Medical fluorophore 1 (MF1), a benzoquinolizinium-based fluorescent dye, as an inflammation imaging agent. J Mater Chem B 2019; 7:7326-7331. [PMID: 31681930 DOI: 10.1039/c9tb01266d] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Structure-based targeting of fluorescent dyes is essential for their use as imaging agents for disease diagnosis. Here, we describe the development of the benzoquinolizinium compound Medical fluorophore 1 (MF1) as a novel biomedical imaging agent that allows the visualization of inflammation by virtue of its unique chemical structure. Lipopolysaccharide treatment stimulated the uptake of MF1 by bone marrow-derived macrophages, with no adverse effects on cell proliferation. In vivo fluorescence lifetime imaging revealed the accumulation of MF1 in carrageenan-induced acute inflammatory lesions in mice, which peaked at 6 h. MF1-based imaging also allowed monitoring of the response to the anti-inflammatory drugs dexamethasone and sulfasalazine. Thus, MF1 can be used to diagnose diseases characterized by inflammation as well as treatment efficacy.
Collapse
Affiliation(s)
- Sang Bong Lee
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, 80 Chembok-ro Dong-gu Daegu, Republic of Korea.
| | - Ye Ri Han
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, 80 Chembok-ro Dong-gu Daegu, Republic of Korea.
| | - Hui-Jeon Jeon
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, 80 Chembok-ro Dong-gu Daegu, Republic of Korea.
| | - Chul-Ho Jun
- Department of Chemistry, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea and Center for NanoMedicine, Institute for Basic Science (IBS), Seoul, Republic of Korea and Yonsei-IBS Institute, Yonsei University, Seoul 03722, Republic of Korea
| | - Sang-Kyoon Kim
- Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation, 80 Chembok-ro Dong-gu Daegu, Republic of Korea.
| | - Jungwook Chin
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, 80 Chembok-ro Dong-gu Daegu, Republic of Korea.
| | - Su-Jeong Lee
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, 80 Chembok-ro Dong-gu Daegu, Republic of Korea.
| | - Minseon Jeong
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, 80 Chembok-ro Dong-gu Daegu, Republic of Korea.
| | - Jae-Eon Lee
- Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation, 80 Chembok-ro Dong-gu Daegu, Republic of Korea. and Department of Biomaterials Science, College of Natural Resources and Life Science/Life and Industry Convergence Research Institute, Pusan National University, Pusan, Republic of Korea
| | - Chang-Hee Lee
- Department of Chemistry, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea and Center for NanoMedicine, Institute for Basic Science (IBS), Seoul, Republic of Korea and Yonsei-IBS Institute, Yonsei University, Seoul 03722, Republic of Korea
| | - Sung Jin Cho
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, 80 Chembok-ro Dong-gu Daegu, Republic of Korea.
| | - Dong-Su Kim
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, 80 Chembok-ro Dong-gu Daegu, Republic of Korea.
| | - Yong Hyun Jeon
- Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation, 80 Chembok-ro Dong-gu Daegu, Republic of Korea.
| |
Collapse
|
21
|
Renga G, Bellet MM, Stincardini C, Pariano M, Oikonomou V, Villella VR, Brancorsini S, Clerici C, Romani L, Costantini C. To Be or Not to Be a Pathogen: Candida albicans and Celiac Disease. Front Immunol 2019; 10:2844. [PMID: 31867008 PMCID: PMC6906151 DOI: 10.3389/fimmu.2019.02844] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 11/19/2019] [Indexed: 12/20/2022] Open
Abstract
Celiac disease (CD) is an immune-mediated disorder triggered by the ingestion of gluten and characterized by reversible small-bowel mucosal atrophy in genetically predisposed subjects. Although the prevalence of CD has increased, many aspects of this pathology are still unrecognized. Candida albicans, a commensal of the human gastrointestinal tract, has been linked to CD for a long time based, among others, upon the observation of similarity between the fungal wall component, hyphal wall protein 1, and CD-related gliadin T-cell epitopes. We have recently demonstrated that Candida may switch from commensal to pathogen contingent upon several players, including mast cells, key sentinels of the immune system at the interface between the environment and the host, and the pleiotropic cytokine IL-9. However, other factors are likely to play a role by altering the balance between inflammation and tolerance. In this regard, tryptophan and its metabolites are increasingly being recognized in promoting mucosal homeostasis by balancing the immune response to external cues. Based on these premises, we will discuss how the output of Candida colonization in the gut is highly contextual, being determined at the intersection of many immunological (IL-9/mast cells) and metabolic (tryptophan) pathways that ultimately dictate the Candida commensalism vs. pathogenicity in CD, thus paving the way for novel therapeutic opportunities in CD.
Collapse
Affiliation(s)
- Giorgia Renga
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Marina M Bellet
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | | | - Marilena Pariano
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Vasilis Oikonomou
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Valeria R Villella
- Division of Genetics and Cell Biology, European Institute for Research in Cystic Fibrosis, San Raffaele Scientific Institute, Milan, Italy
| | | | - Carlo Clerici
- Gastroenterology Unit, Santa Maria della Misericordia Hospital of Perugia, Perugia, Italy
| | - Luigina Romani
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Claudio Costantini
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| |
Collapse
|
22
|
Taleb S. Tryptophan Dietary Impacts Gut Barrier and Metabolic Diseases. Front Immunol 2019; 10:2113. [PMID: 31552046 PMCID: PMC6746884 DOI: 10.3389/fimmu.2019.02113] [Citation(s) in RCA: 127] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 08/21/2019] [Indexed: 12/13/2022] Open
Abstract
The intestine has a major role in the digestion and absorption of nutrients, and gut barrier is the first defense line against harmful pathogens. Alteration of the intestinal barrier is associated with enhanced intestinal permeability and development of numerous pathological diseases including gastrointestinal and cardiometabolic diseases. Among the metabolites that play an important role within intestinal health, L Tryptophan (Trp) is one of the nine essential amino acids supplied by diet, whose metabolism appears as a key modulator of gut microbiota, with major impacts on physiological, and pathological pathways. Recently, emerging evidence showed that the Trp catabolism through one major enzyme indoleamine 2,3-dioxygenase 1 (IDO1) expressed by the host affects Trp metabolism by gut microbiota to generate indole metabolites, thereby altering gut function and health in mice and humans. In this mini review, I summarize the most recent advances concerning the role of Trp metabolism in host–microbiota cross-talk in health, and metabolic diseases. This novel aspect of IDO1 function in intestine will better explain its complex roles in a broad range of disease states where the gut function affects local as well as systemic health, and will open new therapeutic strategies.
Collapse
Affiliation(s)
- Soraya Taleb
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unit 970, Paris Cardiovascular Research Center, and Université Paris-Descartes, Paris, France
| |
Collapse
|
23
|
Mirkov I, Popov Aleksandrov A, Lazovic B, Glamoclija J, Kataranovski M. Usefulness of animal models of aspergillosis in studying immunity against Aspergillus infections. J Mycol Med 2019; 29:84-96. [DOI: 10.1016/j.mycmed.2019.01.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 11/28/2018] [Accepted: 01/14/2019] [Indexed: 01/08/2023]
|
24
|
Gupta AK, Carviel J, Shear NH. Onychomycosis and Chronic Fungal Disease: Exploiting a Commensal Disguise to Stage a Covert Invasion. J Cutan Med Surg 2017; 22:318-322. [PMID: 29191054 DOI: 10.1177/1203475417745827] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Onychomycosis is a chronic fungal infection that is recalcitrant to treatment and often results in relapse. New evidence suggests that disease prognosis may be linked to pathogens manipulating host immune responses. Therefore, individuals with specific mutations, including those affecting pattern recognition receptors or the interleukin (IL)-17 and IL-22 pathways, may be more susceptible to infection. Moreover, it is recommended that those with a family history of immune mutations or predisposition to fungal disease be treated aggressively for onychomycosis prior to symptom progression. In addition, incorporating genetic testing and new investigational therapy such as IL-33 and interferon-γ may improve treatment outcome.
Collapse
Affiliation(s)
- Aditya K Gupta
- 1 Department of Medicine, University of Toronto School of Medicine, Toronto, Ontario, Canada.,2 Mediprobe Research, London, Ontario, Canada
| | | | - Neil H Shear
- 3 Division of Dermatology, Department of Medicine, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Canada
| |
Collapse
|
25
|
Colliou N, Ge Y, Sahay B, Gong M, Zadeh M, Owen JL, Neu J, Farmerie WG, Alonzo F, Liu K, Jones DP, Li S, Mohamadzadeh M. Commensal Propionibacterium strain UF1 mitigates intestinal inflammation via Th17 cell regulation. J Clin Invest 2017; 127:3970-3986. [PMID: 28945202 PMCID: PMC5663347 DOI: 10.1172/jci95376] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 08/02/2017] [Indexed: 12/21/2022] Open
Abstract
Consumption of human breast milk (HBM) attenuates the incidence of necrotizing enterocolitis (NEC), which remains a leading and intractable cause of mortality in preterm infants. Here, we report that this diminution correlates with alterations in the gut microbiota, particularly enrichment of Propionibacterium species. Transfaunation of microbiota from HBM-fed preterm infants or a newly identified and cultured Propionibacterium strain, P. UF1, to germfree mice conferred protection against pathogen infection and correlated with profound increases in intestinal Th17 cells. The induction of Th17 cells was dependent on bacterial dihydrolipoamide acetyltransferase (DlaT), a major protein expressed on the P. UF1 surface layer (S-layer). Binding of P. UF1 to its cognate receptor, SIGNR1, on dendritic cells resulted in the regulation of intestinal phagocytes. Importantly, transfer of P. UF1 profoundly mitigated induced NEC-like injury in neonatal mice. Together, these results mechanistically elucidate the protective effects of HBM and P. UF1-induced immunoregulation, which safeguard against proinflammatory diseases, including NEC.
Collapse
Affiliation(s)
- Natacha Colliou
- Department of Infectious Diseases and Immunology
- Division of Gastroenterology, Hepatology & Nutrition, Department of Medicine
| | - Yong Ge
- Department of Infectious Diseases and Immunology
- Division of Gastroenterology, Hepatology & Nutrition, Department of Medicine
| | - Bikash Sahay
- Department of Infectious Diseases and Immunology
| | - Minghao Gong
- Department of Infectious Diseases and Immunology
- Division of Gastroenterology, Hepatology & Nutrition, Department of Medicine
| | - Mojgan Zadeh
- Department of Infectious Diseases and Immunology
- Division of Gastroenterology, Hepatology & Nutrition, Department of Medicine
| | | | - Josef Neu
- Division of Neonatology, Department of Pediatrics, and
| | - William G. Farmerie
- Interdisciplinary Center for Biotechnology Research, University of Florida, Gainesville, Florida, USA
| | - Francis Alonzo
- Department of Microbiology and Immunology, Loyola University Chicago, Maywood, Illinois, USA
| | - Ken Liu
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Dean P. Jones
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Shuzhao Li
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Mansour Mohamadzadeh
- Department of Infectious Diseases and Immunology
- Division of Gastroenterology, Hepatology & Nutrition, Department of Medicine
| |
Collapse
|
26
|
Xu K, Liu H, Bai M, Gao J, Wu X, Yin Y. Redox Properties of Tryptophan Metabolism and the Concept of Tryptophan Use in Pregnancy. Int J Mol Sci 2017; 18:E1595. [PMID: 28737706 PMCID: PMC5536082 DOI: 10.3390/ijms18071595] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 07/11/2017] [Accepted: 07/19/2017] [Indexed: 12/30/2022] Open
Abstract
During pregnancy, tryptophan (Trp) is required for several purposes, and Trp metabolism varies over time in the mother and fetus. Increased oxidative stress (OS) with high metabolic, energy and oxygen demands during normal pregnancy or in pregnancy-associated disorders has been reported. Taking the antioxidant properties of Trp and its metabolites into consideration, we made four hypotheses. First, the use of Trp and its metabolites is optional based on their antioxidant properties during pregnancy. Second, dynamic Trp metabolism is an accommodation mechanism in response to OS. Third, regulation of Trp metabolism could be used to control/attenuate OS according to variations in Trp metabolism during pregnancy. Fourth, OS-mediated injury could be alleviated by regulation of Trp metabolism in pregnancy-associated disorders. Future studies in normal/abnormal pregnancies and in associated disorders should include measurements of free Trp, total Trp, Trp metabolites, and activities of Trp-degrading enzymes in plasma. Abnormal pregnancies and some associated disorders may be associated with disordered Trp metabolism related to OS. Mounting evidence suggests that the investigation of the use of Trp and its metabolites in pregnancy will be meanful.
Collapse
Affiliation(s)
- Kang Xu
- Chinese Academy of Sciences, Institute of Subtropical Agriculture, Key Laboratory of Agroecological Processes in Subtropical Region, Changsha 410125, China.
- National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Changsha 410125, China.
- Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha 410125, China.
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South Central, Ministry of Agriculture, Changsha 410125, China.
| | - Hongnan Liu
- Chinese Academy of Sciences, Institute of Subtropical Agriculture, Key Laboratory of Agroecological Processes in Subtropical Region, Changsha 410125, China.
- National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Changsha 410125, China.
- Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha 410125, China.
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South Central, Ministry of Agriculture, Changsha 410125, China.
| | - Miaomiao Bai
- Chinese Academy of Sciences, Institute of Subtropical Agriculture, Key Laboratory of Agroecological Processes in Subtropical Region, Changsha 410125, China.
- National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Changsha 410125, China.
- Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha 410125, China.
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South Central, Ministry of Agriculture, Changsha 410125, China.
| | - Jing Gao
- Chinese Academy of Sciences, Institute of Subtropical Agriculture, Key Laboratory of Agroecological Processes in Subtropical Region, Changsha 410125, China.
- National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Changsha 410125, China.
- Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha 410125, China.
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South Central, Ministry of Agriculture, Changsha 410125, China.
| | - Xin Wu
- Chinese Academy of Sciences, Institute of Subtropical Agriculture, Key Laboratory of Agroecological Processes in Subtropical Region, Changsha 410125, China.
- National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Changsha 410125, China.
- Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha 410125, China.
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South Central, Ministry of Agriculture, Changsha 410125, China.
| | - Yulong Yin
- Chinese Academy of Sciences, Institute of Subtropical Agriculture, Key Laboratory of Agroecological Processes in Subtropical Region, Changsha 410125, China.
- National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Changsha 410125, China.
- Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha 410125, China.
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South Central, Ministry of Agriculture, Changsha 410125, China.
| |
Collapse
|
27
|
The Role of Interleukin-23 in the Early Development of Emphysema in HIV1(+) Smokers. J Immunol Res 2016; 2016:3463104. [PMID: 27446965 PMCID: PMC4942665 DOI: 10.1155/2016/3463104] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 05/19/2016] [Indexed: 12/28/2022] Open
Abstract
Rationale. Matrix metalloproteinase-9 (MMP-9) expression is upregulated in alveolar macrophages (AM) of HIV1+ smokers who develop emphysema. Knowing that lung epithelial lining fluid (ELF) of HIV1+ smokers contains increased levels of inflammatory cytokines compared to HIV1− smokers, we hypothesized that upregulation of lung cytokines in HIV1+ smokers may be functionally related to increased MMP-9 expression. Methods. Cytokine arrays evaluated cytokine protein levels in ELF obtained from 5 groups of individuals: HIV1− healthy nonsmokers, HIV1− healthy smokers, HIV1− smokers with low diffusing capacity (DLCO), HIV1+ nonsmokers, and HIV1+ smokers with low DLCO. Results. Increased levels of the Th17 related cytokine IL-23 were found in HIV1− smokers with low DLCO and HIV1+ smokers and nonsmokers. Relative IL-23 gene expression was increased in AM of HIV1+ individuals, with greater expression in AM of HIV1+ smokers with low DLCO. Infection with HIV1 in vitro induced IL-23 expression in normal AM. IL-23 stimulation of AM/lymphocyte cocultures in vitro induced upregulation of MMP-9. Lung T lymphocytes express receptor IL-23R and interact with AM in order to upregulate MMP-9. Conclusion. This mechanism may contribute to the increased tissue destruction in the lungs of HIV1+ smokers and suggests that Th17 related inflammation may play a role.
Collapse
|
28
|
Morris G, Berk M, Carvalho A, Caso JR, Sanz Y, Walder K, Maes M. The Role of the Microbial Metabolites Including Tryptophan Catabolites and Short Chain Fatty Acids in the Pathophysiology of Immune-Inflammatory and Neuroimmune Disease. Mol Neurobiol 2016; 54:4432-4451. [PMID: 27349436 DOI: 10.1007/s12035-016-0004-2] [Citation(s) in RCA: 183] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 06/14/2016] [Indexed: 12/19/2022]
Abstract
There is a growing awareness that gut commensal metabolites play a major role in host physiology and indeed the pathophysiology of several illnesses. The composition of the microbiota largely determines the levels of tryptophan in the systemic circulation and hence, indirectly, the levels of serotonin in the brain. Some microbiota synthesize neurotransmitters directly, e.g., gamma-amino butyric acid, while modulating the synthesis of neurotransmitters, such as dopamine and norepinephrine, and brain-derived neurotropic factor (BDNF). The composition of the microbiota determines the levels and nature of tryptophan catabolites (TRYCATs) which in turn has profound effects on aryl hydrocarbon receptors, thereby influencing epithelial barrier integrity and the presence of an inflammatory or tolerogenic environment in the intestine and beyond. The composition of the microbiota also determines the levels and ratios of short chain fatty acids (SCFAs) such as butyrate and propionate. Butyrate is a key energy source for colonocytes. Dysbiosis leading to reduced levels of SCFAs, notably butyrate, therefore may have adverse effects on epithelial barrier integrity, energy homeostasis, and the T helper 17/regulatory/T cell balance. Moreover, dysbiosis leading to reduced butyrate levels may increase bacterial translocation into the systemic circulation. As examples, we describe the role of microbial metabolites in the pathophysiology of diabetes type 2 and autism.
Collapse
Affiliation(s)
- Gerwyn Morris
- Tir Na Nog, Bryn Road seaside 87, Llanelli, SA152LW, Wales, UK
| | - Michael Berk
- IMPACT Strategic Research Centre, School of Medicine, Barwon Health, Deakin University, P.O. Box 291, Geelong, VIC, 3220, Australia.,Orygen Youth Health Research Centre and the Centre of Youth Mental Health, The Florey Institute for Neuroscience and Mental Health and the Department of Psychiatry, University of Melbourne, Parkville, 3052, Australia
| | - Andre Carvalho
- Department of Clinical Medicine and Translational Psychiatry Research Group, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, 60430-040, Brazil
| | - Javier R Caso
- Department of Pharmacology, School of Medicine, University Complutense of Madrid, Avda. Complutense s/n, 28040, Madrid, Spain.,Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Avda. Complutense s/n, 28040, Madrid, Spain.,Instituto de Investigación Hospital 12 de Octubre (Imas12), Avda. Complutense s/n, 28040, Madrid, Spain
| | - Yolanda Sanz
- Microbial Ecology, Nutrition & Health Research Unit, Institute of Agrochemistry and Food Technology, National Research Council (IATA-CSIC), Av. Agustin Escardino 7, 46980, Paterna, Valencia, Spain
| | - Ken Walder
- Centre for Molecular and Medical Research, School of Medicine, Deakin University, Geelong, Australia
| | - Michael Maes
- IMPACT Strategic Research Centre, School of Medicine, Barwon Health, Deakin University, P.O. Box 291, Geelong, VIC, 3220, Australia. .,Health Sciences Postgraduate Program, Health Sciences Center, State University of Londrina, Londrina, Parana, Brazil.
| |
Collapse
|
29
|
Jung CC, Su HJ, Liang HH. Association between indoor air pollutant exposure and blood pressure and heart rate in subjects according to body mass index. THE SCIENCE OF THE TOTAL ENVIRONMENT 2016; 539:271-276. [PMID: 26363400 DOI: 10.1016/j.scitotenv.2015.08.158] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Revised: 08/11/2015] [Accepted: 08/31/2015] [Indexed: 05/11/2023]
Abstract
This study investigates the effects of high body mass index (BMI) of subjects on individual who exhibited high cardiovascular disease indexes with blood pressure (BP) and heart rate (HR) when exposed to high levels of indoor air pollutants. We collected 115 office workers, and measured their systolic blood pressure (SBP), diastolic blood pressure (DBP) and HR at the end of the workday. The subjects were divided into three groups according to BMI: 18-24 (normal weight), 24-27 (overweight) and >27 (obese). This study also measured the levels of carbon dioxide (CO2), total volatile organic compounds (TVOC), particulate matter with an aerodynamic diameter less than 2.5μm (PM2.5), as well as the bacteria and fungi in the subjects' work-places. The pollutant effects were divided by median. Two-way analysis of variance (ANOVA) was used to analyze the health effects of indoor air pollution exposure according to BMI. Our study showed that higher levels of SBP, DBP and HR occurred in subjects who were overweight or obese as compared to those with normal weight. Moreover, there was higher level of SBP in subjects who were overweight or obese when they were exposed to higher levels of TVOC and fungi (p<0.05). We also found higher value for DBP and HR with increasing BMI to be associated with exposure to higher TVOC levels. This study suggests that individuals with higher BMI have higher cardiovascular disease risk when they are exposed to poor indoor air quality (IAQ), and specifically in terms of TVOC.
Collapse
Affiliation(s)
- Chien-Cheng Jung
- Dept. of Environmental and Occupational Health, College of Medicine, National Cheng Kung University, Tainan City, Taiwan
| | - Huey-Jen Su
- Dept. of Environmental and Occupational Health, College of Medicine, National Cheng Kung University, Tainan City, Taiwan.
| | - Hsiu-Hao Liang
- Dept. of Environmental and Occupational Health, College of Medicine, National Cheng Kung University, Tainan City, Taiwan
| |
Collapse
|
30
|
Learning from other diseases: protection and pathology in chronic fungal infections. Semin Immunopathol 2015; 38:239-48. [DOI: 10.1007/s00281-015-0523-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 08/18/2015] [Indexed: 12/11/2022]
|
31
|
Araújo EF, Loures FV, Bazan SB, Feriotti C, Pina A, Schanoski AS, Costa TA, Calich VLG. Indoleamine 2,3-dioxygenase controls fungal loads and immunity in Paracoccidioidomicosis but is more important to susceptible than resistant hosts. PLoS Negl Trop Dis 2014; 8:e3330. [PMID: 25411790 PMCID: PMC4238999 DOI: 10.1371/journal.pntd.0003330] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Accepted: 10/08/2014] [Indexed: 11/29/2022] Open
Abstract
Background Paracoccidioidomycosis, a primary fungal infection restricted to Latin America, is acquired by inhalation of fungal particles. The immunoregulatory mechanisms that control the severe and mild forms of paracoccidioidomycosis are still unclear. Indoleamine 2,3-dioxygenase (IDO), an IFN-γ induced enzyme that catalyzes tryptophan metabolism, can control host-pathogen interaction by inhibiting pathogen growth, T cell immunity and tissue inflammation. Methodology/Principal Findings In this study, we investigated the role of IDO in pulmonary paracoccidioidomycosis of susceptible and resistant mice. IDO was blocked by 1-methyl-dl-tryptophan (1MT), and fungal infection studied in vitro and in vivo. Paracoccidioides brasiliensis infection was more severe in 1MT treated than untreated macrophages of resistant and susceptible mice, concurrently with decreased production of kynurenines and IDO mRNA. Similar results were observed in the pulmonary infection. Independent of the host genetic pattern, IDO inhibition reduced fungal clearance but enhanced T cell immunity. The early IDO inhibition resulted in increased differentiation of dendritic and Th17 cells, accompanied by reduced responses of Th1 and Treg cells. Despite these equivalent biological effects, only in susceptible mice the temporary IDO blockade caused sustained fungal growth, increased tissue pathology and mortality rates. In contrast, resistant mice were able to recover the transitory IDO blockade by the late control of fungal burdens without enhanced tissue pathology. Conclusions/Significance Our studies demonstrate for the first time that in pulmonary paracoccidioidomycosis, IDO is an important immunoregulatory enzyme that promotes fungal clearance and inhibits T cell immunity and inflammation, with prominent importance to susceptible hosts. In fact, only in the susceptible background IDO inhibition resulted in uncontrolled tissue pathology and mortality rates. Our findings open new perspectives to understand the immunopathology of paracoccidioidomycosis, and suggest that an insufficient IDO activity could be associated with the severe cases of human PCM characterized by inefficient fungal clearance and excessive inflammation. Immunoprotection to paracoccidiodomycosis, a systemic mycosis endemic in Latin America, is mediated by T cell immunity whereas immunosuppression characterizes the severe forms of the disease. Indoleamine 2,3-dioxygenase (IDO), an enzyme mainly induced by IFN-γ, catabolizes tryptophan along the kynurenines pathway. Tryptophan deficiency has been associated with reduced pathogen growth, while elevated levels of kynurenines with suppressed immune responses. In this study, the role of IDO in pulmonary paracoccidioidomycosis was investigated using resistant and susceptible mice. In both mouse strains, IDO blockade by 1-methyl tryptophan resulted in inefficient fungal clearance accompanied by enhanced T cell immunity. Despite these equivalent biological effects, only in susceptible mice IDO inhibition caused progressive fungal growth and tissue pathology resulting in increased mortality. Our findings demonstrate for the first time that IDO exert a yet unexplored immunoregulatory role in pulmonary paracoccidioidomycosis that can be particularly important in the severe cases of the disease.
Collapse
Affiliation(s)
- Eliseu F. Araújo
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, São Paulo, Brazil
| | - Flávio V. Loures
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, São Paulo, Brazil
| | - Silvia B. Bazan
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, São Paulo, Brazil
| | - Claudia Feriotti
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, São Paulo, Brazil
| | - Adriana Pina
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, São Paulo, Brazil
| | - Alessandra S. Schanoski
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, São Paulo, Brazil
| | - Tânia A. Costa
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, São Paulo, Brazil
| | - Vera L. G. Calich
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, São Paulo, Brazil
- * E-mail:
| |
Collapse
|
32
|
Romani L, Zelante T, De Luca A, Iannitti RG, Moretti S, Bartoli A, Aversa F, Puccetti P. Microbiota control of a tryptophan-AhR pathway in disease tolerance to fungi. Eur J Immunol 2014; 44:3192-200. [PMID: 25256754 DOI: 10.1002/eji.201344406] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Revised: 07/30/2014] [Accepted: 09/23/2014] [Indexed: 01/09/2023]
Abstract
An increased understanding of the importance of microbiota in shaping the host's immune and metabolic activities has rendered fungal interactions with their hosts more complex than previously appreciated. The aryl hydrocarbon receptor (AhR) has a pivotal role in connecting tryptophan catabolism by microbial communities and the host's own pathway of tryptophan metabolite production with the orchestration of T-cell function. AhR activation by a Lactobacillus-derived AhR ligand leads to the production of IL-22 to the benefit of mucosal defense mechanisms, an activity upregulated in the absence of the host tryptophan catabolic enzyme, indoleamine 2,3-dioxygenase 1 (IDO1), which is required for protection from fungal diseases ("disease tolerance"). As AhR activation in turn leads to the activation-in a feedback fashion-of IDO1, the regulatory loop involving AhR and IDO1 may have driven the coevolution of commensal fungi with the mammalian immune system and the microbiota, to the benefit of host survival and fungal commensalism. This review will discuss the essential help the microbiota provides in controlling the balance between the dual nature of the fungal-host relationship, namely, commensalism vs. infection.
Collapse
Affiliation(s)
- Luigina Romani
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Borghi M, Renga G, Puccetti M, Oikonomou V, Palmieri M, Galosi C, Bartoli A, Romani L. Antifungal Th Immunity: Growing up in Family. Front Immunol 2014; 5:506. [PMID: 25360137 PMCID: PMC4197763 DOI: 10.3389/fimmu.2014.00506] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Accepted: 09/28/2014] [Indexed: 12/25/2022] Open
Abstract
Fungal diseases represent an important paradigm in immunology since they can result from either the lack of recognition or over-activation of the inflammatory response. Current understanding of the pathophysiology underlying fungal infections and diseases highlights the multiple cell populations and cell-signaling pathways involved in these conditions. A systems biology approach that integrates investigations of immunity at the systems-level is required to generate novel insights into this complexity and to decipher the dynamics of the host–fungus interaction. It is becoming clear that a three-way interaction between the host, microbiota, and fungi dictates the types of host–fungus relationship. Tryptophan metabolism helps support this interaction, being exploited by the mammalian host and commensals to increase fitness in response to fungi via resistance and tolerance mechanisms of antifungal immunity. The cellular and molecular mechanisms that provide immune homeostasis with the fungal biota and its possible rupture in fungal infections and diseases will be discussed within the expanding role of antifungal Th cell responses.
Collapse
Affiliation(s)
- Monica Borghi
- Pathology Section, Department of Experimental Medicine, University of Perugia , Perugia , Italy
| | - Giorgia Renga
- Pathology Section, Department of Experimental Medicine, University of Perugia , Perugia , Italy
| | | | - Vasileios Oikonomou
- Pathology Section, Department of Experimental Medicine, University of Perugia , Perugia , Italy
| | - Melissa Palmieri
- Pathology Section, Department of Experimental Medicine, University of Perugia , Perugia , Italy
| | - Claudia Galosi
- Pathology Section, Department of Experimental Medicine, University of Perugia , Perugia , Italy
| | - Andrea Bartoli
- Pathology Section, Department of Experimental Medicine, University of Perugia , Perugia , Italy
| | - Luigina Romani
- Pathology Section, Department of Experimental Medicine, University of Perugia , Perugia , Italy
| |
Collapse
|
34
|
Bacher P, Kniemeyer O, Teutschbein J, Thön M, Vödisch M, Wartenberg D, Scharf DH, Koester-Eiserfunke N, Schütte M, Dübel S, Assenmacher M, Brakhage AA, Scheffold A. Identification of Immunogenic Antigens fromAspergillus fumigatusby Direct Multiparameter Characterization of Specific Conventional and Regulatory CD4+T Cells. THE JOURNAL OF IMMUNOLOGY 2014; 193:3332-43. [DOI: 10.4049/jimmunol.1400776] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
35
|
Denning DW, Pashley C, Hartl D, Wardlaw A, Godet C, Del Giacco S, Delhaes L, Sergejeva S. Fungal allergy in asthma-state of the art and research needs. Clin Transl Allergy 2014; 4:14. [PMID: 24735832 PMCID: PMC4005466 DOI: 10.1186/2045-7022-4-14] [Citation(s) in RCA: 215] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Accepted: 03/19/2014] [Indexed: 01/31/2023] Open
Abstract
Sensitization to fungi and long term or uncontrolled fungal infection are associated with poor control of asthma, the likelihood of more severe disease and complications such as bronchiectasis and chronic pulmonary aspergillosis. Modelling suggests that >6.5 million people have severe asthma with fungal sensitizations (SAFS), up to 50% of adult asthmatics attending secondary care have fungal sensitization, and an estimated 4.8 million adults have allergic bronchopulmonary aspergillosis (ABPA). There is much uncertainty about which fungi and fungal allergens are relevant to asthma, the natural history of sensitisation to fungi, if there is an exposure response relationship for fungal allergy, and the pathogenesis and frequency of exacerbations and complications. Genetic associations have been described but only weakly linked to phenotypes. The evidence base for most management strategies in ABPA, SAFS and related conditions is weak. Yet straightforward clinical practice guidelines for management are required. The role of environmental monitoring and optimal means of controlling disease to prevent disability and complications are not yet clear. In this paper we set out the key evidence supporting the role of fungal exposure, sensitisation and infection in asthmatics, what is understood about pathogenesis and natural history and identify the numerous areas for research studies.
Collapse
Affiliation(s)
- David W Denning
- The National Aspergillosis Centre, University Hospital of South Manchester, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK ; Education and Research Centre, UHSM, Southmoor Road, Manchester M23 9LT, UK
| | - Catherine Pashley
- Leicester Institute for Lung Health and Respiratory Biomedical Research Unit, Department of Infection Immunity and Inflammation, University of Leicester, Glenfield Hospital, Groby Road, Leicester LE3 9QP, UK
| | - Domink Hartl
- Department of Pediatrics, Infectious Diseases & Immunology, University of Tübingen, Tübingen, Germany
| | - Andrew Wardlaw
- Leicester Institute for Lung Health and Respiratory Biomedical Research Unit, Department of Infection Immunity and Inflammation, University of Leicester, Glenfield Hospital, Groby Road, Leicester LE3 9QP, UK
| | - Cendrine Godet
- Department of Infectious Diseases, CHU la Milétrie, Poitiers, France
| | - Stefano Del Giacco
- Department of Medical Sciences "M. Aresu", University of Cagliari, Cagliari, Italy
| | - Laurence Delhaes
- Biology & Diversity of Emerging Eukaryotic Pathogens (BDEEP), Center for Infection and Immunity of Lille (CIIL), INSERM U1019, CNRS UMR8204, IFR142, Lille Pasteur Institute, Lille Nord de France University (EA4547), Lille, France ; Department of Parasitology-Mycology, Regional Hospital Center, Faculty of Medicine, Lille, France
| | - Svetlana Sergejeva
- Translational Immunology Group, Institute of Technology, Tartu University, Tartu, Estonia ; North Estonia Medical Centre, Tallinn, Estonia
| |
Collapse
|
36
|
Lowe MM, Mold JE, Kanwar B, Huang Y, Louie A, Pollastri MP, Wang C, Patel G, Franks DG, Schlezinger J, Sherr DH, Silverstone AE, Hahn ME, McCune JM. Identification of cinnabarinic acid as a novel endogenous aryl hydrocarbon receptor ligand that drives IL-22 production. PLoS One 2014; 9:e87877. [PMID: 24498387 PMCID: PMC3912126 DOI: 10.1371/journal.pone.0087877] [Citation(s) in RCA: 103] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Accepted: 12/30/2013] [Indexed: 01/17/2023] Open
Abstract
The aryl hydrocarbon receptor (AHR) binds to environmental toxicants including synthetic halogenated aromatic hydrocarbons and is involved in a diverse array of biological processes. Recently, the AHR was shown to control host immunity by affecting the balance between inflammatory T cells that produce IL-17 (Th17) and IL-22 versus regulatory T cells (Treg) involved in tolerance. While environmental AHR ligands can mediate this effect, endogenous ligands are likely to be more relevant in host immune responses. We investigated downstream metabolites of tryptophan as potential AHR ligands because (1) tryptophan metabolites have been implicated in regulating the balance between Th17 and Treg cells and (2) many of the AHR ligands identified thus far are derivatives of tryptophan. We characterized the ability of tryptophan metabolites to bind and activate the AHR and to increase IL-22 production in human T cells. We report that the tryptophan metabolite, cinnabarinic acid (CA), is an AHR ligand that stimulates the differentiation of human and mouse T cells producing IL-22. We compare the IL-22-stimulating activity of CA to that of other tryptophan metabolites and define stimulation conditions that lead to CA production from immune cells. Our findings link tryptophan metabolism to AHR activation and define a novel endogenous AHR agonist with potentially broad biological functions.
Collapse
MESH Headings
- Animals
- Cell Differentiation
- Cells, Cultured
- Chromatography, Liquid
- Cytochrome P-450 CYP1A1/metabolism
- Humans
- Interleukins/metabolism
- Ligands
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Oxazines/metabolism
- Receptors, Aryl Hydrocarbon/physiology
- Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
- T-Lymphocytes, Regulatory/cytology
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- Th17 Cells
- Tryptophan/metabolism
- Interleukin-22
Collapse
Affiliation(s)
- Margaret M. Lowe
- Division of Experimental Medicine, Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
| | - Jeff E. Mold
- Division of Experimental Medicine, Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
| | - Bittoo Kanwar
- Division of Experimental Medicine, Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
- Division of Gastroenterology, Department of Pediatrics, University of California San Francisco, San Francisco, California, United States of America
| | - Yong Huang
- Drug Studies Unit, Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, United States of America
| | - Alexander Louie
- Drug Studies Unit, Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, United States of America
| | - Michael P. Pollastri
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, Massachusetts, United States of America
| | - Cuihua Wang
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, Massachusetts, United States of America
| | - Gautam Patel
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, Massachusetts, United States of America
| | - Diana G. Franks
- Biology Department, Woods Hole Oceanographic Institution, Woods Hole, Massachusetts, United States of America
| | - Jennifer Schlezinger
- Department of Environmental Health, School of Public Health, Boston University, Boston, Massachusetts, United States of America
| | - David H. Sherr
- Department of Environmental Health, School of Public Health, Boston University, Boston, Massachusetts, United States of America
| | - Allen E. Silverstone
- Department of Microbiology and Immunology, State University of New York Upstate Medical University, Syracuse, New York, United States of America
| | - Mark E. Hahn
- Biology Department, Woods Hole Oceanographic Institution, Woods Hole, Massachusetts, United States of America
| | - Joseph M. McCune
- Division of Experimental Medicine, Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
| |
Collapse
|
37
|
High doses of CpG oligodeoxynucleotides stimulate a tolerogenic TLR9-TRIF pathway. Nat Commun 2013; 4:1852. [PMID: 23673637 DOI: 10.1038/ncomms2874] [Citation(s) in RCA: 89] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2012] [Accepted: 04/12/2013] [Indexed: 02/07/2023] Open
Abstract
CpG-rich oligodeoxynucleotides activate the immune system, leading to innate and acquired immune responses. The immune-stimulatory effects of CpG-rich oligodeoxynucleotides are being exploited as a therapeutic approach. Here we show that at high doses, CpG-rich oligodeoxynucleotides promote an opposite, tolerogenic response in mouse plasmacytoid dendritic cells in vivo and in a human in vitro model. Unveiling a previously undescribed role for TRIF and TRAF6 proteins in Toll-like receptor 9 (TLR9) signalling, we demonstrate that physical association of TLR9, TRIF and TRAF6 leads to activation of noncanonical NF-κB signalling and the induction of IRF3- and TGF-β-dependent immune-suppressive tryptophan catabolism. In vivo, the TLR9-TRIF circuit--but not MyD88 signalling--was required for CpG protection against allergic inflammation. Our findings may be relevant to an increased understanding of the complexity of Toll-like receptor signalling and optimal exploitation of CpG-rich oligodeoxynucleotides as immune modulators.
Collapse
|
38
|
Iannitti RG, Carvalho A, Cunha C, De Luca A, Giovannini G, Casagrande A, Zelante T, Vacca C, Fallarino F, Puccetti P, Massi-Benedetti C, Defilippi G, Russo M, Porcaro L, Colombo C, Ratclif L, De Benedictis FM, Romani L. Th17/Treg imbalance in murine cystic fibrosis is linked to indoleamine 2,3-dioxygenase deficiency but corrected by kynurenines. Am J Respir Crit Care Med 2013; 187:609-20. [PMID: 23306541 DOI: 10.1164/rccm.201207-1346oc] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
RATIONALE Mutations in the cystic fibrosis (CF) transmembrane conductance regulator affect the innate epithelial immune function of the lung, resulting in exaggerated and ineffective airway inflammation that fails to eradicate pathogenic fungi. The appreciation of whether such fungi are primarily responsible for or a consequence of ineffective airway inflammation is important for future therapeutics development. OBJECTIVES To characterize the impact of the tryptophan/kynurenine pathway on pathogenic airway inflammation preventing effective fungal clearance in CF. METHODS We studied the expression of indoleamine 2,3-dioxygenase (IDO), the first enzyme in the kynurenine pathway of tryptophan degradation, in human and murine CF, the impact of IDO on lung inflammation and immunity in murine CF, and the potential role of tryptophan catabolism in pathogenesis and therapy of fungus-associated lung inflammation. MEASUREMENTS AND MAIN RESULTS IDO was defective in murine and human CF. Genetic and transcriptional regulatory mechanisms contributed to dysfunctional IDO activity that, in turn, correlated with imbalanced Th17/Treg-cell responses to Aspergillus fumigatus in murine CF. Treatments enhancing IDO function or preventing pathogenic Th17-cell activation restored protective immunity to the fungus and improved lung inflammation in murine CF. CONCLUSIONS This study provides a link between tryptophan catabolism and lung immune homeostasis in murine CF, representing a proof-of-concept that targeting pathogenic inflammation via IDO-mimetic drugs may benefit patients with CF.
Collapse
Affiliation(s)
- Rossana G Iannitti
- Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Via del Giochetto, Perugia, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Depletion of neutrophils in a protective model of pulmonary cryptococcosis results in increased IL-17A production by γδ T cells. BMC Immunol 2012; 13:65. [PMID: 23216912 PMCID: PMC3538069 DOI: 10.1186/1471-2172-13-65] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2012] [Accepted: 11/30/2012] [Indexed: 12/24/2022] Open
Abstract
Protective responses in mice immunized with an interferon-gamma producing strain of Cryptococcus neoformans, H99γ, are associated with IL-17A production by neutrophils. Neutrophil depletion in H99γ-immunized mice did not affect pulmonary fungal burden, indicating that neutrophils are not required for clearance. However, we observed an increase in IL-17A in the lungs of neutrophil-depleted H99γ infected mice, which corresponded to an increase in IL-17A+ γδ+ T cells. Moreover, we observed increased IL-17A+/ CD3+ cells and IL-17A+/γδ+ cells, but decreased IL-17A+/Ly6G+ neutrophils in the lungs of IL-17 receptor (R)A deficient mice compared to wild-type mice. Increased production of IL-17A in neutropenic mice coincided with increased IL-6 and CXCL1, but not Th17 inducing cytokines TGF-β, IL-21 and IL-23. Concurrent depletion of neutrophils and γδ+ T cells reduced IL-17A levels. Our results suggest that γδ+ T cells mediate significant IL-17A production in neutropenic mice during the protective response to C. neoformans infection.
Collapse
|
40
|
Hage CA, Horan DJ, Durkin M, Connolly P, Desta Z, Skaar TC, Knox KS, Wheat LJ. Histoplasma capsulatum preferentially induces IDO in the lung. Med Mycol 2012. [PMID: 23181600 DOI: 10.3109/13693786.2012.710857] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Indoleamine 2,3 dioxygenase (IDO) plays an important role in immunoregulation as it is involved in downregulating immune responses to infections. We sought to characterize IDO activity in histoplasmosis and to do so, C57Bl6 mice were infected intranasally with Histoplasma capsulatum. After infection, lung and spleen IDO activity was assessed by HPLC and IDO expression by qRT-PCR. The distribution of IDO was determined by immunohistochemical staining. Cytokine levels were measured in lung and spleen homogenates using cytokine bead array. Fungal burden was quantified by culture. Subcutaneous pellets containing methyltryptophane (1-MT) were employed to inhibit IDO in vivo. Histoplasma infection strongly induced functional lung IDO, with activity at its highest at weeks 1 and 2 and then decreased thereafter as the mice cleared the infection. Lung IDO activity positively correlated with the fungal burden (Rho = 0.845), interferon-γ (Rho = 0.839) and tumor necrosis factor-α (Rho = 0.791) levels, P < 0.001. In contrast, spleen IDO activity was not induced despite high infection burden and cytokine levels. IDO expressing cells were predominately located at the ring edge of Histoplasma-induced granulomas. IDO inhibition prior to infection reduced fungal burdens and inflammation in lungs and spleen. Histoplasma preferentially induces lung IDO, as early as one week after infection. IDO appears to modulate the immune response to Histoplasma infection.
Collapse
Affiliation(s)
- Chadi A Hage
- Indiana University School of Medicine and Roudebush Veterans' Administration Medical Center, Pulmonary Critical Care Medicine, Indianapolis, Indiana 46202, USA. @iupui.edu
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Munn DH, Mellor AL. Indoleamine 2,3 dioxygenase and metabolic control of immune responses. Trends Immunol 2012; 34:137-43. [PMID: 23103127 DOI: 10.1016/j.it.2012.10.001] [Citation(s) in RCA: 768] [Impact Index Per Article: 59.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2012] [Revised: 09/18/2012] [Accepted: 10/01/2012] [Indexed: 02/07/2023]
Abstract
Sustained access to nutrients is a fundamental biological need, especially for proliferating cells, and controlling nutrient supply is an ancient strategy to regulate cellular responses to stimuli. By catabolizing the essential amino acid TRP, cells expressing the enzyme indoleamine 2,3 dioxygenase (IDO) can mediate potent local effects on innate and adaptive immune responses to inflammatory insults. Here, we discuss recent progress in elucidating how IDO activity promotes local metabolic changes that impact cellular and systemic responses to inflammatory and immunological signals. These recent developments identify potential new targets for therapy in a range of clinical settings, including cancer, chronic infections, autoimmune and allergic syndromes, and transplantation.
Collapse
Affiliation(s)
- David H Munn
- Department of Pediatrics, Cancer Immunology, Inflammation and Tolerance Program, Cancer Center, Georgia Health Sciences University, Augusta GA 30912, USA
| | | |
Collapse
|
42
|
Regulatory T cells in HIV infection: can immunotherapy regulate the regulator? Clin Dev Immunol 2012; 2012:908314. [PMID: 23251223 PMCID: PMC3509372 DOI: 10.1155/2012/908314] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2012] [Accepted: 08/28/2012] [Indexed: 12/25/2022]
Abstract
Regulatory T cells (Tregs) have a dominant role in self-tolerance and control of autoimmune diseases. These cells also play a pivotal role in chronic viral infections and cancer by limiting immune activation and specific immune response. The role of Tregs in HIV pathogenesis remains poorly understood as their function, changes according to the phases of infection. Tregs can suppress anti-HIV specific responses and conversely can have a beneficial role by reducing the deleterious impact of immune activation. We review the frequency, function and homing potential of Tregs in the blood and lymphoid tissues as well as their interaction with dendritic cells in the context of HIV infection. We also examine the new insights generated by recombinant IL-2 and IL-7 clinical trials in HIV-infected adults, including the immunomodulatory effects of Tregs. Based on their detrimental role in limiting anti-HIV responses, we propose Tregs as potential targets for immunotherapeutic strategies aimed at decreasing Tregs frequency and/or immunosuppressive function. However, such approaches require a better understanding of the time upon infection when interfering with Treg function may not cause a deleterious state of hyperimmune activation.
Collapse
|
43
|
Carvalho A, Cunha C, Bozza S, Moretti S, Massi-Benedetti C, Bistoni F, Aversa F, Romani L. Immunity and tolerance to fungi in hematopoietic transplantation: principles and perspectives. Front Immunol 2012; 3:156. [PMID: 22707953 PMCID: PMC3374351 DOI: 10.3389/fimmu.2012.00156] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2012] [Accepted: 05/25/2012] [Indexed: 12/30/2022] Open
Abstract
Resistance and tolerance are two complementary host defense mechanisms that increase fitness in response to low-virulence fungi. Resistance is meant to reduce pathogen burden during infection through innate and adaptive immune mechanisms, whereas tolerance mitigates the substantial cost of resistance to host fitness through a multitude of anti-inflammatory mechanisms, including immunological tolerance. In experimental fungal infections, both defense mechanisms are activated through the delicate equilibrium between Th1/Th17 cells, which provide antifungal resistance, and regulatory T cells limiting the consequences of the ensuing inflammatory pathology. Indoleamine 2,3-dioxygenase (IDO), a rate-limiting enzyme in the tryptophan catabolism, plays a key role in induction of tolerance against fungi. Both hematopoietic and non-hematopoietic compartments contribute to the resistance/tolerance balance against Aspergillus fumigatus via the involvement of selected innate receptors converging on IDO. Several genetic polymorphisms in pattern recognition receptors influence resistance and tolerance to fungal infections in human hematopoietic transplantation. Thus, tolerance mechanisms may be exploited for novel diagnostics and therapeutics against fungal infections and diseases.
Collapse
Affiliation(s)
- Agostinho Carvalho
- Department of Experimental Medicine and Biochemical Sciences, University of Perugia Perugia, Italy
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Blumenthal A, Nagalingam G, Huch JH, Walker L, Guillemin GJ, Smythe GA, Ehrt S, Britton WJ, Saunders BM. M. tuberculosis induces potent activation of IDO-1, but this is not essential for the immunological control of infection. PLoS One 2012; 7:e37314. [PMID: 22649518 PMCID: PMC3359358 DOI: 10.1371/journal.pone.0037314] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2012] [Accepted: 04/18/2012] [Indexed: 11/18/2022] Open
Abstract
Indoleamine 2,3-dioxygenesae-1 (IDO-1) catalyses the initial, rate-limiting step in tryptophan metabolism, thereby regulating tryptophan availability and the formation of downstream metabolites, including picolinic and quinolinic acid. We found that Mycobacterium tuberculosis infection induced marked upregulation of IDO-1 expression in both human and murine macrophages in vitro and in the lungs of mice following aerosol challenge with M. tuberculosis. The absence of IDO-1 in dendritic cells enhanced the activation of mycobacteria-specific T cells in vitro. Interestingly, IDO-1-deficiency during M. tuberculosis infection in mice was not associated with altered mycobacteria-specific T cell responses in vivo. The bacterial burden of infected organs, pulmonary inflammatory responses, and survival were also comparable in M. tuberculosis-infected IDO-1 deficient and wild type animals. Tryptophan is metabolised into either picolinic acid or quinolinic acid, but only picolinic acid inhibited the growth of M. tuberculosis in vitro. By contrast macrophages infected with pathogenic mycobacteria, produced quinolinic, rather than picolinic acid, which did not reduce M. tuberculosis growth in vitro. Therefore, although M. tuberculosis induces robust expression of IDO-1 and activation of tryptophan metabolism, IDO-1-deficiency fails to impact on the immune control and the outcome of the infection in the mouse model of tuberculosis.
Collapse
Affiliation(s)
- Antje Blumenthal
- Department of Microbiology and Immunology, Weill Cornell Medical College, New York, New York, United States of America
- The University of Queensland Diamantina Institute, Brisbane, Queensland, Australia
- * E-mail: (AB); (BMS)
| | - Gayathri Nagalingam
- Mycobacterial Research Program, Centenary Institute Sydney, New South Wales, Australia
| | - Jennifer H. Huch
- Mycobacterial Research Program, Centenary Institute Sydney, New South Wales, Australia
| | - Lara Walker
- Mycobacterial Research Program, Centenary Institute Sydney, New South Wales, Australia
| | - Gilles J. Guillemin
- Department of Pharmacology, School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - George A. Smythe
- Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Sabine Ehrt
- Department of Microbiology and Immunology, Weill Cornell Medical College, New York, New York, United States of America
- Program in Immunology and Microbial Pathogenesis, Weill Graduate School of Medical Sciences of Cornell University, New York, New York, United States of America
| | - Warwick J. Britton
- The University of Queensland Diamantina Institute, Brisbane, Queensland, Australia
- Department of Medicine, University of Sydney, Sydney, New South Wales, Australia
| | - Bernadette M. Saunders
- The University of Queensland Diamantina Institute, Brisbane, Queensland, Australia
- Department of Medicine, University of Sydney, Sydney, New South Wales, Australia
- * E-mail: (AB); (BMS)
| |
Collapse
|
45
|
Iken K, Liu K, Liu H, Bizargity P, Wang L, Hancock WW, Visner GA. Indoleamine 2,3-dioxygenase and metabolites protect murine lung allografts and impair the calcium mobilization of T cells. Am J Respir Cell Mol Biol 2012; 47:405-16. [PMID: 22517796 DOI: 10.1165/rcmb.2011-0438oc] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The enzyme indoleamine 2,3-dioxygenase (IDO) converts tryptophan into kynurenine metabolites that suppress effector T-cell function. In this study, we investigated IDO and its metabolite, 3-hydroxyanthranilic acid (3HAA), in regulating lung allograft rejection, using a murine orthotopic lung transplant model with a major mismatch (BALB/c donor and C57BL6 recipient). IDO was overexpressed in murine donor lungs, using an established nonviral (polyethylenimine carrier)-based gene transfer approach, whereas 3HAA was delivered daily via intraperitoneal injection. Increased IDO expression or its metabolite, 3HAA, resulted in a remarkable therapeutic effect with near normal lung function and little acute rejection, approximately A1, compared with A3 in untreated allografts (grading based on International Society for Heart and Lung Transplantation guidelines). We found that a high IDO environment for 7 days in lung allografts resulted in impaired T-cell activation, the production of multiple effector cytokines (IL-2, IL-4, IL-5, IL-6, IFN-γ, TNF-α, IL-12, and IL-13), and the generation of effector memory T cells (CD62L(lo)CD44(hi) phenotype). In isolated murine splenocytes, we observed that IDO/3HAA impaired T-cell receptor (TCR)-mediated T-cell activation, and more importantly, a decrease of intracellular calcium, phospholipase C-γ1 phosphorylation, and mitochondrial mass was evident. This work further illustrates the potential role of a high IDO environment in lung transplantation, and that the high IDO environment directly impairs TCR activation via the disruption of calcium signaling.
Collapse
Affiliation(s)
- Khadija Iken
- Division of Pulmonary Medicine, Children's Hospital Boston, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | | | |
Collapse
|
46
|
Contribution of Bordetella filamentous hemagglutinin and adenylate cyclase toxin to suppression and evasion of interleukin-17-mediated inflammation. Infect Immun 2012; 80:2061-75. [PMID: 22473603 DOI: 10.1128/iai.00148-12] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Bordetella pertussis and Bordetella bronchiseptica establish respiratory infections with notorious efficiency. Our previous studies showed that the fhaB genes of B. pertussis and B. bronchiseptica, which encode filamentous hemagglutinin (FHA), are functionally interchangeable and provided evidence that FHA-deficient B. bronchiseptica induces more inflammation in the lungs of mice than wild-type B. bronchiseptica. We show here that the robust inflammatory response to FHA-deficient B. bronchiseptica is characterized by the early and sustained influx of interleukin-17 (IL-17)-positive neutrophils and macrophages and, at 72 h postinoculation, IL-17-positive CD4(+) T cells, suggesting that FHA allows the bacteria to suppress the development of an IL-17-mediated inflammatory response. We also show that the cyaA genes of B. pertussis and B. bronchiseptica, which encode adenylate cyclase toxin (ACT), are functionally interchangeable and that ACT, specifically its catalytic activity, is required for B. bronchiseptica to resist phagocytic clearance but is neither required for nor inhibitory of the induction of inflammation if bacteria are present in numbers sufficient to persist during the first 3 days postinoculation. Incubation of bone marrow-derived macrophages with a ΔcyaA strain caused decreased production of IL-1β and increased production of tumor necrosis factor alpha (TNF-α) and IL-12, while incubation with a ΔcyaA ΔfhaB strain caused increased production of IL-23. These data suggest that FHA and ACT both contribute to suppress the recruitment of neutrophils and the development of an IL-17-mediated immune response. To our knowledge, this is the first demonstration of a microbial pathogen suppressing IL-17-mediated inflammation in vivo as a strategy to evade innate immunity.
Collapse
|
47
|
Interleukin-17 drives pulmonary eosinophilia following repeated exposure to Aspergillus fumigatus conidia. Infect Immun 2012; 80:1424-36. [PMID: 22252873 DOI: 10.1128/iai.05529-11] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Previous research in our laboratory has demonstrated that repeated intranasal exposure to Aspergillus fumigatus conidia in C57BL/6 mice results in a chronic pulmonary inflammatory response that reaches its maximal level after four challenges. The inflammatory response is characterized by eosinophilia, goblet cell metaplasia, and T helper T(H)2 cytokine production, which is accompanied by sustained interleukin-17 (IL-17) expression that persists even after the T(H)2 response has begun to resolve. T(H)17 cells could develop in mice deficient in gamma interferon (IFN-γ), IL-4, or IL-10. In the lungs of IL-17 knockout mice repeatedly challenged with A. fumigatus conidia, inflammation was attenuated (with the most significant decrease occurring in eosinophils), conidial clearance was enhanced, and the early transient peak of CD4(+) CD25(+) FoxP3(+) cells blunted. IL-17 appeared to play only a minor role in eosinophil differentiation in the bone marrow but a central role in eosinophil extravasation from the blood into the lungs. These observations point to an expanded role for IL-17 in driving T(H)2-type inflammation to repeated inhalation of fungal conidia.
Collapse
|
48
|
Optimizing a photoallodepletion protocol for adoptive immunotherapy after haploidentical SCT. Bone Marrow Transplant 2011; 47:1196-200. [PMID: 22139067 DOI: 10.1038/bmt.2011.237] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
In adults, one-haplotype-mismatched haematopoietic SCT (haploidentical HSCT) is associated with slow immune recovery due to decaying thymic function and extensive T-cell depletion of the graft. Although essential for preventing GVHD, T-cell depletion underlies the major reasons for transplant failure: leukemia relapse and infections, with infection-related mortality accounting for about 40% of non-leukemic deaths. Adoptive T-cell therapy would be helpful for these patients but to administer it without causing GVHD, alloreactive T cells need to be eliminated from donor T lymphocytes before infusion. In a preclinical study, to address this problem, we determined the efficacy of photodynamic purging of alloreactive T cells, by investigating combinations of parameters in order to achieve maximum allodepletion, preservation of T-regulatory cells and of pathogen and leukemia-specific T-cell responses in donor-vs-recipient MLR. We also needed to identify an optimal method to quantify the Ag-specific T-cell repertoires. Optimal procedures were identified. In particular, we compared limiting-dilution analyses (LDA) of proliferating T cells with H(3)-thymidine incorporation by bulk T cells and with flow cytometry CD25 expression, which is accepted as a T-cell activation marker. This study demonstrated that LDA is a reliable, predictable and sensitive method for measuring alloreactive, pathogen- and leukemia-specific T-cell frequencies.
Collapse
|
49
|
Divanovic S, Sawtell NM, Trompette A, Warning JI, Dias A, Cooper AM, Yap GS, Arditi M, Shimada K, Duhadaway JB, Prendergast GC, Basaraba RJ, Mellor AL, Munn DH, Aliberti J, Karp CL. Opposing biological functions of tryptophan catabolizing enzymes during intracellular infection. J Infect Dis 2011; 205:152-61. [PMID: 21990421 DOI: 10.1093/infdis/jir621] [Citation(s) in RCA: 94] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Recent studies have underscored physiological and pathophysiological roles for the tryptophan-degrading enzyme indolamine 2,3-dioxygenase (IDO) in immune counterregulation. However, IDO was first recognized as an antimicrobial effector, restricting tryptophan availability to Toxoplasma gondii and other pathogens in vitro. The biological relevance of these findings came under question when infectious phenotypes were not forthcoming in IDO-deficient mice. The recent discovery of an IDO homolog, IDO-2, suggested that the issue deserved reexamination. IDO inhibition during murine toxoplasmosis led to 100% mortality, with increased parasite burdens and no evident effects on the immune response. Similar studies revealed a counterregulatory role for IDO during leishmaniasis (restraining effector immune responses and parasite clearance), and no evident role for IDO in herpes simplex virus type 1 (HSV-1) infection. Thus, IDO plays biologically important roles in the host response to diverse intracellular infections, but the dominant nature of this role--antimicrobial or immunoregulatory--is pathogen-specific.
Collapse
Affiliation(s)
- Senad Divanovic
- Division of Molecular Immunology, Cincinnati Children's Hospital Research Foundation, and the University of Cincinnati College of Medicine, Ohio, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Zwilling D, Huang SY, Sathyasaikumar KV, Notarangelo FM, Guidetti P, Wu HQ, Lee J, Truong J, Andrews-Zwilling Y, Hsieh EW, Louie JY, Wu T, Scearce-Levie K, Patrick C, Adame A, Giorgini F, Moussaoui S, Laue G, Rassoulpour A, Flik G, Huang Y, Muchowski JM, Masliah E, Schwarcz R, Muchowski PJ. Kynurenine 3-monooxygenase inhibition in blood ameliorates neurodegeneration. Cell 2011; 145:863-74. [PMID: 21640374 DOI: 10.1016/j.cell.2011.05.020] [Citation(s) in RCA: 314] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2011] [Revised: 04/30/2011] [Accepted: 05/12/2011] [Indexed: 10/18/2022]
Abstract
Metabolites in the kynurenine pathway, generated by tryptophan degradation, are thought to play an important role in neurodegenerative disorders, including Alzheimer's and Huntington's diseases. In these disorders, glutamate receptor-mediated excitotoxicity and free radical formation have been correlated with decreased levels of the neuroprotective metabolite kynurenic acid. Here, we describe the synthesis and characterization of JM6, a small-molecule prodrug inhibitor of kynurenine 3-monooxygenase (KMO). Chronic oral administration of JM6 inhibits KMO in the blood, increasing kynurenic acid levels and reducing extracellular glutamate in the brain. In a transgenic mouse model of Alzheimer's disease, JM6 prevents spatial memory deficits, anxiety-related behavior, and synaptic loss. JM6 also extends life span, prevents synaptic loss, and decreases microglial activation in a mouse model of Huntington's disease. These findings support a critical link between tryptophan metabolism in the blood and neurodegeneration, and they provide a foundation for treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Daniel Zwilling
- Gladstone Institute of Neurological Disease, University of California, San Francisco, San Francisco, CA 94158, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|