451
|
Key Factor Regulating Inflammatory Microenvironment, Metastasis, and Resistance in Breast Cancer: Interleukin-1 Signaling. Mediators Inflamm 2021; 2021:7785890. [PMID: 34602858 PMCID: PMC8486558 DOI: 10.1155/2021/7785890] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 08/20/2021] [Indexed: 02/06/2023] Open
Abstract
Breast cancer is one of the top-ranked cancers for incidence and mortality worldwide. The biggest challenges in breast cancer treatment are metastasis and drug resistance, for which work on molecular evaluation, mechanism studies, and screening of therapeutic targets is ongoing. Factors that lead to inflammatory infiltration and immune system suppression in the tumor microenvironment are potential therapeutic targets. Interleukin-1 is known as a proinflammatory and immunostimulatory cytokine, which plays important roles in inflammatory diseases. Recent studies have shown that interleukin-1 cytokines drive the formation and maintenance of an inflammatory/immunosuppressive microenvironment through complex intercellular signal crosstalk and tight intracellular signal transduction, which were found to be potentially involved in the mechanism of metastasis and drug resistance of breast cancer. Some preclinical and clinical treatments or interventions to block the interleukin-1/interleukin-1 receptor system and its up- and downstream signaling cascades have also been proven effective. This study provides an overview of IL-1-mediated signal communication in breast cancer and discusses the potential of IL-1 as a therapeutic target especially for metastatic breast cancer and combination therapy and current problems, aiming at enlightening new ideas in the study of inflammatory cytokines and immune networks in the tumor microenvironment.
Collapse
|
452
|
Guo H, Qian L, Cui J. Focused evaluation of the roles of macrophages in chimeric antigen receptor (CAR) T cell therapy associated cytokine release syndrome. Cancer Biol Med 2021; 19:j.issn.2095-3941.2021.0087. [PMID: 34570442 PMCID: PMC8958886 DOI: 10.20892/j.issn.2095-3941.2021.0087] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 05/17/2021] [Indexed: 11/11/2022] Open
Abstract
Cytokine release syndrome (CRS) is a major obstacle to the widespread clinical application of chimeric antigen receptor (CAR) T cell therapies. CRS can also be induced by infections (such as SARS-CoV-2), drugs (such as therapeutic antibodies), and some autoimmune diseases. Myeloid-derived macrophages play key roles in the pathogenesis of CRS, and participate in the production and release of the core CRS cytokines, including interleukin (IL)-1, IL-6, and interferon-γ. In this review, we summarize the roles of macrophages in CRS and discuss new developments in macrophage activation and the related mechanisms of cytokine regulation in CRS.
Collapse
Affiliation(s)
- Hanfei Guo
- Cancer Center, the First Hospital of Jilin University, Changchun 130021, China
| | - Lei Qian
- Cancer Center, the First Hospital of Jilin University, Changchun 130021, China
| | - Jiuwei Cui
- Cancer Center, the First Hospital of Jilin University, Changchun 130021, China
| |
Collapse
|
453
|
Bozkurt HS, Bilen Ö. Gastrointestinal symptoms in COVID-19 could be associated with severe lung involvement and increased readmission rates. EUR J INFLAMM 2021. [DOI: 10.1177/20587392211048259] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Introduction SARS-CoV-2 virus manifests itself with primary lung damage but also has intestinal involvement. In this study, we aimed to investigate the frequency of gastrointestinal symptoms (GIS) and the relationship of GIS with readmission to the hospital within 30 days in SARS-CoV-2 infected patients who were hospitalized in a specified pandemic hospital. Materials and Methods Symptomatic patients diagnosed with rapid antibody positivity with real-time polymerase chain reaction and typical thorax computed tomography findings were included in this retrospective cohort observational study. Demographic and clinical data were obtained from electronic medical records. Hospital-associated GIS were considered as experiencing at least one of the GIS such as gas, bloating, diarrhea, and constipation developing within72 h after hospital admission. Results The mean age of the patients was 58 ± 14.4 years and 60.7% were men. 82% of hospitalizations were a moderate and severe disease. 71.4% of patients without GIS had at least one of the GIS after hospitalization. As the severity of the disease increased, the frequency of the severity of gastrointestinal symptom increased. GIS bowel disorders were more prominent in patients with moderate and severe disease. Antibiotic and specific treatment (anti-Il-1, anti-Il-6) contributed to the occurrence of gastrointestinal symptom in SARS-CoV-2 inpatients. Conclusion According to our observations of the second wave of the pandemic, the presence, frequency, and severity of gastrointestinal symptom in inpatient is associated with severity of lung disease and increased readmission rate after discharge.
Collapse
Affiliation(s)
- Hüseyin S Bozkurt
- Clinic of Gastroenterology, Istanbul Maltepe University Faculty Of Medicine, Istanbul, Turkey
| | - Ömer Bilen
- Bursa Techinical University, Bursa, Turkey
| |
Collapse
|
454
|
Nandi D, Farid NSS, Karuppiah HAR, Kulkarni A. Imaging Approaches to Monitor Inflammasome Activation. J Mol Biol 2021; 434:167251. [PMID: 34537231 DOI: 10.1016/j.jmb.2021.167251] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 09/09/2021] [Accepted: 09/10/2021] [Indexed: 10/20/2022]
Abstract
Inflammasomes are a critical component of innate immune response which plays an important role in the pathogenesis of various chronic and acute inflammatory disease conditions. An inflammasome complex consists of a multimeric protein assembly triggered by any form of pathogenic or sterile insult, resulting in caspase-1 activation. This active enzyme is further known to activate downstream pro-inflammatory cytokines along with a pore-forming protein, eventually leading to a lytic cell death called pyroptosis. Understanding the spatiotemporal kinetics of essential inflammasome components provides a better interpretation of the complex signaling underlying inflammation during several disease pathologies. This can be attained via in-vitro and in-vivo imaging platforms, which not only provide a basic understanding of molecular signaling but are also crucial to develop and screen targeted therapeutics. To date, numerous studies have reported platforms to image different signaling components participating in inflammasome activation. Here, we review several elements of inflammasome signaling, a common molecular mechanism combining these elements and their respective imaging tools. We anticipate that future needs will include developing new inflammasome imaging systems that can be utilized as clinical tools for diagnostics and monitoring treatment responses.
Collapse
Affiliation(s)
- Dipika Nandi
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA, USA; Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, USA. https://twitter.com/dipikanandi24
| | - Noorul Shaheen Sheikh Farid
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA, USA. https://twitter.com/Shaheen30n
| | - Hayat Anu Ranjani Karuppiah
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA, USA. https://twitter.com/AnuHayat
| | - Ashish Kulkarni
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA, USA; Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, USA; Department of Biomedical Engineering, University of Massachusetts, Amherst, MA, USA; Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003, USA.
| |
Collapse
|
455
|
Imazio M, Lazaros G, Gattorno M, LeWinter M, Abbate A, Brucato A, Klein A. Anti-interleukin-1 agents for pericarditis: a primer for cardiologists. Eur Heart J 2021; 43:2946-2957. [PMID: 34528670 PMCID: PMC9375710 DOI: 10.1093/eurheartj/ehab452] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 02/21/2021] [Accepted: 07/02/2021] [Indexed: 01/24/2023] Open
Abstract
Anti-interleukin (IL)-1 agents have been developed for the treatment of autoinflammatory and rheumatic conditions, where overproduction of IL-1 is an important pathophysiologic process. IL-1α and IL-1β are the most studied members of the IL-1 family of cytokines and have the strongest proinflammatory effects. A naturally occurring antagonist (IL-1Ra) mitigates their proinflammatory effects. Overproduction of both IL-1α (released by inflamed/damaged pericardial cells) and IL-1β (released by inflammatory cells) is now a well-recognized therapeutic target in patients with recurrent idiopathic pericarditis. Currently, there are three available anti-IL-1 agents: anakinra (recombinant human IL-1Ra), rilonacept (a soluble decoy receptor ‘trap’, binding both IL-1α and IL-1β), and canakinumab (human monoclonal anti-IL-1β antibody). For patients with corticosteroid-dependent and colchicine-resistant recurrent pericarditis with evidence of systemic inflammation, as evidenced by elevated C-reactive protein, the efficacy and safety of anakinra (2 mg/kg/day up to 100 mg/day subcutaneously usually for at least 6 months, then tapered) and rilonacept (320 mg subcutaneously for the first day followed by 160 mg subcutaneously weekly) have been clearly demonstrated in observational studies and randomized controlled clinical trials. Severe side effects are rare and discontinuation rates are very low (<4%). The most common reported side effect is injection site reactions (>50% of patients). In this article, we describe the historical and pathophysiological background and provide a comprehensive review of these agents, which appear to be the most significant advance in medical therapy of recurrent pericarditis in the last 5 years.
Collapse
Affiliation(s)
- Massimo Imazio
- Head of Cardiology, Cardiothoracic Department, University Hospital "Santa Maria della Misericordia", ASUFC, Piazzale Santa Maria della Misericordia 15, Udine 33100, Italy
| | - George Lazaros
- 1st Cardiology Clinic, National and Kapodistrian University of Athens, School of Medicine, Hippokration General Hospital, Athens, Greece
| | - Marco Gattorno
- Center for Autoinflammatory Diseases and Immunodeficiencies, IRCCS G. Gaslini, Genova, Italy
| | - Martin LeWinter
- Cardiology Unit, University of Vermont Medical Center, Burlington, VT, USA
| | - Antonio Abbate
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, USA
| | - Antonio Brucato
- Department of Biomedical and Clinical Sciences "Sacco", Fatebenefratelli Hospital, Università di Milano, Milan, Italy
| | - Allan Klein
- Center for the Diagnosis and Treatment of Pericardial Diseases, Section of Cardiovascular Imaging, Department of Cardiovascular Medicine, Heart, Vascular, and Thoracic Institute, Cleveland Clinic, USA
| |
Collapse
|
456
|
Serafim CADL, Araruna MEC, Alves Júnior EB, Silva LMO, Silva AO, da Silva MS, Alves AF, Araújo AA, Batista LM. (-)-Carveol Prevents Gastric Ulcers via Cytoprotective, Antioxidant, Antisecretory and Immunoregulatory Mechanisms in Animal Models. Front Pharmacol 2021; 12:736829. [PMID: 34497525 PMCID: PMC8419343 DOI: 10.3389/fphar.2021.736829] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 08/05/2021] [Indexed: 12/12/2022] Open
Abstract
Background: (-)-Carveol (p-Mentha-6,8-dien-2-ol) is a monocyclic monoterpenic alcohol, present in essential oils of plant species such as Cymbopogon giganteus, Illicium pachyphyllum and in spices such as Carum carvi (cumin). Pharmacological studies report its antitumor, antimicrobial, neuroprotective, vasorelaxant, antioxidant and anti-inflammatory activity. Hypothesis/Purpose: The objective of this study was to evaluate the acute non-clinical oral toxicity, gastroprotective activity of monoterpene (-)-Carveol in animal models and the related mechanisms of action. Methods: Acute toxicity was assessed according to OECD guide 423 in mice. Ethanol, stress, NSAIDs and pylorus ligation-induced gastric ulcer models were used to investigate antiulcer properties. The related mechanisms of action were using the ethanol-gastric lesions protocol. Results: (-)-Carveol has low toxicity, with a lethal dose 50% (LD50) equal to or greater than 2,500 mg/kg according to OECD guide nº 423. In all gastric ulcer induction methods evaluated, (-)-Carveol (25, 50, 100 and 200 mg/kg, p.o.) significantly reduced the ulcerative lesion in comparison with the respective control groups. To investigate the mechanisms involved in the gastroprotective activity, the antisecretory or neutralizing of gastric secretion, cytoprotective, antioxidant and immunoregulatory effects were evaluated. In the experimental protocol of pylorus ligation-induced gastric ulcer, (-)-Carveol (100 mg/kg) reduced (p < 0.001) the volume of gastric secretion in both routes (oral and intraduodenal). The previous administration of blockers NEM (sulfhydryl groups blocker), L-NAME (nitric oxide synthesis inhibitor), glibenclamide (KATP channel blocker) and indomethacin (cyclo-oxygenase inhibitor), significantly reduced the gastroprotection exercised by (-)-Carveol, suggesting the participation of these pathways in its gastroprotective activity. In addition, treatment with (-)-Carveol (100 mg/kg) increased (p < 0.001) mucus adhered to the gastric wall. Treatment also increased (p < 0.001) levels of reduced glutathione (GSH), superoxide dismutase (SOD) and interleukin-10 (IL-10). It also reduced (p < 0.001) malondialdehyde (MDA), myeloperoxidase (MPO), interleukin-1 beta (IL-1β) and tumor necrosis factor-alpha (TNF-α) levels. Conclusion: Thus, it is possible to infer that (-)-Carveol presents gastroprotective activity related to antisecretory, cytoprotective, antioxidant and immunomodulatory mechanisms.
Collapse
Affiliation(s)
- Catarina Alves de Lima Serafim
- Postgraduate Program in Natural and Synthetic Bioactive Products, Health Sciences Center, Federal University of Paraíba (UFPB), João Pessoa, Brazil
| | - Maria Elaine Cristina Araruna
- Postgraduate Program in Natural and Synthetic Bioactive Products, Health Sciences Center, Federal University of Paraíba (UFPB), João Pessoa, Brazil
| | - Edvaldo Balbino Alves Júnior
- Postgraduate Program in Natural and Synthetic Bioactive Products, Health Sciences Center, Federal University of Paraíba (UFPB), João Pessoa, Brazil
| | - Leiliane Macena Oliveira Silva
- Postgraduate Program in Natural and Synthetic Bioactive Products, Health Sciences Center, Federal University of Paraíba (UFPB), João Pessoa, Brazil
| | - Alessa Oliveira Silva
- Postgraduate Program in Natural and Synthetic Bioactive Products, Health Sciences Center, Federal University of Paraíba (UFPB), João Pessoa, Brazil
| | - Marcelo Sobral da Silva
- Postgraduate Program in Natural and Synthetic Bioactive Products, Health Sciences Center, Federal University of Paraíba (UFPB), João Pessoa, Brazil
| | - Adriano Francisco Alves
- Department of Physiology and Pathology, Health Sciences Center, Federal University of Paraíba (UFPB), João Pessoa, Brazil
| | - Aurigena Antunes Araújo
- Department of Morphology, Histology and Basic Pathology, Biosciences Center, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Leônia Maria Batista
- Postgraduate Program in Natural and Synthetic Bioactive Products, Health Sciences Center, Federal University of Paraíba (UFPB), João Pessoa, Brazil
| |
Collapse
|
457
|
Elias M, Zhao S, Le HT, Wang J, Neurath MF, Neufert C, Fiocchi C, Rieder F. IL-36 in chronic inflammation and fibrosis - bridging the gap? J Clin Invest 2021; 131:144336. [PMID: 33463541 DOI: 10.1172/jci144336] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
IL-36 is a member of the IL-1 superfamily and consists of three agonists and one receptor antagonist (IL-36Ra). The three endogenous agonists, IL-36α, -β, and -γ, act primarily as proinflammatory cytokines, and their signaling through the IL-36 receptor (IL-36R) promotes immune cell infiltration and secretion of inflammatory and chemotactic molecules. However, IL-36 signaling also fosters secretion of profibrotic soluble mediators, suggesting a role in fibrotic disorders. IL-36 isoforms and IL-36 have been implicated in inflammatory diseases including psoriasis, arthritis, inflammatory bowel diseases, and allergic rhinitis. Moreover, IL-36 has been connected to fibrotic disorders affecting the kidney, lung, and intestines. This review summarizes the expression, cellular source, and function of IL-36 in inflammation and fibrosis in various organs, and proposes that IL-36 modulation may prove valuable in preventing or treating inflammatory and fibrotic diseases and may reveal a mechanistic link between inflammation and fibrosis.
Collapse
Affiliation(s)
- Michael Elias
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA.,Department of Gastroenterology, Hepatology and Nutrition, Digestive Diseases and Surgery Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Shuai Zhao
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA.,Department of Gastroenterology, Hepatology and Nutrition, Digestive Diseases and Surgery Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Hongnga T Le
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA.,Department of Gastroenterology, Hepatology and Nutrition, Digestive Diseases and Surgery Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Jie Wang
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA.,Department of Gastroenterology, Hepatology and Nutrition, Digestive Diseases and Surgery Institute, Cleveland Clinic, Cleveland, Ohio, USA.,Henan Key Laboratory of Immunology and Targeted Drug, Xinxiang Medical University, Xinxiang, Henan Province, China
| | - Markus F Neurath
- Department of Medicine 1 and Deutsches Zentrum Immuntherapie DZI, Universitaetsklinikum Erlangen, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Clemens Neufert
- Department of Medicine 1 and Deutsches Zentrum Immuntherapie DZI, Universitaetsklinikum Erlangen, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Claudio Fiocchi
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA.,Department of Gastroenterology, Hepatology and Nutrition, Digestive Diseases and Surgery Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Florian Rieder
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA.,Department of Gastroenterology, Hepatology and Nutrition, Digestive Diseases and Surgery Institute, Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
458
|
Boechat JL, Chora I, Morais A, Delgado L. The immune response to SARS-CoV-2 and COVID-19 immunopathology - Current perspectives. Pulmonology 2021; 27:423-437. [PMID: 33867315 PMCID: PMC8040543 DOI: 10.1016/j.pulmoe.2021.03.008] [Citation(s) in RCA: 110] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 02/26/2021] [Accepted: 03/13/2021] [Indexed: 12/15/2022] Open
Abstract
SARS-CoV-2 is a new beta coronavirus, similar to SARS-CoV-1, that emerged at the end of 2019 in the Hubei province of China. It is responsible for coronavirus disease 2019 (COVID-19), which was declared a pandemic by the World Health Organization on March 11, 2020. The ability to gain quick control of the pandemic has been hampered by a lack of detailed knowledge about SARS-CoV-2-host interactions, mainly in relation to viral biology and host immune response. The rapid clinical course seen in COVID-19 indicates that infection control in asymptomatic patients or patients with mild disease is probably due to the innate immune response, as, considering that SARS-CoV-2 is new to humans, an effective adaptive response would not be expected to occur until approximately 2-3 weeks after contact with the virus. Antiviral innate immunity has humoral components (complement and coagulation-fibrinolysis systems, soluble proteins that recognize glycans on cell surface, interferons, chemokines, and naturally occurring antibodies) and cellular components (natural killer cells and other innate lymphocytes). Failure of this system would pave the way for uncontrolled viral replication in the airways and the mounting of an adaptive immune response, potentially amplified by an inflammatory cascade. Severe COVID-19 appears to be due not only to viral infection but also to a dysregulated immune and inflammatory response. In this paper, the authors review the most recent publications on the immunobiology of SARS-CoV-2, virus interactions with target cells, and host immune responses, and highlight possible associations between deficient innate and acquired immune responses and disease progression and mortality. Immunotherapeutic strategies targeting both the virus and dysfunctional immune responses are also addressed.
Collapse
Affiliation(s)
- J L Boechat
- Clinical Immunology Service, Internal Medicine Department, Faculty of Medicine, Universidade Federal Fluminense, Niterói, RJ, Brazil; Basic and Clinical Immunology Unit, Department of Pathology, Faculty of Medicine, University of Porto, Portugal; Center for Health Technology and Services Research (CINTESIS), Faculty of Medicine, University of Porto, Portugal
| | - I Chora
- Internal Medicine Service, Department of Medicine, Hospital Pedro Hispano, Unidade Local de Saúde de Matosinhos, Senhora da Hora, Portugal; Department of Medicine, Faculty of Medicine, University of Porto, Portugal
| | - A Morais
- Department of Medicine, Faculty of Medicine, University of Porto, Portugal; Pulmonology Department, Centro Hospitalar e Universitario de Sao Joao, Porto, Portugal
| | - L Delgado
- Basic and Clinical Immunology Unit, Department of Pathology, Faculty of Medicine, University of Porto, Portugal; Center for Health Technology and Services Research (CINTESIS), Faculty of Medicine, University of Porto, Portugal
| |
Collapse
|
459
|
Gowri Sankar S, Mowna Sundari T, Alwin Prem Anand A. Emergence of Dengue 4 as Dominant Serotype During 2017 Outbreak in South India and Associated Cytokine Expression Profile. Front Cell Infect Microbiol 2021; 11:681937. [PMID: 34447698 PMCID: PMC8382982 DOI: 10.3389/fcimb.2021.681937] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 07/12/2021] [Indexed: 12/23/2022] Open
Abstract
Dengue virus (DENV) infection is prevalent in tropical and subtropical regions of the world, which is fatal if untreated symptomatically. Emergence of new genotype within serotypes led to enhanced severity. The objective of the study is to identify the molecular characteristics of the DENV circulated during 2017 outbreak in Tamil Nadu, India, and to investigate the role of inflammatory cytokines in different “serotypes” and in “dengue severity”. A total of 135 suspected samples were tested for DENV infection using IgM, IgG, and qPCR assay; where 76 samples were positive for DENV and analyzed for 12 inflammatory cytokines using ELISA. Serotyping shows 14 DENV-1, 22 DENV-2, 7 DENV-3, and 33 DENV-4, where DENV-4 was predominant. Among 76, 42 isolates were successfully sequenced for C-prM region and grouped. A lineage shift was observed in DENV-4 genotype. Irrespective of serotypes, IFNγ was significantly elevated in all serotypes than control as well as in primary infection than secondary, indicating its role in immune response. GM-CSF and IP-10 were significantly elevated in secondary infection and could be used as prognostic biomarkers for secondary infection. Our observation shows differential cytokine expression profile varied with each serotype, indicating serotype/genotype-specific viral proteins might play a major role in dengue severity. DENV-4 as dominant serotype was reported in Tamil Nadu for the first time during an outbreak with a mixed Th1/Th17 cytokine expression profile that correlated with disease severity. We conclude it is essential to identify circulating viral genotype and their fitness by mutational analysis to correlate with disease severity and immune status, as this correlation will be helpful in diagnostics and therapeutics applications.
Collapse
Affiliation(s)
- S Gowri Sankar
- Department of Molecular Biology, Indian Council of Medical Research (ICMR)-Vector Control Research Center - Field Station, Madurai, India
| | - T Mowna Sundari
- Department of Biotechnology - Bioinformatics Infrastructure Facilities (DBT-BIF) Centre (Under DBT Biotechnology Information System Network (BTISNet) Scheme), Lady Doak College, Madurai, India.,Department of Biotechnology, Lady Doak College, Madurai, India
| | - A Alwin Prem Anand
- Institute of Clinical Anatomy and Cell Analysis, University of Tuebingen, Tuebingen, Germany
| |
Collapse
|
460
|
Adverse effects of biologic anti-inflammatory agents on the respiratory system: A review. Afr J Thorac Crit Care Med 2021; 27. [PMID: 34430870 PMCID: PMC8327682 DOI: 10.7196/ajtccm.2021.v27i2.117] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/09/2021] [Indexed: 12/12/2022] Open
Abstract
The therapy of autoimmune rheumatological conditions has undergone significant changes with the introduction of biologic antiinflammatory agents including cytokine antagonists and agents that interfere with the function of T and B cells or those that inhibit
intracellular enzymes such as Janus kinase (JAK). Although useful to control inflammation, these agents may be associated with druginduced lung disease, which may be difficult to differentiate from pulmonary disorders caused by the underlying autoimmune diseases.
This review aims to provide a description of lung disease, both infectious and non-infectious, that may be induced by the administration of
biologic anti-inflammatory agents with emphasis on inhibitors of tumour necrosis factor, interleukin-1, interleukin-6 and JAK.
Collapse
|
461
|
Niklander SE, Murdoch C, Hunter KD. IL-1/IL-1R Signaling in Head and Neck Cancer. FRONTIERS IN ORAL HEALTH 2021; 2:722676. [PMID: 35048046 PMCID: PMC8757896 DOI: 10.3389/froh.2021.722676] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 08/04/2021] [Indexed: 01/22/2023] Open
Abstract
Decades ago, the study of cancer biology was mainly focused on the tumor itself, paying little attention to the tumor microenvironment (TME). Currently, it is well recognized that the TME plays a vital role in cancer development and progression, with emerging treatment strategies focusing on different components of the TME, including tumoral cells, blood vessels, fibroblasts, senescent cells, inflammatory cells, inflammatory factors, among others. There is a well-accepted relationship between chronic inflammation and cancer development. Interleukin-1 (IL-1), a potent pro-inflammatory cytokine commonly found at tumor sites, is considered one of the most important inflammatory factors in cancer, and has been related with carcinogenesis, tumor growth and metastasis. Increasing evidence has linked development of head and neck squamous cell carcinoma (HNSCC) with chronic inflammation, and particularly, with IL-1 signaling. This review focuses on the most important members of the IL-1 family, with emphasis on how their aberrant expression can promote HNSCC development and metastasis, highlighting possible clinical applications.
Collapse
Affiliation(s)
- Sven E. Niklander
- Unidad de Patología y Medicina Oral, Facultad de Odontologia, Universidad Andres Bello, Viña del Mar, Chile
| | - Craig Murdoch
- Unit of Oral and Maxillofacial Medicine, Pathology and Surgery, School of Clinical Dentistry, University of Sheffield, Sheffield, United Kingdom
| | - Keith D. Hunter
- Unit of Oral and Maxillofacial Medicine, Pathology and Surgery, School of Clinical Dentistry, University of Sheffield, Sheffield, United Kingdom
- Oral Biology and Pathology, University of Pretoria, Pretoria, South Africa
| |
Collapse
|
462
|
Hicks KC, Chariou PL, Ozawa Y, Minnar CM, Knudson KM, Meyer TJ, Bian J, Cam M, Schlom J, Gameiro SR. Tumour-targeted interleukin-12 and entinostat combination therapy improves cancer survival by reprogramming the tumour immune cell landscape. Nat Commun 2021; 12:5151. [PMID: 34446712 PMCID: PMC8390765 DOI: 10.1038/s41467-021-25393-x] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 07/30/2021] [Indexed: 01/01/2023] Open
Abstract
Poorly inflamed carcinomas do not respond well to immune checkpoint blockade. Converting the tumour microenvironment into a functionally inflamed immune hub would extend the clinical benefit of immune therapy to a larger proportion of cancer patients. Here we show, by using comprehensive single-cell transcriptome, proteome, and immune cell analysis, that Entinostat, a class I histone deacetylase inhibitor, facilitates accumulation of the necrosis-targeted recombinant murine immune-cytokine, NHS-rmIL12, in experimental mouse colon carcinomas and poorly immunogenic breast tumours. This combination therapy reprograms the tumour innate and adaptive immune milieu to an inflamed landscape, where the concerted action of highly functional CD8+ T cells and activated neutrophils drive macrophage M1-like polarization, leading to complete tumour eradication in 41.7%-100% of cases. Biomarker signature of favourable overall survival in multiple human tumor types shows close resemblance to the immune pattern generated by Entinostat/NHS-rmIL12 combination therapy. Collectively, these findings provide a rationale for combining NHS-IL12 with Entinostat in the clinical setting.
Collapse
Affiliation(s)
- Kristin C Hicks
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Paul L Chariou
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Yohei Ozawa
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Christine M Minnar
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Karin M Knudson
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Thomas J Meyer
- CCR Collaborative Bioinformatics Resource (CCBR), Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jing Bian
- CCR Collaborative Bioinformatics Resource (CCBR), Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Margaret Cam
- CCR Collaborative Bioinformatics Resource (CCBR), Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jeffrey Schlom
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| | - Sofia R Gameiro
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
463
|
Das B, Sarkar C, Rawat VS, Kalita D, Deka S, Agnihotri A. Promise of the NLRP3 Inflammasome Inhibitors in In Vivo Disease Models. Molecules 2021; 26:4996. [PMID: 34443594 PMCID: PMC8399941 DOI: 10.3390/molecules26164996] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/28/2021] [Accepted: 08/10/2021] [Indexed: 12/26/2022] Open
Abstract
Nucleotide-binding oligomerization domain NOD-like receptors (NLRs) are conserved cytosolic pattern recognition receptors (PRRs) that track the intracellular milieu for the existence of infection, disease-causing microbes, as well as metabolic distresses. The NLRP3 inflammasome agglomerates are consequent to sensing a wide spectrum of pathogen-associated molecular patterns (PAMPs) and danger-associated molecular patterns (DAMPs). Certain members of the NLR family have been documented to lump into multimolecular conglomerates called inflammasomes, which are inherently linked to stimulation of the cysteine protease caspase-1. Following activation, caspase-1 severs the proinflammatory cytokines interleukin (IL)-1β and IL-18 to their biologically active forms, with consequent commencement of caspase-1-associated pyroptosis. This type of cell death by pyroptosis epitomizes a leading pathway of inflammation. Accumulating scientific documentation has recorded overstimulation of NLRP3 (NOD-like receptor protein 3) inflammasome involvement in a wide array of inflammatory conditions. IL-1β is an archetypic inflammatory cytokine implicated in multiple types of inflammatory maladies. Approaches to impede IL-1β's actions are possible, and their therapeutic effects have been clinically demonstrated; nevertheless, such strategies are associated with certain constraints. For instance, treatments that focus on systemically negating IL-1β (i.e., anakinra, rilonacept, and canakinumab) have been reported to result in an escalated peril of infections. Therefore, given the therapeutic promise of an NLRP3 inhibitor, the concerted escalated venture of the scientific sorority in the advancement of small molecules focusing on direct NLRP3 inflammasome inhibition is quite predictable.
Collapse
Affiliation(s)
- Biswadeep Das
- Department of Pharmacology, All India Institute of Medical Sciences (AIIMS), Virbhadra Road, Rishikesh 249203, Uttarakhand, India;
| | - Chayna Sarkar
- Department of Clinical Pharmacology & Therapeutics, North Eastern Indira Gandhi Regional Institute of Health and Medical Sciences (NEIGRIHMS), Mawdiangdiang, Shillong 793018, Meghalaya, India;
| | - Vikram Singh Rawat
- Department of Psychiatry, All India Institute of Medical Sciences (AIIMS), Virbhadra Road, Rishikesh 249203, Uttarakhand, India;
| | - Deepjyoti Kalita
- Department of Microbiology, All India Institute of Medical Sciences (AIIMS), Virbhadra Road, Rishikesh 249203, Uttarakhand, India; (D.K.); (S.D.)
| | - Sangeeta Deka
- Department of Microbiology, All India Institute of Medical Sciences (AIIMS), Virbhadra Road, Rishikesh 249203, Uttarakhand, India; (D.K.); (S.D.)
| | - Akash Agnihotri
- Department of Pharmacology, All India Institute of Medical Sciences (AIIMS), Virbhadra Road, Rishikesh 249203, Uttarakhand, India;
| |
Collapse
|
464
|
Ciaglia E, Lopardo V, Montella F, Sellitto C, Manzo V, De Bellis E, Iannaccone T, Franci G, Zannella C, Pagliano P, Di Pietro P, Carrizzo A, Vecchione C, Conti V, Filippelli A, Puca AA. BPIFB4 Circulating Levels and Its Prognostic Relevance in COVID-19. J Gerontol A Biol Sci Med Sci 2021; 76:1775-1783. [PMID: 34396395 PMCID: PMC8436991 DOI: 10.1093/gerona/glab208] [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: 04/27/2021] [Indexed: 02/06/2023] Open
Abstract
Aging and comorbidities make individuals at greatest risk of COVID-19 serious illness and mortality due to senescence-related events and deleterious inflammation. Long-living individuals (LLIs) are less susceptible to inflammation and develop more resiliency to COVID-19. As demonstrated, LLIs are characterized by high circulating levels of BPIFB4, a protein involved in homeostatic response to inflammatory stimuli. Also, LLIs show enrichment of homozygous genotype for the minor alleles of a 4 missense single-nucleotide polymorphism haplotype (longevity-associated variant [LAV]) in BPIFB4, able to counteract progression of diseases in animal models. Thus, the present study was designed to assess the presence and significance of BPIFB4 level in COVID-19 patients and the potential therapeutic use of LAV-BPIFB4 in fighting COVID-19. BPIFB4 plasma concentration was found significantly higher in LLIs compared to old healthy controls while it significantly decreased in 64 COVID-19 patients. Further, the drop in BPIFB4 values correlated with disease severity. Accordingly to the LAV-BPIFB4 immunomodulatory role, while lysates of SARS-CoV-2-infected cells induced an inflammatory response in healthy peripheral blood mononuclear cells in vitro, the co-treatment with recombinant protein (rh) LAV-BPIFB4 resulted in a protective and self-limiting reaction, culminating in the downregulation of CD69 activating-marker for T cells (both TCD4+ and TCD8+) and in MCP-1 reduction. On the contrary, rhLAV-BPIFB4 induced a rapid increase in IL-18 and IL-1b levels, shown largely protective during the early stages of the virus infection. This evidence, along with the ability of rhLAV-BPIFB4 to counteract the cytotoxicity induced by SARS-CoV-2 lysate in selected target cell lines, corroborates BPIFB4 prognostic value and open new therapeutic possibilities in more vulnerable people.
Collapse
Affiliation(s)
- Elena Ciaglia
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana," University of Salerno, Baronissi, Italy
| | - Valentina Lopardo
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana," University of Salerno, Baronissi, Italy
| | - Francesco Montella
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana," University of Salerno, Baronissi, Italy
| | - Carmine Sellitto
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana," University of Salerno, Baronissi, Italy.,Department of Clinical Pharmacology and Pharmacogenetics Unit, University Hospital "San Giovanni di Dio e Ruggi d'Aragona," Salerno, Italy
| | - Valentina Manzo
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana," University of Salerno, Baronissi, Italy.,Department of Clinical Pharmacology and Pharmacogenetics Unit, University Hospital "San Giovanni di Dio e Ruggi d'Aragona," Salerno, Italy
| | - Emanuela De Bellis
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana," University of Salerno, Baronissi, Italy.,Department of Clinical Pharmacology and Pharmacogenetics Unit, University Hospital "San Giovanni di Dio e Ruggi d'Aragona," Salerno, Italy
| | - Teresa Iannaccone
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana," University of Salerno, Baronissi, Italy
| | - Gianluigi Franci
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana," University of Salerno, Baronissi, Italy.,Department of Experimental Medicine, University of Campania "Luigi Vanvitelli," Naples, Italy
| | - Carla Zannella
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli," Naples, Italy
| | - Pasquale Pagliano
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana," University of Salerno, Baronissi, Italy.,Infectious Diseases Unit, University Hospital "San Giovanni di Dio e Ruggi d'Aragona," Salerno, Italy
| | - Paola Di Pietro
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana," University of Salerno, Baronissi, Italy
| | - Albino Carrizzo
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana," University of Salerno, Baronissi, Italy.,Department of AngioCardioNeurology, IRCCS Neuromed, Pozzilli, Isernia, Italy
| | - Carmine Vecchione
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana," University of Salerno, Baronissi, Italy.,Department of AngioCardioNeurology, IRCCS Neuromed, Pozzilli, Isernia, Italy
| | - Valeria Conti
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana," University of Salerno, Baronissi, Italy.,Department of Clinical Pharmacology and Pharmacogenetics Unit, University Hospital "San Giovanni di Dio e Ruggi d'Aragona," Salerno, Italy
| | - Amelia Filippelli
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana," University of Salerno, Baronissi, Italy.,Department of Clinical Pharmacology and Pharmacogenetics Unit, University Hospital "San Giovanni di Dio e Ruggi d'Aragona," Salerno, Italy
| | - Annibale Alessandro Puca
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana," University of Salerno, Baronissi, Italy.,Cardiovascular Research Unit, IRCCS MultiMedica, Milan, Italy
| |
Collapse
|
465
|
Chen R, Zhang S, Su S, Ye H, Shu H. Interactions Between Specific Immune Status of Pregnant Women and SARS-CoV-2 Infection. Front Cell Infect Microbiol 2021; 11:721309. [PMID: 34458162 PMCID: PMC8387674 DOI: 10.3389/fcimb.2021.721309] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Accepted: 07/29/2021] [Indexed: 12/18/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the pathogen responsible for the Coronavirus Disease 2019 (COVID-19) global pandemic. Because it is a new and highly contagious coronavirus, most people, especially pregnant women, lack immunity. It is therefore important to understand the interaction between why pregnant women are susceptible to SARS-CoV-2 and the specific immune systems of pregnant women. Here, we provide an overview of the changes that occur in the immune system during pregnancy, the activation and response of the immune system in pregnant women with COVID-19, adverse pregnancy outcomes in pregnant women with COVID-19, and the treatment and prevention of COVID-19 in this population.
Collapse
Affiliation(s)
- Ruirong Chen
- Department of Anesthesiology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Shaofen Zhang
- Department of Gynaecology and Obstetrics, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangzhou, China
| | - Sheng Su
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Haiyan Ye
- Department of Gynaecology and Obstetrics, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Haihua Shu
- Department of Anesthesiology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| |
Collapse
|
466
|
Hsu SK, Li CY, Lin IL, Syue WJ, Chen YF, Cheng KC, Teng YN, Lin YH, Yen CH, Chiu CC. Inflammation-related pyroptosis, a novel programmed cell death pathway, and its crosstalk with immune therapy in cancer treatment. Theranostics 2021; 11:8813-8835. [PMID: 34522213 PMCID: PMC8419056 DOI: 10.7150/thno.62521] [Citation(s) in RCA: 223] [Impact Index Per Article: 55.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Accepted: 07/12/2021] [Indexed: 12/12/2022] Open
Abstract
In recent decades, chemotherapies targeting apoptosis have emerged and demonstrated remarkable achievements. However, emerging evidence has shown that chemoresistance is mediated by impairing or bypassing apoptotic cell death. Several novel types of programmed cell death, such as ferroptosis, necroptosis, and pyroptosis, have recently been reported to play significant roles in the modulation of cancer progression and are considered a promising strategy for cancer treatment. Thus, the switch between apoptosis and pyroptosis is also discussed. Cancer immunotherapy has gained increasing attention due to breakthroughs in immune checkpoint inhibitors; moreover, ferroptosis, necroptosis, and pyroptosis are highly correlated with the modulation of immunity in the tumor microenvironment. Compared with necroptosis and ferroptosis, pyroptosis is the primary mechanism for host defense and is crucial for bridging innate and adaptive immunity. Furthermore, recent evidence has demonstrated that pyroptosis exerts benefits on cancer immunotherapies, including immune checkpoint inhibitors (ICIs) and chimeric antigen receptor T-cell therapy (CAR-T). Hence, in this review, we elucidate the role of pyroptosis in cancer progression and the modulation of immunity. We also summarize the potential small molecules and nanomaterials that target pyroptotic cell death mechanisms and their therapeutic effects on cancer.
Collapse
Affiliation(s)
- Sheng-Kai Hsu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Chia-Yang Li
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - I-Ling Lin
- Department of Medical Laboratory Science and Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Wun-Jyun Syue
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Yih-Fung Chen
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Kai-Chun Cheng
- Department of Ophthalmology, Kaohsiung Municipal Hsiaokang Hospital, Kaohsiung 812, Taiwan
- Department of Ophthalmology, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| | - Yen-Ni Teng
- Department of Biological Sciences and Technology, National University of Tainan, Tainan 700, Taiwan
| | - Yi-Hsiung Lin
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Medical University Hospital
- Center for Lipid Biosciences, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Lipid Science and Aging Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chia-Hung Yen
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Chien-Chih Chiu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung 804, Taiwan
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| |
Collapse
|
467
|
Soh J, Hamada A, Fujino T, Mitsudomi T. Perioperative Therapy for Non-Small Cell Lung Cancer with Immune Checkpoint Inhibitors. Cancers (Basel) 2021; 13:4035. [PMID: 34439189 PMCID: PMC8391213 DOI: 10.3390/cancers13164035] [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: 07/15/2021] [Revised: 08/04/2021] [Accepted: 08/04/2021] [Indexed: 12/25/2022] Open
Abstract
The emergence of immune checkpoint inhibitors (ICIs) has dramatically changed the treatment landscape for patients with metastatic non-small cell lung cancer (NSCLC). These achievements inspired investigators and pharmaceutical companies to conduct clinical trials in patients with early-stage NSCLC because both adjuvant and neoadjuvant platinum-based doublet chemotherapies (PT-DCs) showed only a 5% improvement in 5-year overall survival. IMpower010, a phase 3 trial (P3), showed that adjuvant PT-DC followed by maintenance atezolitumab significantly prolonged disease-free survival over adjuvant PT-DC alone (hazard ratio, 0.79; stage II to IIIA). Since conventional therapies, including chemotherapy and radiotherapy, can promote immunogenic cell death, releasing tumour antigens from dead tumour cells, ICI combination therapies with conventional therapies are widely proposed. The Checkmate 816 trial (P3) indicated a significantly higher pathological complete response rate of neoadjuvant nivolumab/PT-DC combination therapy than of neoadjuvant PT-DC alone (odds ratio, 13.9, for stage IB to IIIA). Detection of circulating tumour DNA is highly anticipated for the evaluation of minimal residual disease. Multimodal approaches and new ICI agents are being attempted to improve the efficacy of ICI treatment in phase 2 trials. This review presents the development of perioperative treatment using ICIs in patients with NSCLC while discussing problems and perspectives.
Collapse
Affiliation(s)
- Junichi Soh
- Division of Thoracic Surgery, Department of Surgery, Kindai University Faculty of Medicine, 377-2 Ohno-higashi, Osaka-Sayama, Osaka 589-8511, Japan; (A.H.); (T.F.); (T.M.)
| | | | | | | |
Collapse
|
468
|
Abstract
Interleukin-1 (IL-1) is a key orchestrator of inflammation and plays an important role in tumor progression. Based on preclinical models and human genetic associations, we surmise that targeting IL-1 should be considered in treating selected human tumors as well as in a prevention and/or interception setting.
Collapse
Affiliation(s)
- Cecilia Garlanda
- Humanitas Research Hospital-IRCCS, Via Manzoni 56, 20089 Rozzano, Milan, Italy; Humanitas University, Department of Biomedical Sciences, Via Rita Levi Montalcini 4, 20090 Pieve Emanuele, Milan, Italy.
| | - Alberto Mantovani
- Humanitas Research Hospital-IRCCS, Via Manzoni 56, 20089 Rozzano, Milan, Italy; Humanitas University, Department of Biomedical Sciences, Via Rita Levi Montalcini 4, 20090 Pieve Emanuele, Milan, Italy; The William Harvey Research Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK.
| |
Collapse
|
469
|
Yang Z, Schooling CM, Kwok MK. Genetic Evidence on the Association of Interleukin (IL)-1-Mediated Chronic Inflammation with Airflow Obstruction: A Mendelian Randomization Study. COPD 2021; 18:432-442. [PMID: 34348529 DOI: 10.1080/15412555.2021.1955848] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Preclinical studies suggest interleukin (IL)-1α/β is involved in the pathogenesis of chronic obstructive pulmonary disease (COPD). However, recent trials of anti-IL-1 therapies showed limited benefit for COPD. To clarify, we primarily examined total and direct effects of IL-1 and its receptors/coreceptors/receptor antagonists (IL-1/IL-1Rs) on airflow obstruction (AO) using Mendelian randomization (MR), and secondarily explored reverse causation using bidirectional MR. We selected independent cis protein quantitative trait loci (cis-pQTLs) as genetic instruments for IL-1/IL-1Rs from two proteomic genome-wide association studies (n = 11,594) of European ancestry (mean age ∼47 years). We applied those cis-pQTLs to the International COPD Genetics Consortium (n = 15,256 cases, 47,936 controls) of ∼81.9% European descent (∼57 years). No IL-1/IL-1Rs were significantly associated with AO after correction for multiple testing. However, a higher genetically predicted IL-1 receptor antagonist (IL-1Ra) was nominally associated with a 20% reduction in AO risk using univariable MR, with a larger direct effect (∼31%, i.e. not via IL-1α/β) using multivariable MR. Furthermore, higher total IL-18 binding protein (IL-18BP) was nominally associated with lower AO. Nominal total effects were also noted for higher IL-1α with lower AO and higher IL-1R1 with higher AO. Higher IL-1Ra and IL-18BP might have a role in preventing AO, but need to be contextualized.Supplemental data for this article is available online at https://doi.org/10.1080/15412555.2021.1955848 .
Collapse
Affiliation(s)
- Zhao Yang
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special administrative Region, China
| | - C Mary Schooling
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special administrative Region, China.,Graduate School of Public Health and Health Policy, City University of New York, New York, USA
| | - Man Ki Kwok
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special administrative Region, China
| |
Collapse
|
470
|
Levescot A, Chang MH, Schnell J, Nelson-Maney N, Yan J, Martínez-Bonet M, Grieshaber-Bouyer R, Lee PY, Wei K, Blaustein RB, Morris A, Wactor A, Iwakura Y, Lederer JA, Rao DA, Charles JF, Nigrovic PA. IL-1β-driven osteoclastogenic T regulatory cells accelerate bone erosion in arthritis. J Clin Invest 2021; 131:e141008. [PMID: 34343136 DOI: 10.1172/jci141008] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 07/22/2021] [Indexed: 11/17/2022] Open
Abstract
IL-1β is a pro-inflammatory mediator with roles in innate and adaptive immunity. Here we show that IL-1β contributes to autoimmune arthritis by inducing osteoclastogenic capacity in T regulatory cells (Tregs). Using mice with joint inflammation arising through deficiency of the IL-1 receptor antagonist (Il1rn-/-), we observed that IL-1β blockade attenuated disease more effectively in early arthritis than in established arthritis, especially with respect to bone erosion. Protection was accompanied by a reduction in synovial CD4+Foxp3+ Tregs that displayed preserved suppressive capacity and aerobic metabolism but aberrant expression of RANKL and a striking capacity to drive RANKL-dependent osteoclast differentiation. Both Il1rn-/- Tregs and wild-type Tregs differentiated with IL-1β accelerated bone erosion upon adoptive transfer. Human Tregs exhibited analogous differentiation, and corresponding RANKLhiFoxp3+ T cells could be identified in rheumatoid arthritis synovial tissue. Together, these findings identify IL-1β-induced osteoclastogenic Tregs (O-Tregs) as a contributor to bone erosion in arthritis.
Collapse
Affiliation(s)
- Anaïs Levescot
- Department of Medicine, Brigham and Women's Hospital, Boston, United States of America
| | - Margaret H Chang
- Department of Medicine, Brigham and Women's Hospital, Boston, United States of America
| | - Julia Schnell
- Department of Medicine V, Hematology, Oncology and Rheumatology, University of Heidelberg, Heidelberg, Germany
| | - Nathan Nelson-Maney
- Department of Medicine, Brigham and Women's Hospital, Boston, United States of America
| | - Jing Yan
- Department of Medicine, Brigham and Women's Hospital, Boston, United States of America
| | - Marta Martínez-Bonet
- Department of Medicine, Brigham and Women's Hospital, Boston, United States of America
| | | | - Pui Y Lee
- Division of Immunology, Boston's Children Hospital, Boston, United States of America
| | - Kevin Wei
- Department of Medicine, Brigham and Women's Hospital, Boston, United States of America
| | - Rachel B Blaustein
- Department of Medicine, Brigham and Women's Hospital, Boston, United States of America
| | - Allyn Morris
- Department of Medicine, Brigham and Women's Hospital, Boston, United States of America
| | - Alexandra Wactor
- Department of Medicine, Brigham and Women's Hospital, Boston, United States of America
| | - Yoichiro Iwakura
- Research Institute for Science and Technology, Tokyo University of Science, Tokyo, Japan
| | - James A Lederer
- Department of Surgery, Brigham and Women's Hospital, Boston, United States of America
| | - Deepak A Rao
- Department of Medicine, Brigham and Women's Hospital, Boston, United States of America
| | - Julia F Charles
- Department of Medicine, Brigham and Women's Hospital, Boston, United States of America
| | - Peter A Nigrovic
- Division of Immunology, Boston's Children Hospital, Boston, United States of America
| |
Collapse
|
471
|
Liu G, Särén L, Douglasson H, Zhou XH, Åberg PM, Ollerstam A, Betts CJ, Balogh Sivars K. Precision cut lung slices: an ex vivo model for assessing the impact of immunomodulatory therapeutics on lung immune responses. Arch Toxicol 2021; 95:2871-2877. [PMID: 34191076 DOI: 10.1007/s00204-021-03096-y] [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: 04/24/2021] [Accepted: 06/15/2021] [Indexed: 11/28/2022]
Abstract
Chronic inflammatory diseases of the respiratory tract, such as chronic obstructive pulmonary disease (COPD) and asthma, are severe lung diseases that require effective treatments. In search for new medicines for these diseases, there is an unmet need for predictive and translatable disease-relevant in vitro/ex vivo models to determine the safety and efficacy of novel drug candidates. Here, we report the use of precision cut lung slices (PCLS) as a potential ex vivo platform to study compound effects in a physiologically relevant environment. PCLS derived from an elastase-challenged mouse model display key characteristics of increased inflammation ex vivo, which is exacerbated further upon challenge with LPS, mimicking the immune insult of a pathogen triggering disease exacerbation. Such LPS-induced inflammatory conditions are significantly abrogated by immunomodulatory agents targeting specific inflammatory signaling pathways in the absence of cytotoxic effects in lung slices. Thus, an ex vivo model of PCLS with a simulated pathogenic insult can replicate proposed in vivo pharmacological effects and thus could potentially act as a valuable tool to investigate the underlying mechanisms associated with lung safety, therapeutic efficacy and exacerbations with infection.
Collapse
Affiliation(s)
- Guanghui Liu
- Respiratory and Immunology Safety, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Linnea Särén
- Animal Sciences and Technologies, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Helena Douglasson
- Bioscience Cough and in Vivo, Early Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Xiao-Hong Zhou
- Patient Safety, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Per M Åberg
- Respiratory and Immunology Safety, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Anna Ollerstam
- Respiratory and Immunology Safety, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Catherine J Betts
- Functional and Mechanistic Safety, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Cambridge, UK.
| | - Kinga Balogh Sivars
- Clinical Testing, Global Procurement, Operations, AstraZeneca, Gothenburg, Sweden.
| |
Collapse
|
472
|
Briukhovetska D, Dörr J, Endres S, Libby P, Dinarello CA, Kobold S. Interleukins in cancer: from biology to therapy. Nat Rev Cancer 2021; 21:481-499. [PMID: 34083781 PMCID: PMC8173513 DOI: 10.1038/s41568-021-00363-z] [Citation(s) in RCA: 389] [Impact Index Per Article: 97.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/13/2021] [Indexed: 02/06/2023]
Abstract
Interleukins and associated cytokines serve as the means of communication for innate and adaptive immune cells as well as non-immune cells and tissues. Thus, interleukins have a critical role in cancer development, progression and control. Interleukins can nurture an environment enabling and favouring cancer growth while simultaneously being essential for a productive tumour-directed immune response. These properties of interleukins can be exploited to improve immunotherapies to promote effectiveness as well as to limit side effects. This Review aims to unravel some of these complex interactions.
Collapse
Affiliation(s)
- Daria Briukhovetska
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, LMU, Munich, Germany
| | - Janina Dörr
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, LMU, Munich, Germany
| | - Stefan Endres
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, LMU, Munich, Germany
- German Center for Translational Cancer Research (DKTK), Munich, Germany
- Einheit für Klinische Pharmakologie (EKLiP), Helmholtz Zentrum München, German Research Center for Environmental Health (HMGU), Neuherberg, Germany
| | - Peter Libby
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Sebastian Kobold
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, LMU, Munich, Germany.
- German Center for Translational Cancer Research (DKTK), Munich, Germany.
- Einheit für Klinische Pharmakologie (EKLiP), Helmholtz Zentrum München, German Research Center for Environmental Health (HMGU), Neuherberg, Germany.
| |
Collapse
|
473
|
Conti P, Pregliasco FE, Bellomo RG, Gallenga CE, Caraffa A, Kritas SK, Lauritano D, Ronconi G. Mast Cell Cytokines IL-1, IL-33, and IL-36 Mediate Skin Inflammation in Psoriasis: A Novel Therapeutic Approach with the Anti-Inflammatory Cytokines IL-37, IL-38, and IL-1Ra. Int J Mol Sci 2021; 22:ijms22158076. [PMID: 34360845 PMCID: PMC8348737 DOI: 10.3390/ijms22158076] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 07/22/2021] [Accepted: 07/25/2021] [Indexed: 12/18/2022] Open
Abstract
Psoriasis (PS) is a skin disease with autoimmune features mediated by immune cells, which typically presents inflammatory erythematous plaques, and is associated with many comorbidities. PS exhibits excessive keratinocyte proliferation, and a high number of immune cells, including macrophages, neutrophils, Th1 and Th17 lymphocytes, and mast cells (MCs). MCs are of hematopoietic origin, derived from bone marrow cells, which migrate, mature, and reside in vascularized tissues. They can be activated by antigen-provoking overexpression of proinflammatory cytokines, and release a number of mediators including interleukin (IL)-1 and IL-33. IL-1, released by activated keratinocytes and MCs, stimulates skin macrophages to release IL-36—a powerful proinflammatory IL-1 family member. IL-36 mediates both innate and adaptive immunity, including chronic proinflammatory diseases such as psoriasis. Suppression of IL-36 could result in a dramatic improvement in the treatment of psoriasis. IL-36 is inhibited by IL-36Ra, which binds to IL-36 receptor ligands, but suppression can also occur by binding IL-38 to the IL-36 receptor (IL-36R). IL-38 specifically binds only to IL-36R, and inhibits human mononuclear cells stimulated with IL-36 in vitro, sharing the effect with IL-36Ra. Here, we report that inflammation in psoriasis is mediated by IL-1 generated by MCs—a process that activates macrophages to secrete proinflammatory IL-36 inhibited by IL-38. IL-37 belongs to the IL-1 family, and broadly suppresses innate inflammation via IL-1 inhibition. IL-37, in murine models of inflammatory arthritis, causes the suppression of joint inflammation through the inhibition of IL-1. Therefore, it is pertinent to think that IL-37 can play an inhibitory role in inflammatory psoriasis. In this article, we confirm that IL-38 and IL-37 cytokines emerge as inhibitors of inflammation in psoriasis, and hold promise as an innovative therapeutic tool.
Collapse
Affiliation(s)
- Pio Conti
- Postgraduate Medical School, University of Chieti, 66100 Chieti, Italy
- Correspondence: ; Tel.: +39-0871-574136
| | | | - Rosa G. Bellomo
- Facoltà di Scienze dell’Educazione Motoria, Università “Carlo Bo”, 61029 Urbino, Italy;
| | - Carla E. Gallenga
- Department of Biomedical Sciences and Specialist Surgery, University of Ferrara, 44100 Ferrara, Italy;
| | | | - Spyros K. Kritas
- Department of Microbiology and Infectious Diseases, Aristotle University of Thessaloniki, 54250 Macedonia, Greece;
| | - Dorina Lauritano
- Medicine and Surgery Centre of Neuroscience of Milan, University of Milan-Bicocca, 20100 Milano, Italy;
| | - Gianpaolo Ronconi
- Clinica dei Pazienti del Territorio, Fondazione Policlinico Gemelli, 00168 Rome, Italy;
| |
Collapse
|
474
|
Waldron JL, Schworer SA, Kwan M. Hypersensitivity and Immune-related Adverse Events in Biologic Therapy. Clin Rev Allergy Immunol 2021; 62:413-431. [PMID: 34319562 DOI: 10.1007/s12016-021-08879-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/13/2021] [Indexed: 12/13/2022]
Abstract
Biologic medications are an expanding field of therapeutics for various medical conditions including cancer and inflammatory diseases. Due to their targeted approach to therapy, biologics can be less toxic than traditional systemic medications. However, as use becomes more widespread, adverse effects from biologic administration have also become apparent. Immune-related adverse events are a common mechanism by which biologics can cause on-target immune-related toxicities and both immediate and delayed-type hypersensitivity reactions. Immediate hypersensitivity reactions can be mediated by cytokine release or antibody mediated reactions, while delayed-type hypersensitivity is most often caused by serum sickness-like reactions. Additionally, biologics used for treatment of cancer using checkpoint blockade and rheumatologic disease using cytokine blockade can result in autoimmunity. Finally, when inflammatory cytokines are targeted for treatment of autoimmune or autoinflammatory disease, the host immune defense can be compromised predisposing to secondary immunodeficiency. This review will discuss the mechanisms of these reactions and discuss examples of biologics implicated in each of these adverse events.
Collapse
Affiliation(s)
- Jamie L Waldron
- Department of Medicine, Division of Rheumatology, Allergy, and Immunology, UNC School of Medicine, Chapel Hill, NC, USA
| | - Stephen A Schworer
- Department of Pediatrics, Division of Allergy & Immunology, UNC School of Medicine, Chapel Hill, NC, USA
| | - Mildred Kwan
- Department of Medicine, Division of Rheumatology, Allergy, and Immunology, UNC School of Medicine, Chapel Hill, NC, USA.
| |
Collapse
|
475
|
Grauers Wiktorin H, Aydin E, Christenson K, Issdisai N, Thorén FB, Hellstrand K, Martner A. Impact of IL-1β and the IL-1R antagonist on relapse risk and survival in AML patients undergoing immunotherapy for remission maintenance. Oncoimmunology 2021; 10:1944538. [PMID: 34367728 PMCID: PMC8317920 DOI: 10.1080/2162402x.2021.1944538] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Interleukin-1 beta (IL-1β), a pro-inflammatory cytokine, has been ascribed a role in the expansion of myeloid progenitors in acute myeloid leukemia (AML) and in promoting myeloid cell-induced suppression of lymphocyte-mediated immunity against malignant cells. This study aimed at defining the potential impact of IL-1β in the post-remission phase of AML patients receiving immunotherapy for relapse prevention in an international phase IV trial of 84 patients (ClinicalTrials.gov; NCT01347996). Consecutive serum samples were collected from AML patients in first complete remission (CR) who received cycles of relapse-preventive immunotherapy with histamine dihydrochloride (HDC) and low-dose interleukin-2 (IL-2). Low IL-1β serum levels before and after the first HDC/IL-2 treatment cycle favorably prognosticated leukemia-free survival and overall survival. Serum levels of IL-1β were significantly reduced in patients receiving HDC/IL-2. HDC also reduced the formation of IL-1β from activated human PBMCs in vitro. Additionally, high serum levels of the IL-1 receptor antagonist IL-1RA were associated with favorable outcome, and AML patients with low IL-1β along with high IL-1RA levels were strikingly protected against leukemic relapse. Our results suggest that strategies to target IL-1β might impact on relapse risk and survival in AML.
Collapse
Affiliation(s)
- Hanna Grauers Wiktorin
- TIMM Laboratory, Sahlgrenska Center for Cancer Research, Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Ebru Aydin
- TIMM Laboratory, Sahlgrenska Center for Cancer Research, Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Sweden.,Molecular Genetics, Germany Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Karin Christenson
- TIMM Laboratory, Sahlgrenska Center for Cancer Research, Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Sweden.,Department of Oral Microbiology and Immunology, Institute of Odontology, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Nuttida Issdisai
- TIMM Laboratory, Sahlgrenska Center for Cancer Research, Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Fredrik B Thorén
- TIMM Laboratory, Sahlgrenska Center for Cancer Research, Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Kristoffer Hellstrand
- TIMM Laboratory, Sahlgrenska Center for Cancer Research, Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Anna Martner
- TIMM Laboratory, Sahlgrenska Center for Cancer Research, Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Sweden
| |
Collapse
|
476
|
Systemic IL-15, IFN-γ, and IP-10/CXCL10 signature associated with effective immune response to SARS-CoV-2 in BNT162b2 mRNA vaccine recipients. Cell Rep 2021; 36:109504. [PMID: 34352226 PMCID: PMC8299183 DOI: 10.1016/j.celrep.2021.109504] [Citation(s) in RCA: 133] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 06/16/2021] [Accepted: 07/19/2021] [Indexed: 12/29/2022] Open
Abstract
Early responses to vaccination are important for shaping both humoral and cellular protective immunity. Dissecting innate vaccine signatures may predict immunogenicity to help optimize the efficacy of mRNA and other vaccine strategies. Here, we characterize the cytokine and chemokine responses to the 1st and 2nd dose of the BNT162b2 mRNA (Pfizer/BioNtech) vaccine in antigen-naive and in previously coronavirus disease 2019 (COVID-19)-infected individuals (NCT04743388). Transient increases in interleukin-15 (IL-15) and interferon gamma (IFN-γ) levels early after boost correlate with Spike antibody levels, supporting their use as biomarkers of effective humoral immunity development in response to vaccination. We identify a systemic signature including increases in IL-15, IFN-γ, and IP-10/CXCL10 after the 1st vaccination, which were enriched by tumor necrosis factor alpha (TNF-α) and IL-6 after the 2nd vaccination. In previously COVID-19-infected individuals, a single vaccination results in both strong cytokine induction and antibody titers similar to the ones observed upon booster vaccination in antigen-naive individuals, a result with potential implication for future public health recommendations.
Collapse
|
477
|
Cytokine-Laden Extracellular Vesicles Predict Patient Prognosis after Cerebrovascular Accident. Int J Mol Sci 2021; 22:ijms22157847. [PMID: 34360613 PMCID: PMC8345931 DOI: 10.3390/ijms22157847] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 07/18/2021] [Accepted: 07/20/2021] [Indexed: 12/20/2022] Open
Abstract
Background: A major contributor to disability after hemorrhagic stroke is secondary brain damage induced by the inflammatory response. Following stroke, global increases in numerous cytokines—many associated with worse outcomes—occur within the brain, cerebrospinal fluid, and peripheral blood. Extracellular vesicles (EVs) may traffic inflammatory cytokines from damaged tissue within the brain, as well as peripheral sources, across the blood–brain barrier, and they may be a critical component of post-stroke neuroinflammatory signaling. Methods: We performed a comprehensive analysis of cytokine concentrations bound to plasma EV surfaces and/or sequestered within the vesicles themselves. These concentrations were correlated to patient acute neurological condition by the Glasgow Coma Scale (GCS) and to chronic, long-term outcome via the Glasgow Outcome Scale-Extended (GOS-E). Results: Pro-inflammatory cytokines detected from plasma EVs were correlated to worse outcomes in hemorrhagic stroke patients. Anti-inflammatory cytokines detected within EVs were still correlated to poor outcomes despite their putative neuroprotective properties. Inflammatory cytokines macrophage-derived chemokine (MDC/CCL2), colony stimulating factor 1 (CSF1), interleukin 7 (IL7), and monokine induced by gamma interferon (MIG/CXCL9) were significantly correlated to both negative GCS and GOS-E when bound to plasma EV membranes. Conclusions: These findings correlate plasma-derived EV cytokine content with detrimental outcomes after stroke, highlighting the potential for EVs to provide cytokines with a means of long-range delivery of inflammatory signals that perpetuate neuroinflammation after stroke, thus hindering recovery.
Collapse
|
478
|
Ren AQ, Wang HJ, Zhu HY, Ye G, Li K, Chen DF, Zeng T, Li H. Glycoproteins From Rabdosia japonica var. glaucocalyx Regulate Macrophage Polarization and Alleviate Lipopolysaccharide-Induced Acute Lung Injury in Mice via TLR4/NF-κB Pathway. Front Pharmacol 2021; 12:693298. [PMID: 34366849 PMCID: PMC8333617 DOI: 10.3389/fphar.2021.693298] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Accepted: 06/24/2021] [Indexed: 01/04/2023] Open
Abstract
Background and Aims:Rabdosia japonica var. glaucocalyx is a traditional Chinese medicine (TCM) for various inflammatory diseases. This present work aimed to investigate the protective effects of R. japonica var. glaucocalyx glycoproteins on lipopolysaccharide (LPS)-induced acute lung injury (ALI) and the potential mechanism. Methods: Glycoproteins (XPS) were isolated from R. japonica var. glaucocalyx, and homogeneous glycoprotein (XPS5-1) was purified from XPS. ANA-1 cells were used to observe the effect of glycoproteins on the secretion of inflammatory mediators by enzyme-linked immunosorbent assay (ELISA). Flow cytometry assay, immunofluorescence assay, and Western blot analysis were performed to detect macrophage polarization in vitro. The ALI model was induced by LPS via intratracheal instillation, and XPS (20, 40, and 80 mg/kg) was administered intragastrically 2 h later. The mechanisms of XPS against ALI were investigated by Western blot, ELISA, and immunohistochemistry. Results:In vitro, XPS and XPS5-1 downregulated LPS-induced proinflammatory mediators production including tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), IL-6, and nitric oxide (NO) and upregulated LPS-induced IL-10 secretion. The LPS-stimulated macrophage polarization was also modulated from M1 to M2. In vivo, XPS maintained pulmonary histology with significantly reducing protein concentration and numbers of mononuclear cells in bronchoalveolar lavage fluid (BALF). The level of IL-10 in BALF was upregulated by XPS treatment. The level of cytokines including TNF-α, IL-1β, and IL-6 was downregulated. XPS also decreased infiltration of macrophages and polymorphonuclear leukocytes (PMNs) in lung. XPS suppressed the expression of key proteins in the TLR4/NF-κB signal pathway. Conclusion: XPS was demonstrated to be a potential agent for treating ALI. Our findings might provide evidence supporting the traditional application of R. japonica var. glaucocalyx in inflammation-linked diseases.
Collapse
Affiliation(s)
- An-Qi Ren
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Hui-Jun Wang
- The MOE Key Laboratory for Standardization of Chinese Medicines and the SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hai-Yan Zhu
- Department of Biological Medicines and Shanghai Engineering Research Center of Immuno Therapeutics, School of Pharmacy, Fudan University, Shanghai, China
| | - Guan Ye
- Central Research Institute, Shanghai Pharmaceuticals Holding Co., Ltd., Shanghai, China
| | - Kun Li
- Central Research Institute, Shanghai Pharmaceuticals Holding Co., Ltd., Shanghai, China
| | - Dao-Feng Chen
- Department of Pharmacognosy, School of Pharmacy, Fudan University, Shanghai, China
| | - Tao Zeng
- Clinical Trial Institution, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Hong Li
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| |
Collapse
|
479
|
Namba T, Ichii O, Nakamura T, Masum MA, Otani Y, Hosotani M, Elewa YHA, Kon Y. Compartmentalization of interleukin 36 subfamily according to inducible and constitutive expression in the kidneys of a murine autoimmune nephritis model. Cell Tissue Res 2021; 386:59-77. [PMID: 34287716 DOI: 10.1007/s00441-021-03495-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 06/21/2021] [Indexed: 12/22/2022]
Abstract
The interleukin (IL) 36 subfamily belongs to the IL-1 family and is comprised of agonists (IL-36α, IL-36β, IL-36γ) and antagonists (IL-36Ra, IL-38). We previously reported IL-36α overexpression in renal tubules of chronic nephritis mice. To understand the localization status and biological relationships among each member of the IL-36 subfamily in the kidneys, MRL/MpJ-Faslpr/lpr mice were investigated as autoimmune nephritis models using pathology-based techniques. MRL/MpJ-Faslpr/lpr mice exhibited disease onset from 3 months and severe nephritis at 6-7 months (early and late stages, respectively). Briefly, IL-36γ and IL-36Ra were constitutively expressed in murine kidneys, while the expression of IL-36α, IL-36β, IL-36Ra, and IL-38 was induced in MRL/MpJ-Faslpr/lpr mice. IL-36α expression was significantly increased and localized to injured tubular epithelial cells (TECs). CD44+-activated parietal epithelial cells (PECs) also exhibited higher IL-36α-positive rates, particularly in males. IL-36β and IL-38 are expressed in interstitial plasma cells. Quantitative indices for IL-36α and IL-38 positively correlated with nephritis severity. Similar to IL-36α, IL-36Ra localized to TECs and PECs at the late stage; however, MRL/MpJ-Faslpr/lpr and healthy MRL/MpJ mice possessed IL-36Ra+ smooth muscle cells in kidney arterial tunica media at both stages. IL-36γ was constitutively expressed in renal sympathetic axons regardless of strain and stage. IL-36 receptor gene was ubiquitously expressed in the kidneys and was induced proportional to disease severity. MRL/MpJ-Faslpr/lpr mice kidneys possessed significantly upregulated IL-36 downstream candidates, including NF-κB- or MAPK-pathway organizing molecules. Thus, the IL-36 subfamily contributes to homeostasis and inflammation in the kidneys, and especially, an IL-36α-dominant imbalance could strongly impact nephritis deterioration.
Collapse
Affiliation(s)
- Takashi Namba
- Laboratory of Anatomy, Department of Basic Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido, 060-0818, Japan
| | - Osamu Ichii
- Laboratory of Anatomy, Department of Basic Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido, 060-0818, Japan.
- Laboratory of Agrobiomedical Science, Faculty of Agriculture, Hokkaido University, Sapporo, Hokkaido, 060-8589, Japan.
| | - Teppei Nakamura
- Laboratory of Anatomy, Department of Basic Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido, 060-0818, Japan
- Section of Biological Safety Research, Chitose Laboratory, Japan Food Research Laboratories, Chitose, Hokkaido, 066-0052, Japan
| | - Md Abdul Masum
- Laboratory of Anatomy, Department of Basic Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido, 060-0818, Japan
- Department of Anatomy, Histology and Physiology, Faculty of Animal Science and Veterinary Medicine, Sher-E-Bangla Agricultural University, Dhaka, 1207, Bangladesh
| | - Yuki Otani
- Laboratory of Anatomy, Department of Basic Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido, 060-0818, Japan
| | - Marina Hosotani
- Laboratory of Veterinary Anatomy, Department of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Hokkaido, 069-8501, Japan
| | - Yaser Hosny Ali Elewa
- Laboratory of Anatomy, Department of Basic Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido, 060-0818, Japan
- Department of Histology and Cytology, Faculty of Veterinary Medicine, Zagazig University, Zigazig, 44519, Egypt
| | - Yasuhiro Kon
- Laboratory of Anatomy, Department of Basic Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido, 060-0818, Japan
| |
Collapse
|
480
|
Ferreira BA, Toyama D, Henrique-Silva F, Araújo FDA. Recombinant sugarcane cystatin CaneCPI-5 down regulates inflammation and promotes angiogenesis and collagen deposition in a mouse subcutaneous sponge model. Int Immunopharmacol 2021; 96:107801. [PMID: 34162162 DOI: 10.1016/j.intimp.2021.107801] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 05/17/2021] [Accepted: 05/17/2021] [Indexed: 11/30/2022]
Abstract
Cystatins are natural inhibitors of cysteine peptidases that are found practically in all living organisms. CaneCPI-5 is a sugarcane cystatin with inhibitory activity against human cathepsins B, K and L, which are cysteine proteases highly expressed in a variety of pathological conditions, usually marked by persistent inflammation and processing of the extracellular matrix. This work evaluated the effects of daily administration of the recombinant cystatin CaneCPI-5 [0.01, 0.1 or 1.0 μg in 10 μL of Phosphate-Buffered Saline (PBS)] on the inflammatory, angiogenic and fibrogenic components during chronic inflammatory response induced by subcutaneous sponge implants. The anti-inflammatory effect of treatment with CaneCPI-5 was confirmed by reduction of the levels of the pro-inflammatory mediators TNF-α, CXCL1 and CCL2/JE/MCP-1, as well as the activity of the myeloperoxidase and n-acetyl-β-D-glucosaminidase. Treatment with CaneCPI-5 promoted angiogenesis in the implants, increasing the production of cytokines VEGF and FGF and the formation of new blood vessels. Finally, the administration of the recombinant cystatin favored the production of the pro-fibrogenic cytokine TGF-β1 and collagen deposition next to the implants. Together, these results show the potential therapeutic application of CaneCPI-5 as an anti-inflammatory agent, capable of favoring angiogenesis and fibrogenesis processes, necessary for tissue repair.
Collapse
Affiliation(s)
- Bruno Antonio Ferreira
- Programa de Pós-graduação em Genética e Bioquímica, Instituto de Biotecnologia, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil; Departamento de Ciências Fisiológicas, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | - Danyelle Toyama
- Departamento de Genética e Evolução, Universidade Federal de São Carlos, São Paulo, Brazil
| | - Flávio Henrique-Silva
- Departamento de Genética e Evolução, Universidade Federal de São Carlos, São Paulo, Brazil
| | - Fernanda de Assis Araújo
- Departamento de Ciências Fisiológicas, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil.
| |
Collapse
|
481
|
Wang EY, Mao T, Klein J, Dai Y, Huck JD, Jaycox JR, Liu F, Zhou T, Israelow B, Wong P, Coppi A, Lucas C, Silva J, Oh JE, Song E, Perotti ES, Zheng NS, Fischer S, Campbell M, Fournier JB, Wyllie AL, Vogels CBF, Ott IM, Kalinich CC, Petrone ME, Watkins AE, Dela Cruz C, Farhadian SF, Schulz WL, Ma S, Grubaugh ND, Ko AI, Iwasaki A, Ring AM. Diverse functional autoantibodies in patients with COVID-19. Nature 2021; 595:283-288. [PMID: 34010947 DOI: 10.1038/s41586-021-03631-y] [Citation(s) in RCA: 566] [Impact Index Per Article: 141.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 05/11/2021] [Indexed: 12/15/2022]
Abstract
COVID-19 manifests with a wide spectrum of clinical phenotypes that are characterized by exaggerated and misdirected host immune responses1-6. Although pathological innate immune activation is well-documented in severe disease1, the effect of autoantibodies on disease progression is less well-defined. Here we use a high-throughput autoantibody discovery technique known as rapid extracellular antigen profiling7 to screen a cohort of 194 individuals infected with SARS-CoV-2, comprising 172 patients with COVID-19 and 22 healthcare workers with mild disease or asymptomatic infection, for autoantibodies against 2,770 extracellular and secreted proteins (members of the exoproteome). We found that patients with COVID-19 exhibit marked increases in autoantibody reactivities as compared to uninfected individuals, and show a high prevalence of autoantibodies against immunomodulatory proteins (including cytokines, chemokines, complement components and cell-surface proteins). We established that these autoantibodies perturb immune function and impair virological control by inhibiting immunoreceptor signalling and by altering peripheral immune cell composition, and found that mouse surrogates of these autoantibodies increase disease severity in a mouse model of SARS-CoV-2 infection. Our analysis of autoantibodies against tissue-associated antigens revealed associations with specific clinical characteristics. Our findings suggest a pathological role for exoproteome-directed autoantibodies in COVID-19, with diverse effects on immune functionality and associations with clinical outcomes.
Collapse
Affiliation(s)
- Eric Y Wang
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Tianyang Mao
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Jon Klein
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Yile Dai
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - John D Huck
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Jillian R Jaycox
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Feimei Liu
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Ting Zhou
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Benjamin Israelow
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Patrick Wong
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Andreas Coppi
- Center for Outcomes Research and Evaluation, Yale-New Haven Hospital, New Haven, CT, USA
| | - Carolina Lucas
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Julio Silva
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Ji Eun Oh
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Eric Song
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Emily S Perotti
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Neil S Zheng
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Suzanne Fischer
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Melissa Campbell
- Department of Internal Medicine (Infectious Diseases), Yale School of Medicine, New Haven, CT, USA
| | - John B Fournier
- Department of Internal Medicine (Infectious Diseases), Yale School of Medicine, New Haven, CT, USA
| | - Anne L Wyllie
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| | - Chantal B F Vogels
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| | - Isabel M Ott
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| | - Chaney C Kalinich
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| | - Mary E Petrone
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| | - Anne E Watkins
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| | - Charles Dela Cruz
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Shelli F Farhadian
- Department of Internal Medicine (Infectious Diseases), Yale School of Medicine, New Haven, CT, USA
| | - Wade L Schulz
- Center for Outcomes Research and Evaluation, Yale-New Haven Hospital, New Haven, CT, USA
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Shuangge Ma
- Department of Biostatistics, Yale School of Public Health, New Haven, CT, USA
| | - Nathan D Grubaugh
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| | - Albert I Ko
- Department of Internal Medicine (Infectious Diseases), Yale School of Medicine, New Haven, CT, USA
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| | - Akiko Iwasaki
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA.
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA.
- Howard Hughes Medical Institute, Chevy Chase, MD, USA.
| | - Aaron M Ring
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA.
- Department of Pharmacology, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
482
|
Zheng J, Chen D, Xu J, Ding X, Wu Y, Shen HC, Tan X. Small molecule approaches to treat autoimmune and inflammatory diseases (Part III): Targeting cytokines and cytokine receptor complexes. Bioorg Med Chem Lett 2021; 48:128229. [PMID: 34214508 DOI: 10.1016/j.bmcl.2021.128229] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 06/21/2021] [Accepted: 06/22/2021] [Indexed: 01/14/2023]
Abstract
Chronic and dysregulated cytokine signaling plays an important role in the pathogenic development of many autoimmune and inflammatory diseases. Despite intrinsic challenges in the disruption of interactions between cytokines and cytokine receptors, many first-in-class small-molecule inhibitors have been discovered over the past few years. The third part of the digest series presents recent progress in identifying such inhibitors and highlights the application of novel research tools in the fields of structural biology, computational analysis, screening methods, biophysical/biochemical assays and medicinal chemistry strategy.
Collapse
Affiliation(s)
- Jiamin Zheng
- Department of Medicinal Chemistry, Roche Innovation Center Shanghai, Roche Pharma Research and Early Development, Shanghai 201203, China
| | - Dongdong Chen
- Department of Medicinal Chemistry, Roche Innovation Center Shanghai, Roche Pharma Research and Early Development, Shanghai 201203, China
| | - Jie Xu
- Department of Immunology, Infectious Disease and Ophthalmology, Roche Innovation Center Shanghai, Roche Pharma Research and Early Development, Shanghai 201203, China
| | - Xiao Ding
- Department of Medicinal Chemistry, Roche Innovation Center Shanghai, Roche Pharma Research and Early Development, Shanghai 201203, China
| | - Yao Wu
- Computer Aided Drug Design, Roche Innovation Center Shanghai, Roche Pharma Research and Early Development, Shanghai 201203, China
| | - Hong C Shen
- Department of Medicinal Chemistry, Roche Innovation Center Shanghai, Roche Pharma Research and Early Development, Shanghai 201203, China
| | - Xuefei Tan
- Department of Medicinal Chemistry, Roche Innovation Center Shanghai, Roche Pharma Research and Early Development, Shanghai 201203, China.
| |
Collapse
|
483
|
Evaluation of periostin levels in gingival crevicular fluid in association between coronary heart disease and chronic periodontitis. Dent Res J (Isfahan) 2021; 18:46. [PMID: 34429866 PMCID: PMC8352264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 03/18/2020] [Accepted: 05/03/2020] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Periostin is a protein, which is essential for periodontal tissue integrity, development and maturity. The aim of this study was to evaluate the role of gingival crevicular fluid (GCF) periostin levels in the association between coronary heart disease (CHD) and chronic periodontitis (CP). MATERIALS AND METHODS This matched case-control study was conducted on 116 participants. The participants were matched for age, gender, and body mass index and divided into four groups as follows: (1) 29 patients with CHD and sever CP (CHD-CP), (2) 29 patients with CHD and without CP (CHD-H), (3) 29 patients without CHD and with sever CP (H-CP), and (4) 29 healthy participants (H-H). The GCF periostin was collected and evaluated using the enzyme-linked immunosorbent assay (ELASA). Finally, the data were analyzed by analysis of variance using the stata software. Significance was assigned at P < 0.05. RESULTS The results showed that there was a significant difference in the GCF periostin levels in the four groups (P < 0.05). Moreover, according to the results of the Bonferroni's test, differences in the mean periostin levels were significant (P < 0.001) between CHD-CP and CHD-H, CHD-CP and H-CP, CHD-CP and H-H, CHD-H and H-H, and also between H-CP and H-H. CONCLUSION The periostin levels reduced in the CHD patients, especially in the CHD-CP group. The findings reveal a probable role of periostin in the association between CHD and CP.
Collapse
|
484
|
Ji DX, Witt KC, Kotov DI, Margolis SR, Louie A, Chevée V, Chen KJ, Gaidt MM, Dhaliwal HS, Lee AY, Nishimura SL, Zamboni DS, Kramnik I, Portnoy DA, Darwin KH, Vance RE. Role of the transcriptional regulator SP140 in resistance to bacterial infections via repression of type I interferons. eLife 2021; 10:67290. [PMID: 34151776 PMCID: PMC8248984 DOI: 10.7554/elife.67290] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Accepted: 06/20/2021] [Indexed: 12/18/2022] Open
Abstract
Type I interferons (IFNs) are essential for anti-viral immunity, but often impair protective immune responses during bacterial infections. An important question is how type I IFNs are strongly induced during viral infections, and yet are appropriately restrained during bacterial infections. The Super susceptibility to tuberculosis 1 (Sst1) locus in mice confers resistance to diverse bacterial infections. Here we provide evidence that Sp140 is a gene encoded within the Sst1 locus that represses type I IFN transcription during bacterial infections. We generated Sp140–/– mice and found that they are susceptible to infection by Legionella pneumophila and Mycobacterium tuberculosis. Susceptibility of Sp140–/– mice to bacterial infection was rescued by crosses to mice lacking the type I IFN receptor (Ifnar–/–). Our results implicate Sp140 as an important negative regulator of type I IFNs that is essential for resistance to bacterial infections.
Collapse
Affiliation(s)
- Daisy X Ji
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States
| | - Kristen C Witt
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States
| | - Dmitri I Kotov
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States.,Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, United States
| | - Shally R Margolis
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States
| | - Alexander Louie
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States
| | - Victoria Chevée
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States
| | - Katherine J Chen
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States.,Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, United States
| | - Moritz M Gaidt
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States
| | - Harmandeep S Dhaliwal
- Cancer Research Laboratory, University of California, Berkeley, Berkeley, United States
| | - Angus Y Lee
- Cancer Research Laboratory, University of California, Berkeley, Berkeley, United States
| | - Stephen L Nishimura
- Department of Pathology, University of California, San Francisco, San Francisco, United States
| | - Dario S Zamboni
- Department of Cell Biology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Igor Kramnik
- The National Emerging Infectious Diseases Laboratory, Department of Medicine (Pulmonary Center), and Department of Microbiology, Boston University School of Medicine, Boston, United States
| | - Daniel A Portnoy
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States.,Division of Biochemistry, Biophysics and Structural Biology, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States.,Department of Plant and Microbial Biology, University of California, Berkeley, Berkeley, United States
| | - K Heran Darwin
- Department of Microbiology, New York University Grossman School of Medicine, New York, United States
| | - Russell E Vance
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States.,Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, United States.,Cancer Research Laboratory, University of California, Berkeley, Berkeley, United States
| |
Collapse
|
485
|
Pariano M, Pieroni S, De Luca A, Iannitti RG, Borghi M, Puccetti M, Giovagnoli S, Renga G, D’Onofrio F, Bellet MM, Stincardini C, Della-Fazia MA, Servillo G, van de Veerdonk FL, Costantini C, Romani L. Anakinra Activates Superoxide Dismutase 2 to Mitigate Inflammasome Activity. Int J Mol Sci 2021; 22:ijms22126531. [PMID: 34207085 PMCID: PMC8234597 DOI: 10.3390/ijms22126531] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 05/27/2021] [Accepted: 06/14/2021] [Indexed: 01/03/2023] Open
Abstract
Inflammasomes are powerful cytosolic sensors of environmental stressors and are critical for triggering interleukin-1 (IL-1)-mediated inflammatory responses. However, dysregulation of inflammasome activation may lead to pathological conditions, and the identification of negative regulators for therapeutic purposes is increasingly being recognized. Anakinra, the recombinant form of the IL-1 receptor antagonist, proved effective by preventing the binding of IL-1 to its receptor, IL-1R1, thus restoring autophagy and dampening NLR family pyrin domain containing 3 (NLRP3) activity. As the generation of mitochondrial reactive oxidative species (ROS) is a critical upstream event in the activation of NLRP3, we investigated whether anakinra would regulate mitochondrial ROS production. By profiling the activation of transcription factors induced in murine alveolar macrophages, we found a mitochondrial antioxidative pathway induced by anakinra involving the manganese-dependent superoxide dismutase (MnSOD) or SOD2. Molecularly, anakinra promotes the binding of SOD2 with the deubiquitinase Ubiquitin Specific Peptidase 36 (USP36) and Constitutive photomorphogenesis 9 (COP9) signalosome, thus increasing SOD2 protein longevity. Functionally, anakinra and SOD2 protects mice from pulmonary oxidative inflammation and infection. On a preclinical level, anakinra upregulates SOD2 in murine models of chronic granulomatous disease (CGD) and cystic fibrosis (CF). These data suggest that protection from mitochondrial oxidative stress may represent an additional mechanism underlying the clinical benefit of anakinra and identifies SOD2 as a potential therapeutic target.
Collapse
Affiliation(s)
- Marilena Pariano
- Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy; (M.P.); (S.P.); (A.D.L.); (R.G.I.); (M.B.); (G.R.); (F.D.); (M.M.B.); (C.S.); (M.A.D.-F.); (G.S.); (C.C.)
| | - Stefania Pieroni
- Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy; (M.P.); (S.P.); (A.D.L.); (R.G.I.); (M.B.); (G.R.); (F.D.); (M.M.B.); (C.S.); (M.A.D.-F.); (G.S.); (C.C.)
| | - Antonella De Luca
- Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy; (M.P.); (S.P.); (A.D.L.); (R.G.I.); (M.B.); (G.R.); (F.D.); (M.M.B.); (C.S.); (M.A.D.-F.); (G.S.); (C.C.)
| | - Rossana G. Iannitti
- Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy; (M.P.); (S.P.); (A.D.L.); (R.G.I.); (M.B.); (G.R.); (F.D.); (M.M.B.); (C.S.); (M.A.D.-F.); (G.S.); (C.C.)
| | - Monica Borghi
- Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy; (M.P.); (S.P.); (A.D.L.); (R.G.I.); (M.B.); (G.R.); (F.D.); (M.M.B.); (C.S.); (M.A.D.-F.); (G.S.); (C.C.)
| | - Matteo Puccetti
- Department of Pharmaceutical Science, University of Perugia, 06132 Perugia, Italy; (M.P.); (S.G.)
| | - Stefano Giovagnoli
- Department of Pharmaceutical Science, University of Perugia, 06132 Perugia, Italy; (M.P.); (S.G.)
| | - Giorgia Renga
- Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy; (M.P.); (S.P.); (A.D.L.); (R.G.I.); (M.B.); (G.R.); (F.D.); (M.M.B.); (C.S.); (M.A.D.-F.); (G.S.); (C.C.)
| | - Fiorella D’Onofrio
- Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy; (M.P.); (S.P.); (A.D.L.); (R.G.I.); (M.B.); (G.R.); (F.D.); (M.M.B.); (C.S.); (M.A.D.-F.); (G.S.); (C.C.)
| | - Marina M. Bellet
- Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy; (M.P.); (S.P.); (A.D.L.); (R.G.I.); (M.B.); (G.R.); (F.D.); (M.M.B.); (C.S.); (M.A.D.-F.); (G.S.); (C.C.)
| | - Claudia Stincardini
- Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy; (M.P.); (S.P.); (A.D.L.); (R.G.I.); (M.B.); (G.R.); (F.D.); (M.M.B.); (C.S.); (M.A.D.-F.); (G.S.); (C.C.)
| | - Maria Agnese Della-Fazia
- Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy; (M.P.); (S.P.); (A.D.L.); (R.G.I.); (M.B.); (G.R.); (F.D.); (M.M.B.); (C.S.); (M.A.D.-F.); (G.S.); (C.C.)
| | - Giuseppe Servillo
- Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy; (M.P.); (S.P.); (A.D.L.); (R.G.I.); (M.B.); (G.R.); (F.D.); (M.M.B.); (C.S.); (M.A.D.-F.); (G.S.); (C.C.)
| | | | - Claudio Costantini
- Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy; (M.P.); (S.P.); (A.D.L.); (R.G.I.); (M.B.); (G.R.); (F.D.); (M.M.B.); (C.S.); (M.A.D.-F.); (G.S.); (C.C.)
| | - Luigina Romani
- Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy; (M.P.); (S.P.); (A.D.L.); (R.G.I.); (M.B.); (G.R.); (F.D.); (M.M.B.); (C.S.); (M.A.D.-F.); (G.S.); (C.C.)
- Correspondence:
| |
Collapse
|
486
|
Tomita T, Kato M, Mishima T, Matsunaga Y, Sanjo H, Ito KI, Minagawa K, Matsui T, Oikawa H, Takahashi S, Takao T, Iwai N, Mino T, Takeuchi O, Maru Y, Hiratsuka S. Extracellular mRNA transported to the nucleus exerts translation-independent function. Nat Commun 2021; 12:3655. [PMID: 34135341 PMCID: PMC8208975 DOI: 10.1038/s41467-021-23969-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 05/27/2021] [Indexed: 12/19/2022] Open
Abstract
RNA in extracellular vesicles (EVs) are uptaken by cells, where they regulate fundamental cellular functions. EV-derived mRNA in recipient cells can be translated. However, it is still elusive whether “naked nonvesicular extracellular mRNA” (nex-mRNA) that are not packed in EVs can be uptaken by cells and, if so, whether they have any functions in recipient cells. Here, we show the entrance of nex-mRNA in the nucleus, where they exert a translation-independent function. Human nex-interleukin-1β (IL1β)-mRNA outside cells proved to be captured by RNA-binding zinc finger CCCH domain containing protein 12D (ZC3H12D)-expressing human natural killer (NK) cells. ZC3H12D recruited to the cell membrane binds to the 3′-untranslated region of nex-IL1β-mRNA and transports it to the nucleus. The nex-IL1β-mRNA in the NK cell nucleus upregulates antiapoptotic gene expression, migration activity, and interferon-γ production, leading to the killing of cancer cells and antimetastasis in mice. These results implicate the diverse actions of mRNA. Nonvesicular extracellular RNA (nex-RNA) that are not packed in extracellular vesicles is detected outside the cell, but it is poorly understood. Here the authors report that nex-RNA is captured by a zinc finger protein and transported to the nucleus to enhance antimetastatic characters of the cell.
Collapse
Affiliation(s)
- Takeshi Tomita
- Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, School of Medicine, Matsumoto, Nagano, Japan.,Department of Biochemistry and Molecular Biology, Shinshu University, School of Medicine, Matsumoto, Nagano, Japan
| | - Masayoshi Kato
- Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, School of Medicine, Matsumoto, Nagano, Japan.,Department of Biochemistry and Molecular Biology, Shinshu University, School of Medicine, Matsumoto, Nagano, Japan
| | - Taishi Mishima
- Department of Pharmacology, Tokyo Women's Medical University, Shinjuku, Tokyo, Japan
| | - Yuta Matsunaga
- Department of Pharmacology, Tokyo Women's Medical University, Shinjuku, Tokyo, Japan
| | - Hideki Sanjo
- Department of Molecular and Cellular Immunology, Shinshu University, School of Medicine, Matsumoto, Nagano, Japan
| | - Ken-Ichi Ito
- Division of Breast, Endocrine and Respiratory Surgery, Department of Surgery, Shinshu University, School of Medicine, Matsumoto, Nagano, Japan
| | - Kentaro Minagawa
- Department of Hematology/Oncology, Penn State College of Medicine, Hershey, PA, USA
| | - Toshimitsu Matsui
- Department of Hematology, Nishiwaki Municipal Hospital, Nishiwaki, Hyogo, Japan
| | - Hiroyuki Oikawa
- Institute of Multidisciplinary Research for Advanced Materials, Tohoku University, Sendai, Japan
| | - Satoshi Takahashi
- Institute of Multidisciplinary Research for Advanced Materials, Tohoku University, Sendai, Japan
| | - Toshifumi Takao
- Institute for Protein Research, Osaka University, Suita, Osaka, Japan
| | - Noriki Iwai
- Department of Medical Chemistry, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takashi Mino
- Department of Medical Chemistry, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Osamu Takeuchi
- Department of Medical Chemistry, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yoshiro Maru
- Department of Pharmacology, Tokyo Women's Medical University, Shinjuku, Tokyo, Japan.
| | - Sachie Hiratsuka
- Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, School of Medicine, Matsumoto, Nagano, Japan. .,Department of Biochemistry and Molecular Biology, Shinshu University, School of Medicine, Matsumoto, Nagano, Japan.
| |
Collapse
|
487
|
Kim YW, Tonti E, Hickey P, Ellis AK, Neighbour H, Larché M, Tebbutt SJ. Immunological changes in peripheral blood following nasal allergen challenge in subjects with allergic rhinitis pre- and post-peptide immunotherapy: An open-label clinical study. Allergy 2021; 76:1907-1911. [PMID: 33320968 DOI: 10.1111/all.14710] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 11/27/2020] [Accepted: 12/09/2020] [Indexed: 11/30/2022]
Affiliation(s)
- Young Woong Kim
- Experimental Medicine University of British Columbia Vancouver BC Canada
- Prevention of Organ Failure (PROOF) Centre of Excellence Vancouver BC Canada
- Centre for Heart Lung Innovation St. Paul’s Hospital Vancouver BC Canada
| | - Elena Tonti
- Department of Medicine McMaster University Hamilton ON Canada
| | | | - Anne K. Ellis
- Departments of Medicine and Biomedical & Molecular Science Queen’s University Kingston ON Canada
- Allergy Research Unit Kingston General Hospital Kingston ON Canada
| | - Helen Neighbour
- Department of Medicine McMaster University Hamilton ON Canada
- Firestone Institute for Respiratory Health, St. Joseph’s Healthcare Hamilton ON Canada
| | - Mark Larché
- Department of Medicine McMaster University Hamilton ON Canada
- Firestone Institute for Respiratory Health, St. Joseph’s Healthcare Hamilton ON Canada
| | - Scott J. Tebbutt
- Experimental Medicine University of British Columbia Vancouver BC Canada
- Prevention of Organ Failure (PROOF) Centre of Excellence Vancouver BC Canada
- Centre for Heart Lung Innovation St. Paul’s Hospital Vancouver BC Canada
- Department of Medicine (Respiratory Division) University of British Columbia Vancouver BC Canada
| |
Collapse
|
488
|
Kim G, Choi KH, Kim H, Chung DK. Alleviation of LPS-Induced Inflammation and Septic Shock by Lactiplantibacillus plantarum K8 Lysates. Int J Mol Sci 2021; 22:ijms22115921. [PMID: 34072918 PMCID: PMC8197946 DOI: 10.3390/ijms22115921] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/18/2021] [Accepted: 05/26/2021] [Indexed: 01/08/2023] Open
Abstract
We previously showed that Lactiplantibacillus plantarum K8 and its cell wall components have immunoregulatory effects. In this study, we demonstrate that pre-treatment of L. plantarum K8 lysates reduced LPS-induced TNF-α production in THP-1 cells by down-regulating the early signals of mitogen-activated protein kinase (MAPK) and nuclear factor-κB (NF-κB). The down-regulation of signals may be caused by the induction of negative regulators involved in toll-like receptor (TLR)-mediated signaling. However, co-treatment with high concentrations of L. plantarum K8 lysates and lipopolysaccharide (LPS) activated the late signaling of extracellular signal-regulated kinase (ERK), c-Jun N-terminal kinase (JNK), and NF-κB pathways and resulted in the induction of absent in melanoma 2 (AIM2) inflammasome-mediated interleukin (IL)-1β secretion. Intraperitoneal injection of L. plantarum K8 lysates in LPS-induced endotoxin shock mice alleviated mortality and reduced serum tumor-necrosis factor (TNF)-α, IL-1β, aspartate aminotransferase (AST) and alanine aminotransferase (ALT) levels. In addition, the mRNA levels of TNF-α, IL-1β, and IL-6 decreased in livers from mice injected with L. plantarum K8 followed by LPS. Hematoxylin and eosin (H&E) staining of the liver showed that the cell size was enlarged by LPS injection and slightly reduced by L. plantarum K8 lysate pre-injection followed by LPS injection. Macrophage infiltration of the liver also decreased in response to the combination injection compared with mice injected with only LPS. Taken together, our results show that although L. plantarum K8 lysates differentially regulated the production of LPS-induced inflammatory cytokines in THP-1 cells, the lysates inhibited overall inflammation in mice. Thus, this study suggests that L. plantarum K8 lysates could be developed as a substance that modulates immune homeostasis by regulating inflammation.
Collapse
Affiliation(s)
- Gayoung Kim
- Graduate School of Biotechnology, Kyung Hee University, Yongin 17104, Korea; (G.K.); (K.-H.C.)
| | - Kyeong-Hun Choi
- Graduate School of Biotechnology, Kyung Hee University, Yongin 17104, Korea; (G.K.); (K.-H.C.)
| | - Hangeun Kim
- Research and Development Center, Skin Biotechnology Center Inc., Yongin 17104, Korea
- Correspondence: (H.K.); (D.-K.C.); Tel.: +82-31-201-2465 (H.K.); +82-31-888-6170 (D.-K.C.); Fax: +82-31-888-6173 (D.-K.C.)
| | - Dae-Kyun Chung
- Graduate School of Biotechnology, Kyung Hee University, Yongin 17104, Korea; (G.K.); (K.-H.C.)
- Research and Development Center, Skin Biotechnology Center Inc., Yongin 17104, Korea
- Skin Biotechnology Center, Kyung Hee University, Suwon 16229, Korea
- Correspondence: (H.K.); (D.-K.C.); Tel.: +82-31-201-2465 (H.K.); +82-31-888-6170 (D.-K.C.); Fax: +82-31-888-6173 (D.-K.C.)
| |
Collapse
|
489
|
Devi-Marulkar P, Moraes-Cabe C, Campagne P, Corre B, Meghraoui-Kheddar A, Bondet V, Llibre A, Duffy D, Maillart E, Papeix C, Pellegrini S, Michel F. Altered Immune Phenotypes and HLA-DQB1 Gene Variation in Multiple Sclerosis Patients Failing Interferon β Treatment. Front Immunol 2021; 12:628375. [PMID: 34113337 PMCID: PMC8185344 DOI: 10.3389/fimmu.2021.628375] [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: 11/11/2020] [Accepted: 04/23/2021] [Indexed: 11/25/2022] Open
Abstract
Background Interferon beta (IFNβ) has been prescribed as a first-line disease-modifying therapy for relapsing-remitting multiple sclerosis (RRMS) for nearly three decades. However, there is still a lack of treatment response markers that correlate with the clinical outcome of patients. Aim To determine a combination of cellular and molecular blood signatures associated with the efficacy of IFNβ treatment using an integrated approach. Methods The immune status of 40 RRMS patients, 15 of whom were untreated and 25 that received IFNβ1a treatment (15 responders, 10 non-responders), was investigated by phenotyping regulatory CD4+ T cells and naïve/memory T cell subsets, by measurement of circulating IFNα/β proteins with digital ELISA (Simoa) and analysis of ~600 immune related genes including 159 interferon-stimulated genes (ISGs) with the Nanostring technology. The potential impact of HLA class II gene variation in treatment responsiveness was investigated by genotyping HLA-DRB1, -DRB3,4,5, -DQA1, and -DQB1, using as a control population the Milieu Interieur cohort of 1,000 French healthy donors. Results Clinical responders and non-responders displayed similar plasma levels of IFNβ and similar ISG profiles. However, non-responders mainly differed from other subject groups with reduced circulating naïve regulatory T cells, enhanced terminally differentiated effector memory CD4+ TEMRA cells, and altered expression of at least six genes with immunoregulatory function. Moreover, non-responders were enriched for HLA-DQB1 genotypes encoding DQ8 and DQ2 serotypes. Interestingly, these two serotypes are associated with type 1 diabetes and celiac disease. Overall, the immune signatures of non-responders suggest an active disease that is resistant to therapeutic IFNβ, and in which CD4+ T cells, likely restricted by DQ8 and/or DQ2, exert enhanced autoreactive and bystander inflammatory activities.
Collapse
Affiliation(s)
- Priyanka Devi-Marulkar
- Cytokine Signaling Unit, Department of Immunology, Institut Pasteur, Paris, France.,INSERM U1221, Department of Immunology, Institut Pasteur, Paris, France
| | - Carolina Moraes-Cabe
- Cytokine Signaling Unit, Department of Immunology, Institut Pasteur, Paris, France.,INSERM U1221, Department of Immunology, Institut Pasteur, Paris, France
| | - Pascal Campagne
- Center of Bioinformatics, Biostatistics and Integrative Biology, Institut Pasteur, Paris, France
| | - Béatrice Corre
- Cytokine Signaling Unit, Department of Immunology, Institut Pasteur, Paris, France.,INSERM U1221, Department of Immunology, Institut Pasteur, Paris, France
| | - Aida Meghraoui-Kheddar
- Cytokine Signaling Unit, Department of Immunology, Institut Pasteur, Paris, France.,INSERM U1221, Department of Immunology, Institut Pasteur, Paris, France
| | - Vincent Bondet
- Translational Immunology Laboratory, Department of Immunology, Institut Pasteur, Paris, France
| | - Alba Llibre
- Translational Immunology Laboratory, Department of Immunology, Institut Pasteur, Paris, France
| | - Darragh Duffy
- Translational Immunology Laboratory, Department of Immunology, Institut Pasteur, Paris, France
| | | | - Caroline Papeix
- Department of Neurology, Pitié-Salpêtrière Hospital, Paris, France
| | - Sandra Pellegrini
- Cytokine Signaling Unit, Department of Immunology, Institut Pasteur, Paris, France.,INSERM U1221, Department of Immunology, Institut Pasteur, Paris, France
| | - Frédérique Michel
- Cytokine Signaling Unit, Department of Immunology, Institut Pasteur, Paris, France.,INSERM U1221, Department of Immunology, Institut Pasteur, Paris, France
| |
Collapse
|
490
|
Rolfes V, Ribeiro LS, Hawwari I, Böttcher L, Rosero N, Maasewerd S, Santos MLS, Próchnicki T, Silva CMDS, Wanderley CWDS, Rothe M, Schmidt SV, Stunden HJ, Bertheloot D, Rivas MN, Fontes CJ, Carvalho LH, Cunha FQ, Latz E, Arditi M, Franklin BS. Platelets Fuel the Inflammasome Activation of Innate Immune Cells. Cell Rep 2021; 31:107615. [PMID: 32402278 PMCID: PMC7225754 DOI: 10.1016/j.celrep.2020.107615] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 03/12/2020] [Accepted: 04/14/2020] [Indexed: 12/12/2022] Open
Abstract
The inflammasomes control the bioactivity of pro-inflammatory cytokines of the interleukin (IL)-1 family. The inflammasome assembled by NLRP3 has been predominantly studied in homogeneous cell populations in vitro, neglecting the influence of cellular interactions that occur in vivo. Here, we show that platelets boost the inflammasome capacity of human macrophages and neutrophils and are critical for IL-1 production by monocytes. Platelets license NLRP3 transcription, thereby enhancing ASC oligomerization, caspase-1 activity, and IL-1β secretion. Platelets influence IL-1β production in vivo, and blood platelet counts correlate with plasmatic IL-1β levels in malaria. Furthermore, we reveal an enriched platelet gene signature among the highest-expressed transcripts in IL-1β-driven autoinflammatory diseases. The platelet effect is independent of cell-to-cell contact, platelet-derived lipid mediators, purines, nucleic acids, and a host of platelet cytokines, and it involves the triggering of calcium-sensing receptors on macrophages. Hence, platelets provide an additional layer of regulation of inflammasomes and IL-1-driven inflammation. Platelets license NLRP3 for inflammasome activattion in innate immune cells Platelets are required for optimal monocyte inflammasome activation Platelets shape IL-1β in vivo, and platelet counts correlate with IL-1β in plasma A constitutive, heat-sensitive soluble platelet-factor boost IL-1β in macrophages
Collapse
Affiliation(s)
- Verena Rolfes
- Institute of Innate Immunity, Medical Faculty, University of Bonn, 53127 Bonn, NRW, Germany
| | - Lucas Secchim Ribeiro
- Institute of Innate Immunity, Medical Faculty, University of Bonn, 53127 Bonn, NRW, Germany.
| | - Ibrahim Hawwari
- Institute of Innate Immunity, Medical Faculty, University of Bonn, 53127 Bonn, NRW, Germany
| | - Lisa Böttcher
- Institute of Innate Immunity, Medical Faculty, University of Bonn, 53127 Bonn, NRW, Germany
| | - Nathalia Rosero
- Institute of Innate Immunity, Medical Faculty, University of Bonn, 53127 Bonn, NRW, Germany
| | - Salie Maasewerd
- Institute of Innate Immunity, Medical Faculty, University of Bonn, 53127 Bonn, NRW, Germany
| | - Marina Lima Silva Santos
- Laboratório de Malária, Instituto René Rachou, Fundação Oswaldo Cruz, 30190-002 Belo Horizonte, MG, Brazil
| | - Tomasz Próchnicki
- Institute of Innate Immunity, Medical Faculty, University of Bonn, 53127 Bonn, NRW, Germany
| | - Camila Meirelles de Souza Silva
- Center for Research in Inflammatory Diseases, School of Medicine of Ribeirão Preto, University of Sao Paulo, 14049-900 Ribeirão Preto, SP, Brazil
| | - Carlos Wagner de Souza Wanderley
- Center for Research in Inflammatory Diseases, School of Medicine of Ribeirão Preto, University of Sao Paulo, 14049-900 Ribeirão Preto, SP, Brazil
| | - Maximilian Rothe
- Institute of Innate Immunity, Medical Faculty, University of Bonn, 53127 Bonn, NRW, Germany
| | - Susanne V Schmidt
- Institute of Innate Immunity, Medical Faculty, University of Bonn, 53127 Bonn, NRW, Germany
| | - H James Stunden
- Institute of Innate Immunity, Medical Faculty, University of Bonn, 53127 Bonn, NRW, Germany
| | - Damien Bertheloot
- Institute of Innate Immunity, Medical Faculty, University of Bonn, 53127 Bonn, NRW, Germany
| | - Magali Noval Rivas
- Departments of Pediatrics, Division of Infectious Diseases and Immunology, and Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Cor Jesus Fontes
- Departamento de Clínica Médica, Universidade Federal de Mato Grosso, 78060-900 Cuiabá, MT, Brazil
| | - Luzia Helena Carvalho
- Laboratório de Malária, Instituto René Rachou, Fundação Oswaldo Cruz, 30190-002 Belo Horizonte, MG, Brazil
| | - Fernando Queiroz Cunha
- Center for Research in Inflammatory Diseases, School of Medicine of Ribeirão Preto, University of Sao Paulo, 14049-900 Ribeirão Preto, SP, Brazil
| | - Eicke Latz
- Institute of Innate Immunity, Medical Faculty, University of Bonn, 53127 Bonn, NRW, Germany; Department of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, MA 01655, USA; German Center for Neurodegenerative Diseases, 53127 Bonn, NRW, Germany
| | - Moshe Arditi
- Departments of Pediatrics, Division of Infectious Diseases and Immunology, and Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA.
| | | |
Collapse
|
491
|
Mado H, Adamczyk-Sowa M, Bartman W, Wierzbicki K, Tadeusiak B, Sowa P. Plasma Interleukin-33 level in relapsing-remitting multiple sclerosis. Is it negatively correlated with central nervous system lesions in patients with mild disability? Clin Neurol Neurosurg 2021; 206:106700. [PMID: 34030079 DOI: 10.1016/j.clineuro.2021.106700] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 04/06/2021] [Accepted: 05/12/2021] [Indexed: 01/02/2023]
Abstract
BACKGROUND Cytokines and chemokines are undoubtedly involved in the pathogenesis of multiple sclerosis (MS). There are many reports that suggest a significant role for Interleukin-33 (IL-33) in the course of MS development, but it is not clear whether negative or positive. We therefore investigated plasma IL-33 levels in patients with relapsing-remitting MS (RRMS). METHODS The study consisted of RRMS patients (n = 73) and healthy subjects (n = 54). Blood samples were taken from all and plasma IL-33 levels were then determined using an enzyme-linked immunosorbent assay method. Patients also underwent laboratory and imaging tests and their disability status was assessed. RESULTS Plasma IL-33 levels were marginally significantly higher in patients with RRMS (p = 0.07). Higher IL-33 levels are significantly associated with higher age (p = 0.01). There was also a statistically significant negative correlation between plasma IL-33 levels and the number of high signal intensity lesions in T2-weighted MRI (p = 0.03). After dividing the number of lesions into groups < 9 and ≥ 9 T2-weighted lesions, the Student's t-test for unrelated variables showed a negative correlation, but not statistically significant (p = 0.22), while the Spearman's correlation showed a marginally significant correlation (p = 0.06) between IL-33 level and number of T2-weighted lesions. IL-33 was also shown to have a significant ability to differentiate RRMS patients from healthy subjects with a sensitivity of 99% and specificity of 70% (p = 0.00). CONCLUSIONS Patients with RRMS have elevated plasma IL-33 levels. In RRMS patients with mild disability, high plasma levels of IL-33 may have neuroprotective effects potentially by stimulating remyelination and/or suppressing autoimmune inflammation and damage. Further studies on this matter on a larger number of patients are needed.
Collapse
Affiliation(s)
- Hubert Mado
- Department of Neurology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, Poland.
| | - Monika Adamczyk-Sowa
- Department of Neurology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, Poland
| | - Wojciech Bartman
- Department of Neurology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, Poland
| | - Krzysztof Wierzbicki
- Department of Neurology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, Poland
| | - Bartosz Tadeusiak
- Department of Neurology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, Poland
| | - Paweł Sowa
- Department of Otorhinolaryngology and Oncological Laryngology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, Poland
| |
Collapse
|
492
|
Sernoskie SC, Jee A, Uetrecht JP. The Emerging Role of the Innate Immune Response in Idiosyncratic Drug Reactions. Pharmacol Rev 2021; 73:861-896. [PMID: 34016669 DOI: 10.1124/pharmrev.120.000090] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Idiosyncratic drug reactions (IDRs) range from relatively common, mild reactions to rarer, potentially life-threatening adverse effects that pose significant risks to both human health and successful drug discovery. Most frequently, IDRs target the liver, skin, and blood or bone marrow. Clinical data indicate that most IDRs are mediated by an adaptive immune response against drug-modified proteins, formed when chemically reactive species of a drug bind to self-proteins, making them appear foreign to the immune system. Although much emphasis has been placed on characterizing the clinical presentation of IDRs and noting implicated drugs, limited research has focused on the mechanisms preceding the manifestations of these severe responses. Therefore, we propose that to address the knowledge gap between drug administration and onset of a severe IDR, more research is required to understand IDR-initiating mechanisms; namely, the role of the innate immune response. In this review, we outline the immune processes involved from neoantigen formation to the result of the formation of the immunologic synapse and suggest that this framework be applied to IDR research. Using four drugs associated with severe IDRs as examples (amoxicillin, amodiaquine, clozapine, and nevirapine), we also summarize clinical and animal model data that are supportive of an early innate immune response. Finally, we discuss how understanding the early steps in innate immune activation in the development of an adaptive IDR will be fundamental in risk assessment during drug development. SIGNIFICANCE STATEMENT: Although there is some understanding that certain adaptive immune mechanisms are involved in the development of idiosyncratic drug reactions, the early phase of these immune responses remains largely uncharacterized. The presented framework refocuses the investigation of IDR pathogenesis from severe clinical manifestations to the initiating innate immune mechanisms that, in contrast, may be quite mild or clinically silent. A comprehensive understanding of these early influences on IDR onset is crucial for accurate risk prediction, IDR prevention, and therapeutic intervention.
Collapse
Affiliation(s)
- Samantha Christine Sernoskie
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy (S.C.S., J.P.U.), and Department of Pharmacology and Toxicology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada (A.J., J.P.U.)
| | - Alison Jee
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy (S.C.S., J.P.U.), and Department of Pharmacology and Toxicology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada (A.J., J.P.U.)
| | - Jack Paul Uetrecht
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy (S.C.S., J.P.U.), and Department of Pharmacology and Toxicology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada (A.J., J.P.U.)
| |
Collapse
|
493
|
Cox CB, Storm EE, Kapoor VN, Chavarria-Smith J, Lin DL, Wang L, Li Y, Kljavin N, Ota N, Bainbridge TW, Anderson K, Roose-Girma M, Warming S, Arron JR, Turley SJ, de Sauvage FJ, van Lookeren Campagne M. IL-1R1-dependent signaling coordinates epithelial regeneration in response to intestinal damage. Sci Immunol 2021; 6:eabe8856. [PMID: 33963061 DOI: 10.1126/sciimmunol.abe8856] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 04/08/2021] [Indexed: 12/29/2022]
Abstract
Repair of the intestinal epithelium is tightly regulated to maintain homeostasis. The response after epithelial damage needs to be local and proportional to the insult. How different types of damage are coupled to repair remains incompletely understood. We report that after distinct types of intestinal epithelial damage, IL-1R1 signaling in GREM1+ mesenchymal cells increases production of R-spondin 3 (RSPO3), a Wnt agonist required for intestinal stem cell self-renewal. In parallel, IL-1R1 signaling regulates IL-22 production by innate lymphoid cells and promotes epithelial hyperplasia and regeneration. Although the regulation of both RSPO3 and IL-22 is critical for epithelial recovery from Citrobacter rodentium infection, IL-1R1-dependent RSPO3 production by GREM1+ mesenchymal cells alone is sufficient and required for recovery after dextran sulfate sodium-induced colitis. These data demonstrate how IL-1R1-dependent signaling orchestrates distinct repair programs tailored to the type of injury sustained that are required to restore intestinal epithelial barrier function.
Collapse
Affiliation(s)
- Christian B Cox
- Department of Immunology, Genentech Inc., South San Francisco, CA 94080, USA
| | - Elaine E Storm
- Department of Molecular Oncology, Genentech Inc., South San Francisco, CA 94080, USA
| | - Varun N Kapoor
- Department of Cancer Immunology, Genentech Inc., South San Francisco, CA 94080, USA
| | | | - David L Lin
- Department of Immunology, Genentech Inc., South San Francisco, CA 94080, USA
| | - Lifen Wang
- Department of Immunology, Genentech Inc., South San Francisco, CA 94080, USA
| | - Yun Li
- Department of Immunology, Genentech Inc., South San Francisco, CA 94080, USA
| | - Noelyn Kljavin
- Department of Molecular Oncology, Genentech Inc., South San Francisco, CA 94080, USA
| | - Naruhisa Ota
- Department of Immunology, Genentech Inc., South San Francisco, CA 94080, USA
| | - Travis W Bainbridge
- Department of Protein Chemistry, Genentech Inc., South San Francisco, CA 94080, USA
| | - Keith Anderson
- Department of Molecular Biology, Genentech Inc., South San Francisco, CA 94080, USA
| | - Merone Roose-Girma
- Department of Molecular Biology, Genentech Inc., South San Francisco, CA 94080, USA
| | - Søren Warming
- Department of Molecular Biology, Genentech Inc., South San Francisco, CA 94080, USA
| | - Joseph R Arron
- Department of Immunology, Genentech Inc., South San Francisco, CA 94080, USA
| | - Shannon J Turley
- Department of Cancer Immunology, Genentech Inc., South San Francisco, CA 94080, USA
| | - Frederic J de Sauvage
- Department of Molecular Oncology, Genentech Inc., South San Francisco, CA 94080, USA.
| | | |
Collapse
|
494
|
Kim H, Hong JY, Jeon WJ, Lee J, Baek SH, Ha IH. Lycopus lucidus Turcz Exerts Neuroprotective Effects Against H 2O 2-Induced Neuroinflammation by Inhibiting NLRP3 Inflammasome Activation in Cortical Neurons. J Inflamm Res 2021; 14:1759-1773. [PMID: 33981154 PMCID: PMC8109151 DOI: 10.2147/jir.s305031] [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: 02/05/2021] [Accepted: 04/07/2021] [Indexed: 12/20/2022] Open
Abstract
Purpose Lycopus lucidus Turcz (LLT) is a potent traditional medicinal herb that exerts therapeutic effects, regulating inflammatory disorders. However, the precise mechanisms by which LLT plays a potent role as an anti-inflammatory agent are still unknown, and in particular, the effects of LLT on cortical neurons and related mechanisms of neuroinflammation have not been studied. The NLRP3 inflammasome pathway is one of the most well known as an important driver of inflammation. We therefore hypothesized that LLT, as an effective anti-inflammatory agent, might have neurotherapeutic potential by inhibiting the NLRP3 inflammasome pathway in cortical neurons. Materials and Methods Primary cortical neurons were isolated from the embryonic rat cerebral cortex, and H2O2 was used to stimulate neuron damage in vitro. After treatment with LLT at three concentrations (10, 25, and 50 µg/mL), the expression of iNOS, NLRP3, ASC, caspase-1, IL-1β, IL-18, IL-6, and IL-10 was determined by immunocytochemistry, qPCR, and ELISA. Neuron apoptosis was also evaluated using Annexin V-FITC/PI double staining FACS analysis. Neural regeneration-related factors (BDNF, NGF, synaptophysin, NT3, AKT, and mTOR) were analyzed by immunocytochemistry and qPCR. Results LLT effectively protected cultured rat cortical neurons from H2O2-induced neuronal injury by significantly inhibiting NLRP3 inflammasome activation. In addition, it significantly reduced caspase-1 activation, which is induced by inflammasome formation and regulated the secretion of IL-1β/IL-18. We demonstrated that LLT enhances axonal elongation and synaptic connectivity upon H2O2-induced neuronal injury in rat primary cortical neurons. Conclusion It was first demonstrated in vitro that LLT suppresses NLRP3 inflammasome activation, attenuates inflammation and apoptosis, and consequently promotes neuroprotection and the stimulation of neuron repair, suggesting that it is a promising therapeutic for neurological diseases.
Collapse
Affiliation(s)
- Hyunseong Kim
- Jaseng Spine and Joint Research Institute, Jaseng Medical Foundation, Seoul, 135-896, Republic of Korea
| | - Jin Young Hong
- Jaseng Spine and Joint Research Institute, Jaseng Medical Foundation, Seoul, 135-896, Republic of Korea
| | - Wan-Jin Jeon
- Jaseng Spine and Joint Research Institute, Jaseng Medical Foundation, Seoul, 135-896, Republic of Korea
| | - Junseon Lee
- Jaseng Spine and Joint Research Institute, Jaseng Medical Foundation, Seoul, 135-896, Republic of Korea
| | - Seung Ho Baek
- College of Korean Medicine, Dongguk University, Goyang-si, 10326, Republic of Korea
| | - In-Hyuk Ha
- Jaseng Spine and Joint Research Institute, Jaseng Medical Foundation, Seoul, 135-896, Republic of Korea
| |
Collapse
|
495
|
Kitiyodom S, Trullàs C, Rodkhum C, Thompson KD, Katagiri T, Temisak S, Namdee K, Yata T, Pirarat N. Modulation of the mucosal immune response of red tilapia (Oreochromis sp.) against columnaris disease using a biomimetic-mucoadhesive nanovaccine. FISH & SHELLFISH IMMUNOLOGY 2021; 112:81-91. [PMID: 33675991 DOI: 10.1016/j.fsi.2021.02.017] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 02/27/2021] [Accepted: 02/28/2021] [Indexed: 06/12/2023]
Abstract
Columnaris, a highly contagious bacterial disease caused by Flavobacterium columnare, is recognized as one of the most important infectious diseases in farmed tilapia, especially during the fry and fingerling stages of production. The disease is associated with characteristic lesions in the mucosa of affected fish, particularly their skin and gills. Vaccines delivered via the mucosa are therefore of great interest to scientists developing vaccines for this disease. In the present study, we characterized field isolates of F. columnare obtained from clinical columnaris outbreaks in red tilapia to select an isolate to use as a candidate for our vaccine study. This included characterizing its colony morphology, genotype and virulence status. The isolate was incorporated into a mucoadhesive polymer chitosan-complexed nanovaccine (CS-NE), the efficacy of which was determined by experimentally infecting red tilapia that had been vaccinated with the nanoparticles by immersion. The experimental infection was performed 30-days post-vaccination (dpv), which resulted in 89% of the unvaccinated control fish dying, while the relative percentage survival (RPS) of the CS-NE vaccinated group was 78%. Histology of the mucosal associated lymphoid tissue (MALT) showed a significantly higher presence of leucocytes and a greater antigen uptake by the mucosal epithelium in CS-NE vaccinated fish compared to control fish and whole cell vaccinated fish, respectively, and there was statistically significant up-regulation of IgT, IgM, TNF α, IL1-β and MHC-1 genes in the gill of the CS-NE vaccinated group. Overall, the results of our study confirmed that the CS-NE particles achieved better adsorption onto the mucosal surfaces of the fish, elicited great vaccine efficacy and modulated the MALT immune response better than the conventional whole cell-killed vaccine, demonstrating the feasibility of the mucoadhesive nano-immersion vaccine as an effective delivery system for the induction of a mucosal immune response against columnaris disease in tilapia.
Collapse
Affiliation(s)
- Sirikorn Kitiyodom
- Wildlife Exotic Aquatic Animal Pathology-Research Unit, Department of Pathology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Clara Trullàs
- Wildlife Exotic Aquatic Animal Pathology-Research Unit, Department of Pathology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Channarong Rodkhum
- Department of Microbiology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Kim D Thompson
- Moredun Research Institute, Pentlands Science Park, Penicuik, UK
| | - Takayuki Katagiri
- Laboratory of Fish Health Management, Course of Aquatic Biosciences, Tokyo University of Marine Science and Technology, Tokyo, Japan
| | - Sasithon Temisak
- Bio Analysis Group, Chemical Metrology and Biometry Department, National Institute of Metrology (NIMT), Pathum Thani, 12120, Thailand
| | - Katawut Namdee
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency (NSTDA), Pathum Thani, 12120, Thailand
| | - Teerapong Yata
- Biochemistry Unit, Department of Physiology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330, Thailand.
| | - Nopadon Pirarat
- Wildlife Exotic Aquatic Animal Pathology-Research Unit, Department of Pathology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330, Thailand.
| |
Collapse
|
496
|
Linh NV, Dien LT, Panphut W, Thapinta A, Senapin S, St-Hilaire S, Rodkhum C, Dong HT. Ozone nanobubble modulates the innate defense system of Nile tilapia (Oreochromis niloticus) against Streptococcus agalactiae. FISH & SHELLFISH IMMUNOLOGY 2021; 112:64-73. [PMID: 33667674 DOI: 10.1016/j.fsi.2021.02.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 02/06/2021] [Accepted: 02/25/2021] [Indexed: 06/12/2023]
Abstract
Ozone nanobubble (NB-O3) is a promising technology for improving dissolved oxygen and reducing bacterial concentration in aquaculture systems. Here, we investigated the effects of NB-O3 on the innate immunity of fish by monitoring the expression levels of nonspecific immune-related genes (IL-1β, IL-2β, TNF-α), heat-shock protein genes (HSP70, HSP90-α), and a bacteriolytic enzyme, C-type lysozyme, gene (LYZ) post-treatment with this technology. Following exposure to NB-O3, the different tissues of Nile tilapia (Oreochromis niloticus) were collected over time for quantitative real-time PCR (qPCR) analysis. The expression of all the genes evaluated in the gills, the head kidney, and the spleen of the NB-O3 treated group was significantly up-regulated compared to that in the untreated control group. The expression levels were the highest (approx. 2 to 4-fold) at 15 min and 3 h post-exposure and then decreased from 6 to 24 h. These findings suggested that NB-O3 could switch on the innate immunity genes of Nile tilapia. Thus, we hypothesized that the NB-O3-immune-activated fish would respond more effectively to subsequent bacterial infections, thereby improving survivability compared to that of untreated fish. To test this hypothesis, 3 h post NB-O3 exposed fish and unexposed fish were challenged with a lethal dose of Streptococcus agalactiae. Interestingly, the survival rate of the NB-O3 group was significantly higher than that of the non-treated controls, with a relative percent survival (RPS) of 60-70%. Together, these findings indicate, for the first time, that NB-O3 may trigger the nonspecific defense system of the fish, thereby improving fish survivability during subsequent bacterial infections. This research identified another potential benefit of NB-O3 in aquaculture for preventing infectious bacterial diseases.
Collapse
Affiliation(s)
- Nguyen Vu Linh
- Fish Infectious Diseases Research Unit (FID RU), Department of Veterinary Microbiology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Le Thanh Dien
- Fish Infectious Diseases Research Unit (FID RU), Department of Veterinary Microbiology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330, Thailand; Department of Biotechnology and Crop Science, Faculty of Agriculture and Food Technology, Tien Giang University, Tien Giang, Viet Nam
| | - Wattana Panphut
- Faculty of Science and Technology, Suan Sunandha Rajabhat University, Bangkok, 10300, Thailand
| | - Anat Thapinta
- Faculty of Science and Technology, Suan Sunandha Rajabhat University, Bangkok, 10300, Thailand
| | - Saengchan Senapin
- Fish Health Platform, Centex of Excellence for Shrimp Molecular Biology and Biotechnology (Centex Shrimp), Faculty of Science, Mahidol University, Bangkok, 10400, Thailand; National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathum Thani, 12120, Thailand
| | - Sophie St-Hilaire
- Department of Infectious Diseases and Public Health, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon Tong, Hong Kong
| | - Channarong Rodkhum
- Fish Infectious Diseases Research Unit (FID RU), Department of Veterinary Microbiology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330, Thailand.
| | - Ha Thanh Dong
- Faculty of Science and Technology, Suan Sunandha Rajabhat University, Bangkok, 10300, Thailand.
| |
Collapse
|
497
|
Lanzillotta M, Sant'Angelo M, Kaneko N, Pillai S, Ponzoni M, Della-Torre E. Treating life-threatening TAFRO syndrome with interleukin-1 inhibition. Eur J Intern Med 2021; 87:121-123. [PMID: 33608160 DOI: 10.1016/j.ejim.2021.02.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 02/06/2021] [Indexed: 01/11/2023]
Affiliation(s)
- Marco Lanzillotta
- Università Vita-Salute San Raffaele, IRCCS San Raffaele Scientific Institute, Milan, Italy; Unit of Immunology, Rheumatology, Allergy and Rare Diseases (UnIRAR); IRCCS San Raffaele Scientific Institute, Milan, Italy.
| | - Miriam Sant'Angelo
- Università Vita-Salute San Raffaele, IRCCS San Raffaele Scientific Institute, Milan, Italy; Pathology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Naoki Kaneko
- Ragon Institute of MGH, MIT, and Harvard, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Section of Oral and Maxillofacial Oncology, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Shiv Pillai
- Ragon Institute of MGH, MIT, and Harvard, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Maurilio Ponzoni
- Università Vita-Salute San Raffaele, IRCCS San Raffaele Scientific Institute, Milan, Italy; Pathology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Emanuel Della-Torre
- Università Vita-Salute San Raffaele, IRCCS San Raffaele Scientific Institute, Milan, Italy; Unit of Immunology, Rheumatology, Allergy and Rare Diseases (UnIRAR); IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
498
|
Higa KC, Goodspeed A, Chavez JS, De Dominici M, Danis E, Zaberezhnyy V, Rabe JL, Tenen DG, Pietras EM, DeGregori J. Chronic interleukin-1 exposure triggers selection for Cebpa-knockout multipotent hematopoietic progenitors. J Exp Med 2021; 218:212039. [PMID: 33914855 PMCID: PMC8094119 DOI: 10.1084/jem.20200560] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 02/11/2021] [Accepted: 03/24/2021] [Indexed: 12/14/2022] Open
Abstract
The early events that drive myeloid oncogenesis are not well understood. Most studies focus on the cell-intrinsic genetic changes and how they impact cell fate decisions. We consider how chronic exposure to the proinflammatory cytokine, interleukin-1β (IL-1β), impacts Cebpa-knockout hematopoietic stem and progenitor cells (HSPCs) in competitive settings. Surprisingly, we found that Cebpa loss did not confer a hematopoietic cell–intrinsic competitive advantage; rather chronic IL-1β exposure engendered potent selection for Cebpa loss. Chronic IL-1β augments myeloid lineage output by activating differentiation and repressing stem cell gene expression programs in a Cebpa-dependent manner. As a result, Cebpa-knockout HSPCs are resistant to the prodifferentiative effects of chronic IL-1β, and competitively expand. We further show that ectopic CEBPA expression reduces the fitness of established human acute myeloid leukemias, coinciding with increased differentiation. These findings have important implications for the earliest events that drive hematologic disorders, suggesting that chronic inflammation could be an important driver of leukemogenesis and a potential target for intervention.
Collapse
Affiliation(s)
- Kelly C Higa
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO.,Integrated Department of Immunology, University of Colorado Anschutz Medical Campus, Aurora, CO.,Medical Scientist Training Program, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Andrew Goodspeed
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO.,University of Colorado Comprehensive Cancer Center, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - James S Chavez
- Division of Hematology, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Marco De Dominici
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Etienne Danis
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO.,University of Colorado Comprehensive Cancer Center, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Vadym Zaberezhnyy
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Jennifer L Rabe
- Division of Hematology, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Daniel G Tenen
- Cancer Science Institute, National University of Singapore, Singapore.,Harvard Stem Cell Institute, Harvard Medical School, Boston, MA
| | - Eric M Pietras
- Integrated Department of Immunology, University of Colorado Anschutz Medical Campus, Aurora, CO.,University of Colorado Comprehensive Cancer Center, University of Colorado Anschutz Medical Campus, Aurora, CO.,Division of Hematology, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - James DeGregori
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO.,Integrated Department of Immunology, University of Colorado Anschutz Medical Campus, Aurora, CO.,University of Colorado Comprehensive Cancer Center, University of Colorado Anschutz Medical Campus, Aurora, CO.,Division of Hematology, University of Colorado Anschutz Medical Campus, Aurora, CO
| |
Collapse
|
499
|
Wedzinska A, Figiel-Dabrowska A, Kozlowska H, Sarnowska A. The Effect of Proinflammatory Cytokines on the Proliferation, Migration and Secretory Activity of Mesenchymal Stem/Stromal Cells (WJ-MSCs) under 5% O 2 and 21% O 2 Culture Conditions. J Clin Med 2021; 10:1813. [PMID: 33919308 PMCID: PMC8122617 DOI: 10.3390/jcm10091813] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 03/24/2021] [Accepted: 04/17/2021] [Indexed: 11/16/2022] Open
Abstract
Treatment with Mesenchymal Stem/Stromal Cells (MSCs) in clinical trials is becoming one of the most-popular and fast-developing branches of modern regenerative medicine, as it is still in an experimental phase. The cross-section of diseases to which these cells are applied is very wide, ranging from degenerative diseases, through autoimmune processes and to acute inflammatory diseases, e.g., viral infections. Indeed, now that first clinical trials applying MSCs against COVID-19 have started, important questions concern not only the therapeutic properties of MSCs, but also the changes that might occur in the cell features as a response to the "cytokine storm" present in the acute phase of an infection and capable of posing a risk to a patient. The aim of our study was thus to assess changes potentially occurring in the biology of MSCs in the active inflammatory environment, e.g., in regards to the cell cycle, cell migration and secretory capacity. The study using MSCs derived from Wharton's jelly (WJ-MSCs) was conducted under two aerobic conditions: 21% O2 vs. 5% O2, since oxygen concentration is one of the key factors in inflammation. Under both oxygen conditions cells were exposed to proinflammatory cytokines involved significantly in acute inflammation, i.e., IFNγ, TNFα and IL-1β at different concentrations. Regardless of the aerobic conditions, WJ-MSCs in the inflammatory environment do not lose features typical for mesenchymal cells, and their proliferation dynamic remains unchanged. Sudden fluctuations in proliferation, the early indicator of potential genetic disturbance, were not observed, while the cells' migration activity increased. The presence of pro-inflammatory factors was also found to increase the secretion of such anti-inflammatory cytokines as IL-4 and IL-10. It is concluded that the inflammatory milieu in vitro does not cause phenotype changes or give rise to proliferation disruption of WJ-MSCs, and nor does it inhibit the secretory properties providing for their use against acute inflammation.
Collapse
Affiliation(s)
- Aleksandra Wedzinska
- Mossakowski Medical Research Centre, Translational Platform for Regenerative Medicine, Polish Academy of Sciences, 02-106 Warsaw, Poland; (A.W.); (A.F.-D.)
| | - Anna Figiel-Dabrowska
- Mossakowski Medical Research Centre, Translational Platform for Regenerative Medicine, Polish Academy of Sciences, 02-106 Warsaw, Poland; (A.W.); (A.F.-D.)
| | - Hanna Kozlowska
- Mossakowski Medical Research Centre, Laboratory of Advanced Microscopy Techniques, Polish Academy of Sciences, 02-106 Warsaw, Poland;
| | - Anna Sarnowska
- Mossakowski Medical Research Centre, Translational Platform for Regenerative Medicine, Polish Academy of Sciences, 02-106 Warsaw, Poland; (A.W.); (A.F.-D.)
- Mossakowski Medical Research Centre, Stem Cell Bioengineering Unit, Polish Academy of Sciences, 02-106 Warsaw, Poland
| |
Collapse
|
500
|
Li W, Meng X, Hao Y, Chen M, Jia Y, Gao P. Elevated sputum IL-36 levels are associated with neutrophil-related inflammation in COPD patients. CLINICAL RESPIRATORY JOURNAL 2021; 15:648-656. [PMID: 33559376 DOI: 10.1111/crj.13338] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 08/30/2020] [Accepted: 02/02/2021] [Indexed: 12/29/2022]
Abstract
BACKGROUND Interleukin (IL)-36, including IL-36α, IL-36β, and IL-36γ in the IL-1 family, are agonists of their receptors. IL-36 expression is associated with inflammation, including lung infection in humans. However, there is no information on its role in the inflammation of different types of chronic obstructive pulmonary disease (COPD). OBJECTIVE This study focused on the sputum IL-36α, IL-36β, and IL-36γ levels in stable COPD patients and their relationship with lung function and other cytokines in different inflammatory types of COPD patients. METHODS Sputum specimens were collected from 73 stable COPD patients and 20 age- and gender-matched healthy controls. The levels of sputum IL-36α, IL-36β, and IL-36γ and other cytokines were quantified and sputum cells were characterized. The potential relationship between the levels of sputum IL-36α, IL-36β, or IL-36γ and lung functional measures, inflammatory cells, and cytokines was analyzed. RESULTS In comparison with the healthy controls, sputum IL-36α and IL-36γ levels significantly increased in COPD (106.8 pg/mL vs. 76.9 pg/mL P =.001, 397.9 pg/mL vs. 359.5 pg/mL P =.006). The sputum IL-36α and IL-36γ levels were significantly higher in the neutrophilic and mixed granulocytic types than that in the eosinophilic and paucigranulocytic types of COPD patients. The sputum IL-36α levels were positively correlated with sputum IL-36γ levels and the numbers of sputum neutrophils, and the sputum IL-36γ levels were positively correlated with the numbers of sputum lymphocytes in COPD patients. CONCLUSIONS Elevated levels of sputum IL-36α and IL-36γ were detected in COPD patients and may provide insights into the inflammatory pathways in neutrophilic COPD.
Collapse
Affiliation(s)
- Wei Li
- Department of Respiratory and Critical Care Medicine, the Second Hospital of Jilin University, Changchun, Jilin, 130041, China
| | - Xiaoli Meng
- Department of Respiratory and Critical Care Medicine, the Second Hospital of Jilin University, Changchun, Jilin, 130041, China
| | - Yuqiu Hao
- Department of Respiratory and Critical Care Medicine, the Second Hospital of Jilin University, Changchun, Jilin, 130041, China
| | - Mo Chen
- Respiratory Medicine, Changchun Center Hospital, Changchun, Jilin, China
| | - Yuxi Jia
- Department of Orthopedics| Application Demonstration Center of Precision Medicine Molecular Diagnosis, the Second Hospital of Jilin University, Changchun, Jilin, China
| | - Peng Gao
- Department of Respiratory and Critical Care Medicine, the Second Hospital of Jilin University, Changchun, Jilin, 130041, China
| |
Collapse
|