1
|
Chauhan R, Mohan M, Mannan A, Devi S, Singh TG. Unravelling the role of Interleukin-12 in Neuroinflammatory mechanisms: Pathogenic pathways linking Neuroinflammation to neuropsychiatric disorders. Int Immunopharmacol 2025; 156:114654. [PMID: 40294470 DOI: 10.1016/j.intimp.2025.114654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 04/08/2025] [Accepted: 04/08/2025] [Indexed: 04/30/2025]
Abstract
Neuropsychiatric disorders are clinically characterized conditions involving both neurology and psychiatry, arising from dysfunctioning of cerebral function, or indirect effects of extra cerebral disease. Neuropsychiatric disorders tend to influence emotions, mood, and brain functioning. Growing evidence indicates that the etiology of these disorders is not confined to neuronal abnormalities but extends to include inflammation. While the underlying mechanism of these disorders is still in its infancy, recent data highlights the significant role of neuroinflammation in their pathophysiology. Neuroinflammation concerns the inflammation within the neural tissue characterized by alteration in astrocytes, cytokines, microglia, and chemokines within the central nervous system. The cytokines include IFN-γ, IL-1β, IL-2, IL4, IL-6, IL-8, IL-10, and IL-12. This review focuses on interleukin-12 (IL-12), a key mediator of neuroinflammation, and its potential involvement in neuropsychiatric disorders. IL-12 promotes neuroinflammation and influences neurotransmitter systems. Additionally, it also affects the HPA axis, impairs neuroplasticity, and activates microglia by interacting with TLR, JAK-STAT, PI3K/Akt, GSK-3, NMDA, MAPK, PKC, VEGFR, ROCK, and Wnt signaling pathways and elicit its role in ND. In this review, we dwell on the current evidence supporting IL-12's pathogenic role and explore the possible mechanisms by which it contributes to the development and progression of these conditions. This review aims to provide insights that may aid in future therapeutic strategies by illuminating the interplay between neuroinflammation and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Rupali Chauhan
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India
| | - Maneesh Mohan
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India
| | - Ashi Mannan
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India.
| | - Sushma Devi
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India
| | - Thakur Gurjeet Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India.
| |
Collapse
|
2
|
Ma D, Feng Y, Lin X. Immune and non-immune mediators in the fibrosis pathogenesis of salivary gland in Sjögren's syndrome. Front Immunol 2024; 15:1421436. [PMID: 39469708 PMCID: PMC11513355 DOI: 10.3389/fimmu.2024.1421436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 09/30/2024] [Indexed: 10/30/2024] Open
Abstract
Sjögren's syndrome (SS) or Sjögren's disease (SjD) is a systemic autoimmune disease clinically manifested as sicca symptoms. This disease primarily impacts the functionality of exocrine glands, specifically the lacrimal and salivary glands (SG). SG fibrosis, an irreversible morphological change, is a severe consequence that occurs in the later stages of the disease due to sustained inflammation. However, the mechanism underlying SG fibrosis in SS remains under-investigated. Glandular fibrosis may arise from chronic sialadenitis, in which the interactions between infiltrating lymphocytes and epithelial cells potentially contributes to fibrotic pathogenesis. Thus, both immune and non-immune cells are closely involved in this process, while their interplays are not fully understood. The molecular mechanism of tissue fibrosis is partly associated with an imbalance of immune responses, in which the transforming growth factor-beta (TGF-β)-dependent epithelial-mesenchymal transition (EMT) and extracellular matrix remodeling are recently investigated. In addition, viral infection has been implicated in the pathogenesis of SS. Viral-specific innate immune response could exacerbate the autoimmune progression, resulting in overt inflammation in SG. Notably, post-COVID patients exhibit typical SS symptoms and severe inflammatory sialadenitis, which are positively correlated with SG damage. In this review, we discuss the immune and non-immune risk factors in SG fibrosis and summarize the evidence to understand the mechanisms upon autoimmune progression in SS.
Collapse
Affiliation(s)
- Danbao Ma
- School of Chinese Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Yun Feng
- Department of Ophthalmology, Peking University Third Hospital, Beijing, China
| | - Xiang Lin
- School of Chinese Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Department of Chinese Medicine, the University of Hong Kong-Shenzhen Hospital (HKU-SZH), Shenzhen, China
| |
Collapse
|
3
|
Boukouaci W, Lajnef M, Wu CL, Bouassida J, Saitoh K, Sugunasabesan S, Richard JR, Apavou M, Lamy A, Henensal A, Nkam I, Hasty L, Sayous R, Bengoufa D, Barau C, Le Corvoisier P, Honnorat J, Maskos U, Yolken R, Leboyer M, Tamouza R. B Cell-activating factor (BAFF): A promising trans-nosographic biomarker of inflammation and autoimmunity in bipolar disorder and schizophrenia. Brain Behav Immun 2024; 121:178-188. [PMID: 39047848 DOI: 10.1016/j.bbi.2024.07.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 06/27/2024] [Accepted: 07/20/2024] [Indexed: 07/27/2024] Open
Abstract
Immune dysregulation is an important aspect of schizophrenia (SZ) and bipolar disorders (BD) pathophysiology, including not only inflammatory but also autoimmune process reflective of abnormal humoral immune responses. Given that B cell-activating factor (BAFF) is an integral aspect of B lymphocyte regulation, the current study investigated BAFF in SZ and BD. 255 SZ patients, 407 BD patients and 185 healthy controls (HC) were investigated across three aspects of soluble BAFF (sBAFF) by (i) comparing sBAFF circulatory levels across SZ, BD and HC, (ii) determining potential correlations between the circulating levels of sBAFF and the genotype distribution of a functionally relevant polymorphism, namely the TNFSF13B 3'UTR insertion-deletion polymorphism (GCTGT>A), (iii) analyzing relationships between both sBAFF levels and 3'UTR insertion-deletion genotypes and disease risk, patients clinical characteristics and circulating levels of potent inflammatory molecules. In addition, in subsets of patients, we also searched for possible correlations between sBAFF levels and stigma of past infectious events as well as positivity for circulating systemic autoantibodies or those directed against central nervous system (CNS) structures. Studying blood derived serum and DNA, weobserved that circulating sBAFF levels were significantly higher in SZ and BD patients, versus HC (p = 5.3*10-10and p = 4.4*10-09). Patients experiencing acute episodes, versus stable patients, in between acute episodes, exhibited higher sBAFF levels (p = 0.017).In SZ patients, positive correlations were observed between elevated sBAFF levels and: (i) elevated positive psychotic symptoms (PANSS pos), (ii) history of childhood trauma (physical abuse), and (iii) low scores on global functioning (GAF) (p = 0.024, p = 0.024, and p = 0.041).We also found that the distribution of the BAFF Ins/Del genotypes was significantly correlated with circulating sBAFF levels in SZ and BD patients (p = 0.0004). Elevated sBAFF levels were also correlated with increased levels of pro-inflammatory markers in both SZ and BD cohorts (p < 0.001). Regarding infectious stigma, only patients seropositive, versus seronegative, for herpes simplex virus (HSV)1 immunoglobulin (Ig)G antibodies exhibited a significant association with high sBAFF levels (p = 0.013). In contrast, positivity for systemic or CNS autoantibodies was significantly associated with reduced sBAFF levels, compared to patients without autoantibodies (p = 0.0017). Overall, our findings indicate that BAFF may be a promising trans-nosographic biomarker of inflammation that is likely to offer predictive, diagnostic, and prognostic tools for the management of SZ and BD. The results therefore have practicable clinical utility given the availability of immunotherapeutic treatment options including targeted monoclonal antibodies against BAFF.
Collapse
Affiliation(s)
- Wahid Boukouaci
- Univ Paris Est Créteil (UPEC), INSERM, IMRB, Translational Neuropsychiatry laboratory, AP-HP, DMU IMPACT, Fédération Hospitalo-Universitaire de Médecine de Précision en Psychiatrie (FHU ADAPT), Fondation FondaMental, Créteil F-94010, France
| | - Mohamed Lajnef
- Univ Paris Est Créteil (UPEC), INSERM, IMRB, Translational Neuropsychiatry laboratory, AP-HP, DMU IMPACT, Fédération Hospitalo-Universitaire de Médecine de Précision en Psychiatrie (FHU ADAPT), Fondation FondaMental, Créteil F-94010, France
| | - Ching-Lien Wu
- Univ Paris Est Créteil (UPEC), INSERM, IMRB, Translational Neuropsychiatry laboratory, AP-HP, DMU IMPACT, Fédération Hospitalo-Universitaire de Médecine de Précision en Psychiatrie (FHU ADAPT), Fondation FondaMental, Créteil F-94010, France
| | - Jihène Bouassida
- Univ Paris Est Créteil (UPEC), INSERM, IMRB, Translational Neuropsychiatry laboratory, AP-HP, DMU IMPACT, Fédération Hospitalo-Universitaire de Médecine de Précision en Psychiatrie (FHU ADAPT), Fondation FondaMental, Créteil F-94010, France
| | - Kaori Saitoh
- Univ Paris Est Créteil (UPEC), INSERM, IMRB, Translational Neuropsychiatry laboratory, AP-HP, DMU IMPACT, Fédération Hospitalo-Universitaire de Médecine de Précision en Psychiatrie (FHU ADAPT), Fondation FondaMental, Créteil F-94010, France
| | - Sobika Sugunasabesan
- Univ Paris Est Créteil (UPEC), INSERM, IMRB, Translational Neuropsychiatry laboratory, AP-HP, DMU IMPACT, Fédération Hospitalo-Universitaire de Médecine de Précision en Psychiatrie (FHU ADAPT), Fondation FondaMental, Créteil F-94010, France
| | - Jean-Romain Richard
- Univ Paris Est Créteil (UPEC), INSERM, IMRB, Translational Neuropsychiatry laboratory, AP-HP, DMU IMPACT, Fédération Hospitalo-Universitaire de Médecine de Précision en Psychiatrie (FHU ADAPT), Fondation FondaMental, Créteil F-94010, France
| | - Maud Apavou
- Univ Paris Est Créteil (UPEC), INSERM, IMRB, Translational Neuropsychiatry laboratory, AP-HP, DMU IMPACT, Fédération Hospitalo-Universitaire de Médecine de Précision en Psychiatrie (FHU ADAPT), Fondation FondaMental, Créteil F-94010, France
| | - Anais Lamy
- Univ Paris Est Créteil (UPEC), INSERM, IMRB, Translational Neuropsychiatry laboratory, AP-HP, DMU IMPACT, Fédération Hospitalo-Universitaire de Médecine de Précision en Psychiatrie (FHU ADAPT), Fondation FondaMental, Créteil F-94010, France
| | - Adèle Henensal
- Univ Paris Est Créteil (UPEC), INSERM, IMRB, Translational Neuropsychiatry laboratory, AP-HP, DMU IMPACT, Fédération Hospitalo-Universitaire de Médecine de Précision en Psychiatrie (FHU ADAPT), Fondation FondaMental, Créteil F-94010, France
| | - Irène Nkam
- Univ Paris Est Créteil (UPEC), INSERM, IMRB, Translational Neuropsychiatry laboratory, AP-HP, DMU IMPACT, Fédération Hospitalo-Universitaire de Médecine de Précision en Psychiatrie (FHU ADAPT), Fondation FondaMental, Créteil F-94010, France
| | - Lauren Hasty
- Univ Paris Est Créteil (UPEC), INSERM, IMRB, Translational Neuropsychiatry laboratory, AP-HP, DMU IMPACT, Fédération Hospitalo-Universitaire de Médecine de Précision en Psychiatrie (FHU ADAPT), Fondation FondaMental, Créteil F-94010, France
| | - Romain Sayous
- Univ Paris Est Créteil (UPEC), INSERM, IMRB, Translational Neuropsychiatry laboratory, AP-HP, DMU IMPACT, Fédération Hospitalo-Universitaire de Médecine de Précision en Psychiatrie (FHU ADAPT), Fondation FondaMental, Créteil F-94010, France
| | - Djaouida Bengoufa
- Univ Paris Est Créteil (UPEC), INSERM, IMRB, Translational Neuropsychiatry laboratory, AP-HP, DMU IMPACT, Fédération Hospitalo-Universitaire de Médecine de Précision en Psychiatrie (FHU ADAPT), Fondation FondaMental, Créteil F-94010, France
| | - Caroline Barau
- Plateforme de Ressources Biologiques, HU Henri Mondor, AP-HP, Créteil F94010, France
| | - Philippe Le Corvoisier
- Inserm, Centre d'Investigation Clinique 1430 et AP-HP, Hôpitaux Universitaires Henri Mondor, Univ Paris Est Creteil, Créteil F-94010, France
| | - Jérome Honnorat
- French Reference Centre on Paraneoplastic Neurological Syndromes and Autoimmune Encephalitis, Hospices Civils de Lyon, MeLiS-UCBL-CNRS UMR 5284. INSERM U1314, University Claude Bernard Lyon 1, Lyon, France
| | - Uwe Maskos
- Institut Pasteur, Université de Paris, Integrative Neurobiology of Cholinergic Systems, CNRS UMR 3571, Paris, France
| | - Robert Yolken
- Johns Hopkins school of medicine, Baltimore, MD, USA
| | - Marion Leboyer
- Univ Paris Est Créteil (UPEC), INSERM, IMRB, Translational Neuropsychiatry laboratory, AP-HP, DMU IMPACT, Fédération Hospitalo-Universitaire de Médecine de Précision en Psychiatrie (FHU ADAPT), Fondation FondaMental, Créteil F-94010, France
| | - Ryad Tamouza
- Univ Paris Est Créteil (UPEC), INSERM, IMRB, Translational Neuropsychiatry laboratory, AP-HP, DMU IMPACT, Fédération Hospitalo-Universitaire de Médecine de Précision en Psychiatrie (FHU ADAPT), Fondation FondaMental, Créteil F-94010, France.
| |
Collapse
|
4
|
Abstract
Pouchitis is an acute or chronic inflammatory disease of the ileal reservoir. It is common after restorative proctocolectomy with ileal pouch-anal anastomosis, and treatment of chronic antibiotic-refractory pouchitis has proven challenging. Most cases of acute pouchitis evolve into chronic pouchitis. The aetiology of acute pouchitis is likely to be partly related to the gut microbiota, whereas the pathophysiology of chronic pouchitis involves abnormal interactions between genetic disposition, faecal stasis, the gut microbiota, dysregulated host immunity, surgical techniques, ischaemia and mesentery-related factors. Pouchoscopy with biopsy is the most valuable modality for diagnosis, disease monitoring, assessment of treatment response, dysplasia surveillance and delivery of endoscopic therapy. Triggering or risk factors, such as Clostridioides difficile infection and use of non-steroidal anti-inflammatory drugs, should be modified or eradicated. In terms of treatment, acute pouchitis usually responds to oral antibiotics, whereas chronic antibiotic-refractory pouchitis often requires induction and maintenance therapy with integrin, interleukin or tumour necrosis factor inhibitors. Chronic pouchitis with ischaemic features, fistulae or abscesses can be treated with hyperbaric oxygen therapy.
Collapse
Affiliation(s)
- Bo Shen
- Center for Inflammatory Bowel Diseases and the Global Center for Integrated Colorectal Surgery and IBD Interventional Endoscopy, Columbia University Irving Medical Center/New York Presbyterian Hospital, New York, NY, USA.
| |
Collapse
|
5
|
Andrejčinová I, Blažková G, Papatheodorou I, Bendíčková K, Bosáková V, Skotáková M, Panovský R, Opatřil L, Vymazal O, Kovačovicová P, Šrámek V, Helán M, Hortová-Kohoutková M, Frič J. Persisting IL-18 levels after COVID-19 correlate with markers of cardiovascular inflammation reflecting potential risk of CVDs development. Heliyon 2024; 10:e25938. [PMID: 38404862 PMCID: PMC10884808 DOI: 10.1016/j.heliyon.2024.e25938] [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: 11/10/2023] [Revised: 02/04/2024] [Accepted: 02/05/2024] [Indexed: 02/27/2024] Open
Abstract
COVID-19 manifestation is associated with a strong immune system activation leading to inflammation and subsequently affecting the cardiovascular system. The objective of the study was to reveal possible interconnection between prolongated inflammation and the development or exacerbation of long-term cardiovascular complications after COVID-19. We investigated correlations between humoral and cellular immune system markers together with markers of cardiovascular inflammation/dysfunction during COVID-19 onset and subsequent recovery. We analyzed 22 hospitalized patients with severe COVID-19 within three timepoints (acute, 1 and 6 months after COVID-19) in order to track the impact of COVID-19 on the long-term decline of the cardiovascular system fitness and eventual development of CVDs. Among the cytokines dysregulated during COVID-19 changes, we showed significant correlations of IL-18 as a key driver of several pathophysiological changes with markers of cardiovascular inflammation/dysfunction. Our findings established novel immune-related markers, which can be used for the stratification of patients at high risk of CVDs for further therapy.
Collapse
Affiliation(s)
- Ivana Andrejčinová
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Gabriela Blažková
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
| | - Ioanna Papatheodorou
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Kamila Bendíčková
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
- International Clinical Research Center, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Veronika Bosáková
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Monika Skotáková
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
| | - Roman Panovský
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
- 1st Department of Internal Medicine/Cardioangiology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Lukáš Opatřil
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
- 1st Department of Internal Medicine/Cardioangiology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Ondřej Vymazal
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Petra Kovačovicová
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Vladimír Šrámek
- Department of Anesthesiology and Intensive Care, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Martin Helán
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
- Department of Anesthesiology and Intensive Care, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Marcela Hortová-Kohoutková
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
- International Clinical Research Center, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Jan Frič
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
- International Clinical Research Center, Faculty of Medicine, Masaryk University, Brno, Czech Republic
- Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| |
Collapse
|
6
|
Li F, Wang H, Li YQ, Gu Y, Jia XM. C-type lectin receptor 2d forms homodimers and heterodimers with TLR2 to negatively regulate IRF5-mediated antifungal immunity. Nat Commun 2023; 14:6718. [PMID: 37872182 PMCID: PMC10593818 DOI: 10.1038/s41467-023-42216-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 10/02/2023] [Indexed: 10/25/2023] Open
Abstract
Dimerization of C-type lectin receptors (CLRs) or Toll-like receptors (TLRs) can alter their ligand binding ability, thereby modulating immune responses. However, the possibilities and roles of dimerization between CLRs and TLRs remain unclear. Here we show that C-type lectin receptor-2d (CLEC2D) forms homodimers, as well as heterodimers with TLR2. Quantitative ligand binding assays reveal that both CLEC2D homodimers and CLEC2D/TLR2 heterodimers have a higher binding ability to fungi-derived β-glucans than TLR2 homodimers. Moreover, homo- or hetero-dimeric CLEC2D mediates β-glucan-induced ubiquitination and degradation of MyD88 to inhibit the activation of transcription factor IRF5 and subsequent IL-12 production. Clec2d-deficient female mice are resistant to infection with Candida albicans, a human fungal pathogen, owing to the increase of IL-12 production and subsequent generation of IFN-γ-producing NK cells. Together, these data indicate that CLEC2D forms homodimers or heterodimers with TLR2, which negatively regulate antifungal immunity through suppression of IRF5-mediated IL-12 production. These homo- and hetero-dimers of CLEC2D and TLR2 provide an example of receptor dimerization to regulate host innate immunity against microbial infections.
Collapse
Affiliation(s)
- Fan Li
- Department of Stomatology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
- Clinical Medicine Scientific and Technical Innovation Center, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
- Key Laboratory of Pathogen-Host Interactions of the Ministry of Education of China, Tongji University, Shanghai, 200092, China
| | - Hui Wang
- Clinical Medicine Scientific and Technical Innovation Center, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
- Key Laboratory of Pathogen-Host Interactions of the Ministry of Education of China, Tongji University, Shanghai, 200092, China
| | - Yan-Qi Li
- Clinical Medicine Scientific and Technical Innovation Center, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
- Key Laboratory of Pathogen-Host Interactions of the Ministry of Education of China, Tongji University, Shanghai, 200092, China
| | - Yebo Gu
- Department of Stomatology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China.
| | - Xin-Ming Jia
- Clinical Medicine Scientific and Technical Innovation Center, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China.
- Key Laboratory of Pathogen-Host Interactions of the Ministry of Education of China, Tongji University, Shanghai, 200092, China.
| |
Collapse
|
7
|
Bayraktar N, Eren MA, Bayraktar M, Öztürk A, Erdoğdu H. Analysis of Interleukin-17, Interleukin-23, neopterin and Nesfatin-1 levels in the sera of Hashimoto patients. J Med Biochem 2023; 42:460-468. [PMID: 37790207 PMCID: PMC10542705 DOI: 10.5937/jomb0-40683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Accepted: 12/27/2022] [Indexed: 10/05/2023] Open
Abstract
Background Hashimoto's thyroiditis (HT) is an autoimmune disorder affecting the thyroid gland and may present as goiter or atrophic thyroiditis that may result in various metabolic and inflammatory disorders. The aim of this study is to determine the changes in serum levels of interleukin-17 (IL-17), IL-23, neopterin, and nesfatin-1 parameters in HT patients and to evaluate the possible relationship among these parameters. Methods 90 HT patients and 30 healthy individuals were included in this study. Demographic data of the patients included in the study were recorded and detailed physical examinations were performed. IL-17, IL-23, neopterin, and nesfatin-1 levels were measured in the serum samples of the participants by the ELISA method.
Collapse
Affiliation(s)
- Nihayet Bayraktar
- Harran University, Faculty of Medicine, Department of Medical Biochemistry, Şanlıurfa, Turkey
| | - Mehmet Ali Eren
- Harran University, Faculty of Medicine, Department of Endocrinology, Şanlıurfa, Turkey
| | - Mustafa Bayraktar
- Yıldırım Beyazıt University, Faculty of Medicine, Department of Internal Medicine, Ankara, Turkey
| | - Ali Öztürk
- Niğde Ömer Halisdemir University, Faculty of Medicine, Department of Medical Microbiology, Niğde, Turkey
| | - Hamza Erdoğdu
- Harran University, Faculty of Business Administration, Department of Statistics, Şanlıurfa, Turkey
| |
Collapse
|
8
|
Møller DL, Sørensen SS, Rezahosseini O, Rasmussen DB, Arentoft NS, Loft JA, Perch M, Gustafsson F, Lundgren J, Scheike T, Knudsen JD, Ostrowski SR, Rasmussen A, Nielsen SD. Prediction of herpes virus infections after solid organ transplantation: a prospective study of immune function. Front Immunol 2023; 14:1183703. [PMID: 37465673 PMCID: PMC10351284 DOI: 10.3389/fimmu.2023.1183703] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 05/24/2023] [Indexed: 07/20/2023] Open
Abstract
Introduction Herpes virus infections are a major concern after solid organ transplantation and linked to the immune function of the recipient. We aimed to determine the incidence of positive herpes virus (cytomegalovirus (CMV), Epstein-Barr virus (EBV), herpes simplex virus type 1/2 (HSV-1/2), and varicella zoster virus (VZV)) PCR tests during the first year post-transplantation and assess whether a model including immune function pre-transplantation and three months post-transplantation could predict a subsequent positive herpes virus PCR. Methods All participants were preemptively screened for CMV, and EBV IgG-negative participants were screened for EBV during the first year post-transplantation. Herpes virus PCR tests for all included herpes viruses (CMV, EBV, HSV-1/2, and VZV) were retrieved from the Danish Microbiology database containing nationwide PCR results from both hospitals and outpatient clinics. Immune function was assessed by whole blood stimulation with A) LPS, B) R848, C) Poly I:C, and D) a blank control. Cytokine concentrations (TNF-α, IL-1β, IL-6, IL-8, IL-10, IL-12p40, IL-17A, IFN-α, and IFN-γ) were measured using Luminex. Results We included 123 liver (54%), kidney (26%), and lung (20%) transplant recipients. The cumulative incidence of positive herpes virus PCR tests was 36.6% (95% CI: 28.1-45.1) during the first year post-transplantation. The final prediction model included recipient age, type of transplantation, CMV serostatus, and change in Poly I:C-induced IL-12p40 from pre-transplantation to three months post-transplantation. The prediction model had an AUC of 77% (95% CI: 61-92). Risk scores were extracted from the prediction model, and the participants were divided into three risk groups. Participants with a risk score <5 (28% of the cohort), 5-10 (45% of the cohort), and >10 (27% of the cohort) had a cumulative incidence of having a positive herpes virus PCR test at 5.8%, 25%, and 73%, respectively (p < 0.001). Conclusion In conclusion, the incidence of positive herpes virus PCR tests was high, and a risk model including immune function allowed the prediction of positive herpes virus PCR and may be used to identify recipients at higher risk.
Collapse
Affiliation(s)
- Dina Leth Møller
- Viro-immunology Research Unit, Department of Infectious Diseases 8632, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Søren Schwartz Sørensen
- Department of Nephrology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Omid Rezahosseini
- Viro-immunology Research Unit, Department of Infectious Diseases 8632, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Daniel Bräuner Rasmussen
- Viro-immunology Research Unit, Department of Infectious Diseases 8632, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Nicoline Stender Arentoft
- Viro-immunology Research Unit, Department of Infectious Diseases 8632, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Josefine Amalie Loft
- Viro-immunology Research Unit, Department of Infectious Diseases 8632, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Michael Perch
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Department of Cardiology, Section for Lung Transplantation, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Finn Gustafsson
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Department of Cardiology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Jens Lundgren
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Centre of Excellence for Health, Immunity, and Infections, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Thomas Scheike
- Department of Biostatistics, University of Copenhagen, Copenhagen, Denmark
| | - Jenny Dahl Knudsen
- Department of Clinical Microbiology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Sisse Rye Ostrowski
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Immunology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Allan Rasmussen
- Department of Surgical Gastroenterology and Transplantation, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Susanne Dam Nielsen
- Viro-immunology Research Unit, Department of Infectious Diseases 8632, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Department of Surgical Gastroenterology and Transplantation, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
9
|
Purzycka-Bohdan D, Nedoszytko B, Sobalska-Kwapis M, Zabłotna M, Żmijewski MA, Wierzbicka J, Gleń J, Strapagiel D, Szczerkowska-Dobosz A, Nowicki RJ. Assessment of the Potential Role of Selected Single Nucleotide Polymorphisms (SNPs) of Genes Related to the Functioning of Regulatory T Cells in the Pathogenesis of Psoriasis. Int J Mol Sci 2023; 24:ijms24076061. [PMID: 37047033 PMCID: PMC10094301 DOI: 10.3390/ijms24076061] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 03/19/2023] [Accepted: 03/21/2023] [Indexed: 04/14/2023] Open
Abstract
Recent studies have indicated a key role of the impaired suppressive capacity of regulatory T cells (Tregs) in psoriasis (PsO) pathogenesis. However, the genetic background of Treg dysfunctions remains unknown. The aim of this study was to evaluate the association of PsO development with selected single nucleotide polymorphisms (SNPs) of genes in which protein products play a significant role in the regulation of differentiation and function of Tregs. There were three study groups in our research and each consisted of different unrelated patients and controls: 192 PsO patients and 5605 healthy volunteers in the microarray genotyping group, 150 PsO patients and 173 controls in the ARMS-PCR method group, and 6 PsO patients and 6 healthy volunteers in the expression analysis group. The DNA microarrays analysis (283 SNPs of 57 genes) and ARMS-PCR method (8 SNPs in 7 genes) were used to determine the frequency of occurrence of SNPs in selected genes. The mRNA expression of selected genes was determined in skin samples. There were statistically significant differences in the allele frequencies of four SNPs in three genes (TNF, IL12RB2, and IL12B) between early-onset PsO patients and controls. The lowest p-value was observed for rs3093662 (TNF), and the G allele carriers had a 2.73 times higher risk of developing early-onset PsO. Moreover, the study revealed significant differences in the frequency of SNPs and their influence on PsO development between early- and late-onset PsO. Based on the ARMS-PCR method, the association between some polymorphisms of four genes (IL4, IL10, TGFB1, and STAT3) and the risk of developing PsO was noticed. Psoriatic lesions were characterized with a lower mRNA expression of FOXP3, CTLA4, and IL2, and a higher expression of TNF and IL1A in comparison with unaffected skin. In conclusion, the genetic background associated with properly functioning Tregs seems to play a significant role in PsO pathogenesis and could have diagnostic value.
Collapse
Affiliation(s)
- Dorota Purzycka-Bohdan
- Department of Dermatology, Venereology and Allergology, Medical University of Gdansk, 80-210 Gdansk, Poland
| | - Bogusław Nedoszytko
- Department of Dermatology, Venereology and Allergology, Medical University of Gdansk, 80-210 Gdansk, Poland
- Molecular Laboratory, Invicta Fertility and Reproductive Centre, 81-740 Sopot, Poland
| | - Marta Sobalska-Kwapis
- Biobank Laboratory, Department of Oncobiology and Epigenetics, Faculty of Biology and Environmental Protection, University of Lodz, 90-237 Lodz, Poland
| | - Monika Zabłotna
- Department of Dermatology, Venereology and Allergology, Medical University of Gdansk, 80-210 Gdansk, Poland
| | - Michał A Żmijewski
- Department of Histology, Medical University of Gdansk, 80-211 Gdansk, Poland
| | - Justyna Wierzbicka
- Department of Histology, Medical University of Gdansk, 80-211 Gdansk, Poland
| | - Jolanta Gleń
- Department of Dermatology, Venereology and Allergology, Medical University of Gdansk, 80-210 Gdansk, Poland
| | - Dominik Strapagiel
- Biobank Laboratory, Department of Oncobiology and Epigenetics, Faculty of Biology and Environmental Protection, University of Lodz, 90-237 Lodz, Poland
| | - Aneta Szczerkowska-Dobosz
- Department of Dermatology, Venereology and Allergology, Medical University of Gdansk, 80-210 Gdansk, Poland
| | - Roman J Nowicki
- Department of Dermatology, Venereology and Allergology, Medical University of Gdansk, 80-210 Gdansk, Poland
| |
Collapse
|
10
|
Atluri K, Manne S, Nalamothu V, Mantel A, Sharma PK, Babu RJ. Advances in Current Drugs and Formulations for the Management of Atopic Dermatitis. Crit Rev Ther Drug Carrier Syst 2023; 40:1-87. [PMID: 37585309 DOI: 10.1615/critrevtherdrugcarriersyst.2023042979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/18/2023]
Abstract
Atopic dermatitis (AD) is a chronic, relapsing inflammatory skin disease with a complex pathophysiology. Treatment of AD remains challenging owing to the presence of a wide spectrum of clinical phenotypes and limited response to existing therapies. However, recent genetic, immunological, and pathophysiological insights into the disease mechanism resulted in the invention of novel therapeutic drug candidates. This review provides a comprehensive overview of current therapies and assesses various novel drug delivery strategies currently under clinical investigation. Further, this review majorly emphasizes on various topical treatments including emollient therapies, barrier repair agents, topical corticosteroids (TCS), phosphodiesterase 4 (PDE4) inhibitors, calcineurin inhibitors, and Janus kinase (JAK)-signal transducer and activator of transcription (STAT) pathway inhibitors. It also discusses biological and systemic therapies, upcoming treatments based on ongoing clinical trials. Additionally, this review scrutinized the use of pharmaceutical inactive ingredients in the approved topical dosage forms for AD treatment.
Collapse
Affiliation(s)
| | | | | | | | | | - R Jayachandra Babu
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL 36849, USA
| |
Collapse
|
11
|
Zheng YJ, Ho W, Sanlorenzo M, Vujic I, Daud A, Algazi A, Rappersberger K, Ortiz-Urda S. Melanoma risk during immunomodulating treatment. Melanoma Res 2022; 32:411-418. [PMID: 35993892 DOI: 10.1097/cmr.0000000000000838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
Abstract
Immunosuppressive therapy is standard for the treatment of inflammatory diseases and for minimizing rejection in transplant patients. However, immunosuppressant drugs are associated with an increased risk of certain cancers. In particular, melanoma is an immunogenic tumor and as such, is strongly influenced by the immune system. We performed this literature review to summarize the effects of commonly used immunomodulating agents on melanoma development, recurrence and progression. We outline the mechanism of action of each drug and discuss the available evidence on its influence on melanoma. Based on existing literature, we recommend avoiding the following agents in patients with a history of invasive melanoma: cyclosporine, sirolimus, natalizumab, IL-6 inhibitors, cyclophosphamide, methotrexate and the tumor necrosis factor-alpha inhibitors infliximab and etanercept. If there are no viable alternative agents, we recommend for these patients to see a dermatologist every 6 months for a thorough skin examination.
Collapse
Affiliation(s)
- Yixuan James Zheng
- Department of Dermatology, University of California San Francisco
- School of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Wilson Ho
- Department of Dermatology, University of California San Francisco
| | - Martina Sanlorenzo
- Department of Dermatology, University of California San Francisco
- Department of Oncology, University of Turin, Torino, Italy
- Department of Medicine, Institute of Cancer Research, Medical University of Vienna
| | - Igor Vujic
- Department of Dermatology, University of California San Francisco
- Department of Dermatology and Venereology, The Rudolfstiftung Hospital
- School of Medicine, Sigmund Freud University Vienna, Vienna, Austria
| | - Adil Daud
- Department of Dermatology, University of California San Francisco
| | - Alain Algazi
- Department of Dermatology, University of California San Francisco
| | - Klemens Rappersberger
- Department of Dermatology and Venereology, The Rudolfstiftung Hospital
- School of Medicine, Sigmund Freud University Vienna, Vienna, Austria
| | | |
Collapse
|
12
|
Han Y, Klinger K, Rajpal DK, Zhu C, Teeple E. Empowering the discovery of novel target-disease associations via machine learning approaches in the open targets platform. BMC Bioinformatics 2022; 23:232. [PMID: 35710324 PMCID: PMC9202116 DOI: 10.1186/s12859-022-04753-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 05/26/2022] [Indexed: 11/10/2022] Open
Abstract
Background The Open Targets (OT) Platform integrates a wide range of data sources on target-disease associations to facilitate identification of potential therapeutic drug targets to treat human diseases. However, due to the complexity that targets are usually functionally pleiotropic and efficacious for multiple indications, challenges in identifying novel target to indication associations remain. Specifically, persistent need exists for new methods for integration of novel target-disease association evidence and biological knowledge bases via advanced computational methods. These offer promise for increasing power for identification of the most promising target-disease pairs for therapeutic development. Here we introduce a novel approach by integrating additional target-disease features with machine learning models to further uncover druggable disease to target indications. Results We derived novel target-disease associations as supplemental features to OT platform-based associations using three data sources: (1) target tissue specificity from GTEx expression profiles; (2) target semantic similarities based on gene ontology; and (3) functional interactions among targets by embedding them from protein–protein interaction (PPI) networks. Machine learning models were applied to evaluate feature importance and performance benchmarks for predicting targets with known drug indications. The evaluation results show the newly integrated features demonstrate higher importance than current features in OT. In addition, these also show superior performance over association benchmarks and may support discovery of novel therapeutic indications for highly pursued targets. Conclusion Our newly generated features can be used to represent additional underlying biological relatedness among targets and diseases to further empower improved performance for predicting novel indications for drug targets through advanced machine learning models. The proposed methodology enables a powerful new approach for systematic evaluation of drug targets with novel indications. Supplementary Information The online version contains supplementary material available at 10.1186/s12859-022-04753-4.
Collapse
Affiliation(s)
- Yingnan Han
- Translational Sciences, Sanofi US, Framingham, MA, 01701, USA
| | | | - Deepak K Rajpal
- Translational Sciences, Sanofi US, Framingham, MA, 01701, USA
| | - Cheng Zhu
- Translational Sciences, Sanofi US, Framingham, MA, 01701, USA.
| | - Erin Teeple
- Translational Sciences, Sanofi US, Framingham, MA, 01701, USA.
| |
Collapse
|
13
|
Shah AM, Zamora R, Korff S, Barclay D, Yin J, El-Dehaibi F, Billiar TR, Vodovotz Y. Inferring Tissue-Specific, TLR4-Dependent Type 17 Immune Interactions in Experimental Trauma/Hemorrhagic Shock and Resuscitation Using Computational Modeling. Front Immunol 2022; 13:908618. [PMID: 35663944 PMCID: PMC9160183 DOI: 10.3389/fimmu.2022.908618] [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: 03/30/2022] [Accepted: 04/21/2022] [Indexed: 11/13/2022] Open
Abstract
Trauma/hemorrhagic shock followed by resuscitation (T/HS-R) results in multi-system inflammation and organ dysfunction, in part driven by binding of damage-associated molecular pattern molecules to Toll-like Receptor 4 (TLR4). We carried out experimental T/HS-R (pseudo-fracture plus 2 h of shock followed by 0-22 h of resuscitation) in C57BL/6 (wild type [WT]) and TLR4-null (TLR4-/-) mice, and then defined the dynamics of 20 protein-level inflammatory mediators in the heart, gut, lung, liver, spleen, kidney, and systemic circulation. Cross-correlation and Principal Component Analysis (PCA) on data from the 7 tissues sampled suggested that TLR4-/- samples express multiple inflammatory mediators in a small subset of tissue compartments as compared to the WT samples, in which many inflammatory mediators were localized non-specifically to nearly all compartments. We and others have previously defined a central role for type 17 immune cells in human trauma. Accordingly, correlations between IL-17A and GM-CSF (indicative of pathogenic Th17 cells); between IL-17A and IL-10 (indicative of non-pathogenic Th17 cells); and IL-17A and TNF (indicative of memory/effector T cells) were assessed across all tissues studied. In both WT and TLR4-/- mice, positive correlations were observed between IL-17A and GM-CSF, IL-10, and TNF in the kidney and gut. In contrast, the variable and dynamic presence of both pathogenic and non-pathogenic Th17 cells was inferred in the systemic circulation of TLR4-/- mice over time, suggesting a role for TLR4 in efflux of these cells into peripheral tissues. Hypergraph analysis - used to define dynamic, cross compartment networks - in concert with PCA-suggested that IL-17A was present persistently in all tissues at all sampled time points except for its absence in the plasma at 0.5h in the WT group, supporting the hypothesis that T/HS-R induces efflux of Th17 cells from the circulation and into specific tissues. These analyses suggest a complex, context-specific role for TLR4 and type 17 immunity following T/HS-R.
Collapse
Affiliation(s)
- Ashti M Shah
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Ruben Zamora
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States.,Center for Inflammation and Regeneration Modeling, McGowan Institute for Regenerative Medicine, Pittsburgh, PA, United States
| | - Sebastian Korff
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Derek Barclay
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Jinling Yin
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Fayten El-Dehaibi
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Timothy R Billiar
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Yoram Vodovotz
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States.,Center for Inflammation and Regeneration Modeling, McGowan Institute for Regenerative Medicine, Pittsburgh, PA, United States.,Center for Systems Immunology, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
14
|
Saber S, Alomar SY, Yahya G. Blocking prostanoid receptors switches on multiple immune responses and cascades of inflammatory signaling against larval stages in snail fever. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:43546-43555. [PMID: 35396684 PMCID: PMC9200668 DOI: 10.1007/s11356-022-20108-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 04/01/2022] [Indexed: 05/27/2023]
Abstract
Schistosomiasis, also known as snail fever or bilharziasis, is a worm infection caused by trematode called schistosomes that affects humans and animals worldwide. Schistosomiasis endemically exists in developing countries. Inflammatory responses elicited in the early phase of infection represent the rate limiting step for parasite migration and pathogenesis and could be a valuable target for therapeutic interventions. Prostaglandin E2 (PGE2) and interleukin (IL)-10 were found to be differentially affected in case of immune-modulation studies and cytokine analysis of hosts infected with either normal or radiation-attenuated parasite (RA) which switches off the development of an effective immune response against the migrating parasite in the early phase of schistosomiasis. Normal parasites induce predominantly a T helper 2 (Th2)-type cytokine response (IL-4 and IL-5) which is essential for parasite survival; here, we discuss in detail the downstream effects and cascades of inflammatory signaling of PGE2 and IL10 induced by normal parasites and the effect of blocking PGE2 receptors. We suggest that by selectively constraining the production of PGE2 during vaccination or therapy of susceptible persons or infected patients of schistosomiasis, this would boost IL-12 and reduce IL-10 production leading to a polarization toward the anti-worm Thl cytokine synthesis (IL-2 and Interferon (IFN)-γ).
Collapse
Affiliation(s)
- Sameh Saber
- Department of Pharmacology, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, Egypt
| | - Suliman Y. Alomar
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, Riyadh, 11451 Saudi Arabia
| | - Galal Yahya
- Department of Microbiology and Immunology, Faculty of Pharmacy, Zagazig University, Al Sharkia, 44519 Egypt
| |
Collapse
|
15
|
Shao J, Xu Z, Xu Y. Integrated Population Pharmacokinetic Analysis of Ustekinumab Across Multiple Immune-Mediated Inflammatory Disease Populations and Healthy Subjects. Eur J Drug Metab Pharmacokinet 2022; 47:537-548. [PMID: 35442011 DOI: 10.1007/s13318-022-00768-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/16/2022] [Indexed: 11/30/2022]
Abstract
BACKGROUND AND OBJECTIVES Ustekinumab has been approved for the treatment of patients with plaque psoriasis (PSO), psoriatic arthritis (PSA), Crohn's disease (CD), and ulcerative colitis (UC). This study was performed to investigate whether the pharmacokinetics of ustekinumab differ across different disease populations. METHODS An integrated population pharmacokinetic analysis was conducted to characterize ustekinumab pharmacokinetics across four disease indications (i.e., PSO, PSA, CD, and UC) and healthy subjects. RESULTS The pooled ustekinumab pharmacokinetic data consisted of 46,970 serum concentrations from 3,217 subjects (n = 356 for PSO, 696 for PSA, 1196 for CD, 823 for UC, and 24 for healthy subjects) in 7 clinical trials following subcutaneous or intravenous ustekinumab administrations. The pharmacokinetics of ustekinumab were adequately described by a two-compartment linear model with first-order absorption and elimination. Ustekinumab clearance (CL) and volume of distribution parameters increased nonlinearly with body weight, and the CL was higher in subjects with lower serum albumin, non-Caucasians, and positive antibodies to ustekinumab. Although CL in subjects with PSO and PSA appeared to be slightly different (- 12.7% and + 8.87%, respectively) from CL in other populations (including CD, UC, and healthy subjects), the model-derived post-hoc pharmacokinetic parameters were generally comparable across the 4 disease populations. Simulations also suggested overall comparable ustekinumab exposure (i.e., trough concentration and AUC) at steady state across the disease populations following the same subcutaneous dose regimen. CONCLUSIONS Ustekinumab pharmacokinetics are generally comparable across all approved inflammatory-mediated indications and healthy subjects after accounting for the body weight-related pharmacokinetic difference.
Collapse
Affiliation(s)
- Jie Shao
- Clinical Pharmacology and Pharmacometrics, Janssen Research & Development LLC, 1400 McKean Rd, PA, 19477, Spring House, USA.
| | - Zhenhua Xu
- Clinical Pharmacology and Pharmacometrics, Janssen Research & Development LLC, 1400 McKean Rd, PA, 19477, Spring House, USA
| | - Yan Xu
- Clinical Pharmacology and Pharmacometrics, Janssen Research & Development LLC, 1400 McKean Rd, PA, 19477, Spring House, USA.,Clinical Pharmacology, Simcere Pharmaceuticals, MB, Cambridge, USA
| |
Collapse
|
16
|
Ali Imarah A, Subhi Mohammed M, Ahmed Najm R, Al Zeyadi M, Khayoon SQ, Alhamadani I. Effect of the Concentration Levels of Growth Hormone and Insulin-like Growth Factor I on the Polymorphisms of the Il12p40 Gene in Lung Cancer Patients. ARCHIVES OF RAZI INSTITUTE 2022; 77:413-419. [PMID: 35891724 PMCID: PMC9288594 DOI: 10.22092/ari.2021.356610.1880] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Accepted: 12/04/2021] [Indexed: 11/25/2022]
Abstract
The prevalence of lung cancer as one of the most common cancers with the highest mortality rate is one of the most important health problems in humans across the world. Molecular research can provide valuable information about genetic changes associated with the pathogenesis of the disease that may be used to improve prognosis and treatment. The current study aimed to examine the genotyping utility of the Il12p40 (IL-12B) gene (rs3212227, A>C) polymorphisms and detect its relationship with the concentration levels of HGH and IGF-1 for the non-small cell lung carcinoma (NSCLC). This study investigated 67 cases with NSCLC (60 males and 7 females) and 28 healthy individuals as controls. The serum level of HGH and IGF-1 was determined using an enzyme-linked immunosorbent assay. Genotyping of the IL-12B gene polymorphisms (rs3212227, A>C) was carried out by polymerase chain reaction-restriction fragment length polymorphism. The serum levels of the HGH and IGF-1 were estimated, and the results of the IL-12B genotyping showed an increased risk of NSCLC. The homozygous wild (AA) genotype of the IL-12 gene showed that the risk of NSCLC was higher than that of the heterozygous (AC) and homozygous genotypes (CC). Moreover, a significant elevation was found in the serum levels of the HGH in the NSCLC patients, compared to the control group. The result showed that the IL-12 gene polymorphism was implicated in the pathogenesis of the NSCLC and directed several metabolic changes.
Collapse
Affiliation(s)
- A Ali Imarah
- Faculty of Sciences, University of Kufa, Kufa, Iraq
| | - M Subhi Mohammed
- Al-Furat Al-Awsat Technical University, Technical Institute, Kufa, Iraq
| | - R Ahmed Najm
- Al-Furat Al-Awsat Technical University, Technical Institute, Kufa, Iraq
| | - M Al Zeyadi
- Faculty of Sciences, University of Kufa, Kufa, Iraq
| | - S. Q Khayoon
- Faculty of Sciences, University of Kufa, Kufa, Iraq
| | - I Alhamadani
- Faculty of Sciences, University of Kufa, Kufa, Iraq
| |
Collapse
|
17
|
Zwicky P, Ingelfinger F, Silva de Melo BM, Ruchti F, Schärli S, Puertas N, Lutz M, Phan TS, Kündig TM, Levesque MP, Maul JT, Schlapbach C, LeibundGut-Landmann S, Mundt S, Becher B. IL-12 regulates type 3 immunity through interfollicular keratinocytes in psoriasiform inflammation. Sci Immunol 2021; 6:eabg9012. [PMID: 34678045 DOI: 10.1126/sciimmunol.abg9012] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Pascale Zwicky
- Institute of Experimental Immunology, University of Zurich, 8057 Zurich, Switzerland
| | - Florian Ingelfinger
- Institute of Experimental Immunology, University of Zurich, 8057 Zurich, Switzerland
| | - Bruno Marcel Silva de Melo
- Institute of Experimental Immunology, University of Zurich, 8057 Zurich, Switzerland.,Center for Research in Inflammatory Diseases, Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirão Preto Sao Paulo, Brazil
| | - Fiorella Ruchti
- Institute of Experimental Immunology, University of Zurich, 8057 Zurich, Switzerland.,Section of Immunology, Vetsuisse Faculty, University of Zurich, 8057 Zurich, Switzerland
| | - Stefanie Schärli
- Institute of Experimental Immunology, University of Zurich, 8057 Zurich, Switzerland
| | - Nicole Puertas
- Institute of Experimental Immunology, University of Zurich, 8057 Zurich, Switzerland
| | - Mirjam Lutz
- Institute of Experimental Immunology, University of Zurich, 8057 Zurich, Switzerland
| | - Truong San Phan
- Department of Dermatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Thomas M Kündig
- Department of Dermatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Mitchell P Levesque
- Department of Dermatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Julia-Tatjana Maul
- Department of Dermatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Christoph Schlapbach
- Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Salomé LeibundGut-Landmann
- Institute of Experimental Immunology, University of Zurich, 8057 Zurich, Switzerland.,Section of Immunology, Vetsuisse Faculty, University of Zurich, 8057 Zurich, Switzerland
| | - Sarah Mundt
- Institute of Experimental Immunology, University of Zurich, 8057 Zurich, Switzerland
| | - Burkhard Becher
- Institute of Experimental Immunology, University of Zurich, 8057 Zurich, Switzerland
| |
Collapse
|
18
|
Oxidative Stress and Inflammatory Mediators in Exhaled Breath Condensate of Patients with Pulmonary Tuberculosis. A Pilot Study with a Biomarker Perspective. Antioxidants (Basel) 2021; 10:antiox10101572. [PMID: 34679707 PMCID: PMC8533495 DOI: 10.3390/antiox10101572] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 09/22/2021] [Accepted: 09/23/2021] [Indexed: 12/21/2022] Open
Abstract
Tuberculosis (TB) is one of the highest infectious burdens worldwide. An excess of inflammation and inadequate antioxidant defense mechanisms are believed to lead to chronic inflammation and lung damage in tuberculosis (TB). However, circulating metabolites do not always replicate lung-associated biomarkers that define the pathobiology of the disease. The objective of this study was to determine the utility of exhaled breath condensate (EBC), a non-invasive and straightforward sample, to evaluate alveolar space-derived metabolites of redox state and inflammation. We assessed the levels of exhaled oxidant/antioxidant parameters (8-isoprostane, MDA, GSH), inflammatory markers, such as nucleosomes, cytokines (IL-2, IL-4, IL-6 and IL-8, IL-10, GM-CSF, TNF-α, and IFN-γ) and lipid mediators (PGE2, LTB4, RvD1, and Mar1), in patients with recently diagnosed pulmonary TB and healthy controls’ EBC and serum. The TB patients showed 36% lower GSH levels, and 2-, 1.4-, 1.1-, and 50-fold higher levels of 8-isoprostanes, nucleosomes, IL-6, and LTB4, respectively, in EBC. There was no correlation between EBC and serum, highlighting the importance of measuring local biomarkers. Quantitation of local inflammatory molecules and redox states in EBC would help find biomarkers useful for pharmacological and follow-up studies in pulmonary tuberculosis.
Collapse
|
19
|
Zhang X, Smith SM, Wang X, Zhao B, Wu L, Hu X. Three paralogous clusters of the miR-17~92 family of microRNAs restrain IL-12-mediated immune defense. Cell Mol Immunol 2021; 18:1751-1760. [PMID: 32015501 PMCID: PMC8245425 DOI: 10.1038/s41423-020-0363-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 01/01/2020] [Indexed: 02/06/2023] Open
Abstract
MicroRNAs (miRNAs) have been widely implicated in immune regulation, but evidence for the coordinated function of paralogous miRNA clusters remains scarce. Here, by using genetically modified mice with individual or combined cluster deficiencies, we found that three paralogous clusters of the miR-17~92 family of miRNAs collectively suppressed IL-12 production in macrophages. Accordingly, miR-17~92 family miRNAs deficiencies resulted in heightened production of IL-12 and thus enhanced the host defense against intracellular pathogen Listeria monocytogenes in vivo. Mechanistically, different members of the miR-17~92 family of miRNAs acted on a common target, PTEN, to inhibit IL-12 expression by modulating the PI3K-Akt-GSK3 pathway. In addition, the expression of miR-17~92 family miRNAs was collectively inhibited by the transcription factor RBP-J, and RBP-J-associated macrophage functional defects were genetically rescued by deleting three clusters of miR-17~92 family miRNAs on a RBP-J null background. Thus, our results illustrated key roles of three clusters of miR-17~92 family miRNAs in cooperatively controlling IL-12-mediated immune responses and identified miR-17~92 family miRNAs as functional targets of RBP-J in macrophages.
Collapse
Affiliation(s)
- Xiang Zhang
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, 100084, China
- Tsinghua-Peking Center for Life Sciences, Beijing, 10084, China
- Beijing Key Laboratory for Immunological Research on Chronic Diseases, Beijing, 100084, China
| | - Sinead M Smith
- Department of Clinical Medicine, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Xi Wang
- Department of Immunology, Beijing Key Laboratory for Cancer Invasion and Metastasis, Advanced Innovation Center for Human Brain Protection, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Baohong Zhao
- Arthritis and Tissue Degeneration Program and the David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY, 10021, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, 10021, USA
| | - Li Wu
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, 100084, China
- Tsinghua-Peking Center for Life Sciences, Beijing, 10084, China
- Beijing Key Laboratory for Immunological Research on Chronic Diseases, Beijing, 100084, China
| | - Xiaoyu Hu
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, 100084, China.
- Tsinghua-Peking Center for Life Sciences, Beijing, 10084, China.
- Beijing Key Laboratory for Immunological Research on Chronic Diseases, Beijing, 100084, China.
| |
Collapse
|
20
|
Gezmis H, Doran T, Mayda Domac F, Yucel D, Karaci R, Kirac D. CD4+ and CD25+ T-cell response to short-time interferon-beta therapy on IL10, IL23A and FOXP3 genes in multiple sclerosis patients. Int J Clin Pract 2021; 75:e14238. [PMID: 33884734 DOI: 10.1111/ijcp.14238] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 04/10/2021] [Indexed: 12/22/2022] Open
Abstract
AIM OF THE STUDY Interferon-beta (IFN-β), multiple sclerosis (MS) drug for years, does not have therapeutic effects on each patient. Yet, a considerable portion has experienced no therapeutic response to IFN-β. Therefore, it is necessary to determine disease-specific biomarkers that affect drug response. Here, we aimed to determine the effects of interleukin 10 (IL10) and 23 (IL23A), as well as forkhead box P3 (FOXP3) genes on MS after IFN-β therapy. MATERIALS AND METHODS Peripheral blood mononuclear cells (PBMCs) of 42 MS patients were isolated to obtain CD4+ and CD25+ T cells. Both cell types were characterised by flow cytometry. To determine optimum drug concentration of IFN-β, cytotoxicity assays were assessed on each cell type for 4, 16, 24 and 48 hours respectively. Then, cells were cultured in the presence of 500 IU/mL of IFN-β. cDNA synthesis was performed after mRNA extraction. RT-PCR was performed to measure gene expressions of IL10, IL23A and FOXP3. Results were evaluated statistically. RESULTS It was found that the cytotoxic effect of IFN-β was more efficient as the exposure time was expanded regardless of drug concentration. Moreover, CD25+ T lymphocytes were more resistant to IFN-β. IL23A was down-regulated, whereas FOXP3 was up-regulated at 48 hours in CD4+ T cells. For CD25+ T cells, the graded increase in FOXP3 was obtained while IL10 expression was gradually decreased throughout the drug intake. CONCLUSION Although a considerable change in expression was obtained, the long-term IFN-β effect on both genes and cells should be determined by follow-up at least a year.
Collapse
Affiliation(s)
- Hazal Gezmis
- Department of Medical Biology, Faculty of Medicine, Yeditepe University, Istanbul, Turkey
- Department of Materials, University of Oxford, Oxfordshire, OX1 3PH, UK
| | - Tansu Doran
- Department of Medical Biology, Faculty of Medicine, Yeditepe University, Istanbul, Turkey
| | - Fusun Mayda Domac
- Department of Neurology, University of Health Sciences, Erenkoy Mental and Nervous Diseases Training and Research Hospital, Istanbul, Turkey
| | - Deniz Yucel
- Department of Histology and Embryology, Faculty of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Rahsan Karaci
- Department of Neurology, University of Health Sciences, Erenkoy Mental and Nervous Diseases Training and Research Hospital, Istanbul, Turkey
| | - Deniz Kirac
- Department of Medical Biology, Faculty of Medicine, Yeditepe University, Istanbul, Turkey
| |
Collapse
|
21
|
Sperti M, Malavolta M, Ciniero G, Borrelli S, Cavaglià M, Muscat S, Tuszynski JA, Afeltra A, Margiotta DPE, Navarini L. JAK inhibitors in immune-mediated rheumatic diseases: From a molecular perspective to clinical studies. J Mol Graph Model 2021; 104:107789. [DOI: 10.1016/j.jmgm.2020.107789] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 09/21/2020] [Accepted: 10/20/2020] [Indexed: 12/11/2022]
|
22
|
JAK Inhibitors for Axial Spondyloarthritis: What does the Future Hold? Curr Rheumatol Rep 2021; 23:34. [PMID: 33909185 DOI: 10.1007/s11926-021-01001-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/23/2021] [Indexed: 12/16/2022]
Abstract
PURPOSE OF REVIEW To discuss the potential role of JAK inhibitors (JAKis) as a new therapeutic class for the treatment of axial spondyloarthritis (axSpA, including ankylosing spondylitis [AS] and non-radiographic axSpA [nr-axSpA]). RECENT FINDINGS A phase III randomized controlled trial of tofacitinib (a "pan JAKi") in patients with active AS was found to be superior to placebo in achieving the ASAS20 primary endpoint at week 16 (56.4% and 29.4%, p < 0.0001, phase II trials of AS). Upadacitinib, a JAK1 inhibitor, has also been evaluated in a phase III trial for its efficacy and safety in AS. The primary endpoint, ASAS40 at week 16, was reached by 52% of the patients randomized to upadacitinib and 26% of the patients receiving placebo (p = 0·0003). All the important secondary endpoints also improved with both agents. No new changes in their safety profile were noted. However, the more frequent occurrence of cardiovascular and cancer adverse events associated with tofacitinib than with TNFi observed in the very recent post-marketing "ORAL surveillance" safety study, the results of which were released on January 27, 2021, may lead to safety concerns swirling around the whole class of JAKis. JAKis seem to be effective in treating signs and symptoms of AS but have not been studied in nr-axSpA. Both tofacitinib and upadacitinib have been pre-registered with the FDA for the treatment of AS. Upadacitinib has just recently received approval for this indication in the European Union..
Collapse
|
23
|
Theoharides TC, Conti P. Mast cells to dendritic cells: Let IL-13 shut your IL-12 down. J Allergy Clin Immunol 2021; 147:2073-2074. [PMID: 33831419 DOI: 10.1016/j.jaci.2021.03.032] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 03/12/2021] [Accepted: 03/18/2021] [Indexed: 12/17/2022]
Affiliation(s)
- Theoharis C Theoharides
- Laboratory of Molecular Immunopharmacology and Drug Discovery, Department of Immunology, Tufts University School of Medicine, Boston, Mass; School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, Mass; Department of Internal Medicine, Tufts University School of Medicine and Tufts Medical Center, Boston, Mass.
| | - Pio Conti
- Postgraduate Medical School, University of Chieti, Chieti, Italy
| |
Collapse
|
24
|
Bates FA, Duncan EH, Simmons M, Robinson T, Samineni S, Strbo N, Villasante E, Bergmann-Leitner E, Wijayalath W. Exposure-related, global alterations in innate and adaptive immunity; a consideration for re-use of non-human primates in research. PeerJ 2021; 9:e10955. [PMID: 33732548 PMCID: PMC7950202 DOI: 10.7717/peerj.10955] [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: 11/26/2020] [Accepted: 01/27/2021] [Indexed: 11/20/2022] Open
Abstract
Background Non-human primates (NHPs) play an important role in biomedical research, where they are often being re-used in multiple research studies over the course of their life-time. Researchers employ various study-specific screening criteria to reduce potential variables associated with subsequent re-use of NHPs. However, criteria set for NHP re-assignments largely neglect the impact of previous exposures on overall biology. Since the immune system is a key determinant of overall biological outcome, an altered biological state could be predicted by monitoring global changes in the immune profile. We postulate that every different exposure or a condition can generate a unique global immune profile in NHPs. Methods Changes in the global immune profile were evaluated in three different groups of rhesus macaques previously enrolled in dengue or malaria vaccine studies over six months after their last exposure. Naïve animals served as the baseline. Fresh blood samples were stained with various immune cell surface markers and analyzed by multi-color flow-cytometry to study immune cell dynamics in the peripheral blood. Serum cytokine profile in the pre-exposed animals were analyzed by mesoscale assay using a customized U-PLEX NHP biomarker panel of 12 cytokines/chemokines. Results Pre-exposed macaques showed altered dynamics in circulating cytokines and certain innate and adaptive immune cell subsets such as monocytes, HLA-DR+NKT cells, B cells and T cells. Some of these changes were transient, while some lasted for more than six months. Each group seemed to develop a global immune profile unique to their particular exposure. Conclusion Our data strongly suggest that re-used NHPs should be evaluated for long-term, overall immunological changes and randomly assigned to new studies to avoid study bias.
Collapse
Affiliation(s)
- François A Bates
- Veterinary Services Program, Walter Reed Army Institute of Research, Silver Spring, MD, United States of America
| | - Elizabeth H Duncan
- Immunology Core/Malaria Biologics Branch, Walter Reed Army Institute of Research, Silver Spring, MD, United States of America
| | - Monika Simmons
- Viral and Rickettsial Diseases Department, Naval Medical Research Center, Silver Spring, MD, United States of America
| | - Tanisha Robinson
- Immunology Core/Malaria Biologics Branch, Walter Reed Army Institute of Research, Silver Spring, MD, United States of America.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc. (HJF), Bethesda, MD, United States of America
| | - Sridhar Samineni
- Veterinary Services Program, Walter Reed Army Institute of Research, Silver Spring, MD, United States of America.,SoBran, Inc, Falls Church, VA, United States of America
| | - Natasa Strbo
- Department of Microbiology and Immunology, Miller School of Medicine University of Miami, Miami, FL, United States of America
| | - Eileen Villasante
- Malaria Department, Naval Medical Research Center, Silver Spring, MD, United States of America
| | - Elke Bergmann-Leitner
- Immunology Core/Malaria Biologics Branch, Walter Reed Army Institute of Research, Silver Spring, MD, United States of America
| | - Wathsala Wijayalath
- Malaria Department, Naval Medical Research Center, Silver Spring, MD, United States of America.,CAMRIS International, Bethesda, MD, United States of America
| |
Collapse
|
25
|
Eld HMS, Nielsen EM, Johnsen PR, Marengo M, Kamper IW, Frederiksen L, Bonomi F, Frees D, Iametti S, Frøkiær H. Cefoxitin treatment of MRSA leads to a shift in the IL-12/IL-23 production pattern in dendritic cells by a mechanism involving changes in the MAPK signaling. Mol Immunol 2021; 134:1-12. [PMID: 33676343 DOI: 10.1016/j.molimm.2021.02.025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 02/11/2021] [Accepted: 02/23/2021] [Indexed: 12/25/2022]
Abstract
Methicillin resistant Staphylococcus aureus (MRSA) constitute a serious health care problem worldwide. This study addresses the effect of β-lactam treatment on the ability of clinically relevant MRSA strains to induce IL-12 and IL-23. MRSA strains induced a dose-dependent IL-12 response in murine bone-marrow-derived dendritic cells that was dependent on endocytosis and acidic degradation. Facilitated induction of IL-12 (but not of IL-23) called for activation of the MAP kinase JNK, and was suppressed by p38. Compromised peptidoglycan structure in cefoxitin-treated bacteria - as denoted by increased sensitivity to mutanolysin -caused a shift from IL-12 towards IL-23. Moreover, cefoxitin treatment of MRSA led to a p38 MAPK-dependent early up-regulation of Dual Specificity Phosphatase (DUSP)-1. Compared to common MRSA, characteristics associated with a persister phenotype increased intracellular survival and upon cefoxitin treatment, the peptidoglycan was not equally compromised and the cytokine induction still required phagosomal acidification. Together, these data demonstrate that β-lactam treatment changes the MRSA-induced IL-12/IL-23 pattern determined by the activation of JNK and p38. We suggest that accelerated endosomal degradation of the peptidoglycan in cefoxitin-treated MRSA leads to an early expression of DUSP-1 and accordingly, a reduction in the IL-12/IL-23 ratio in dendritic cells. This may influence the clearance of S. aureus.
Collapse
Affiliation(s)
- Helene M S Eld
- Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Emilie M Nielsen
- Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Peter R Johnsen
- Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mauro Marengo
- Department of Food, Environmental and Nutritional Science, Università degli Studi di Milano, Milan, Italy
| | - Ida W Kamper
- Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lise Frederiksen
- Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Francesco Bonomi
- Department of Food, Environmental and Nutritional Science, Università degli Studi di Milano, Milan, Italy
| | - Dorte Frees
- Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Stefania Iametti
- Department of Food, Environmental and Nutritional Science, Università degli Studi di Milano, Milan, Italy
| | - Hanne Frøkiær
- Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
26
|
Tang PCT, Chan ASW, Zhang CB, García Córdoba CA, Zhang YY, To KF, Leung KT, Lan HY, Tang PMK. TGF-β1 Signaling: Immune Dynamics of Chronic Kidney Diseases. Front Med (Lausanne) 2021; 8:628519. [PMID: 33718407 PMCID: PMC7948440 DOI: 10.3389/fmed.2021.628519] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 01/21/2021] [Indexed: 12/13/2022] Open
Abstract
Chronic kidney disease (CKD) is a major cause of morbidity and mortality worldwide, imposing a great burden on the healthcare system. Regrettably, effective CKD therapeutic strategies are yet available due to their elusive pathogenic mechanisms. CKD is featured by progressive inflammation and fibrosis associated with immune cell dysfunction, leading to the formation of an inflammatory microenvironment, which ultimately exacerbating renal fibrosis. Transforming growth factor β1 (TGF-β1) is an indispensable immunoregulator promoting CKD progression by controlling the activation, proliferation, and apoptosis of immunocytes via both canonical and non-canonical pathways. More importantly, recent studies have uncovered a new mechanism of TGF-β1 for de novo generation of myofibroblast via macrophage-myofibroblast transition (MMT). This review will update the versatile roles of TGF-β signaling in the dynamics of renal immunity, a better understanding may facilitate the discovery of novel therapeutic strategies against CKD.
Collapse
Affiliation(s)
- Philip Chiu-Tsun Tang
- State Key Laboratory of Translational Oncology, Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Alex Siu-Wing Chan
- Department of Applied Social Sciences, The Hong Kong Polytechnic University, Hung Hom, Hong Kong
| | - Cai-Bin Zhang
- State Key Laboratory of Translational Oncology, Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Cristina Alexandra García Córdoba
- State Key Laboratory of Translational Oncology, Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Ying-Ying Zhang
- Department of Nephrology, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ka-Fai To
- State Key Laboratory of Translational Oncology, Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Kam-Tong Leung
- Department of Paediatrics, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Hui-Yao Lan
- Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong.,Guangdong-Hong Kong Joint Laboratory on Immunological and Genetic Kidney Diseases, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Patrick Ming-Kuen Tang
- State Key Laboratory of Translational Oncology, Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Shatin, Hong Kong
| |
Collapse
|
27
|
Ullrich KAM, Schulze LL, Paap EM, Müller TM, Neurath MF, Zundler S. Immunology of IL-12: An update on functional activities and implications for disease. EXCLI JOURNAL 2020; 19:1563-1589. [PMID: 33408595 PMCID: PMC7783470 DOI: 10.17179/excli2020-3104] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 12/07/2020] [Indexed: 12/15/2022]
Abstract
As its first identified member, Interleukin-12 (IL-12) named a whole family of cytokines. In response to pathogens, the heterodimeric protein, consisting of the two subunits p35 and p40, is secreted by phagocytic cells. Binding of IL-12 to the IL-12 receptor (IL-12R) on T and natural killer (NK) cells leads to signaling via signal transducer and activator of transcription 4 (STAT4) and subsequent interferon gamma (IFN-γ) production and secretion. Signaling downstream of IFN-γ includes activation of T-box transcription factor TBX21 (Tbet) and induces pro-inflammatory functions of T helper 1 (TH1) cells, thereby linking innate and adaptive immune responses. Initial views on the role of IL-12 and clinical efforts to translate them into therapeutic approaches had to be re-interpreted following the discovery of other members of the IL-12 family, such as IL-23, sharing a subunit with IL-12. However, the importance of IL-12 with regard to immune processes in the context of infection and (auto-) inflammation is still beyond doubt. In this review, we will provide an update on functional activities of IL-12 and their implications for disease. We will begin with a summary on structure and function of the cytokine itself as well as its receptor and outline the signal transduction and the transcriptional regulation of IL-12 secretion. In the second part of the review, we will depict the involvement of IL-12 in immune-mediated diseases and relevant experimental disease models, while also providing an outlook on potential translational approaches.
Collapse
Affiliation(s)
- Karen A.-M. Ullrich
- Department of Medicine and Deutsches Zentrum Immuntherapie, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg, Germany
| | - Lisa Lou Schulze
- Department of Medicine and Deutsches Zentrum Immuntherapie, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg, Germany
| | - Eva-Maria Paap
- Department of Medicine and Deutsches Zentrum Immuntherapie, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg, Germany
| | - Tanja M. Müller
- Department of Medicine and Deutsches Zentrum Immuntherapie, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg, Germany
| | - Markus F. Neurath
- Department of Medicine and Deutsches Zentrum Immuntherapie, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg, Germany
| | - Sebastian Zundler
- Department of Medicine and Deutsches Zentrum Immuntherapie, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg, Germany
| |
Collapse
|
28
|
Del Duca E, Morelli P, Bennardo L, Di Raimondo C, Nisticò SP. Cytokine Pathways and Investigational Target Therapies in Hidradenitis Suppurativa. Int J Mol Sci 2020; 21:ijms21228436. [PMID: 33182701 PMCID: PMC7696820 DOI: 10.3390/ijms21228436] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 11/05/2020] [Accepted: 11/06/2020] [Indexed: 12/13/2022] Open
Abstract
Background: Hidradenitis suppurativa (HS) is a chronic inflammatory skin disease affecting areas with a high density of apocrine glands and characterized by subcutaneous nodules that may evolve into fistulas with pus secretion. Methods: The aim of this review is to investigate all current knowledge on cytokine regulation in the pathogenesis of HS. A systematic literature research using the words “cytokine”, “interleukin”, “pathway”, and “hidradenitis suppurativa” was performed in PubMed/Medline and Scopus/Embase databases. A search of the clinicaltrials.gov website for interventional recruiting and completed trials including the term “hidradenitis suppurativa” was also performed up to August 2020. We will discuss the pathogenetic role of various cytokines in HS and potential therapeutic targets for this debilitating disease. Results: The pathophysiology underlying this complex condition has not been clearly defined. An upregulation of various cytokines, such as tumor necrosis factor alpha (TNF-α), interleukin (IL)-1, IL-17, IL-23, and other molecules seems to be related to this inflammatory condition. Various cells, such as lymphocytes T Helper 1 and 17 and keratinocytes seem to be involved in the genesis of this condition. Conclusions: Several future studies and clinical trials are necessary in order to have new knowledge about HS and to properly treat this complex condition.
Collapse
Affiliation(s)
- Ester Del Duca
- Department of Health Science, University of Catanzaro Magna Graecia, 88100 Catanzaro, Italy; (P.M.); (L.B.); (S.P.N.)
- Correspondence: ; Tel.: +39-917-9694-386; Fax: +39-0961-369-6150
| | - Paola Morelli
- Department of Health Science, University of Catanzaro Magna Graecia, 88100 Catanzaro, Italy; (P.M.); (L.B.); (S.P.N.)
| | - Luigi Bennardo
- Department of Health Science, University of Catanzaro Magna Graecia, 88100 Catanzaro, Italy; (P.M.); (L.B.); (S.P.N.)
| | - Cosimo Di Raimondo
- Department of Dermatology, University of Rome Tor Vergata, 00133 Rome, Italy;
| | - Steven Paul Nisticò
- Department of Health Science, University of Catanzaro Magna Graecia, 88100 Catanzaro, Italy; (P.M.); (L.B.); (S.P.N.)
| |
Collapse
|
29
|
Faust HJ, Zhang H, Han J, Wolf MT, Jeon OH, Sadtler K, Peña AN, Chung L, Maestas DR, Tam AJ, Pardoll DM, Campisi J, Housseau F, Zhou D, Bingham CO, Elisseeff JH. IL-17 and immunologically induced senescence regulate response to injury in osteoarthritis. J Clin Invest 2020; 130:5493-5507. [PMID: 32955487 PMCID: PMC7524483 DOI: 10.1172/jci134091] [Citation(s) in RCA: 155] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 07/09/2020] [Indexed: 12/17/2022] Open
Abstract
Senescent cells (SnCs) are implicated in the pathogenesis of age-related diseases including osteoarthritis (OA), in part via expression of a senescence-associated secretory phenotype (SASP) that includes immunologically relevant factors and cytokines. In a model of posttraumatic OA (PTOA), anterior cruciate ligament transection (ACLT) induced a type 17 immune response in the articular compartment and draining inguinal lymph nodes (LNs) that paralleled expression of the senescence marker p16INK4a (Cdkn2a) and p21 (Cdkn1a). Innate lymphoid cells, γδ+ T cells, and CD4+ T cells contributed to IL-17 expression. Intra-articular injection of IL-17-neutralizing antibody reduced joint degeneration and decreased expression of the senescence marker Cdkn1a. Local and systemic senolysis was required to attenuate tissue damage in aged animals and was associated with decreased IL-17 and increased IL-4 expression in the articular joint and draining LNs. In vitro, we found that Th17 cells induced senescence in fibroblasts and that SnCs skewed naive T cells toward Th17 or Th1, depending on the presence of TGF-β. The SASP profile of the inflammation-induced SnCs included altered Wnt signaling, tissue remodeling, and cell-cycle pathways not previously implicated in senescence. These findings provide molecular targets and mechanisms for senescence induction and therapeutic strategies to support tissue healing in an aged environment.
Collapse
Affiliation(s)
- Heather J. Faust
- Translational Tissue Engineering Center, Wilmer Eye Institute and Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Hong Zhang
- Translational Tissue Engineering Center, Wilmer Eye Institute and Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jin Han
- Translational Tissue Engineering Center, Wilmer Eye Institute and Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Matthew T. Wolf
- Translational Tissue Engineering Center, Wilmer Eye Institute and Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Ok Hee Jeon
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, South Korea
| | - Kaitlyn Sadtler
- Translational Tissue Engineering Center, Wilmer Eye Institute and Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Alexis N. Peña
- Translational Tissue Engineering Center, Wilmer Eye Institute and Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Liam Chung
- Translational Tissue Engineering Center, Wilmer Eye Institute and Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - David R. Maestas
- Translational Tissue Engineering Center, Wilmer Eye Institute and Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Ada J. Tam
- Bloomberg-Kimmel Institute for Cancer Immunotherapy and
| | - Drew M. Pardoll
- Bloomberg-Kimmel Institute for Cancer Immunotherapy and
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Judith Campisi
- Buck Institute for Research on Aging, Novato, California, USA
| | | | - Daohong Zhou
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida, USA
| | - Clifton O. Bingham
- Division of Rheumatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jennifer H. Elisseeff
- Translational Tissue Engineering Center, Wilmer Eye Institute and Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Bloomberg-Kimmel Institute for Cancer Immunotherapy and
| |
Collapse
|
30
|
Gow J, Yang Y, Govindraj M, Guo C. Nitric Oxide Regulates Macrophage Fungicidal Activity via S-nitrosylation of Dectin-1. ACTA ACUST UNITED AC 2020; 6:90-98. [PMID: 32953945 PMCID: PMC7500157 DOI: 10.1089/aivt.2020.0009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Introduction: Recognition of fungal surface β-glucan by pattern recognition receptor Dectin-1 is a critical process for fungal clearance in the lung. In humans, persistent fungal infection is observed in individuals with particular Dectin-1 polymorphism. We have identified that nitric oxide (NO) modifies critical cysteines in pattern recognition molecules to disassemble and alter protein function. There is a hydrophobic S-nitrosylation motif present in surfactant protein-D (SP-D) that is also present in Dectin-1. We hypothesized that Dectin-1 can be modified by nitrosative stress potentially leading to impairment of fungal clearance. Materials and Methods: Recombinant Dectin-1 was incubated with l-nitrosocysteine (L-SNOC) and S-nitrosylated Dectin-1 was detected by Biotin-switch assay. Cells of a murine macrophage line (Raw 264.7) were incubated with S-nitroso-glutathione (GSNO) and Dectin-1 shedding from the cell surface was determined by Western blot. Dectin-1 quaternary structure was determined by native gel electrophoresis. Dectin-1 function was assayed by NF-κB activity and IL-6 mRNA real-time polymerase chain reaction (PCR). Phagocytic activity was measured by fluorescence labeled zymosan beads. Results: Dectin-1 was S-nitrosylated by l-nitrosocysteine (L-SNOC) in vitro, as determined by Biotin-switch assay, resulting in structural disruption. We used Western blotting and flow cytometry to demonstrate that incubation of a murine macrophage cell line (Raw 264.7 cells) with GSNO reduced the surface Dectin-1 expression as a result of shedding to the media. The shedding of Dectin-1 is due to formation of S-nitrosothiol (SNO)-Dectin-1 and disruption of the Dectin-1 oligomeric complex. GSNO also induces Dectin-1 shedding from the cell surface. The functional significance of GSNO treatment of macrophages is shown by reduced β-glucan-mediated signaling in terms of NF-κB function and IL-6 expression. Finally, it was demonstrated that GSNO treatment reduces the capability of macrophages to phagocytose zymosan. Conclusions: These data provide mechanistic data to support the role of Dectin-1 nitrosylation as a mediator of reduced fungal clearance in the face of increased NO exposure.
Collapse
Affiliation(s)
- James Gow
- Department of Pharmacology & Toxicology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
| | - Yujie Yang
- Department of Pharmacology & Toxicology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
| | - Mohan Govindraj
- Department of Pharmacology & Toxicology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
| | - Changjiang Guo
- Department of Pharmacology & Toxicology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
| |
Collapse
|
31
|
Wu CT, Lee YT, Ku MS, Lue KH. Role of biomarkers and effect of FIP-fve in acute and chronic animal asthma models. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2020; 53:996-1007. [PMID: 32778497 DOI: 10.1016/j.jmii.2020.07.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 06/08/2020] [Accepted: 07/14/2020] [Indexed: 10/23/2022]
Abstract
BACKGROUND Asthma is a consequence of complex gene-environment interactions. Exploring the heterogeneity of asthma in different stages is contributing to our understanding of its pathogenesis and the development of new therapeutic strategies, especially in severe cases. OBJECTIVE This study aimed to further understand the relationship between manifestations of acute and chronic asthma and various endotypes, and explore the severity of lung inflammation, cell types, cytokine/chemokine differences, and the effects of FIP-fve. MATERIALS AND METHODS Acute and chronic OVA-sensitization mouse asthma models, based on our previously published method, were used and FIP-fve was used to evaluate the effect on these two models. BALF cytokines/chemokines were detected according to the manufacturer's protocol. RESULTS Seventeen cytokine/chemokine secretions were higher in the chronic stage than in the acute stage. Whether in acute stage or chronic stage, the FIP-fve treatment groups had reduced airway hyperresponsiveness, infiltration of airway inflammatory cells, secretion of cytokines, chemokines by Th2 cells, and TNF-α, IL-8, IL-17, CXCL-1, CXCL-10, CCL-17, and CCL-22, and it was also found that the Treg cell cytokine IL-10 had increased significantly. PCA (Principal Component Analysis) was also used to compare statistics and laboratory data to find the important biomarkers in different stages and after treatment with FIP-fve. CONCLUSIONS There are many different immune responses in the different stages of the asthma process. Drug treatment at the appropriate times might help reduce the worsening of asthma.
Collapse
Affiliation(s)
- Chia-Ta Wu
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan; Department of Emergency Medicine, Changhua Christian Hospital, Changhua, Taiwan.
| | - Yu-Tzu Lee
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Min-Sho Ku
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan; Department of Pediatrics, Chung Shan Medical University Hospital, Taichung, Taiwan.
| | - Ko-Huang Lue
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan; School of Medicine, Chung Shan Medical University, Taichung, Taiwan; Department of Pediatrics, Chung Shan Medical University Hospital, Taichung, Taiwan; College of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan.
| |
Collapse
|
32
|
Milovanovic J, Arsenijevic A, Stojanovic B, Kanjevac T, Arsenijevic D, Radosavljevic G, Milovanovic M, Arsenijevic N. Interleukin-17 in Chronic Inflammatory Neurological Diseases. Front Immunol 2020; 11:947. [PMID: 32582147 PMCID: PMC7283538 DOI: 10.3389/fimmu.2020.00947] [Citation(s) in RCA: 126] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 04/22/2020] [Indexed: 12/15/2022] Open
Abstract
A critical role for IL-17, a cytokine produced by T helper 17 (Th17) cells, has been indicated in the pathogenesis of chronic inflammatory and autoimmune diseases. A positive effect of blockade of IL-17 secreted by autoreactive T cells has been shown in various inflammatory diseases. Several cytokines, whose production is affected by environmental factors, control Th17 differentiation and its maintenance in tissues during chronic inflammation. The roles of IL-17 in the pathogenesis of chronic neuroinflammatory conditions, multiple sclerosis (MS), experimental autoimmune encephalomyelitis (EAE), Alzheimer's disease, and ischemic brain injury are reviewed here. The role of environmental stimuli in Th17 differentiation is also summarized, highlighting the role of viral infection in the regulation of pathogenic T helper cells in EAE.
Collapse
Affiliation(s)
- Jelena Milovanovic
- Faculty of Medical Sciences, Center for Molecular Medicine and Stem Cell Research, University of Kragujevac, Kragujevac, Serbia
- Department of Histology and Embriology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Aleksandar Arsenijevic
- Faculty of Medical Sciences, Center for Molecular Medicine and Stem Cell Research, University of Kragujevac, Kragujevac, Serbia
| | - Bojana Stojanovic
- Faculty of Medical Sciences, Center for Molecular Medicine and Stem Cell Research, University of Kragujevac, Kragujevac, Serbia
- Department of Pathophysiology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Tatjana Kanjevac
- Department of Dentistry, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Dragana Arsenijevic
- Faculty of Medical Sciences, Center for Molecular Medicine and Stem Cell Research, University of Kragujevac, Kragujevac, Serbia
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Gordana Radosavljevic
- Faculty of Medical Sciences, Center for Molecular Medicine and Stem Cell Research, University of Kragujevac, Kragujevac, Serbia
| | - Marija Milovanovic
- Faculty of Medical Sciences, Center for Molecular Medicine and Stem Cell Research, University of Kragujevac, Kragujevac, Serbia
| | - Nebojsa Arsenijevic
- Faculty of Medical Sciences, Center for Molecular Medicine and Stem Cell Research, University of Kragujevac, Kragujevac, Serbia
| |
Collapse
|
33
|
Fatima S, Kumari A, Das G, Dwivedi VP. Tuberculosis vaccine: A journey from BCG to present. Life Sci 2020; 252:117594. [PMID: 32305522 DOI: 10.1016/j.lfs.2020.117594] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 03/21/2020] [Accepted: 03/26/2020] [Indexed: 12/15/2022]
Abstract
Tuberculosis (TB) is the leading cause of death worldwide due to an infectious disease, causing around 1.6 million deaths each year. This situation has become more complicated by the emergence of drug-resistant Mycobacterium tuberculosis (M.tb) and HIV-TB co-infection, which has significantly worsened TB prognosis and treatment. Despite years of intensive research, Bacille Calmette-Guerin (BCG) remains the only licensed vaccine and has variable efficacy. It provides protection against childhood TB but is not effective in adult pulmonary TB. As a result of intense research in understanding TB vaccinology, there are many new vaccine candidates in clinical development and many more in pre-clinical trials which aim either to replace or boost BCG vaccine. This review discusses the history of BCG vaccine development and summarizes limitations of the current vaccine strategy and recent advances in improving BCG immunization along with other new vaccines in clinical trials which are promising candidates for the future tuberculosis vaccinology program.
Collapse
Affiliation(s)
- Samreen Fatima
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, India
| | - Anjna Kumari
- Immunobiology Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Gobardhan Das
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, India
| | - Ved Prakash Dwivedi
- Immunobiology Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India.
| |
Collapse
|
34
|
Samotij D, Nedoszytko B, Bartosińska J, Batycka-Baran A, Czajkowski R, Dobrucki IT, Dobrucki LW, Górecka-Sokołowska M, Janaszak-Jasienicka A, Krasowska D, Kalinowski L, Macieja-Stawczyk M, Nowicki RJ, Owczarczyk-Saczonek A, Płoska A, Purzycka-Bohdan D, Radulska A, Reszka E, Siekierzycka A, Słomiński A, Słomiński R, Sobalska-Kwapis M, Strapagiel D, Szczerkowska-Dobosz A, Szczęch J, Żmijewski M, Reich A. Pathogenesis of psoriasis in the "omic" era. Part I. Epidemiology, clinical manifestation, immunological and neuroendocrine disturbances. Postepy Dermatol Alergol 2020; 37:135-153. [PMID: 32489346 PMCID: PMC7262814 DOI: 10.5114/ada.2020.94832] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 04/01/2020] [Indexed: 02/06/2023] Open
Abstract
Psoriasis is a common, chronic, inflammatory, immune-mediated skin disease affecting about 2% of the world's population. According to current knowledge, psoriasis is a complex disease that involves various genes and environmental factors, such as stress, injuries, infections and certain medications. The chronic inflammation of psoriasis lesions develops upon epidermal infiltration, activation, and expansion of type 1 and type 17 Th cells. Despite the enormous progress in understanding the mechanisms that cause psoriasis, the target cells and antigens that drive pathogenic T cell responses in psoriatic lesions are still unproven and the autoimmune basis of psoriasis still remains hypothetical. However, since the identification of the Th17 cell subset, the IL-23/Th17 immune axis has been considered a key driver of psoriatic inflammation, which has led to the development of biologic agents that target crucial elements of this pathway. Here we present the current understanding of various aspects in psoriasis pathogenesis.
Collapse
Affiliation(s)
- Dominik Samotij
- Department of Dermatology, University of Rzeszow, Rzeszow, Poland
| | - Bogusław Nedoszytko
- Department of Dermatology, Venereology and Allergology, Medical University of Gdansk, Gdansk, Poland
| | - Joanna Bartosińska
- Department of Dermatology, Venereology and Pediatric Dermatology, Medical University of Lublin, Lublin, Poland
| | - Aleksandra Batycka-Baran
- Department of Dermatology, Venereology and Allergology, Wroclaw Medical University, Wroclaw, Poland
| | - Rafał Czajkowski
- Department of Dermatology and Venereology, Faculty of Medicine, Ludwik Rydygier Medical College in Bydgoszcz, Nicolaus Copernicus University in Torun, Poland
| | - Iwona T. Dobrucki
- Beckman Institute for Advanced Science and Technology, Urbana, IL, USA
| | - Lawrence W. Dobrucki
- Beckman Institute for Advanced Science and Technology, Urbana, IL, USA
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA
- Department of Medical Laboratory Diagnostics, Medical University of Gdansk, Gdansk, Poland
- Biobanking and Biomolecular Resources Research Infrastructure, (BBMRI.PL), Gdansk, Poland
| | - Magdalena Górecka-Sokołowska
- Department of Dermatology, Sexually Transmitted Disorders and Immunodermatology, Jurasz University Hospital No. 1, Bydgoszcz, Poland
| | - Anna Janaszak-Jasienicka
- Department of Medical Laboratory Diagnostics, Medical University of Gdansk, Gdansk, Poland
- Biobanking and Biomolecular Resources Research Infrastructure, (BBMRI.PL), Gdansk, Poland
| | - Dorota Krasowska
- Department of Dermatology, Venereology and Pediatric Dermatology, Medical University of Lublin, Lublin, Poland
| | - Leszek Kalinowski
- Department of Medical Laboratory Diagnostics, Medical University of Gdansk, Gdansk, Poland
- Biobanking and Biomolecular Resources Research Infrastructure, (BBMRI.PL), Gdansk, Poland
| | - Marta Macieja-Stawczyk
- Department of Dermatology, Venereology and Allergology, Medical University of Gdansk, Gdansk, Poland
| | - Roman J. Nowicki
- Department of Dermatology, Venereology and Allergology, Medical University of Gdansk, Gdansk, Poland
| | - Agnieszka Owczarczyk-Saczonek
- Department of Dermatology, Sexually Transmitted Diseases and Clinical Immunology, University of Warmia and Mazury, Olsztyn, Poland
| | - Agata Płoska
- Department of Medical Laboratory Diagnostics, Medical University of Gdansk, Gdansk, Poland
- Biobanking and Biomolecular Resources Research Infrastructure, (BBMRI.PL), Gdansk, Poland
| | - Dorota Purzycka-Bohdan
- Department of Dermatology, Venereology and Allergology, Medical University of Gdansk, Gdansk, Poland
| | - Adrianna Radulska
- Department of Medical Laboratory Diagnostics, Medical University of Gdansk, Gdansk, Poland
- Biobanking and Biomolecular Resources Research Infrastructure, (BBMRI.PL), Gdansk, Poland
| | - Edyta Reszka
- Department of Molecular Genetics and Epigenetics, Nofer Institute of Occupational Medicine, Lodz, Poland
| | - Anna Siekierzycka
- Department of Medical Laboratory Diagnostics, Medical University of Gdansk, Gdansk, Poland
- Biobanking and Biomolecular Resources Research Infrastructure, (BBMRI.PL), Gdansk, Poland
| | - Andrzej Słomiński
- Department of Dermatology, Birmingham, AL, USA
- Comprehensive Cancer Center, Cancer Chemoprevention Program, Birmingham, AL, USA
- VA Medical Center, Birmingham, AL, USA
| | - Radomir Słomiński
- Department of Medicine, Division of Rheumatology, University of Alabama, Birmingham, AL, USA
| | - Marta Sobalska-Kwapis
- Biobank Lab, Department of Molecular Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| | - Dominik Strapagiel
- Biobank Lab, Department of Molecular Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| | - Aneta Szczerkowska-Dobosz
- Department of Dermatology, Venereology and Allergology, Medical University of Gdansk, Gdansk, Poland
| | - Justyna Szczęch
- Department of Dermatology, University of Rzeszow, Rzeszow, Poland
| | - Michał Żmijewski
- Department of Histology, Medical University of Gdansk, Gdansk, Poland
| | - Adam Reich
- Department of Dermatology, University of Rzeszow, Rzeszow, Poland
| |
Collapse
|
35
|
Abdo AIK, Tye GJ. Interleukin 23 and autoimmune diseases: current and possible future therapies. Inflamm Res 2020; 69:463-480. [PMID: 32215665 DOI: 10.1007/s00011-020-01339-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 02/21/2020] [Accepted: 03/16/2020] [Indexed: 12/17/2022] Open
Abstract
PURPOSE IL-23 is a central proinflammatory cytokine with a wide range of influence over immune response. It is implicated in several autoimmune diseases due to the infinite inflammatory loops it can create through the positive feedbacks of both IL-17 and IL-22 arms. This made IL-23 a key target of autoimmune disorders therapy, which indeed was proven to inhibit inflammation and ameliorate diseases. Current autoimmune treatments targeting IL-23 are either by preventing IL-23 ligation to its receptor (IL-23R) via antibodies or inhibiting IL-23 signaling by signaling downstream mediators' inhibitors, with each approach having its own pros and cons. METHODS Literature review was done to further understand the biology of IL-23 and current therapies. RESULTS In this review, we discuss the biological features of IL-23 and its role in the pathogenesis of autoimmune diseases including psoriasis, rheumatoid arthritis and inflammatory bowel diseases. Advantages, limitations and side effects of each concept will be reviewed, suggesting several advanced IL-23-based bio-techniques to generate new and possible future therapies to overcome current treatments problems.
Collapse
Affiliation(s)
- Ahmad Ismail Khaled Abdo
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, 11800, Minden, Penang, Malaysia
| | - Gee Jun Tye
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, 11800, Minden, Penang, Malaysia.
| |
Collapse
|
36
|
Zhang X, Li X, Ning F, Shang Y, Hu X. TLE4 acts as a corepressor of Hes1 to inhibit inflammatory responses in macrophages. Protein Cell 2020; 10:300-305. [PMID: 29869113 PMCID: PMC6418302 DOI: 10.1007/s13238-018-0554-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Affiliation(s)
- Xiang Zhang
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, 100084, China.,Beijing Key Laboratory for Immunological Research on Chronic Diseases, Beijing, 100084, China.,School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Xiaoyu Li
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, 100084, China.,Beijing Key Laboratory for Immunological Research on Chronic Diseases, Beijing, 100084, China
| | - Fei Ning
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, 100084, China.,Beijing Key Laboratory for Immunological Research on Chronic Diseases, Beijing, 100084, China.,School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Yingli Shang
- College of Veterinary Medicine, Shandong Agricultural University, Taian, 271018, China
| | - Xiaoyu Hu
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, 100084, China. .,Beijing Key Laboratory for Immunological Research on Chronic Diseases, Beijing, 100084, China. .,Collaborative Innovation Center for Biotherapy, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
37
|
The Nature of Antibacterial Adaptive Immune Responses against Staphylococcus aureus Is Dependent on the Growth Phase and Extracellular Peptidoglycan. Infect Immun 2019; 88:IAI.00733-19. [PMID: 31685545 DOI: 10.1128/iai.00733-19] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 10/21/2019] [Indexed: 02/07/2023] Open
Abstract
Staphylococcus aureus has evolved different strategies to evade the immune response, which play an important role in its pathogenesis. The bacteria express and shed various cell wall components and toxins during different stages of growth that may affect the protective T cell responses to extracellular and intracellular S. aureus However, if and how the dendritic cell (DC)-mediated T cell response against S. aureus changes during growth of the bacterium remain elusive. In this study, we show that exponential-phase (EP) S. aureus bacteria were endocytosed very efficiently by human DCs, and these DCs strongly promoted production of the T cell polarizing factor interleukin-12 (IL-12). In contrast, stationary-phase (SP) S. aureus bacteria were endocytosed less efficiently by DCs, and these DCs produced small amounts of IL-12. The high level of IL-12 production induced by EP S. aureus led to the development of a T helper 1 (Th1) cell response, which was inhibited after neutralization of IL-12. Furthermore, preincubation with the staphylococcal cell wall component peptidoglycan (PGN), characteristically shed during the exponential growth phase, modulated the DC response to EP S. aureus PGN preincubation appeared to inhibit IL-12p35 expression, leading to downregulation of IL-12 and an increase of IL-23 production by DCs, enhancing Th17 cell development. Taken together, our data indicate that exponential-phase S. aureus bacteria induce a stronger IL-12-dependent Th1 cell response than stationary-phase S. aureus and that this Th1 cell response shifted toward a Th17 cell response in the presence of PGN.
Collapse
|
38
|
Ueno H. The IL-12-STAT4 axis in the pathogenesis of human systemic lupus erythematosus. Eur J Immunol 2019; 50:10-16. [PMID: 31762023 DOI: 10.1002/eji.201948134] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 10/05/2019] [Indexed: 12/28/2022]
Abstract
Generation of autoantibodies is a hallmark of systemic lupus erythematosus (SLE). As demonstrated in a number of lupus mouse models, recent evidence suggests that both GC and extrafollicular pathways contribute to the generation of autoantibodies also in human SLE, and that CD11c+ IgD- CD27- (double negative:DN) B cells play a central role in the latter pathway. In this mini-review, the author will first briefly summarize the features of CD11c+ DN B cells in human SLE, and discuss how the IL-12-STAT4 axis might contribute to the generation of autoantibodies in SLE. In addition, various types of CD4+ helper T cell subsets promoting the generation of autoantibodies in SLE will be described, and finally it will be discussed how these recent discoveries contribute to understanding of SLE pathogenesis and treatment of SLE patients.
Collapse
Affiliation(s)
- Hideki Ueno
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Immunology, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto, Japan.,Institute for the Advanced Study of Human Biology, Kyoto University, Sakyo-ku, Kyoto, Japan
| |
Collapse
|
39
|
Dillman RO, Nistor GI, Poole AJ. Genomic, proteomic, and immunologic associations with a durable complete remission of measurable metastatic melanoma induced by a patient-specific dendritic cell vaccine. Hum Vaccin Immunother 2019; 16:742-755. [PMID: 31625825 PMCID: PMC7227648 DOI: 10.1080/21645515.2019.1680239] [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] [Indexed: 11/07/2022] Open
Abstract
This report describes efforts to understand the immune mechanism of action that led to a complete response in a patient with progressive, refractory, metastatic melanoma after treatment with a therapeutic vaccine consisting of autologous dendritic cells (DC) loaded with autologous tumor antigens (ATA) derived from cells that were self-renewing in cell culture. Her histocompatibility type proved to be HLA B27 with extensive mutations in the HLA-A locus. Exomic analysis of proliferating tumor cells revealed more than 2800 non-synonymous mutations compared to her leukocytes. Histology of resected tumor lesions showed no evidence of an existing or suppressed immune response. In in vitro mixed cell cultures, DC loaded with ATA induced increased IL-22 expression, and a four-fold increase in CD8 + T lymphocytes. Cryopreserved blood samples obtained at week-0, 1 week before the first of three-weekly vaccine injections, and at week-4, 1 week after the third dose, were analyzed by protein array and compared for 110 different serum markers. At baseline, she had marked elevations of amyloid A, IL-12p40, IL21, IL-22, IL-10, IL-16, GROa, TNF-alpha, IL-3, and IL-2, and a lesser elevation of IL-15. One week after 3 weekly vaccinations she had a further 80% increase in amyloid A, a further 66% increase in IL-22, a 92% decrease in IL12p40, a 45% decrease in TGF-β and 26% decrease in IL-10. The data suggested that by 3 weeks after the first DCV injection, vaccine-induced changes in this particular patient were most consistent with enhanced innate and Th1 immune responses rather than Th2 or Th17.
Collapse
Affiliation(s)
- Robert O Dillman
- AIVITA Biomedical, Inc, Irvine, CA, USA.,Hoag Cancer Institute, Newport Beach, CA, USA
| | | | | |
Collapse
|
40
|
Wei Y, Chen S, Sun D, Li X, Wei R, Li X, Nian H. miR-223-3p promotes autoreactive T h17 cell responses in experimental autoimmune uveitis (EAU) by inhibiting transcription factor FOXO3 expression. FASEB J 2019; 33:13951-13965. [PMID: 31645142 DOI: 10.1096/fj.201901446r] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Pathogenic T helper (Th)17 cells are key mediators of autoimmune diseases such as uveitis and its animal model, experimental autoimmune uveitis (EAU). However, the contribution of microRNAs (miRs) to the intrinsic control of pathogenic Th17 cells in EAU remains largely unknown. Here, we have reported that miR-223-3p was significantly up-regulated in interphotoreceptor retinoid-binding protein-specific Th17 cells, and its expression was enhanced by IL-23-signal transducer and activator of transcription 3 signaling. Knockdown of miR-223-3p decreased the pathogenicity of Th17 cells in a T-cell transfer model of EAU. Mechanistic studies showed that miR-223-3p directly repressed the expression of forkhead box O3 (FOXO3), and FOXO3 negatively regulated pathogenic Th17 cell responses partially via suppression of IL-23 receptor expression. Thus, our results reveal an important role for miR-223-3p in autoreactive Th17 cell responses and suggest a potential therapeutic avenue for uveitis.-Wei, Y., Chen, S., Sun, D., Li, X., Wei, R., Li, X., Nian, H. miR-223-3p promotes autoreactive Th17 cell responses in experimental autoimmune uveitis (EAU) by inhibiting transcription factor FOXO3 expression.
Collapse
Affiliation(s)
- Yankai Wei
- Tianjin Key Laboratory of Retinal Functions and Diseases, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, China
| | - Sisi Chen
- Tianjin Key Laboratory of Retinal Functions and Diseases, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, China
| | - Deming Sun
- Department of Ophthalmology, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, California, USA.,Doheny Eye Institute, Los Angeles, California, USA
| | - Xue Li
- Tianjin Key Laboratory of Retinal Functions and Diseases, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, China
| | - Ruihua Wei
- Tianjin Key Laboratory of Retinal Functions and Diseases, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, China
| | - Xiaorong Li
- Tianjin Key Laboratory of Retinal Functions and Diseases, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, China
| | - Hong Nian
- Tianjin Key Laboratory of Retinal Functions and Diseases, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, China
| |
Collapse
|
41
|
Hua Y, Bergers G. Tumors vs. Chronic Wounds: An Immune Cell's Perspective. Front Immunol 2019; 10:2178. [PMID: 31572387 PMCID: PMC6751250 DOI: 10.3389/fimmu.2019.02178] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 08/29/2019] [Indexed: 12/13/2022] Open
Abstract
The wound repair program is tightly regulated and coordinated among different cell constituents including epithelial cells, fibroblasts, immune cells and endothelial cells following consecutive steps to ensure timely, and proper wound closure. Specifically, innate and adaptive immune cells are pivotal participants that also closely interact with the vasculature. Tumors are portrayed as wounds that do not heal because they undergo continuous stromal remodeling and vascular growth with immunosuppressive features to ensure tumor propagation; a stage that is reminiscent of the proliferative resolution phase in wound repair. There is increasing evidence from mouse model systems and clinical trials that targeting both the immune and vascular compartments is an attractive therapeutic approach to reawaken the inflammatory status in the "tumor wound" with the final goal to abrogate tumor cells and invigorate tissue homeostasis. In this review, we compare the implication of immune cells and the vasculature in chronic wounds and tumor wounds to underscore the conceptual idea of transitioning tumors into an inflammatory wound-like state with antiangiogenic immunotherapies to improve beneficial effects in cancer patients.
Collapse
Affiliation(s)
- Yichao Hua
- Laboratory of Tumor Microenvironment and Therapeutic Resistance, Department of Oncology, VIB-Center for Cancer Biology, KU Leuven, Leuven, Belgium
| | - Gabriele Bergers
- Laboratory of Tumor Microenvironment and Therapeutic Resistance, Department of Oncology, VIB-Center for Cancer Biology, KU Leuven, Leuven, Belgium.,Department of Neurological Surgery, UCSF Comprehensive Cancer Center, UCSF, San Francisco, CA, United States
| |
Collapse
|
42
|
Amir Ali A, Vender R, Vender R. The Role of IL-17 in Papulopustular Rosacea and Future Directions. J Cutan Med Surg 2019; 23:635-641. [DOI: 10.1177/1203475419867611] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Rosacea is a chronic, progressive, inflammatory condition phenotypically subtyped into diagnostic features, major features, and minor/secondary features. There is currently no cure for rosacea, and it carries a significant negative psychosocial burden for afflicted patients. While there are a number of treatment modalities at the disposal of the clinician, clinical experience has suggested a need for updated treatments. The pathogenesis of rosacea is multifactorial; however, this paper will focus on the pivotal role of interleukin 17 (IL-17) in the development and progression of the disease. Furthermore, this paper will explore the mechanism of action of standard rosacea treatments and their effect on different stages of the IL-17 pathway. The standard treatments for rosacea are usually effective in controlling the symptoms of the disease in its mild-to-moderate form; however, their efficacy is diminished in the setting of severe and treatment-resistant rosacea. We hypothesize that IL-17 inhibitors, currently used successfully in psoriasis and psoriatic arthritis, could perhaps be used to treat severe and treatment-resistant papulopustular rosacea in the future; however, clinical trials and case reports will be needed to dictate expanded indications of IL-17 inhibitors. Furthermore, the high cost of IL-17 inhibitors presently prevents their use in disease states other than psoriasis or psoriatic arthritis.
Collapse
Affiliation(s)
- Asma Amir Ali
- Schulich School of Medicine & Dentistry, Western University, London, ON, Canada
| | - Reid Vender
- School of Medicine, Queen’s University, Kingston, ON, Canada
| | | |
Collapse
|
43
|
Yuan N, Yu G, Liu D, Wang X, Zhao L. An emerging role of interleukin-23 in rheumatoid arthritis. Immunopharmacol Immunotoxicol 2019; 41:185-191. [PMID: 31072166 DOI: 10.1080/08923973.2019.1610429] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Rheumatoid arthritis (RA) is an autoimmune, chronic inflammatory disease and is characterized by destruction of the articular cartilage. A number of pro-inflammatory cytokines work sequentially and in concert with one another to induce the development of RA. IL-23, a member of IL-12 family, is composed of p19 and p40 subunits and it interacts with IL-23 receptor complex to trigger plethora of biochemical actions. A number of preclinical studies have shown the role of IL-23 in the development of RA in rodents. IL-23 receptor signaling is primarily linked to the activation of JAK-STAT, tyrosine kinase 2, NF-kB, and retinoic acid receptor-related orphan receptors. IL-23 produces its osteoclastogenic effects, mainly through IL-17 and Th17 cells suggesting the importance of IL-23/IL-17/Th17 in the joint inflammation and destruction in RA. Monoclonal antibodies targeted against IL-23, including tildrakizumab and guselkumab have been developed and evaluated in clinical trials. However, there are very limited clinical studies regarding the use of IL-23 modulators in RA patients. The present review discusses the different aspects of IL-23 including its structural features, signal transduction pathway, preclinical, and clinical role in RA.
Collapse
Affiliation(s)
- Na Yuan
- a Department of Rheumatology , The Affiliated Hospital to Changchun University of Chinese Medicine , Changchun , China
| | - Guimei Yu
- a Department of Rheumatology , The Affiliated Hospital to Changchun University of Chinese Medicine , Changchun , China
| | - Di Liu
- a Department of Rheumatology , The Affiliated Hospital to Changchun University of Chinese Medicine , Changchun , China
| | - Xiancheng Wang
- b Department of Cardiology , The Affiliated Hospital to Changchun University of Chinese Medicine , Changchun , China
| | - Ling Zhao
- c Department of Rheumatology , The First Hospital of Jilin University , Changchun , China
| |
Collapse
|
44
|
Oxaliplatin Treatment Alters Systemic Immune Responses. BIOMED RESEARCH INTERNATIONAL 2019; 2019:4650695. [PMID: 30906773 PMCID: PMC6398049 DOI: 10.1155/2019/4650695] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 11/22/2018] [Indexed: 12/14/2022]
Abstract
Purpose Oxaliplatin is a platinum-based chemotherapeutic agent demonstrating significant antitumor efficacy. Unlike conventional anticancer agents which are immunosuppressive, oxaliplatin has the capacity to stimulate immunological effects in response to the presentation of damage associated molecular patterns (DAMPs) elicited upon cell death. However, the effects of oxaliplatin treatment on systemic immune responses remain largely unknown. Aims of this study were to investigate the effects of oxaliplatin treatment on the proportions of (1) splenic T cells, B cells, macrophages, pro-/anti-inflammatory cytokines, gene expression of splenic cytokines, chemokines, and mediators; (2) double-positive and single-positive CD4+ and CD8+ T thymocytes; (3) bone-marrow hematopoietic stem and progenitor cells. Methods Male BALB/c mice received intraperitoneal injections of oxaliplatin (3mg/kg/d) or sterile water tri-weekly for 2 weeks. Leukocyte populations within the spleen, thymus, and bone-marrow were assessed using flow cytometry. RT-PCR was performed to characterise changes in splenic inflammation-associated genes. Results Oxaliplatin treatment reduced spleen size and cellularity (CD45+ cells), increased the proportion of CD4+, CD8+, and Treg cells, and elevated TNF-α expression. Oxaliplatin was selectively cytotoxic to B cells but had no effect on splenic macrophages. Oxaliplatin treatment altered the gene expression of several cytokines, chemokines, and cell mediators. Oxaliplatin did not deplete double-positive thymocytes but increased the single-positive CD8+ subset. There was also an increase in activated (CD69+) CD8+ T cells. Bone-marrow hematopoietic progenitor pool was demonstrably normal following oxaliplatin treatment when compared to the vehicle-treated cohort. Conclusion Oxaliplatin does not cause systemic immunosuppression and, instead, has the capacity to induce beneficial antitumor immune responses.
Collapse
|
45
|
Aggeletopoulou I, Assimakopoulos SF, Konstantakis C, Triantos C. Interleukin 12/interleukin 23 pathway: Biological basis and therapeutic effect in patients with Crohn's disease. World J Gastroenterol 2018; 24:4093-4103. [PMID: 30271076 PMCID: PMC6158482 DOI: 10.3748/wjg.v24.i36.4093] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Revised: 08/02/2018] [Accepted: 08/24/2018] [Indexed: 02/06/2023] Open
Abstract
Considering that both innate and adaptive immune responses are involved in the pathogenesis of Crohn’s disease (CD), novel therapeutic options have significantly been developed. Biological agents represent an important addition to the conventional treatments for immuno-inflammatory conditions, acting as antagonists of adhesion molecules or various inflammatory cytokines. The interleukin 12 (IL-12)/IL-23 common pathway has been found to play a determinant role in the induction of inflammation in adaptive immune responses. In particular, IL-23 promotes the differentiation of naïve T helper cells into Th17 phenotype with the concomitant secretion of several inflammatory cytokines such as IL-17 and IL-22, whereas IL-12 induces the Th1 polarization and production of critical cytokines such as interferon-γ and tumor necrosis factor. Nowadays, there is increased interest regarding the role of IL-23 as a therapeutic target of CD through the blockage of IL-23 mediated pathways. In this editorial, we focus on the role of IL-12/IL-23 pathway in the regulation of mucosal immunity and in the induction and maintenance of chronic inflammation. In parallel, we critically discuss the available data regarding the therapeutic effect of the IL-12/IL-23 inhibitors and especially of ustekinumab, a human monoclonal antibody which has been recently approved by the United States Food and Drug Administration for the management of moderate-to-severe CD and its potential to be used as first-line therapy in everyday clinical practice.
Collapse
Affiliation(s)
- Ioanna Aggeletopoulou
- Division of Gastroenterology, Department of Internal Medicine, Medical School, University of Patras, Patras 26504, Greece
| | | | - Christos Konstantakis
- Division of Gastroenterology, Department of Internal Medicine, Medical School, University of Patras, Patras 26504, Greece
| | - Christos Triantos
- Division of Gastroenterology, Department of Internal Medicine, Medical School, University of Patras, Patras 26504, Greece
| |
Collapse
|
46
|
Horiuchi S, Ueno H. Potential Pathways Associated With Exaggerated T Follicular Helper Response in Human Autoimmune Diseases. Front Immunol 2018; 9:1630. [PMID: 30061896 PMCID: PMC6054970 DOI: 10.3389/fimmu.2018.01630] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 07/02/2018] [Indexed: 12/26/2022] Open
Abstract
Convincing lines of evidence in both mice and humans show that exaggerated T follicular helper (Tfh) responses is pathogenic in autoimmune diseases. However, the cause of exaggerated Tfh response in humans is still much less clear than in mouse models where genetic factors can be manipulated for in vivo testing. Nonetheless, recent advances in our understanding on the mechanisms of human Tfh differentiation and identification of multiple risk loci in genome-wide association studies have revealed several pathways potentially associated with exaggerated Tfh response in human autoimmune diseases. In this review, we will first briefly summarize the differentiation mechanisms of Tfh cells in humans. We describe the features of “Tfh-like” cells recently identified in inflamed tissues of human autoimmune diseases. Then we will discuss how risk loci identified in GWAS are potentially involved in exaggerated Tfh response in human autoimmune diseases.
Collapse
Affiliation(s)
- Shu Horiuchi
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Hideki Ueno
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
47
|
Naturally derived Heme-Oxygenase 1 inducers attenuate inflammatory responses in human dendritic cells and T cells: relevance for psoriasis treatment. Sci Rep 2018; 8:10287. [PMID: 29980703 PMCID: PMC6035209 DOI: 10.1038/s41598-018-28488-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 06/20/2018] [Indexed: 01/04/2023] Open
Abstract
Psoriasis is a chronic autoimmune disease mediated by dysregulated immune responses in dendritic cells (DC) and T cells. The stress-response enzyme heme oxygenase-1 (HO-1) has been described as protective in animal models of psoriasis, however, implementation of HO-1-based therapies is hindered by the lack of clinically-suitable HO-1 inducers. The plant-derived polyphenols, carnosol and curcumin, have been identified as candidate HO-1 inducers however there has been little investigation into their effects on human immune cells. We demonstrate that treatment of human DC with these polyphenols limits DC maturation, reduces pro-inflammatory cytokine production, and prevents induction of allospecific T cell responses, in a manner partially dependent on carbon monoxide (CO). We also characterised their effects in ex-vivo psoriasis PBMC and report that curcumin, but not carnosol, strongly reduces T cell proliferation and cytokine poly-functionality, with reduced expression of psoriatic cytokines IFNγ, IL-17, GM-CSF and IL-22. This study therefore supports reports highlighting the therapeutic potential of curcumin in psoriasis by providing insight into its immunological effects on healthy human DC and psoriasis PBMC. We also demonstrate, for the first time, the anti-inflammatory effects of carnosol in human immune cells.
Collapse
|
48
|
Pavlov O, Selutin A, Pavlova O, Selkov S. Macrophages are a source of IL-17 in the human placenta. Am J Reprod Immunol 2018; 80:e13016. [DOI: 10.1111/aji.13016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 06/12/2018] [Indexed: 12/18/2022] Open
Affiliation(s)
- Oleg Pavlov
- Department of Immunology and Cell Interaction; D.O. Ott Research Institute for Obstetrics, Gynecology and Reproduction; St. Petersburg Russia
| | - Aleksandr Selutin
- Department of Immunology and Cell Interaction; D.O. Ott Research Institute for Obstetrics, Gynecology and Reproduction; St. Petersburg Russia
| | - Oksana Pavlova
- Department of Histology, Cytology and Embryology; Pavlov First Saint Petersburg State Medical University; St. Petersburg Russia
| | - Sergei Selkov
- Department of Immunology and Cell Interaction; D.O. Ott Research Institute for Obstetrics, Gynecology and Reproduction; St. Petersburg Russia
| |
Collapse
|
49
|
Zhang J, Zhang Z, Zhang W, Li X, Wu T, Li T, Cai M, Yu Z, Xiang J, Cai D. Jia-Jian-Di-Huang-Yin-Zi decoction exerts neuroprotective effects on dopaminergic neurons and their microenvironment. Sci Rep 2018; 8:9886. [PMID: 29959371 PMCID: PMC6026152 DOI: 10.1038/s41598-018-27852-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 06/07/2018] [Indexed: 11/09/2022] Open
Abstract
As a classical prescription of Traditional Chinese medicine, the Jia-Jian-Di-Huang-Yin-Zi (JJDHYZ) decoction has long been used to treat movement disorders. The present study evaluated the effects of JJDHYZ on dopaminergic (DA) neurons and their survival-enhancing microenvironment as well as the possible mechanisms involved using a mouse model of Parkinson's disease. In MPTP-lesioned mice, a high dosage of JJDHYZ (34 g/kg/day) attenuated the loss of DA neurons, reversed the dopamine depletion, and improved the expression of glial-derived neurotrophic factor (GDNF) compared to the untreated model group. JJDHYZ also protected the ultrastructure of the blood-brain barrier (BBB) and tight junction proteins by inhibiting the activation of microglia and astrocytes besides the increase in three types of matrix metalloproteinases in the substantia nigra. In conclusion, the JJDHYZ-high dosage (JJDHYZ-H) group exhibited the neuroprotection of DA neurons, and the underlying mechanism may be related to the survival-enhancing microenvironment of the DA neurons.
Collapse
Affiliation(s)
- Jingsi Zhang
- Department of Integrative Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Zhennian Zhang
- Department of Neurology, Nanjing Hospital of Traditional Chinese Medicine, Nanjing, 210000, China
| | - Wen Zhang
- Department of Integrative Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Xiangting Li
- Department of Integrative Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Ting Wu
- Department of Integrative Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Tingting Li
- Department of Neurology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Min Cai
- Department of Integrative Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Zhonghai Yu
- Department of Integrative Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Jun Xiang
- Department of Integrative Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| | - Dingfang Cai
- Department of Integrative Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
50
|
Jiang H, Zhang S, Song T, Guan X, Zhang R, Chen X. Trichostatin a Protects Dendritic Cells Against Oxygen-Glucose Deprivation via the SRSF3/PKM2/Glycolytic Pathway. Front Pharmacol 2018; 9:612. [PMID: 29942258 PMCID: PMC6004525 DOI: 10.3389/fphar.2018.00612] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 05/22/2018] [Indexed: 12/12/2022] Open
Abstract
Dendritic cells (DCs) are important to the immune system and are frequently recruited to hypoxic regions, especially during acute myocardial infarction (AMI). Emerging data indicate that histone deacetylase (HDAC) inhibitors possess immunomodulatory functions. We previously showed in a rat model of AMI that the HDAC inhibitor TSA improved tissue repair, and this was accompanied by increased DC infiltration in the infarct region, suggesting an important role of TSA in modulating DC functions. To study the potential modulatory effect of TSA on DCs, we exploited an in vitro model of hypoxia and glucose deprivation. Culturing of DCs in the presence of 200 nM TSA improved DC survival under hypoxia and glucose deprivation. However, on a phenotypic level, TSA induced the expression of the DC co-stimulatory molecules CD80 and CD86, decreased FITC-dextran uptake, and facilitated DC migration. Moreover, TSA altered cytokine secretion by reducing the pro-inflammatory cytokines IL-1β, IL-10, IL-12, and TGF-β. Furthermore, TSA treatment enhanced HIF-1α-dependent glycolytic gene expression and increased pyruvate kinase M2 by upregulating SRSF3. These results suggest that by TSA alters important DC functions under hypoxia and glucose deprivation, and that TSA is critical for DC function by modulating SRSF3-PKM2-dependent glycolytic pathways.
Collapse
Affiliation(s)
- Hongyun Jiang
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Siwei Zhang
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Tongtong Song
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Xin Guan
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Ruojin Zhang
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Xia Chen
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun, China
| |
Collapse
|