1
|
Liu B, Shen J, Li J, Tian B, Zhou B, Gui J, Li Z, Zhang Y, Hu W, Li Q. Candidate approaches for predicting vitiligo recurrence: an effective model and biomarkers. Front Immunol 2025; 16:1468665. [PMID: 39981245 PMCID: PMC11839629 DOI: 10.3389/fimmu.2025.1468665] [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: 07/22/2024] [Accepted: 01/21/2025] [Indexed: 02/22/2025] Open
Abstract
Background Vitiligo is a challenging chronic condition with unpredictable disease course and high propensity for relapse post-treatment. Recent studies have reported the biomarkers for disease activity, severity, and therapeutic response of vitiligo, yet very few have investigated cytokines as predictive biomarkers for disease recurrence in vitiligo. This study aims to explore cytokines that serve as biomarkers for disease recurrence and extend research on factors related to the disease's activity. Methods 92 patients and 40 healthy controls were recruited at the Air Force Medical Center from September 20, 2023, to November 30, 2023. Ultrasensitive multiplex cytokine array was used to measure plasma concentrations of cytokines, including IFN-γ, CXCL9, CXCL10, CXCL11, IL-6, and IL-15. Results IFN-γ, CXCL9, CXCL10, CXCL11, IL-6, and IL-15 were expressed at higher levels in the circulation of patients with both segmental and non-segmental vitiligo compared to healthy controls (p < 0.001). There were no significant differences in these cytokine levels between the two types of vitiligo. CXCL9 was associated with the activity of vitiligo (p = 0.027). Correlation analysis showed a positive relationship between IFN-γ, CXCL9, CXCL10, CXCL11, IL-6, and IL-15 in the plasma of patients with recurrent vitiligo. The expression of IFN-γ, CXCL9, CXCL10, CXCL11, and IL-6 was significantly higher in recurrent vitiligo than in cases of persistent stable vitiligo (p = 0.001, p = 0.003, p < 0.001, p = 0.002, p = 0.026, respectively), with ROC analysis demonstrating their predictive capability for vitiligo recurrence, with AUC values of 0.806, 0.773, 0.896, 0.785, and 0.709, respectively. Multivariate logistic regression model showed IFN-γ is an independent predictor for vitiligo recurrence [OR (95%CI) =1.051 (1.012~1.116)], with a prediction accuracy of 90.5% (38/42) on the training dataset and 88.9% (16/18) on the testing dataset. Conclusion Plasma IFN-γ, CXCL9, CXCL10, CXCL11 and IL-6 might be potential biomarkers for vitiligo recurrence, with CXCL9 also associated with disease activity. Additionally, multivariate logistic regression model demonstrated that IFN-γ is an independent predictor of vitiligo recurrence and the model could be a candidate approach for predicting vitiligo recurrence.
Collapse
Affiliation(s)
- Binhao Liu
- The Air Force Clinical College, Anhui Medical University, Hefei, China
| | - Jiacheng Shen
- The Air Force Clinical College, Anhui Medical University, Hefei, China
| | - Jiayu Li
- Department of Anesthesiology, Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Bowen Tian
- Graduate School, Air Force Medical University, Xi’an, China
- Department of Dermatology, 960th Hospital of the People's Liberation Army of China (PLA), Jinan, China
| | - Bin Zhou
- Graduate School, Hebei North University, Zhangjiakou, China
| | - Jiachen Gui
- Graduate School, Air Force Medical University, Xi’an, China
| | - Zhimin Li
- Department of Dermatology, Air Force Medical Center, People's Liberation Army of China (PLA), Beijing, China
| | - Yue Zhang
- Department of Dermatology, Air Force Medical Center, People's Liberation Army of China (PLA), Beijing, China
| | - Wenzhi Hu
- Department of Burn and Plastic Surgery, Air Force Medical Center, People's Liberation Army of China (PLA), Beijing, China
| | - Qiang Li
- The Air Force Clinical College, Anhui Medical University, Hefei, China
- Department of Dermatology, Air Force Medical Center, People's Liberation Army of China (PLA), Beijing, China
| |
Collapse
|
2
|
Abstract
PURPOSE OF REVIEW The mounting body of evidence underscores the pivotal role of interferon gamma (IFNγ) in the pathogenesis of hypertension, prompting exploration of the mechanisms by which this cytokine fosters a pro-inflammatory immune milieu, subsequently exacerbating hypertension. In this review, we delve into recent preclinical and clinical studies from the past two years to elucidate how IFNγ participates in the progression of hypertension. RECENT FINDINGS IFNγ promotes renal CD8 + T cell accumulation by upregulating tubular PDL1 and MHC-I, intensifying cell-to-cell interaction. Intriguingly, a nucleotide polymorphism in LNK, predisposing towards hypertension, correlates with augmented T cell IFNγ production. Additionally, anti-IFNγ treatment exhibits protective effects against T cell-mediated inflammation during angiotensin II infusion or transverse aortic constriction. Moreover, knockout of the mineralocorticoid receptor in T cells protects against cardiac dysfunction induced by myocardial infarction, correlating with reduced IFNγ and IL-6, decreased macrophage recruitment, and attenuated fibrosis. Interestingly, increased IFNγ production correlates with elevated blood pressure, impacting individuals with type 2 diabetes, nondiabetics, and obese hypertensive patients. SUMMARY These revelations spotlight IFNγ as the critical mediator bridging the initial phase of blood pressure elevation with the sustained and exacerbated pathology. Consequently, blocking IFNγ signaling emerges as a promising therapeutic target to improve the management of this 'silent killer.'
Collapse
Affiliation(s)
- Lance N. Benson
- Heersink School of Medicine: Department of CardioRenal Physiology and Medicine, Division of Nephrology University of Alabama at Birmingham, Birmingham, Alabama
| | - Shengyu Mu
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| |
Collapse
|
3
|
Shim S, Ha S, Choi J, Kwon HK, Cheon KA. Alterations in Plasma Cytokine Levels in Korean Children with Autism Spectrum Disorder. Yonsei Med J 2024; 65:70-77. [PMID: 38288647 PMCID: PMC10827638 DOI: 10.3349/ymj.2023.0362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 10/11/2023] [Accepted: 10/25/2023] [Indexed: 02/01/2024] Open
Abstract
PURPOSE Numerous studies have supported the role of the immune dysfunction in the pathogenesis of autism spectrum disorder (ASD); however, to our knowledge, no study has been conducted on plasma cytokine levels in children with ASD in South Korea. In this study, we aimed to analyze the immunological characteristics of Korean children with ASD through plasma cytokine analysis. MATERIALS AND METHODS Blood samples were collected from 94 ASD children (mean age 7.1; 81 males and 13 females) and 48 typically developing children (TDC) (mean age 7.3; 30 males and 18 females). Plasma was isolated from 1 mL of blood by clarifying with centrifugation at 8000 rpm at 4℃ for 10 min. Cytokines in plasma were measured with LEGENDplex HU Th cytokine panel (BioLegend, 741028) and LEGENDplex HU cytokine panel 2 (BioLegend, 740102). RESULTS Among 25 cytokines, innate immune cytokine [interleukin (IL)-33] was significantly decreased in ASD children compared with TDC. In acute phase proteins, tumor necrosis factor α (TNF-α) was significantly increased, while IL-6, another inflammation marker, was decreased in ASD children compared with TDC. The cytokines from T cell subsets, including interferon (IFN)-γ, IL-5, IL-13, and IL-17f, were significantly decreased in ASD children compared to TDC. IL-10, a major anti-inflammatory cytokine, and IL-9, which modulates immune cell growth and proliferation, were also significantly decreased in ASD children compared to TDC. CONCLUSION We confirmed that Korean children with ASD showed altered immune function and unique cytokine expression patterns distinct from TDC.
Collapse
Affiliation(s)
- Songjoo Shim
- Department of Psychiatry, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Sungji Ha
- Department of Psychiatry, Institute of Behavioral Science in Medicine, Yonsei University College of Medicine, Seoul, Korea.
| | - Juli Choi
- Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul, Korea
- Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, Korea.
| | - Ho-Keun Kwon
- Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul, Korea
- Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, Korea
- Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Korea
| | - Keun-Ah Cheon
- Department of Psychiatry, Institute of Behavioral Science in Medicine, Yonsei University College of Medicine, Seoul, Korea
- Department of Child and Adolescent Psychiatry, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
4
|
Majerczyk D, Ayad E, Brewton K, Saing P, Hart P. Systemic maternal inflammation promotes ASD via IL-6 and IFN-γ. Biosci Rep 2022; 42:BSR20220713. [PMID: 36300375 PMCID: PMC9670245 DOI: 10.1042/bsr20220713] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 09/30/2022] [Accepted: 10/26/2022] [Indexed: 07/25/2023] Open
Abstract
Autism spectrum disorder (ASD) is a neurological disorder that manifests during early development, impacting individuals through their ways of communicating, social behaviors, and their ability to perform day-to-day activities. There have been different proposed mechanisms on how ASD precipitates within a patient, one of which being the impact cytokines have on fetal development once a mother's immune system has been activated (referred to as maternal immune activation, MIA). The occurrence of ASD has long been associated with elevated levels of several cytokines, including interleukin-6 (IL-6) and interferon gamma (IFN-γ). These proinflammatory cytokines can achieve high systemic levels in response to immune activating pathogens from various extrinsic sources. Transfer of cytokines such as IL-6 across the placental barrier allows accumulation in the fetus, potentially inducing neuroinflammation and consequently altering neurodevelopmental processes. Individuals who have been later diagnosed with ASD have been observed to have elevated levels of IL-6 and other proinflammatory cytokines during gestation. Moreover, the outcome of MIA has been associated with neurological effects such as impaired social interaction and an increase in repetitive behavior in animal models, supporting a mechanistic link between gestational inflammation and development of ASD-like characteristics. The present review attempts to provide a concise overview of the available preclinical and clinical data that suggest cross-talk between IL-6 and IFN-γ through both extrinsic and intrinsic factors as a central mechanism of MIA that may promote the development of ASD.
Collapse
Affiliation(s)
- Daniel Majerczyk
- College of Science, Health and Pharmacy, Roosevelt University, Illinois 60173, U.S.A
- Loyola Medicine, Berwyn, Illinois 60402, U.S.A
| | - Elizabeth G. Ayad
- College of Science, Health and Pharmacy, Roosevelt University, Illinois 60173, U.S.A
| | - Kari L. Brewton
- College of Science, Health and Pharmacy, Roosevelt University, Illinois 60173, U.S.A
| | - Pichrasmei Saing
- College of Science, Health and Pharmacy, Roosevelt University, Illinois 60173, U.S.A
| | - Peter C. Hart
- College of Science, Health and Pharmacy, Roosevelt University, Illinois 60173, U.S.A
| |
Collapse
|
5
|
Fitoussi R, Faure MO, Beauchef G, Achard S. Human skin responses to environmental pollutants: A review of current scientific models. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2022; 306:119316. [PMID: 35469928 DOI: 10.1016/j.envpol.2022.119316] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Revised: 04/11/2022] [Accepted: 04/14/2022] [Indexed: 06/14/2023]
Abstract
Whatever the exposure route, chemical, physical and biological pollutants modify the whole organism response, leading to nerve, cardiac, respiratory, reproductive, and skin system pathologies. Skin acts as a barrier for preventing pollutant modifications. This review aims to present the available scientific models, which help investigate the impact of pollution on the skin. The research question was "Which experimental models illustrate the impact of pollution on the skin in humans?" The review covered a period of 10 years following a PECO statement on in vitro, ex vivo, in vivo and in silico models. Of 582 retrieved articles, 118 articles were eligible. In oral and inhalation routes, dermal exposure had an important impact at both local and systemic levels. Healthy skin models included primary cells, cell lines, co-cultures, reconstructed human epidermis, and skin explants. In silico models estimated skin exposure and permeability. All pollutants affected the skin by altering elasticity, thickness, the structure of epidermal barrier strength, and dermal extracellular integrity. Some specific models concerned wound healing or the skin aging process. Underlying mechanisms were an exacerbated inflammatory skin reaction with the modulation of several cytokines and oxidative stress responses, ending with apoptosis. Pathological skin models revealed the consequences of environmental pollutants on psoriasis, atopic dermatitis, and tumour development. Finally, scientific models were used for evaluating the safety and efficacy of potential skin formulations in preventing the skin aging process or skin irritation after repeated contact. The review gives an overview of scientific skin models used to assess the effects of pollutants. Chemical and physical pollutants were mainly represented while biological contaminants were little studied. In future developments, cell hypoxia and microbiota models may be considered as more representative of clinical situations. Models considering humidity and temperature variations may reflect the impact of these changes.
Collapse
Affiliation(s)
| | - Marie-Odile Faure
- Scientific Consulting For You, 266 avenue Daumesnil, 75012, PARIS, France
| | | | - Sophie Achard
- HERA Team (Health Environmental Risk Assessment), INSERM UMR1153, CRESS-INRAE, Université Paris Cité, Faculté de Pharmacie, 4 avenue de l'Observatoire, 75270 CEDEX 06, PARIS, France.
| |
Collapse
|
6
|
Anka AU, Usman AB, Kaoje AN, Kabir RM, Bala A, Kazem Arki M, Hossein-Khannazer N, Azizi G. Potential mechanisms of some selected heavy metals in the induction of inflammation and autoimmunity. EUR J INFLAMM 2022. [DOI: 10.1177/1721727x221122719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Inflammation is a physiological event that protects tissues from infection and injury. Chronic inflammation causes immune cell over activation and sustained release of inflammatory cytokines and chemokines cause pathologic conditions including autoimmune diseases. Heavy metals exposure affects innate and adaptive immune systems through triggering inflammatory responses. It seems that extended inflammatory responses could accelerate heavy metal-induced autoimmunity. In the present review we discuss the exposure route and toxicity of Cadmium (Cd), Lead (Pb), Mercury (Hg), Vanadium (V) and Platinum (Pt) and their effects on inflammatory responses by innate and adaptive immune system and autoimmunity.
Collapse
Affiliation(s)
- Abubakar U Anka
- Department of Medical Laboratory Science, College of Medical Sciences, Ahmadu Bello University, Zaria, Nigeria
| | - Abubakar B Usman
- Department of Immunology, School of Medical Laboratory Sciences, Usmanu Danfodiyo University, Sokoto, Nigeria
| | - Abubakar N Kaoje
- Department of Health Services, Federal University Birnin Kebbi, Birnin Kebbi, Nigeria
| | - Ramadan M Kabir
- Laboratory Department, Murtala Muhammad Specialist Hospital, Kano, Nigeria
| | - Aliyu Bala
- Hematology Department, Federal Medical Center, Katsina, Nigeria
| | - Mandana Kazem Arki
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nikoo Hossein-Khannazer
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Gholamreza Azizi
- Non-Communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
- Research Center for Immunodeficiencies, Children’s Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
7
|
Which Therapy for Non-Type(T)2/T2-Low Asthma. J Pers Med 2021; 12:jpm12010010. [PMID: 35055325 PMCID: PMC8779705 DOI: 10.3390/jpm12010010] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 12/13/2021] [Accepted: 12/22/2021] [Indexed: 12/17/2022] Open
Abstract
Currently, the asthmatic population is divided into Type 2-high and non-Type 2/Type 2-low asthmatics, with 50% of patients belonging to one of the two groups. Differently from T2-high, T2-low asthma has not been clearly defined yet, and the T2-low patients are identified on the basis of the absence or non-predominant expression of T2-high biomarkers. The information about the molecular mechanisms underpinning T2-low asthma is scarce, but researchers have recognized as T2-low endotypes type 1 and type 3 immune response, and remodeling events occurring without inflammatory processes. In addition, the lack of agreed biomarkers reprents a challenge for the research of an effective therapy. The first-choice medication is represented by inhaled corticosteroids despite a low efficacy is reported for/in T2-low patients. However, macrolides and long-acting anti-muscarinic drugs have been recognized as efficacious. In recent years, clinical trials targeting biomarkers playing key roles in T3 and T1 immune pathways, alarmins, and molecules involved in neutrophil recruitment have provided conflicting results probably misleading (or biased) in patients' selection. However, further studies are warranted to achieve a precise characterization of T2-low asthma with the aim of defining a tailored therapy for each single asthmatic patient.
Collapse
|
8
|
Lefferts AR, Regner EH, Stahly A, O'Rourke B, Gerich ME, Fennimore BP, Scott FI, Freeman AE, Jones K, Kuhn KA. Circulating mature granzyme B+ T cells distinguish Crohn's disease-associated axial spondyloarthritis from axial spondyloarthritis and Crohn's disease. Arthritis Res Ther 2021; 23:147. [PMID: 34022940 PMCID: PMC8140495 DOI: 10.1186/s13075-021-02531-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 05/12/2021] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Axial spondyloarthritis (axSpA) has strong connections with intestinal inflammation as occurs in Crohn's disease (CD). However, the immunologic mechanisms that distinguish axSpA, CD, and those with features of both diseases (CD-axSpA) are unknown. This study aimed to address this question by initial unbiased single cell RNA-sequencing (scRNAseq) on a pilot cohort followed by validating findings using flow cytometry and ELISA in a larger cohort. METHODS Two individuals each with CD, axSpA, CD-axSpA, and healthy controls (HC) were recruited for a pilot discovery scRNAseq cohort, and the validation cohort consisted of 18 axSpA, 24 CD, 13 CD-axSpA, and 17 HC that was evaluated by flow cytometry on PBMCs and ELISAs for plasma cytokines. RESULTS Uniquely, PBMCs from subjects with CD-axSpA demonstrated a significant increase in granzyme B+ T cells of both CD4+ and CD8+ lineages by both scRNAseq and flow cytometry. T cell maturation was also greater in those with CD-axSpA, particularly the CD4+ granzyme B+ population. Pathway analysis suggested increased interferon response genes in all immune cell populations within CD-axSpA. Although IFN-γ was elevated in the plasma of a subset of subjects with CD-axSpA, IL-6 was also significantly elevated. CONCLUSIONS Our findings support the presence of a chronic interferonopathy in subjects with CD-axSpA characterized by interferon signaling by pathway analysis and an expansion of mature, cytotoxic T cells. These data indicate fundamental immunological differences between CD-axSpA and both of the putative "parent" conditions, suggesting that it is a distinct disease with unique natural history and treatment needs.
Collapse
Affiliation(s)
- Adam R Lefferts
- Division of Rheumatology, Department of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Emilie H Regner
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
- Present Address: Division of Gastroenterology, Department of Medicine, Oregon Health Sciences University, Portland, OR, USA
| | - Andrew Stahly
- Division of Rheumatology, Department of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Becky O'Rourke
- Section of Pediatric Hematology/Oncology/Bone Marrow Transplant, Department of Pediatrics, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Mark E Gerich
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Blair P Fennimore
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Frank I Scott
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Alison E Freeman
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
- Present Address: Cascade Gastroenterology, Bend, OR, USA
| | - Ken Jones
- Section of Pediatric Hematology/Oncology/Bone Marrow Transplant, Department of Pediatrics, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
- Present Address: Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Kristine A Kuhn
- Division of Rheumatology, Department of Medicine, Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
9
|
Pollard KM, Cauvi DM, Mayeux JM, Toomey CB, Peiss AK, Hultman P, Kono DH. Mechanisms of Environment-Induced Autoimmunity. Annu Rev Pharmacol Toxicol 2020; 61:135-157. [PMID: 32857688 DOI: 10.1146/annurev-pharmtox-031320-111453] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Although numerous environmental exposures have been suggested as triggers for preclinical autoimmunity, only a few have been confidently linked to autoimmune diseases. For disease-associated exposures, the lung is a common site where chronic exposure results in cellular toxicity, tissue damage, inflammation, and fibrosis. These features are exacerbated by exposures to particulate material, which hampers clearance and degradation, thus facilitating persistent inflammation. Coincident with exposure and resulting pathological processes is the posttranslational modification of self-antigens, which, in concert with the formation of tertiary lymphoid structures containing abundant B cells, is thought to promote the generation of autoantibodies that in some instances demonstrate major histocompatibility complex restriction. Under appropriate gene-environment interactions, these responses can have diagnostic specificity. Greater insight into the molecular and cellular requirements governing this process, especially those that distinguish preclinical autoimmunity from clinical autoimmunedisease, may facilitate determination of the significance of environmental exposures in human autoimmune disease.
Collapse
Affiliation(s)
- K Michael Pollard
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California 92037, USA;
| | - David M Cauvi
- Department of Surgery, University of California San Diego School of Medicine, La Jolla, California 92093, USA
| | - Jessica M Mayeux
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California 92037, USA;
| | - Christopher B Toomey
- Department of Ophthalmology, University of California San Diego, La Jolla, California 92093, USA
| | - Amy K Peiss
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California 92037, USA;
| | - Per Hultman
- Departments of Clinical Pathology and Biomedical and Clinical Sciences, Linköping University, SE-581 85 Linköping, Sweden
| | - Dwight H Kono
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California 92037, USA
| |
Collapse
|
10
|
Grieco T, Porzia A, Paolino G, Chello C, Sernicola A, Faina V, Carnicelli G, Moliterni E, Mainiero F. IFN‐γ/IL‐6 and related cytokines in chronic spontaneous urticaria: evaluation of their pathogenetic role and changes during omalizumab therapy. Int J Dermatol 2020; 59:590-594. [DOI: 10.1111/ijd.14812] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 10/20/2019] [Accepted: 01/12/2020] [Indexed: 11/29/2022]
Affiliation(s)
- Teresa Grieco
- Dermatologic Clinic Sapienza University of Rome Rome Italy
| | - Alessandra Porzia
- Experimental Medicine Department La Sapienza University of Rome Rome Italy
| | - Giovanni Paolino
- Dermatologic Clinic Sapienza University of Rome Rome Italy
- Unit of Dermatology IRCCS San Raffaele Hospital Milan Italy
| | - Camilla Chello
- Dermatologic Clinic Sapienza University of Rome Rome Italy
| | | | | | | | | | - Fabrizio Mainiero
- Experimental Medicine Department La Sapienza University of Rome Rome Italy
| |
Collapse
|
11
|
Abstract
Until recently, autoimmune disease research has primarily been focused on elucidating the role of the adaptive immune system. In the past decade or so, the role of the innate immune system in the pathogenesis of autoimmunity has increasingly been realized. Recent findings have elucidated paradigm-shifting concepts, for example, the implications of "trained immunity" and a dysbiotic microbiome in the susceptibility of predisposed individuals to clinical autoimmunity. In addition, the application of modern technologies such as the quantum dot (Qdot) system and 'Omics' (e.g., genomics, proteomics, and metabolomics) data-processing tools has proven fruitful in revisiting mechanisms underlying autoimmune pathogenesis and in identifying novel therapeutic targets. This review highlights recent findings discussed at the American Autoimmune Related Disease Association (AARDA) 2019 colloquium. The findings covering autoimmune diseases and autoinflammatory diseases illustrate how new developments in common innate immune pathways can contribute to the better understanding and management of these immune-mediated disorders.
Collapse
|
12
|
Vahkal B, Yegorov S, Onyilagha C, Donner J, Reddick D, Shrivastav A, Uzonna J, Good SV. Immune System Effects of Insulin-Like Peptide 5 in a Mouse Model. Front Endocrinol (Lausanne) 2020; 11:610672. [PMID: 33519716 PMCID: PMC7841425 DOI: 10.3389/fendo.2020.610672] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Accepted: 11/16/2020] [Indexed: 12/25/2022] Open
Abstract
INTRODUCTION Insulin-like peptide 5 (INSL5) is a peptide hormone with proposed actions in glucose homeostasis and appetite regulation via its cognate receptor, relaxin family peptide receptor 4 (RXFP4). Here, we look for evidence for their involvement in the immune system using a mouse model. METHODS In silico analyses: we queried public databases for evidence of expression of INSL5-RXFP4 in immune system tissues/cells (NCBI's SRA and GeoProfiles) and disorders (EMBO-EBI) and performed phylogenetic footprinting to look for evidence that they are regulated by immune-associated transcription factors (TFs). Experimental analyses: We characterized the expression and correlation of INSL5/RXFP4 and other immune system markers in central and peripheral immune organs from C57/bl6 mice in seven cohorts. We tested whether fluctuations in circulating INSL5 induce an immune response, by injecting mice with 30 μg/kg of INSL5 peptide in the peritoneum, and examining levels of immune markers and metabolic peptides in plasma. Lastly, we quantified the expression of Rxfp4 in T-cells, dendritic cells and cell lines derived from human and mouse and tested the hypothesis that co-incubation of ANA-1 cells in INSL5 and LPS alters cytokine expression. RESULTS We find Insl5 expression only in thymus (in addition to colon) where its expression was highly correlated with Il-7, a marker of thymocyte development. This result is consistent with our in silico findings that Insl5 is highly expressed in thymic DP, DN thymocytes and cortical TEC's, and with evidence that it is regulated by thymocyte-associated TF's. We find Rxfp4 expression in all immune organs, and moderately high levels in DCs, particularly splenic DCs, and evidence that it is regulated by immune-associated TF's, such as STAT's and GATA. Systemic effects: We observed significantly elevated concentrations of blood GLP-1, GIP, GCG and PYY following intraperitoneal injection of INSL5, and significantly altered expression of cytokines IL-5, IL-7, M-CSF, IL-15, IL-27 and MIP-2. Immune cell effects: Incubation of ANA-1 cells with INSL5 impeded cell growth and led to a transient elevation of IL-15 and sustained reduction in IL-1β, IL-6 and TNFα. CONCLUSION We propose that INSL5-RXFP4 play a novel role in both central and peripheral immune cell signaling.
Collapse
Affiliation(s)
- Brett Vahkal
- Department of Biology, University of Winnipeg, Winnipeg, MB, Canada
- *Correspondence: Brett Vahkal, ; Sara V. Good,
| | - Sergey Yegorov
- Department of Biology, University of Winnipeg, Winnipeg, MB, Canada
| | | | | | - Dean Reddick
- Department of Biology, University of Winnipeg, Winnipeg, MB, Canada
| | | | - Jude Uzonna
- Department of Immunology, University of Manitoba, Winnipeg, MB, Canada
| | - Sara V. Good
- Department of Biology, University of Winnipeg, Winnipeg, MB, Canada
- *Correspondence: Brett Vahkal, ; Sara V. Good,
| |
Collapse
|
13
|
Pollard KM, Cauvi DM, Toomey CB, Hultman P, Kono DH. Mercury-induced inflammation and autoimmunity. Biochim Biophys Acta Gen Subj 2019; 1863:129299. [PMID: 30742953 DOI: 10.1016/j.bbagen.2019.02.001] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Revised: 01/12/2019] [Accepted: 02/01/2019] [Indexed: 11/29/2022]
Abstract
BACKGROUND Human exposure to mercury leads to a variety of pathologies involving numerous organ systems including the immune system. A paucity of epidemiological studies and suitable diagnostic criteria, however, has hampered collection of sufficient data to support a causative role for mercury in autoimmune diseases. Nevertheless, there is evidence that mercury exposure in humans is linked to markers of inflammation and autoimmunity. This is supported by experimental animal model studies, which convincingly demonstrate the biological plausibility of mercury as a factor in the pathogenesis of autoimmune disease. SCOPE OF THE REVIEW In this review, we focus on ability of mercury to elicit inflammatory and autoimmune responses in both humans and experimental animal models. MAJOR CONCLUSIONS Although subtle differences exist, the inflammatory and autoimmune responses elicited by mercury exposure in humans and experimental animal models show many similarities. Proinflammatory cytokine expression, lymphoproliferation, autoantibody production, and nephropathy are common outcomes. Animal studies have revealed significant strain dependent differences in inflammation and autoimmunity suggesting genetic regulation. This has been confirmed by the requirement for individual genes as well as genome wide association studies. Importantly, many of the genes required for mercury-induced inflammation and autoimmunity are also required for idiopathic systemic autoimmunity. A notable difference is that mercury-induced autoimmunity does not require type I IFN. This observation suggests that mercury-induced autoimmunity may arise by both common and specific pathways, thereby raising the possibility of devising criteria for environmentally associated autoimmunity. GENERAL SIGNIFICANCE Mercury exposure likely contributes to the pathogenesis of autoimmunity.
Collapse
Affiliation(s)
- K Michael Pollard
- Department of Molecular Medicine, Scripps Research, 10550 North Torrey Pines Road, La Jolla, CA, 92037, United States of America.
| | - David M Cauvi
- Department of Surgery and Center for Investigations of Health and Education Disparities, School of Medicine, University of California, San Diego, 9500 Gilman Drive #0739, La Jolla, CA 92093, United States of America.
| | - Christopher B Toomey
- Shiley Eye Institute, Department of Ophthalmology, University of California, San Diego, 9500 Gilman Drive #0946, La Jolla, CA 92093.
| | - Per Hultman
- Department of Experimental and Clinical Medicine, Linköping University, Linköping, Sweden.
| | - Dwight H Kono
- Department of Immunology and Microbiology, Scripps Research, 10550 North Torrey Pines Road, La Jolla, CA, 92037, United States of America.
| |
Collapse
|
14
|
Feng Q, Yao J, Zhou G, Xia W, Lyu J, Li X, Zhao T, Zhang G, Zhao N, Yang J. Quantitative Proteomic Analysis Reveals That Arctigenin Alleviates Concanavalin A-Induced Hepatitis Through Suppressing Immune System and Regulating Autophagy. Front Immunol 2018; 9:1881. [PMID: 30177931 PMCID: PMC6109684 DOI: 10.3389/fimmu.2018.01881] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 07/30/2018] [Indexed: 12/24/2022] Open
Abstract
Concanavalin A-induced autoimmune hepatitis is a well-established experimental model for immune-mediated liver injury. It has been widely used in the therapeutic studies of immune hepatitis. The in-depth analysis of dysregulated proteins from comparative proteomic results indicated that the activation of immune system resulted in the deregulation of autophagy. Follow-up studies validated that some immune related proteins, including Stat1, Pkr, Atg7, and Adrm1, were indeed upregulated. The accumulations of LC3B-II and p62 were confirmed by immunohistochemistry and Western blot analyses. Arctigenin pretreatment significantly alleviated the liver injury, as evidenced by biochemical and histopathological investigations, whose protective effects were comparable with Prednisone acetate and Cyclosporin A. Arctigenin pretreatment decreased the levels of IL-6 and IFN-γ, but increased the ones of IL-10. Next, the quantitative proteomic analysis demonstrated that ARC pretreatment suppressed the activation of immune system through the inhibition of IFN-γ signaling, when it downregulated the protein expressions of Stat1, P-Stat1, Pkr, P-Pkr, Bnip3, Beclin1, Atg7, LC3B, Adrm1, and p62. Meanwhile, Arctigenin pretreatment also reduced the gene expressions of Stat1, Pkr, and Atg7. These results suggested that Arctigenin alleviated autophagy as well as apoptosis through inhibiting IFN-γ/IL-6/Stat1 pathway and IL-6/Bnip3 pathway. In summary, the comparative proteomic analysis revealed that the activation of immune system led to Concanavalin A-induced hepatitis. Both autophagy and apoptosis had important clinical implications for the treatment of immune hepatitis. Arctigenin might exert great therapeutic potential in immune-mediated liver injury.
Collapse
Affiliation(s)
- Qin Feng
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China.,Center for New Drug Pharmacological Research of Lunan Pharmaceutical Group, State Key Laboratory, Generic Manufacture Technology of Chinese Traditional Medicine, Linyi, China
| | - Jingchun Yao
- Center for New Drug Pharmacological Research of Lunan Pharmaceutical Group, State Key Laboratory, Generic Manufacture Technology of Chinese Traditional Medicine, Linyi, China
| | - Ge Zhou
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Wenkai Xia
- Center for New Drug Pharmacological Research of Lunan Pharmaceutical Group, State Key Laboratory, Generic Manufacture Technology of Chinese Traditional Medicine, Linyi, China
| | - Jingang Lyu
- Center for New Drug Pharmacological Research of Lunan Pharmaceutical Group, State Key Laboratory, Generic Manufacture Technology of Chinese Traditional Medicine, Linyi, China
| | - Xin Li
- Center for New Drug Pharmacological Research of Lunan Pharmaceutical Group, State Key Laboratory, Generic Manufacture Technology of Chinese Traditional Medicine, Linyi, China
| | - Tao Zhao
- Center for New Drug Pharmacological Research of Lunan Pharmaceutical Group, State Key Laboratory, Generic Manufacture Technology of Chinese Traditional Medicine, Linyi, China
| | - Guimin Zhang
- Center for New Drug Pharmacological Research of Lunan Pharmaceutical Group, State Key Laboratory, Generic Manufacture Technology of Chinese Traditional Medicine, Linyi, China.,School of Pharmacy, Linyi University, Linyi, China
| | - Ningwei Zhao
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.,Shimadzu Biomedical Research Laboratory, Shanghai, China
| | - Jie Yang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China.,State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, China
| |
Collapse
|
15
|
Ludwig MD, Zagon IS, McLaughlin PJ. Featured Article: Modulation of the OGF-OGFr pathway alters cytokine profiles in experimental autoimmune encephalomyelitis and multiple sclerosis. Exp Biol Med (Maywood) 2018; 243:361-369. [PMID: 29307283 DOI: 10.1177/1535370217749830] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The endogenous neuropeptide opioid growth factor, chemically termed [Met5]-enkephalin, has growth inhibitory and immunomodulatory properties. Opioid growth factor is distributed widely throughout most tissues, is autocrine and paracrine produced, and interacts at the nuclear-associated receptor, OGFr. Serum levels of opioid growth factor are decreased in patients with multiple sclerosis and in animals with experimental autoimmune encephalomyelitis suggesting that the OGF-OGFr pathway becomes dysregulated in this disease. This study begins to assess other cytokines that are altered following opioid growth factor or low-dose naltrexone modulation of the OGF-OGFr axis in mice with experimental autoimmune encephalomyelitis using serum samples collected in mice treated for 10 or 20 days and assayed by a multiplex cytokine assay for inflammatory markers. Cytokines of interest were validated in mice at six days following immunization for experimental autoimmune encephalomyelitis. In addition, selected cytokines were validated with serum from MS patients treated with low-dose naltrexone alone or low-dose naltrexone in combination with glatiramer acetate (Copaxone®). Experimental autoimmune encephalomyelitis mice had elevated levels of 7 of 10 cytokines. Treatment with opioid growth factor or low-dose naltrexone resulted in elevated expression levels of the IL-6 cytokine, and significantly reduced IL-10 values, relative to saline-treated experimental autoimmune encephalomyelitis mice. TNF-γ values were increased in experimental autoimmune encephalomyelitis mice relative to normal, but were not altered by opioid growth factor or low-dose naltrexone. IFN-γ levels were reduced in opioid growth factor- or low-dose naltrexone-treated experimental autoimmune encephalomyelitis mice relative to saline-treated mice at 10 days, and elevated relative to normal values at 20 days. Validation studies revealed that within six days of immunization, opioid growth factor or low-dose naltrexone modulated IL-6 and IL-10 cytokine expression. Validation in human serum revealed markedly reduced IL-6 cytokine levels in MS patients taking low-dose naltrexone relative to standard care. In summary, modulation of the OGF-OGFr pathway regulates some inflammatory cytokines, and together with opioid growth factor serum levels, may begin to form a panel of valid biomarkers to monitor progression of multiple sclerosis and response to therapy. Impact statement Modulation of the opioid growth factor (OGF)-OGF receptor (OGFr) alters inflammatory cytokine expression in multiple sclerosis and experimental autoimmune encephalomyelitis (EAE). Multiplex cytokine assays demonstrated that mice with chronic EAE and treated with either OGF or low-dose naltrexone (LDN) had decreased expression of interferon-gamma (IFN-γ), tumor necrosis factor-alpha (TNF-α), and the anti-inflammatory cytokine IL-10 within 10 days or treatment, as well as increased serum expression of the pro-inflammatory cytokine IL-6, relative to immunized mice receiving saline. Multiplex data were validated using ELISA kits and serum from MS patients treated with LDN and revealed decreased in IL-6 levels in patients taking LDN relative to standard care alone. These data, along with serum levels of OGF, begin to formulate a selective biomarker profile for MS that is easily measured and effective at monitoring disease progression and response to therapy.
Collapse
Affiliation(s)
- Michael D Ludwig
- Department of Neural and Behavioral Sciences, College of Medicine, Pennsylvania State University, PA 17033, USA
| | - Ian S Zagon
- Department of Neural and Behavioral Sciences, College of Medicine, Pennsylvania State University, PA 17033, USA
| | - Patricia J McLaughlin
- Department of Neural and Behavioral Sciences, College of Medicine, Pennsylvania State University, PA 17033, USA
| |
Collapse
|
16
|
Pollard KM, Christy JM, Cauvi DM, Kono DH. Environmental Xenobiotic Exposure and Autoimmunity. CURRENT OPINION IN TOXICOLOGY 2017; 10:15-22. [PMID: 29503968 DOI: 10.1016/j.cotox.2017.11.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Susceptibility to autoimmune diseases is dependent on multigenic inheritance, environmental factors, and stochastic events. Although there has been substantial progress in identifying predisposing genetic variants, a significant challenge facing autoimmune disease research is the identification of the specific events that trigger loss of tolerance, autoreactivity and ultimately autoimmune disease. Accordingly, studies have indicated that a wide range of extrinsic factors including drugs, chemicals, microbes, and other environmental factors can induce autoimmunity, particularly systemic autoimmune diseases such as lupus. This review describes a class of environmental factors, namely xenobiotics, epidemiologically linked to human autoimmunity. Mechanisms of xenobiotic autoimmune disease induction are discussed in terms of human and animal model studies with a focus on the role of inflammation and the innate immune response. We argue that localized tissue damage and chronic inflammation elicited by xenobiotic exposure leads to the release of self-antigens and damage-associated molecular patterns as well as the appearance of ectopic lymphoid structures and secondary lymphoid hypertrophy, which provide a milieu for the production of autoreactive B and T cells that contribute to the development and persistence of autoimmunity in predisposed individuals.
Collapse
Affiliation(s)
- K Michael Pollard
- Department of Molecular Medicine, MEM125, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, USA 92037
| | - Joseph M Christy
- Department of Molecular Medicine, MEM125, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, USA 92037
| | - David M Cauvi
- Department of Surgery, School of Medicine, University of California, San Diego, 9500 Gilman Drive #0739, La Jolla, CA, USA 92093
| | - Dwight H Kono
- Department of Immunology and Microbiology, IMM310, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, USA 92037
| |
Collapse
|
17
|
Chasset F, Arnaud L. Targeting interferons and their pathways in systemic lupus erythematosus. Autoimmun Rev 2017; 17:44-52. [PMID: 29108825 DOI: 10.1016/j.autrev.2017.11.009] [Citation(s) in RCA: 103] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 09/28/2017] [Indexed: 01/07/2023]
Abstract
Significant advances in the understanding of the molecular basis of innate immunity have led to the identification of interferons (IFNs), particularly IFN-α, as central mediators in the pathogenesis of Systemic Lupus Erythematosus. Therefore, targeting of IFNs and of their downstream pathways has emerged as important developments for novel drug research in SLE. Based on this, several specific interferon blocking strategies using anti-IFN-α antibodies, anti-type I interferon receptor antibodies, Interferon-α-kinoid, or anti-IFN-γ antibodies have all been assessed in recent clinical trials. Alternative strategies targeting the plasmacytoid dendritic cells (pDCs), Toll-Like Receptors (TLRs)-7/9 or their downstream pathways such as the myeloid differentiation primary-response protein 88 (MYD88), spleen tyrosine kinase (Syk), Janus-kinases (JAKs), interleukin-1 receptor-associated kinase 4 (IRAK4), or the Tyrosine Kinase 2 (TYK2) are also investigated actively in SLE, at more preliminary clinical development stages, except for JAK inhibitors which have reached phase 2 studies. In a near future, in-depth and personalized functional characterization of IFN pathways may provide further guidance for the selection of the most relevant therapeutic strategy in SLE, tailored at the patient-level.
Collapse
Affiliation(s)
- François Chasset
- AP-HP, Service de Dermatologie et d'Allergologie, Hôpital Tenon, F-75020, Paris, France
| | - Laurent Arnaud
- Service de rhumatologie, Centre National de Référence des Maladies Autoimmunes et Systémiques Rares, Université de Strasbourg, INSERM UMR-S 1109, F-67000 Strasbourg, France.
| |
Collapse
|
18
|
Pollard KM, Escalante GM, Huang H, Haraldsson KM, Hultman P, Christy JM, Pawar RD, Mayeux JM, Gonzalez-Quintial R, Baccala R, Beutler B, Theofilopoulos AN, Kono DH. Induction of Systemic Autoimmunity by a Xenobiotic Requires Endosomal TLR Trafficking and Signaling from the Late Endosome and Endolysosome but Not Type I IFN. THE JOURNAL OF IMMUNOLOGY 2017; 199:3739-3747. [PMID: 29055005 DOI: 10.4049/jimmunol.1700332] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 09/25/2017] [Indexed: 12/17/2022]
Abstract
Type I IFN and nucleic acid-sensing TLRs are both strongly implicated in the pathogenesis of lupus, with most patients expressing IFN-induced genes in peripheral blood cells and with TLRs promoting type I IFNs and autoreactive B cells. About a third of systemic lupus erythematosus patients, however, lack the IFN signature, suggesting the possibility of type I IFN-independent mechanisms. In this study, we examined the role of type I IFN and TLR trafficking and signaling in xenobiotic systemic mercury-induced autoimmunity (HgIA). Strikingly, autoantibody production in HgIA was not dependent on the type I IFN receptor even in NZB mice that require type I IFN signaling for spontaneous disease, but was dependent on the endosomal TLR transporter UNC93B1 and the endosomal proton transporter, solute carrier family 15, member 4. HgIA also required the adaptor protein-3 complex, which transports TLRs from the early endosome to the late endolysosomal compartments. Examination of TLR signaling pathways implicated the canonical NF-κB pathway and the proinflammatory cytokine IL-6 in autoantibody production, but not IFN regulatory factor 7. These findings identify HgIA as a novel type I IFN-independent model of systemic autoimmunity and implicate TLR-mediated NF-κB proinflammatory signaling from the late endocytic pathway compartments in autoantibody generation.
Collapse
Affiliation(s)
- K Michael Pollard
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037;
| | - Gabriela M Escalante
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037
| | - Hua Huang
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037
| | - Katarina M Haraldsson
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037
| | - Per Hultman
- Department of Experimental and Clinical Medicine, Linköping University, Linköping 58183, Sweden; and
| | - Joseph M Christy
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037
| | - Rahul D Pawar
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037
| | - Jessica M Mayeux
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037
| | | | - Roberto Baccala
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037
| | - Bruce Beutler
- Center for the Genetics of Host Defense, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | | | - Dwight H Kono
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037
| |
Collapse
|