1
|
Zhukova JV, Lopatnikova JA, Alshevskaya AA, Sennikov SV. Molecular mechanisms of regulation of IL-1 and its receptors. Cytokine Growth Factor Rev 2024:S1359-6101(24)00079-0. [PMID: 39414547 DOI: 10.1016/j.cytogfr.2024.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 09/26/2024] [Accepted: 09/27/2024] [Indexed: 10/18/2024]
Abstract
Interleukin 1 (IL-1) is a pro-inflammatory cytokine that plays a key role in the development and regulation of nonspecific defense and specific immunity. However, its regulatory influence extends beyond inflammation and impacts a range of immune and non-immune processes. The involvement of IL-1 in numerous biological processes, including modulation of inflammation, necessitates strict regulation at multiple levels. This review focuses on these regulatory processes and discusses their underlying mechanisms. IL-1 activity is controlled at various levels, including receptor binding, gene transcription, expression as inactive proforms, and regulated post-translational processing and secretion. Regulation at the level of the receptor expression - alternative splicing, tissue-specific isoforms, and gene polymorphism - is also crucial to IL-1 functional activity. Understanding these regulatory features of IL-1 will not only continue to shape future research directions but will also highlight promising therapeutic strategies to modulate the biological effects of IL-1.
Collapse
Affiliation(s)
- J V Zhukova
- Federal State Budgetary Scientific Institution "Research Institute of Fundamental and Clinical Immunology" (RIFCI), Novosibirsk 630099, Russia; Federal State Autonomous Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russia
| | - J A Lopatnikova
- Federal State Budgetary Scientific Institution "Research Institute of Fundamental and Clinical Immunology" (RIFCI), Novosibirsk 630099, Russia; Federal State Autonomous Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russia
| | - A A Alshevskaya
- Federal State Autonomous Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russia
| | - S V Sennikov
- Federal State Budgetary Scientific Institution "Research Institute of Fundamental and Clinical Immunology" (RIFCI), Novosibirsk 630099, Russia; Federal State Autonomous Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russia.
| |
Collapse
|
2
|
Machado MSG, Rodrigues VF, Barbosa SC, Elias-Oliveira J, Pereira ÍS, Pereira JA, Pacheco TCF, Carlos D. IL-1 Receptor Contributes to the Maintenance of the Intestinal Barrier via IL-22 during Obesity and Metabolic Syndrome in Experimental Model. Microorganisms 2024; 12:1717. [PMID: 39203559 PMCID: PMC11357463 DOI: 10.3390/microorganisms12081717] [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: 07/10/2024] [Revised: 08/09/2024] [Accepted: 08/14/2024] [Indexed: 09/03/2024] Open
Abstract
Intestinal permeability and bacterial translocation are increased in obesity and metabolic syndrome (MS). ILC3 cells contribute to the integrity of intestinal epithelium by producing IL-22 via IL-1β and IL-23. This study investigates the role of IL-1R1 in inducing ILC3 cells and conferring protection during obesity and MS. For this purpose, C57BL/6 wild-type (WT) and IL-1R1-deficient mice were fed a standard diet (SD) or high-fat diet (HFD) for 16 weeks. Weight and blood glucose levels were monitored, and adipose tissue and blood samples were collected to evaluate obesity and metabolic parameters. The small intestine was collected to assess immunological and junction protein parameters through flow cytometry and RT-PCR, respectively. The intestinal permeability was analyzed using the FITC-dextran assay. The composition of the gut microbiota was also analyzed by qPCR. We found that IL-1R1 deficiency exacerbates MS in HFD-fed mice, increasing body fat and promoting glucose intolerance. A worsening of MS in IL-1R1-deficient mice was associated with a reduction in the ILC3 population in the small intestine. In addition, we found decreased IL-22 expression, increased intestinal permeability and bacterial translocation to the visceral adipose tissue of these mice compared to WT mice. Thus, the IL-1R1 receptor plays a critical role in controlling intestinal homeostasis and obesity-induced MS, possibly through the differentiation or activation of IL-22-secreting ILC3s.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Daniela Carlos
- Laboratory of Immunoregulation of Metabolic Disease, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, SP, Brazil; (M.S.G.M.); (V.F.R.); (S.C.B.); (J.E.-O.); (Í.S.P.); (J.A.P.); (T.C.F.P.)
| |
Collapse
|
3
|
Mao YR, Zhu LY, Du RF, Liu XY, Liu Z, Li L. Melittin Alleviates Oxidative Stress Injury in Schwann Cells by Targeting Interleukin-1 Receptor Type 1 to Downregulate Nuclear Factor Kappa B-Mediated Inflammatory Response In Vitro. Cureus 2024; 16:e65721. [PMID: 39211643 PMCID: PMC11358714 DOI: 10.7759/cureus.65721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/29/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND AND OBJECTIVES In ancient China, bee venom was widely used to treat various diseases. Although using bee venom is not currently a mainstream medical method, some have applied it to treat certain conditions, including idiopathic facial paralysis (IFP). Recently, melittin (Mel), the main active component of bee venom, has been shown strong anti-inflammatory and analgesic effects. However, how bee venom improves neurological dysfunction in facial paralysis remains unknown. This study aimed to investigate the anti-neurotraumatic effect of Mel on Schwann cells (SCs), the main cells of the neuron sheath, injured by oxidative stress. METHODS A model of hypoxic SCs was established, and CCK-8 assay, siRNA transfection, enzyme-linked immunosorbent assay, quantitative reverse transcription-polymerase chain reaction, western blot, immunofluorescence, and cell ultrastructure analyses were conducted to investigate the mitigation of hypoxia-induced damage to SCs in vitro, revealing the effects of Mel on oxidative stress injury in SCs. RESULTS The overexpression of HIF-1α in CoCl2-induced SCs (p < 0.05) indicated the establishment of an SCs hypoxia model. The proliferation and regeneration process of the hypoxic SCs enhanced in the Mel-treated group compared to the CoCl2 group has been proven through the CCK-8 experiment (p < 0.0001) and S-100 mRNA expression detection (p < 0.0001). The increased level of reactive oxygen species (ROS) (p < 0.001) and decreased superoxide dismutase (SOD) levels (p < 0.05) in the CoCl2-induced SCs indicated that Mel can alleviate the oxidative stress damage to SCs induced by CoCl2. Mel alleviated oxidative stress and inflammation in hypoxic SCs by reducing pro-inflammatory cytokines IL-1β (p < 0.0001) and TNF-α (p < 0.0001). In addition, Mel augmented cellular vitality and regulated indicators related to oxygen metabolism, cell repair, neurometabolism, and vascular endothelial formation after hypoxia, such as C-JUN (p < 0.05), glial cell line-derived neurotrophic factor (GDNF; p < 0.001), vascular endothelial growth factor (VEGF; p < 0.05), hypoxia-inducible factor 1-alpha (HIF-1α; p < 0.05), interleukin-1 receptor type 1 (IL-1R1; p < 0.05), enolase1 (ENO1; p < 0.05), aldose reductase (AR; p < 0.01), SOD (p < 0.05), nerve growth factor (NGF; p < 0.05), and inducible nitric oxide synthase (iNOS; p < 0.05). In terms of its mechanism, Mel inhibited the expression of proteins associated with the NF-κB pathway such as IKK (p < 0.01), p65 (p < 0.05), p60 (p < 0.001), IRAK1 (p < 0.05), and increased IKB-α (p < 0.0001). Moreover, knocking out of IL-1R1 in the si-IL-1R1 group enhanced the therapeutic effect of Mel compared to the Mel-treated group (all of which p < 0.05). CONCLUSION This research provided evidence of the substantial involvement of IL-1R1 in oxidative stress damage caused by hypoxia in SCs and proved that Mel alleviated oxidative stress injury in SCs by targeting IL-1R1 to downregulate the NF-κB-mediated inflammatory response. Mel could potentially serve as an innovative therapeutic approach for the treatment of IFP.
Collapse
Affiliation(s)
- Ye-Ran Mao
- Rehabilitation, Baoshan Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, CHN
| | - Ling-Yi Zhu
- Integrative/Complementary Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, CHN
| | - Ruo-Fei Du
- Pharmacology, Institute of Innovative Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, CHN
| | - Xiao-Yu Liu
- Rehabilitation, Baoshan Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, CHN
| | - Zhidan Liu
- Rehabilitation, Baoshan Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, CHN
| | - Li Li
- Respiratory Intensive Care Medicine, Baoshan Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, CHN
| |
Collapse
|
4
|
Arellano G, Loda E, Chen Y, Neef T, Cogswell AC, Primer G, Joy G, Kaschke K, Wills S, Podojil JR, Popko B, Balabanov R, Miller SD. Interferon-γ controls aquaporin 4-specific Th17 and B cells in neuromyelitis optica spectrum disorder. Brain 2024; 147:1344-1361. [PMID: 37931066 PMCID: PMC10994540 DOI: 10.1093/brain/awad373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 09/27/2023] [Accepted: 10/21/2023] [Indexed: 11/08/2023] Open
Abstract
Neuromyelitis optica spectrum disorder (NMOSD) is a CNS autoimmune inflammatory disease mediated by T helper 17 (Th17) and antibody responses to the water channel protein, aquaporin 4 (AQP4), and associated with astrocytopathy, demyelination and axonal loss. Knowledge about disease pathogenesis is limited and the search for new therapies impeded by the absence of a reliable animal model. In our work, we determined that NMOSD is characterized by decreased IFN-γ receptor signalling and that IFN-γ depletion in AQP4201-220-immunized C57BL/6 mice results in severe clinical disease resembling human NMOSD. Pathologically, the disease causes autoimmune astrocytic and CNS injury secondary to cellular and humoral inflammation. Immunologically, the absence of IFN-γ allows for increased expression of IL-6 in B cells and activation of Th17 cells, and generation of a robust autoimmune inflammatory response. Consistent with NMOSD, the experimental disease is exacerbated by administration of IFN-β, whereas repletion of IFN-γ, as well as therapeutic targeting of IL-17A, IL-6R and B cells, ameliorates it. We also demonstrate that immune tolerization with AQP4201-220-coupled poly(lactic-co-glycolic acid) nanoparticles could both prevent and effectively treat the disease. Our findings enhance the understanding of NMOSD pathogenesis and provide a platform for the development of immune tolerance-based therapies, avoiding the limitations of the current immunosuppressive therapies.
Collapse
Affiliation(s)
- Gabriel Arellano
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Eileah Loda
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Yanan Chen
- Department of Biology, Loyola University Chicago, Chicago, IL 60660, USA
| | - Tobias Neef
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Andrew C Cogswell
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Grant Primer
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Godwin Joy
- Department of Biology, Loyola University Chicago, Chicago, IL 60660, USA
| | - Kevin Kaschke
- Department of Biology, Loyola University Chicago, Chicago, IL 60660, USA
| | - Samantha Wills
- Department of Biology, Loyola University Chicago, Chicago, IL 60660, USA
| | - Joseph R Podojil
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- COUR Pharmaceutical Development Company, Inc., Northbrook, IL 60077, USA
| | - Brian Popko
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Roumen Balabanov
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Stephen D Miller
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| |
Collapse
|
5
|
Kim DH, Kim HY, Lee WW. Induction of Unique STAT Heterodimers by IL-21 Provokes IL-1RI Expression on CD8 + T Cells, Resulting in Enhanced IL-1β Dependent Effector Function. Immune Netw 2021; 21:e33. [PMID: 34796037 PMCID: PMC8568912 DOI: 10.4110/in.2021.21.e33] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 08/11/2021] [Accepted: 08/23/2021] [Indexed: 12/24/2022] Open
Abstract
IL-1β plays critical roles in the priming and effector phases of immune responses such as the differentiation, commitment, and memory formation of T cells. In this context, several reports have suggested that the IL-1β signal is crucial for CTL-mediated immune responses to viral infections and tumors. However, little is known regarding whether IL-1β acts directly on CD8+ T cells and what the molecular mechanisms underlying expression of IL-1 receptors (IL-1Rs) on CD8+ T cells and features of IL-1R+CD8+ T cells are. Here, we provide evidence that the expression of IL-1R type I (IL-1RI), the functional receptor of IL-1β, is preferentially induced by IL-21 on TCR-stimulated CD8+ T cells. Further, IL-1β enhances the effector function of CD8+ T cells expressing IL-21-induced IL-1RI by increasing cytokine production and release of cytotoxic granules containing granzyme B. The IL-21-IL-1RI-IL-1β axis is involved in an augmented effector function through regulation of transcription factors BATF, Blimp-1, and IRF4. Moreover, this axis confers a unique effector function to CD8+ T cells compared to conventional type 1 cytotoxic T cells differentiated with IL-12. Chemical inhibitor and immunoprecipitation assay demonstrated that IL-21 induces a unique pattern of STAT activation with the formation of both STAT1:STAT3 and STAT3:STAT5 heterodimers, which are critical for the induction of IL-1RI on TCR-stimulated CD8+ T cells. Taken together, we propose that induction of a novel subset of IL-1RI-expressing CD8+ T cells by IL-21 may be beneficial to the protective immune response against viral infections and is therefore important to consider for vaccine design.
Collapse
Affiliation(s)
- Dong Hyun Kim
- Laboratory of Autoimmunity and Inflammation (LAI), Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea.,Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Hee Young Kim
- Laboratory of Autoimmunity and Inflammation (LAI), Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea.,Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul 03080, Korea.,Institute of Infectious Diseases, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Won-Woo Lee
- Laboratory of Autoimmunity and Inflammation (LAI), Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea.,Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul 03080, Korea.,Institute of Infectious Diseases, Seoul National University College of Medicine, Seoul 03080, Korea.,Cancer Research Institute and Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul 03080, Korea.,Seoul National University Hospital Biomedical Research Institute, Seoul 03080, Korea
| |
Collapse
|
6
|
Kuo WC, Lee CC, Chang YW, Pang W, Chen HS, Hou SC, Lo SY, Yang AS, Wang AHJ. Structure-based Development of Human Interleukin-1β-Specific Antibody That Simultaneously Inhibits Binding to Both IL-1RI and IL-1RAcP. J Mol Biol 2020; 433:166766. [PMID: 33359099 DOI: 10.1016/j.jmb.2020.166766] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 12/07/2020] [Accepted: 12/14/2020] [Indexed: 02/05/2023]
Abstract
Interleukin-1β (IL-1β) is a potent pleiotropic cytokine playing a central role in protecting cells from microbial pathogen infection or endogenous stress. After it binds to IL-1RI and recruits IL-1 receptor accessory protein (IL-1RAcP), signaling culminates in activation of NF-κB. Many pathophysiological diseases have been attributed to the derailment of IL-1β regulation. Several blocking reagents have been developed based on two mechanisms: blocking the binding of IL-1β to IL-1RI or inhibiting the recruitment of IL-1RAcP to the IL-1β initial complex. In order to simultaneously fulfill these two actions, a human anti-IL-1β neutralizing antibody IgG26 was screened from human genetic phage-display library and furthered structure-optimized to final version, IgG26AW. IgG26AW has a sub-nanomolar binding affinity for human IL-1β. We validated IgG26AW-neutralizing antibodies specific for IL-1β in vivo to prevent human IL-1β-driving IL-6 elevation in C56BL/6 mice. Mice underwent treatments with IgG26AW in A549 and MDA-MB-231 xenograft mouse cancer models have also been observed with tumor shrank and inhibition of tumor metastasis. The region where IgG26 binds to IL-1β also overlaps with the position where IL-1RI and IL-1RAcP bind, as revealed by the 26-Fab/IL-1β complex structure. Meanwhile, SPR experiments showed that IL-1β bound by IgG26AW prevented the further binding of IL-1RI and IL-1RAcP, which confirmed our inference from the result of protein structure. Therefore, the inhibitory mechanism of IgG26AW is to block the assembly of the IL-1β/IL-1RI/IL-1RAcP ternary complex which further inhibits downstream signaling. Based on its high affinity, high neutralizing potency, and novel binding epitope simultaneously occupying both IL-1RI and IL-1RAcP residues that bind to IL-1β, IgG26AW may be a new candidate for treatments of inflammation-related diseases or for complementary treatments of cancers in which the role of IL-1β is critical to pathogenesis.
Collapse
Affiliation(s)
- Wen-Chih Kuo
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Cheng-Chung Lee
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Ya-Wen Chang
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Wei Pang
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Hong-Sen Chen
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Shin-Chen Hou
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Shin-Yi Lo
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - An-Suei Yang
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Andrew H-J Wang
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan.
| |
Collapse
|
7
|
Gyorke CE, Kollipara A, Allen J, Zhang Y, Ezzell JA, Darville T, Montgomery SA, Nagarajan UM. IL-1α Is Essential for Oviduct Pathology during Genital Chlamydial Infection in Mice. THE JOURNAL OF IMMUNOLOGY 2020; 205:3037-3049. [PMID: 33087404 DOI: 10.4049/jimmunol.2000600] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 09/22/2020] [Indexed: 12/30/2022]
Abstract
Chlamydia trachomatis infection of the female genital tract can lead to irreversible fallopian tube scarring. In the mouse model of genital infection using Chlamydia muridarum, IL-1R signaling plays a critical role in oviduct tissue damage. In this study, we investigated the pathologic role of IL-1α, one of the two proinflammatory cytokines that bind to IL-1R. Il1a-/- mice infected with C. muridarum cleared infection at their cervix at the same rate as wild-type (WT) mice, but were significantly protected from end point oviduct damage and fibrosis. The contribution of IL-1α to oviduct pathology was more dramatic than observed in mice deficient for IL-1β. Although chlamydial burden was similar in WT and Il1a-/- oviduct during peak days of infection, levels of IL-1β, IL-6, CSF3, and CXCL2 were reduced in Il1a-/- oviduct lysates. During infection, Il1a-/- oviducts and uterine horns exhibited reduced neutrophil infiltration, and this reduction persisted after the infection resolved. The absence of IL-1α did not compromise CD4 T cell recruitment or function during primary or secondary chlamydial infection. IL-1α is expressed predominantly by luminal cells of the genital tract in response to infection, and low levels of expression persisted after the infection cleared. Ab-mediated depletion of IL-1α in WT mice prevented infection-induced oviduct damage, further supporting a key role for IL-1α in oviduct pathology.
Collapse
Affiliation(s)
- Clare E Gyorke
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Avinash Kollipara
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599.,Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599.,Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - John Allen
- Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Yugen Zhang
- Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - J Ashley Ezzell
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599; and.,Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Toni Darville
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599.,Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Stephanie A Montgomery
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Uma M Nagarajan
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599; .,Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| |
Collapse
|
8
|
Microbial therapeutics for acute colitis based on genetically modified Lactococcus lactis hypersecreting IL-1Ra in mice. Exp Mol Med 2020; 52:1627-1636. [PMID: 32989233 PMCID: PMC7520878 DOI: 10.1038/s12276-020-00507-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 07/07/2020] [Accepted: 07/13/2020] [Indexed: 12/21/2022] Open
Abstract
The increased incidence of inflammatory bowel disease (IBD) in Western and rapidly Westernizing developing countries poses a global pandemic threat. The development of affordable drugs for treating IBD worldwide is thus a priority. Genetically modified lactic acid bacteria (gmLAB) as microbial therapeutics are inexpensive protein producers suitable for use as carriers of protein to the intestinal mucosa. Here, we successfully constructed gmLAB hypersecreting interleukin 1 receptor antagonist (IL-1Ra). Oral administration of these gmLAB suppressed body weight reduction and exacerbation of the disease activity index score in mice with acute colitis and decreased the number of CD4+ IL-17A+ cells in the mesenteric lymph nodes. These data suggest that the gmLAB deliver IL-1Ra to the colon, where it inhibits IL-1 signaling. We thus developed a novel IBD therapeutic that blocks IL-1 signaling using a gmLAB protein delivery system. This system could be an inexpensive oral microbial therapeutic. Genetically reprogrammed bacteria can facilitate the efficient delivery of a therapeutic protein for treating inflammatory bowel disease (IBD). Interleukin 1 receptor antagonist (IL-1Ra) inhibits the immune cells that attack the intestinal lining in IBD patients, but current administration strategies are associated with serious side effects. Takeshi Shimosato and colleagues at Shinshu University in Nagano, Japan, have engineered the microbe Lactococcus lactis to secrete high levels of IL-1Ra. The researchers dosed mice orally with these bacteria, which released IL-1Ra into the intestinal tissue. This treatment proved safe and effectively reduced inflammation and associated symptoms in a mouse model of ulcerative colitis. L. lactis has previously been tested as a probiotic in clinical trials, and may therefore offer an appealing alternative to subcutaneous administration of IBD drugs in human patients.
Collapse
|
9
|
Shikonin Inhibits Der p 2-Induced Cytokine and Chemokine Expression in Dendritic Cells in Patients with Atopic Dermatitis. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:9506363. [PMID: 32802141 PMCID: PMC7417924 DOI: 10.1155/2020/9506363] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 07/07/2020] [Accepted: 07/18/2020] [Indexed: 11/17/2022]
Abstract
Atopic dermatitis (AD) is a common inflammatory skin disorder. Shikonin, the active component of Lithospermum erythrorhizon extract, exhibits anti-inflammatory effects. The objective of the present study was to investigate the effect of shikonin on proinflammatory cytokines and chemokine in patients with AD. Ten patients with AD who were allergic to house dust mite (HDM) and seven healthy controls were recruited in this study. Peripheral blood mononuclear cells were isolated, and CD14+ cells were further selected and differentiated to dendritic cells. Dendritic cells stimulated using Der p 2, the major HDM allergen, were cotreated with shikonin for 24 hours, and dexamethasone was used as a control. Culture supernatants were collected, and proinflammatory cytokine and chemokine concentrations were analyzed using a multiplex assay system. Shikonin significantly inhibited Der p 2-induced expression of interleukin (IL)-6, IL-9, and IL-17A; monocyte chemoattractant protein (MCP)-1; macrophage inflammatory protein (MIP)-1α; MIP-1β; and Chemokine (C-C motif) ligand 5 (RANTES). The inhibitory effects of shikonin on IL-9, MIP-1β, and RANTES expression were stronger than those of dexamethasone. Therefore, Shikonin can be considered a promising drug for AD treatment because it inhibits different inflammatory cytokines expression.
Collapse
|
10
|
Chiricosta L, Gugliandolo A, Bramanti P, Mazzon E. Could the Heat Shock Proteins 70 Family Members Exacerbate the Immune Response in Multiple Sclerosis? An in Silico Study. Genes (Basel) 2020; 11:genes11060615. [PMID: 32503176 PMCID: PMC7348765 DOI: 10.3390/genes11060615] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 05/27/2020] [Accepted: 06/01/2020] [Indexed: 01/08/2023] Open
Abstract
Multiple sclerosis (MS) is a chronic autoimmune demyelinating disease of the central nervous system. It represents one of the main causes of neurological disability in young people. In MS, the autoimmune response is directed against myelin antigens but other possible bio-molecular markers are investigated. The aim of this work was, through an in silico study, the evaluation of the transcriptional modifications between healthy subjects and MS patients in six brain areas (corpus callosum, hippocampus, internal capsule, optic chiasm, frontal and parietal cortex) in order to identify genes representative of the disease. Our results show the upregulation of the Heat Shock Proteins (HSPs) HSPA1A, HSPA1B, HSPA7, HSPA6, HSPH1 and HSPA4L of the HSP70 family, among which HSPA1A and HSPA1B are upregulated in all the brain areas. HSP70s are molecular chaperones indispensable for protein folding, recently associated with immune system maintenance. The little overexpression of the HSPs protects the cells from stress but extreme upregulation can contribute to the MS pathogenesis. We also investigated the genes involved in the immune system that result in overall upregulation in the corpus callosum, hippocampus, internal capsule, optic chiasm and are absent in the cortex. Interestingly, the genes of the immune system and the HSP70s have comparable levels of expression.
Collapse
|
11
|
Paryuni AD, Indarjulianto S, Widyarini S. Dermatophytosis in companion animals: A review. Vet World 2020; 13:1174-1181. [PMID: 32801570 PMCID: PMC7396343 DOI: 10.14202/vetworld.2020.1174-1181] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 04/29/2020] [Indexed: 11/17/2022] Open
Abstract
Dermatophytosis, a zoonotic disease, is caused by fungi of three main genera, namely, Micropsorum, Trichophyton, and Epidermophyton. Specific lesions of dermatophyte infections are localized in the face, legs, and/or tail. Skin lesions in infected animals demonstrate localized alopecia, erythema, and crust, which are more commonly known as ringworm. Factors that affect dermatophytosis include the dermatophyte species; virulence factors of the agent; and the immune status, age, and sex of the host. High levels of cortisol and pro-inflammatory cytokines have also been reported to play an important role in dermatophyte infection. This review aims to explore and understand factors that affect dermatophyte infection with an emphasis on the prevalence, clinical signs, pathogenesis, immune response, and the roles of cortisol and cytokines in companion animals infected by a dermatophyte.
Collapse
Affiliation(s)
- Alsi Dara Paryuni
- Department of Pathology, Faculty of Veterinary Medicine, Universitas Gadjah Mada, Yogyakarta 55281, Indonesia
| | - Soedarmanto Indarjulianto
- Department of Internal Medicine, Faculty of Veterinary Medicine, Universitas Gadjah Mada, Yogyakarta 55281, Indonesia
| | - Sitarina Widyarini
- Department of Pathology, Faculty of Veterinary Medicine, Universitas Gadjah Mada, Yogyakarta 55281, Indonesia
| |
Collapse
|
12
|
GSDMD membrane pore is critical for IL-1β release and antagonizing IL-1β by hepatocyte-specific nanobiologics is a promising therapeutics for murine alcoholic steatohepatitis. Biomaterials 2020; 227:119570. [DOI: 10.1016/j.biomaterials.2019.119570] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 10/16/2019] [Accepted: 10/18/2019] [Indexed: 12/15/2022]
|
13
|
Sabeti S, Kheirkhah A, Yin J, Dana R. Management of meibomian gland dysfunction: a review. Surv Ophthalmol 2019; 65:205-217. [PMID: 31494111 DOI: 10.1016/j.survophthal.2019.08.007] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 08/20/2019] [Accepted: 08/23/2019] [Indexed: 11/15/2022]
Abstract
Meibomian gland dysfunction is the leading cause of evaporative dry eye disease and is one of the most common conditions encountered by eye care providers. The disorder is characterized by obstruction of the meibomian gland terminal ducts and/or changes in their glandular secretion, resulting in changes in tear film stability, inflammation, and symptoms of irritation. There is no gold standard treatment for meibomian gland dysfunction, but rather a diversity of options. Conservative measures include warm compresses and lid hygiene, but there is growing interest and need for medical treatments and procedures. Potential medical treatments include antibiotics, nonsteroidal and steroidal anti-inflammatory agents, essential fatty acid supplementation, hormone therapy, and control of Demodex infestation. Procedures include intraductal meibomian gland probing, the use of electronic heating devices, intense pulsed light therapy, and intranasal neurostimulation. We provide an update on meibomian gland dysfunction treatments based on recent studies.
Collapse
Affiliation(s)
- Saama Sabeti
- University of Ottawa Eye Institute, The Ottawa Hospital, Ottawa, Ontario, Canada; Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Ahmad Kheirkhah
- Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, USA; Department of Ophthalmology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Jia Yin
- Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, USA
| | - Reza Dana
- Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, USA.
| |
Collapse
|
14
|
Weiss DI, Ma F, Merleev AA, Maverakis E, Gilliet M, Balin SJ, Bryson BD, Ochoa MT, Pellegrini M, Bloom BR, Modlin RL. IL-1β Induces the Rapid Secretion of the Antimicrobial Protein IL-26 from Th17 Cells. THE JOURNAL OF IMMUNOLOGY 2019; 203:911-921. [PMID: 31235553 DOI: 10.4049/jimmunol.1900318] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 06/06/2019] [Indexed: 12/18/2022]
Abstract
Th17 cells play a critical role in the adaptive immune response against extracellular bacteria, and the possible mechanisms by which they can protect against infection are of particular interest. In this study, we describe, to our knowledge, a novel IL-1β dependent pathway for secretion of the antimicrobial peptide IL-26 from human Th17 cells that is independent of and more rapid than classical TCR activation. We find that IL-26 is secreted 3 hours after treating PBMCs with Mycobacterium leprae as compared with 48 hours for IFN-γ and IL-17A. IL-1β was required for microbial ligand induction of IL-26 and was sufficient to stimulate IL-26 release from Th17 cells. Only IL-1RI+ Th17 cells responded to IL-1β, inducing an NF-κB-regulated transcriptome. Finally, supernatants from IL-1β-treated memory T cells killed Escherichia coli in an IL-26-dependent manner. These results identify a mechanism by which human IL-1RI+ "antimicrobial Th17 cells" can be rapidly activated by IL-1β as part of the innate immune response to produce IL-26 to kill extracellular bacteria.
Collapse
Affiliation(s)
- David I Weiss
- Division of Dermatology, Department of Medicine, David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, CA 90095.,Molecular Biology Interdepartmental Graduate Program, University of California, Los Angeles, Los Angeles, CA 90095
| | - Feiyang Ma
- Molecular Biology Interdepartmental Graduate Program, University of California, Los Angeles, Los Angeles, CA 90095.,Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095
| | - Alexander A Merleev
- Department of Dermatology, University of California, Davis, Sacramento, CA 95817
| | - Emanual Maverakis
- Department of Dermatology, University of California, Davis, Sacramento, CA 95817
| | - Michel Gilliet
- Department of Dermatology, University Hospital Lausanne, 1005 Lausanne, Switzerland
| | - Samuel J Balin
- Division of Dermatology, Department of Medicine, David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, CA 90095
| | - Bryan D Bryson
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02142
| | - Maria Teresa Ochoa
- Department of Dermatology, University of Southern California School of Medicine, Los Angeles, CA 90033
| | - Matteo Pellegrini
- Division of Dermatology, Department of Medicine, David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, CA 90095.,Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095
| | - Barry R Bloom
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115; and
| | - Robert L Modlin
- Division of Dermatology, Department of Medicine, David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, CA 90095; .,Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095
| |
Collapse
|
15
|
NLRP3 Inflammasome and IL-33: Novel Players in Sterile Liver Inflammation. Int J Mol Sci 2018; 19:ijms19092732. [PMID: 30213101 PMCID: PMC6163521 DOI: 10.3390/ijms19092732] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 09/09/2018] [Accepted: 09/10/2018] [Indexed: 12/11/2022] Open
Abstract
In sterile liver inflammation, danger signals are released in response to tissue injury to alert the immune system; e.g., by activation of the NLRP3 inflammasome. Recently, IL-33 has been identified as a novel type of danger signal or “alarmin”, which is released from damaged and necrotic cells. IL-33 is a pleiotropic cytokine that targets a broad range of immune cells and exhibits pro- and anti-inflammatory properties dependent on the disease. This review summarizes the immunomodulatory roles of the NLRP3 inflammasome and IL-33 in sterile liver inflammation and highlights potential therapeutic strategies targeting these pathways in liver disease.
Collapse
|
16
|
Xiao JP, Wang DY, Wang XR, Yuan L, Hao L, Wang DG. Increased ratio of Th17 cells to SIGIRR +CD4 + T cells in peripheral blood of patients with SLE is associated with disease activity. Biomed Rep 2018; 9:339-344. [PMID: 30233787 DOI: 10.3892/br.2018.1139] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 08/01/2018] [Indexed: 11/05/2022] Open
Abstract
To investigate the clinical significance of the ratio of T helper cell 17 (Th17) cells to single immunoglobulin IL-1-related receptor (SIGIRR)+ cluster of differentiation (CD4)+ T cells in patients with systemic lupus erythematosus (SLE), novel data and data from previous studies were analyzed. The frequency of Th17 cells in peripheral blood mononuclear cells (PBMCs) and their correlation with clinical data were evaluated in 48 patients with SLE and 38 healthy controls through flow cytometry. Compared with healthy controls, the percentage of Th17 cells was significantly increased in the PBMCs of patients with SLE (Z=-5.82, P<0.001). Compared with inactive SLE (ISLE), the percentage of Th17 cells in active SLE (ASLE) were significantly increased (Z=-4.26, P<0.0001). Compared with patients without lupus nephritis, the frequency of Th17 cells was significant increased (Z=-2.20, P=0.028). The frequency of Th17 cells was inversely correlated with the frequency of SIGIRR+CD4+ T cells (r=-0.61, P<0.001). The ratio of Th17 cells to SIGIRR+CD4+ T cells in ASLE was significantly increased compared with healthy controls or patients with ISLE (P<0.001) and was inversely correlated with complement component 3 and complement component 4, and positively correlated with SLE disease activity index and 24-h proteinuria (P<0.05). In summary, increased numbers of Th17 cells and decreased numbers of SIGIRR+CD4+ T cells in patients with SLE suggested that SIGIRR+CD4+ T and Th17 cells may be involved in the pathogenesis of SLE.
Collapse
Affiliation(s)
- Jian-Ping Xiao
- Department of Nephrology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, P.R. China
| | - Dao-Yang Wang
- Department of Nephrology, The Second Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233000, P.R. China
| | - Xue-Rong Wang
- Department of Nephrology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, P.R. China
| | - Liang Yuan
- Department of Nephrology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, P.R. China
| | - Li Hao
- Department of Nephrology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, P.R. China
| | - De-Guang Wang
- Department of Nephrology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, P.R. China
| |
Collapse
|
17
|
Worth L, Michel S, Gaertner VD, Kabesch M, Schieck M. Asthma- and IgE-associated polymorphisms affect expression of T H 17 genes. Allergy 2018; 73:1342-1347. [PMID: 29380867 DOI: 10.1111/all.13422] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- L. Worth
- Department of Pediatric Pneumology and Allergy University Children's Hospital Regensburg (KUNO) Regensburg Germany
| | - S. Michel
- Department of Pediatric Pneumology and Allergy University Children's Hospital Regensburg (KUNO) Regensburg Germany
| | - V. D. Gaertner
- Department of Pediatric Pneumology and Allergy University Children's Hospital Regensburg (KUNO) Regensburg Germany
| | - M. Kabesch
- Department of Pediatric Pneumology and Allergy University Children's Hospital Regensburg (KUNO) Regensburg Germany
| | - M. Schieck
- Department of Pediatric Pneumology and Allergy University Children's Hospital Regensburg (KUNO) Regensburg Germany
- Department of Human Genetics Hannover Medical School Hannover Germany
| |
Collapse
|
18
|
Impact of Fungi on Immune Responses. Clin Ther 2018; 40:885-888. [PMID: 29752039 DOI: 10.1016/j.clinthera.2018.04.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 04/09/2018] [Accepted: 04/18/2018] [Indexed: 12/25/2022]
Abstract
Spores and fungal fragments found in indoor and outdoor environments originate from opportunistic fungi and they can contribute to inflammatory responses, causing a broad range of symptoms. Papers were selected and reviewed with an emphasis on the molecular mechanisms involved in the effect of fungi on immune cells, especially mast cells (MCs). Fungi can bind to antibodies and complement them, allowing them to be recognized by cells of the innate immune system, including macrophages, dendritic cells, and MCs, which are then stimulated via Toll-like receptor signaling. Fungi can cause diseases mediated by MCs and aggravate allergic inflammation. Immunosuppressed subjects can be particularly susceptible to developing diseases caused by opportunistic fungi. Mold also liberates mycotoxins that could be on volatile spores and stimulate MCs to secrete pro-inflammatory cytokines/chemokines, but this mechanism is not known. Fungi can activate the immune system directly or through mycotoxins, leading to stimulation of immune cells and chronic neuroinflammatory symptoms. Some of these processes may be inhibited by the new anti-inflammatory cytokine interleukin 37.
Collapse
|
19
|
Luan J, Zhang X, Wang S, Li Y, Fan J, Chen W, Zai W, Wang S, Wang Y, Chen M, Meng G, Ju D. NOD-Like Receptor Protein 3 Inflammasome-Dependent IL-1β Accelerated ConA-Induced Hepatitis. Front Immunol 2018; 9:758. [PMID: 29692782 PMCID: PMC5902503 DOI: 10.3389/fimmu.2018.00758] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 03/27/2018] [Indexed: 12/20/2022] Open
Abstract
Autoimmune hepatitis (AIH) is a progressive inflammatory disorders of unknown etiology, characterized by immune-mediated destruction of hepatocytes and massive production of cytokines. Interleukin-1β is a pleiotropic proinflammatory cytokine and well known to be critical in a variety of autoimmune diseases. However, the role of interleukin-1β (IL-1β) in AIH is still indistinct. Here, we first investigated the significance of NOD-like receptor protein 3 (NLRP3) inflammasome-dependent IL-1β in the pathogenesis of AIH with a murine model of immune-mediated hepatitis induced by Concanavalin A (ConA). In ConA-treated mice, pathogenic elevated NLRP3, Cleaved caspase-1 and IL-1β levels, as well as an inflammatory cell death known as pyroptosis predominantly occurred in the livers. Strikingly, NLRP3−/− and caspase-1−/− mice were broadly protected from hepatitis as determined by decreased histological liver injury, serum aminotransferase (ALT)/aspartate transaminase levels, and pyroptosis. In vivo intervention with recombinant human interleukin-1 receptor antagonist (rhIL-1Ra) strongly suppressed ConA-induced hepatitis by decreasing tumor necrosis factor-alpha (TNF-α) and interleukin-17 (IL-17) secretion, and inflammatory cell infiltration into livers. Additionally, rhIL-1Ra-pretreated mice developed significantly reduced NLRP3 inflammasome activation and reactive oxygen species (ROS) generation. Scavenging of ROS by N-acetyl-cysteine also attenuated NLRP3 inflammasome activation and liver inflammation, indicating that the essential role of ROS in mediating NLRP3 inflammasome activation in ConA-induced hepatitis. In conclusion, our results demonstrated that NLRP3 inflammasome-dependent IL-1β production was crucial in the pathogenesis of ConA-induced hepatitis, which shed light on the development of promising therapeutic strategies for AIH by blocking NLRP3 inflammasome and IL-1β.
Collapse
Affiliation(s)
- Jingyun Luan
- Department of Microbiological and Biochemical Pharmacy, The Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, China
| | - Xuyao Zhang
- Department of Microbiological and Biochemical Pharmacy, The Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, China
| | - Shaofei Wang
- Department of Microbiological and Biochemical Pharmacy, The Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, China
| | - Yubin Li
- Department of Microbiological and Biochemical Pharmacy, The Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, China
| | - Jiajun Fan
- Department of Microbiological and Biochemical Pharmacy, The Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, China
| | - Wei Chen
- Department of Microbiological and Biochemical Pharmacy, The Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, China
| | - Wenjing Zai
- Department of Microbiological and Biochemical Pharmacy, The Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, China
| | - Sijia Wang
- Department of Microbiological and Biochemical Pharmacy, The Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, China
| | - Yichen Wang
- Department of Microbiological and Biochemical Pharmacy, The Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, China
| | - Mingkuan Chen
- Unit of Innate Immunity, Key Laboratory of Molecular Virology and Immunology, Institute Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Guangxun Meng
- Unit of Innate Immunity, Key Laboratory of Molecular Virology and Immunology, Institute Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Dianwen Ju
- Department of Microbiological and Biochemical Pharmacy, The Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, China
| |
Collapse
|
20
|
Sha Y, Markovic-Plese S. Activated IL-1RI Signaling Pathway Induces Th17 Cell Differentiation via Interferon Regulatory Factor 4 Signaling in Patients with Relapsing-Remitting Multiple Sclerosis. Front Immunol 2016; 7:543. [PMID: 27965670 PMCID: PMC5126112 DOI: 10.3389/fimmu.2016.00543] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Accepted: 11/16/2016] [Indexed: 12/30/2022] Open
Abstract
IL-1β plays a crucial role in the differentiation of human Th17 cells. We report here that IL-1RI expression is significantly increased in both naive and memory CD4+ T cells derived from relapsing-remitting multiple sclerosis (RR MS) patients in comparison to healthy controls. Interleukin 1 receptor (IL-1R)I expression is upregulated in the in vitro-differentiated Th17 cells from RR MS patients in comparison to the Th1 and Th2 cell subsets, indicating the role of IL-1R signaling in the Th17 cell differentiation in RR MS. When IL-1RI gene expression was silenced using siRNA, human naive CD4+ T cells cultured in the presence of Th17-polarizing cytokines had a significantly decreased expression of interleukin regulatory factor 4 (IRF4), RORc, IL-17A, IL-17F, IL-21, IL-22, and IL-23R genes, confirming that IL-1RI signaling induces Th17 cell differentiation. Since IL-1R gene expression silencing inhibited IRF4 expression and Th17 differentiation, and IRF4 gene expression silencing inhibited Th17 cell differentiation, our results indicate that IL-1RI induces human Th17 cell differentiation in an IRF4-dependant manner. Our study has identified that IL-1RI-mediated signaling pathway is constitutively activated, leading to an increased Th17 cell differentiation in IRF4-dependent manner in patients with RR MS.
Collapse
Affiliation(s)
- Yonggang Sha
- Department of Neurology, University of North Carolina at Chapel Hill , Chapel Hill, NC , USA
| | - Silva Markovic-Plese
- Department of Neurology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
21
|
|
22
|
Stürner KH, Verse N, Yousef S, Martin R, Sospedra M. Boswellic acids reduce Th17 differentiation via blockade of IL-1β-mediated IRAK1 signaling. Eur J Immunol 2014; 44:1200-12. [PMID: 24469975 DOI: 10.1002/eji.201343629] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2013] [Revised: 12/19/2013] [Accepted: 01/20/2014] [Indexed: 01/29/2023]
Abstract
Interferon-gamma producing CD4(+) T (Th1) cells and IL-17-producing CD4(+) T (Th17) cells are involved in the pathogenesis of several autoimmune diseases including multiple sclerosis. Therefore, the development of treatment strategies controlling the generation and expansion of these effector cells is of high interest. Frankincense, the resin from trees of the genus Boswellia, and particularly its prominent bioactive compound acetyl-11-keto-β-boswellic acid (AKBA), have potent anti-inflammatory properties. Here, we demonstrate that AKBA is able to reduce the differentiation of human CD4(+) T cells to Th17 cells, while slightly increasing Th2- and Treg-cell differentiation. Furthermore, AKBA reduces the IL-1β-triggered IL-17A release of memory Th17 cells. AKBA may affect IL-1β signaling by preventing IL-1 receptor-associated kinase 1 phosphorylation and subsequently decreasing STAT3 phosphorylation at Ser727, which is required for Th17-cell differentiation. The effects of AKBA on Th17 differentiation and IL-17A release make the compound a good candidate for potential treatment of Th17-driven diseases.
Collapse
Affiliation(s)
- Klarissa Hanja Stürner
- Institute for Neuroimmunology and Clinical Multiple Sclerosis Research (INIMS) and Clinic for Neurology, Center for Molecular Neurobiology, Universitätsklinikum Hamburg Eppendorf, Hamburg, Germany
| | | | | | | | | |
Collapse
|
23
|
Tominaga A, Konishi Y, Taguchi T, Fukuoka S, Kawaguchi T, Noda T, Shimizu K. Autonomous cure of damaged human intestinal epithelial cells by TLR2 and TLR4-dependent production of IL-22 in response to Spirulina polysaccharides. Int Immunopharmacol 2013; 17:1009-19. [PMID: 24126111 DOI: 10.1016/j.intimp.2013.09.023] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Revised: 09/14/2013] [Accepted: 09/26/2013] [Indexed: 02/01/2023]
Abstract
In order to analyze the damage of human epithelial cells, we used human quasi-normal FPCK-1-1 cells derived from a colonic polyp in a patient with familial adenomatous polyposis as a monolayer, which is co-cultured with peptidoglycan (PGN)-stimulated THP-1 cells. Co-cultured FPCK-1-1 cells showed a decreased transepithelial electrical resistance (TER) and the lower level of claudin-2. When Spirulina complex polysaccharides were added one day before the start of the co-culture, there was no decrease of TER and claudin-2 (early phase damage). In contrast, when Spirulina complex polysaccharides were added to FPCK-1-1 cells after the level of TER had decreased, there was no recovery at the level of claudin-2, though the TER level recovered (late phase damage). The mucosa reconstitution is suggested to be involved in the recovery from the damaged status. Interestingly, autonomous recovery of FPCK-1-1 cells from both the early and late phase damage requires the production of IL-22, because anti-IL-22 antibodies inhibited recovery in these cases. Antibodies against either TLR2 or TLR4 inhibited the production of IL-22 from FPCK-1-1 colon epithelial cells, suggesting that signals through TLR2 and TLR4 are necessary for autonomous recovery of FPCK-1-1 colon epithelial cells by producing IL-22. In conclusion, we have established a useful model for the study of intestinal damage and recovery using human colon epithelial cells and our data suggest that damage to human colon epithelial cells can, at least in part, be recovered by the autonomous production of IL-22 in response to Spirulina complex polysaccharides.
Collapse
Affiliation(s)
- Akira Tominaga
- Laboratory of Human Health and Medical Science, Graduate School of Kuroshio Science, Kochi University, Kohasu, Okoh-cho, Nankoku, Kochi 783-8505, Japan; Department of Molecular and Cellular Biology, Kochi Medical School, Kochi University, Kohasu, Okoh-cho, Nankoku, Kochi 783-8505, Japan.
| | | | | | | | | | | | | |
Collapse
|
24
|
Amparo F, Dana R. IL-1 receptor antagonist in the treatment of dry eye disease. EXPERT REVIEW OF OPHTHALMOLOGY 2013. [DOI: 10.1586/17469899.2013.851004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
25
|
Lim MA, Lee J, Park JS, Jhun JY, Moon YM, Cho ML, Kim HY. Increased Th17 differentiation in aged mice is significantly associated with high IL-1β level and low IL-2 expression. Exp Gerontol 2013; 49:55-62. [PMID: 24140620 DOI: 10.1016/j.exger.2013.10.006] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Accepted: 10/09/2013] [Indexed: 10/26/2022]
Abstract
OBJECTIVE Aging has been reported to be associated with changes in immune function. Although frequent infection and the development of malignancy suggest the decline of immune function with aging, changes toward proinflammatory conditions also develop at the same time. Th17 cells are well known CD4(+) T cell subpopulation closely linked to chronic inflammation and autoimmunity. In this study, changes in the Th17 population were investigated to elucidate a possible mechanism for this response with aging. METHODS Splenocytes were isolated from 2-month-old (young) and 20-month-old (aged) mice. CD4(+)CD44(+) memory T cells and CD4(+)CD62L(+) naïve T cells were isolated and sorted using magnetic beads and flow cytometry. The frequency of IL-17-producing cells was measured using flow cytometry. The expression of IL-17 and Th17-related factors at the mRNA level was measured with RT-PCR. IL-17 and Il-1β expression in spleen tissues was additionally assessed using confocal microscopy. RESULTS The proportion of IL-17-producing CD4(+) T cells was higher in the splenocytes among the old mice than those of the young mice. When splenocytes were cultured in Th17 polarizing conditions, the proportion of IL-17 producing CD4(+) T cells was higher in aged mice as well. This was consistently observed when naïve and memory cells were isolated and differentiated into Th17 respectively. In addition, the expression of retinoic acid receptor-related orphan nuclear receptor gamma t (RORγt) and other Th17-related factors (AhR, CCR6, and CCL20) increased in the splenocytes of aged mice compared to the young mice. The expression of IL-1β, showing to promote Th17 differentiation, was higher in the aged mice. Likewise, CD4(+) T cell expression of IL-1R was higher in the aged mice, suggesting that the CD4(+) T cells of the aged mice are readily prepared to differentiate into Th17 cells in response to IL-1β. Confocal microscopy showed that cells positive for IL-1R or IL-1β were more frequent in the spleens of the aged mice. When an anti-IL-2 antibody was applied, the proportion of IL-17-producing cells increased more prominently in the young mice. We observed that IL-2 production and IL-2R expression were reduced in the aged mice, respectively, explaining the blunted response to the anti-IL-2 antibody treatment and the consequent minimal change in the Th17 population. CONCLUSION We demonstrated that the proportion of Th17 cells increased in the aged mice both in naïve and memory cell populations. Elevation of IL-1R and IL-1β expression and the reduction in IL-2 and IL-2R expression in aged mice seemed to promote Th17 differentiation. Our results suggest that enhanced Th17 differentiation in aging may have a pathogenic role in the development of Th17-mediated autoimmune diseases.
Collapse
Affiliation(s)
- Mi-Ae Lim
- Rheumatism Research Center, The Catholic University of Korea, Seoul, South Korea
| | - Jennifer Lee
- Rheumatism Research Center, The Catholic University of Korea, Seoul, South Korea
| | - Jin-Sil Park
- Rheumatism Research Center, The Catholic University of Korea, Seoul, South Korea
| | - Joo-Yeon Jhun
- Rheumatism Research Center, The Catholic University of Korea, Seoul, South Korea
| | - Young-Mi Moon
- Rheumatism Research Center, The Catholic University of Korea, Seoul, South Korea
| | - Mi-La Cho
- Rheumatism Research Center, The Catholic University of Korea, Seoul, South Korea.
| | - Ho-Youn Kim
- Division of Rheumatology, Department of Internal Medicine, Konkuk University, Seoul, South Korea.
| |
Collapse
|
26
|
Amparo F, Dastjerdi MH, Okanobo A, Ferrari G, Smaga L, Hamrah P, Jurkunas U, Schaumberg DA, Dana R. Topical interleukin 1 receptor antagonist for treatment of dry eye disease: a randomized clinical trial. JAMA Ophthalmol 2013; 131:715-723. [PMID: 23599118 DOI: 10.1001/jamaophthalmol.2013.195] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
IMPORTANCE The immunopathogenic mechanisms of dry eye disease (DED), one of the most common ophthalmic conditions, is incompletely understood. Data from this prospective, double-masked, randomized trial demonstrate that targeting interleukin 1 (IL-1) by topical application of an IL-1 antagonist is efficacious in significantly reducing DED-related patient symptoms and corneal epitheliopathy. OBJECTIVE To evaluate the safety and efficacy of treatment with the topical IL-1 receptor antagonist anakinra (Kineret; Amgen Inc) in patients having DED associated with meibomian gland dysfunction. DESIGN AND SETTING Prospective phase 1/2, randomized, double-masked, vehicle-controlled clinical trial. PARTICIPANTS Seventy-five patients with refractory DED. INTERVENTIONS Participants were randomized to receive treatment with topical anakinra, 2.5% (n = 30), anakinra, 5% (n = 15), or vehicle (1% carboxymethylcellulose) (n = 30) 3 times daily for 12 weeks. MAIN OUTCOMES AND MEASURES Primary outcomes were corneal fluorescein staining (CFS), complete bilateral CFS clearance, dry eye-related symptoms as measured by the Ocular Surface Disease Index, tear film breakup time, and meibomian gland secretion quality. RESULTS Topical anakinra was well tolerated compared with vehicle, with no reports of serious adverse reactions attributable to the therapy. After 12 weeks of therapy, participants treated with anakinra, 2.5%, achieved a 46% reduction in their mean CFS score (P = .12 compared with vehicle and P < .001 compared with baseline); participants treated with anakinra, 5%, achieved a 17% reduction in their mean CFS score (P = .88 compared with vehicle and P = .33 compared with baseline); and patients treated with vehicle achieved a 19% reduction in their mean CFS score (P = .11). Complete bilateral CFS clearance was noted in 8 of 28 patients (29%) treated with anakinra, 2.5%, vs in 2 of 29 patients (7%) treated with vehicle (P = .03). By week 12, treatment with anakinra, 2.5%, and treatment with anakinra, 5%, led to significant reductions in symptoms of 30% and 35%, respectively (P = .02 and P = .01, respectively, compared with vehicle); treatment with vehicle led to a 5% reduction in symptoms. CONCLUSIONS AND RELEVANCE Treatment with topical anakinra, 2.5%, for 12 weeks was safe and significantly reduced symptoms and corneal epitheliopathy in patients with DED. These data suggest that the use of an IL-1 antagonist may have a role as a novel therapeutic option for patients with DED. TRIAL REGISTRATION clinicaltrials.gov Identifier: NCT00681109.
Collapse
Affiliation(s)
- Francisco Amparo
- Department of Ophthalmology, Massachusetts Eye and Ear Infirmary (Drs Amparo, Dastjerdi, Okanobo, Ferrari, Hamrah, Jurkunas, and Dana and Ms Smaga), and Division of Preventive Medicine, Brigham and Women's Hospital (Dr Schaumberg), Harvard Medical School, Boston
| | - Mohammad H Dastjerdi
- Department of Ophthalmology, Massachusetts Eye and Ear Infirmary (Drs Amparo, Dastjerdi, Okanobo, Ferrari, Hamrah, Jurkunas, and Dana and Ms Smaga), and Division of Preventive Medicine, Brigham and Women's Hospital (Dr Schaumberg), Harvard Medical School, Boston
| | - Andre Okanobo
- Department of Ophthalmology, Massachusetts Eye and Ear Infirmary (Drs Amparo, Dastjerdi, Okanobo, Ferrari, Hamrah, Jurkunas, and Dana and Ms Smaga), and Division of Preventive Medicine, Brigham and Women's Hospital (Dr Schaumberg), Harvard Medical School, Boston
| | - Giulio Ferrari
- Department of Ophthalmology, Massachusetts Eye and Ear Infirmary (Drs Amparo, Dastjerdi, Okanobo, Ferrari, Hamrah, Jurkunas, and Dana and Ms Smaga), and Division of Preventive Medicine, Brigham and Women's Hospital (Dr Schaumberg), Harvard Medical School, Boston
| | - Leila Smaga
- Department of Ophthalmology, Massachusetts Eye and Ear Infirmary (Drs Amparo, Dastjerdi, Okanobo, Ferrari, Hamrah, Jurkunas, and Dana and Ms Smaga), and Division of Preventive Medicine, Brigham and Women's Hospital (Dr Schaumberg), Harvard Medical School, Boston
| | - Pedram Hamrah
- Department of Ophthalmology, Massachusetts Eye and Ear Infirmary (Drs Amparo, Dastjerdi, Okanobo, Ferrari, Hamrah, Jurkunas, and Dana and Ms Smaga), and Division of Preventive Medicine, Brigham and Women's Hospital (Dr Schaumberg), Harvard Medical School, Boston
| | - Ula Jurkunas
- Department of Ophthalmology, Massachusetts Eye and Ear Infirmary (Drs Amparo, Dastjerdi, Okanobo, Ferrari, Hamrah, Jurkunas, and Dana and Ms Smaga), and Division of Preventive Medicine, Brigham and Women's Hospital (Dr Schaumberg), Harvard Medical School, Boston
| | - Debra A Schaumberg
- Department of Ophthalmology, Massachusetts Eye and Ear Infirmary (Drs Amparo, Dastjerdi, Okanobo, Ferrari, Hamrah, Jurkunas, and Dana and Ms Smaga), and Division of Preventive Medicine, Brigham and Women's Hospital (Dr Schaumberg), Harvard Medical School, Boston
| | - Reza Dana
- Department of Ophthalmology, Massachusetts Eye and Ear Infirmary (Drs Amparo, Dastjerdi, Okanobo, Ferrari, Hamrah, Jurkunas, and Dana and Ms Smaga), and Division of Preventive Medicine, Brigham and Women's Hospital (Dr Schaumberg), Harvard Medical School, Boston
| |
Collapse
|
27
|
Thomson TM, Sewer A, Martin F, Belcastro V, Frushour BP, Gebel S, Park J, Schlage WK, Talikka M, Vasilyev DM, Westra JW, Hoeng J, Peitsch MC. Quantitative assessment of biological impact using transcriptomic data and mechanistic network models. Toxicol Appl Pharmacol 2013; 272:863-78. [PMID: 23933166 DOI: 10.1016/j.taap.2013.07.007] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Revised: 07/01/2013] [Accepted: 07/13/2013] [Indexed: 12/29/2022]
Abstract
Exposure to biologically active substances such as therapeutic drugs or environmental toxicants can impact biological systems at various levels, affecting individual molecules, signaling pathways, and overall cellular processes. The ability to derive mechanistic insights from the resulting system responses requires the integration of experimental measures with a priori knowledge about the system and the interacting molecules therein. We developed a novel systems biology-based methodology that leverages mechanistic network models and transcriptomic data to quantitatively assess the biological impact of exposures to active substances. Hierarchically organized network models were first constructed to provide a coherent framework for investigating the impact of exposures at the molecular, pathway and process levels. We then validated our methodology using novel and previously published experiments. For both in vitro systems with simple exposure and in vivo systems with complex exposures, our methodology was able to recapitulate known biological responses matching expected or measured phenotypes. In addition, the quantitative results were in agreement with experimental endpoint data for many of the mechanistic effects that were assessed, providing further objective confirmation of the approach. We conclude that our methodology evaluates the biological impact of exposures in an objective, systematic, and quantifiable manner, enabling the computation of a systems-wide and pan-mechanistic biological impact measure for a given active substance or mixture. Our results suggest that various fields of human disease research, from drug development to consumer product testing and environmental impact analysis, could benefit from using this methodology.
Collapse
Affiliation(s)
- Ty M Thomson
- Selventa, One Alewife Center, Cambridge, MA 02140, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Santarlasci V, Cosmi L, Maggi L, Liotta F, Annunziato F. IL-1 and T Helper Immune Responses. Front Immunol 2013; 4:182. [PMID: 23874332 PMCID: PMC3711056 DOI: 10.3389/fimmu.2013.00182] [Citation(s) in RCA: 102] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Accepted: 06/24/2013] [Indexed: 01/13/2023] Open
Abstract
CD4 T cells play a critical role in mediating adaptive immunity to a variety of pathogens as well as in tumor immunity. If not adequately regulated, CD4 T cells can be also involved in autoimmunity, asthma, and allergic responses. During TCR activation in a particular cytokine milieu, naïve CD4 T cells may differentiate into one of several lineages of T helper (Th) cells, including Th1, Th2, and Th17, as defined by their pattern of cytokine production and function. IL-1, the prototypic proinflammatory cytokine, has been shown to influence growth and differentiation of immunocompetent lymphocytes. The differential expression of IL-1RI on human CD4 T cell subsets confers distinct capacities to acquire specific effector functions. In this review, we summarize the role of IL-1 on CD4 T cells, in terms of differentiation, activation, and maintenance or survival.
Collapse
Affiliation(s)
- Veronica Santarlasci
- Department of Experimental and Clinical Medicine, University of Florence , Florence , Italy
| | | | | | | | | |
Collapse
|
29
|
Gonzalez-Lombana C, Gimblet C, Bacellar O, Oliveira WW, Passos S, Carvalho LP, Goldschmidt M, Carvalho EM, Scott P. IL-17 mediates immunopathology in the absence of IL-10 following Leishmania major infection. PLoS Pathog 2013; 9:e1003243. [PMID: 23555256 PMCID: PMC3605236 DOI: 10.1371/journal.ppat.1003243] [Citation(s) in RCA: 123] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Accepted: 01/29/2013] [Indexed: 12/31/2022] Open
Abstract
Leishmaniasis, resulting from infection with the protozoan parasite Leishmania, consists of a wide spectrum of clinical manifestations, from healing cutaneous lesions to fatal visceral infections. A particularly severe form of cutaneous leishmaniasis, termed mucosal leishmaniasis, exhibits decreased IL-10 levels and an exaggerated inflammatory response that perpetuates the disease. Using a mouse model of leishmaniasis, we investigated what cytokines contribute to increased pathology when IL-10-mediated regulation is absent. Leishmania major infected C57BL/6 mice lacking IL-10 regulation developed larger lesions than controls, but fewer parasites. Both IFN-γ and IL-17 levels were substantially elevated in mice lacking the capacity to respond to IL-10. IFN-γ promoted an increased infiltration of monocytes, while IL-17 contributed to an increase in neutrophils. Surprisingly, however, we found that IFN-γ did not contribute to increased pathology, but instead regulated the IL-17 response. Thus, blocking IFN-γ led to a significant increase in IL-17, neutrophils and disease. Similarly, the production of IL-17 by cells from leishmaniasis patients was also regulated by IL-10 and IFN-γ. Additional studies found that the IL-1 receptor was required for both the IL-17 response and increased pathology. Therefore, we propose that regulating IL-17, possibly by downregulating IL-1β, may be a useful approach for controlling immunopathology in leishmaniasis. Leishmaniasis is a tropical disease transmitted by sand flies that causes visceral and cutaneous lesions. In humans, the most severe form of cutaneous leishmaniasis is the mucosal form, causing disfiguring lesions in the nasal and oral mucosa. Why these patients develop severe disease is not clear. It is known, however, that the severe disease is not due to an overwhelming number of parasites, but rather appears to be due to an uncontrolled inflammatory response that includes elevated production of IFN-γ and IL-17. Here, we used a murine model of leishmaniasis to identify the factors involved in this pathology, and found that mice infected with Leishmania major developed severe lesions in the absence of IL-10 or IL-10 signaling, and similar to patients, contained high levels of IFN-γ and IL-17. While both of these cytokines have the potential to induce pathology, we found that IL-17 was responsible for the severe pathology seen in the absence of IL-10 regulation, and furthermore that IL-17 levels were higher and pathology greater in the absence of IFN-γ. Thus, our study suggests that IL-17, but not the IFN-γ, is a strong candidate to be targeted in strategies to control the severe immunopathology observed in mucosal leishmaniasis patients.
Collapse
Affiliation(s)
- Claudia Gonzalez-Lombana
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Ciara Gimblet
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Olivia Bacellar
- Instituto Nacional de Ciência e Tecnologia de Doenças Tropicais-INCT-DT(CNPq/MCT), Serviço de Imunologia, Hospital Universitario Prof. Edgard Santos, Universidade Federal da Bahia Salvador, Bahia, Brasil
| | - Walker W. Oliveira
- Instituto Nacional de Ciência e Tecnologia de Doenças Tropicais-INCT-DT(CNPq/MCT), Serviço de Imunologia, Hospital Universitario Prof. Edgard Santos, Universidade Federal da Bahia Salvador, Bahia, Brasil
| | - Sara Passos
- Instituto Nacional de Ciência e Tecnologia de Doenças Tropicais-INCT-DT(CNPq/MCT), Serviço de Imunologia, Hospital Universitario Prof. Edgard Santos, Universidade Federal da Bahia Salvador, Bahia, Brasil
| | - Lucas P. Carvalho
- Instituto Nacional de Ciência e Tecnologia de Doenças Tropicais-INCT-DT(CNPq/MCT), Serviço de Imunologia, Hospital Universitario Prof. Edgard Santos, Universidade Federal da Bahia Salvador, Bahia, Brasil
| | - Michael Goldschmidt
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Edgar M. Carvalho
- Instituto Nacional de Ciência e Tecnologia de Doenças Tropicais-INCT-DT(CNPq/MCT), Serviço de Imunologia, Hospital Universitario Prof. Edgard Santos, Universidade Federal da Bahia Salvador, Bahia, Brasil
| | - Phillip Scott
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
30
|
Halappanavar S, Nikota J, Wu D, Williams A, Yauk CL, Stampfli M. IL-1 receptor regulates microRNA-135b expression in a negative feedback mechanism during cigarette smoke-induced inflammation. THE JOURNAL OF IMMUNOLOGY 2013; 190:3679-86. [PMID: 23440414 DOI: 10.4049/jimmunol.1202456] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Although microRNA-135b (miR-135b) is known to be associated with cancer, with recent work showing that it is massively induced in the pulmonary tissues of mice challenged with nanoparticles suggests a critical role for this microRNA in mediating inflammatory response. In this study, we investigated the expression and function of miR-135b in mice exposed to cigarette smoke or nontypeable Haemophilus influenzae (NTHi). Exposure to both cigarette smoke and NTHi elicited robust lung inflammation, but increased miR-135b expression was observed only in the lungs of cigarette smoke-exposed mice. Using IL-1R 1 knockout mice, we show that miR-135b expression is IL-1R1 dependent. A series of in vitro experiments confirmed the role of IL-1R1 in regulating miR-135b expression. In vitro activation of the IL-1R1 pathway in mouse embryonic fibroblast (NIH3T3) and lung epithelial (FE1) cells resulted in increased miR-135b, which was blocked by IL-1R1 antagonists or small interfering RNA-mediated silencing of IL-1R1 expression. Overexpression of mature miR-135b in NIH3T3 cells (pEGP-mmu-mir-135b) resulted in the suppression of endogenous levels of IL-1R1 expression. pEGP-mmu-miR-135b cells transiently transfected with luciferase reporter vector containing the 3'UTR of mouse IL-1R1 showed reduced luciferase activity. Finally, we demonstrate that miR-135b targets IL-1-stimulated activation of Caspase-1, the IL-1R1 downstream activator of IL-1β leading to suppressed synthesis of the active form of IL-1β protein. These results suggest that miR-135b expression during cigarette smoke-induced inflammation is regulated by IL-1R1 in a regulatory feedback mechanism to resolve inflammation.
Collapse
Affiliation(s)
- Sabina Halappanavar
- Environmental and Radiation Health Sciences Directorate, Health Canada, Ottawa, Ontario K1A 0K9, Canada.
| | | | | | | | | | | |
Collapse
|
31
|
Abstract
There is increasing evidence in the literature pointing to an important role of inflammation during initiation and progression of cancer. Glioblastoma is the most common malignant primary brain tumor with approximately 23,000 newly-diagnosed cases each year in the United States, and has a dismal median survival of only 15 months. Although the blood-brain barrier maintains an immune-privileged status of the brain under steady state, intracranial tumors including gliomas are invariably infiltrated with various types of immune cells. The T helper 17 (Th17) cells, a recently discovered interleukin (IL)-17-producing T cell subtype, have been reported in several extracranial and some intracranial tumors, where they have been implicated in either pro- or antitumor activity depending on the tumor type. Here, we present a succinct review of the current literature on the prevalence and potential role of IL-17 in malignant gliomas. Further mechanistic studies on IL-17 mediated inflammatory pathway in gliomas may provide with opportunities for novel immunotherapeutic interventions.
Collapse
Affiliation(s)
- Prahlad Parajuli
- Department of Neurosurgery, Wayne State University and Karmanos Cancer Institute, Detroit, USA
| | - Sandeep Mittal
- Department of Neurosurgery, Wayne State University and Karmanos Cancer Institute, Detroit, USA
| |
Collapse
|
32
|
Lee YS, Lee KA, Yoon HB, Yoo SA, Park YW, Chung Y, Kim WU, Kang CY. The Wnt inhibitor secreted Frizzled-Related Protein 1 (sFRP1) promotes human Th17 differentiation. Eur J Immunol 2012; 42:2564-73. [PMID: 22740051 DOI: 10.1002/eji.201242445] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2012] [Revised: 05/28/2012] [Accepted: 06/21/2012] [Indexed: 01/06/2023]
Abstract
Wnt/β-catenin signaling plays a crucial role during embryogenesis and tumorigenesis, and in T cells, promotes the differentiation of Th2 cells. However, the role of Wnt signals in the differentiation and maintenance of human Th17 cells remains poorly understood. We found that the higher levels of IL-17 in the synovial fluid of rheumatoid arthritis (RA) patients compared with that of osteoarthritis (OA) patients were associated with a higher concentration of sFRP1 (secreted Frizzled-Related Protein 1), an inhibitor of the Wnt/β-catenin pathway. The addition of sFRP1 during TCR-mediated stimulation induced a significant increase in IL-17 production by both naïve and memory CD4(+) T cells. Moreover, under Th17-differentiation conditions, the addition of sFRP1 significantly reduced the requirement for TGF-β. Mechanistically, we observed that sFRP1 significantly enhanced the phosphorylation of Smad2/3 in CD4(+) T cells upon TGF-β stimulation and that blocking TGF-β signaling abolished the Th17-promoting activity of sFRP1. Our findings reveal a novel function for sFRP1 as a potent inducer of human Th17-cell differentiation. Consequently, sFRP1 may represent a promising target for the treatment of Th17-mediated disease in humans.
Collapse
Affiliation(s)
- Yoon-Sook Lee
- Laboratory of Immunology, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, Korea
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Buchholz BM, Chanthaphavong RS, Billiar TR, Bauer AJ. Role of interleukin-6 in hemopoietic and non-hemopoietic synergy mediating TLR4-triggered late murine ileus and endotoxic shock. Neurogastroenterol Motil 2012; 24:658-69, e294. [PMID: 22489868 PMCID: PMC3378783 DOI: 10.1111/j.1365-2982.2012.01914.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
BACKGROUND Early murine endotoxin-induced ileus at 6 h is exclusively mediated by non-hemopoietic TLR4/MyD88 signaling despite molecular activation of hemopoietic cells which included a significant IL-6 mRNA induction. Our objective was to define the role of hemopoietic cells in LPS/TLR4-triggered ileus and inflammation over time, and identify mechanisms of ileus. METHODS CSF-1(-/-) , TLR4 non-chimera and TLR4 chimera mice were single-shot intraperitoneal injected with ultrapure lipopolysaccharide (UP-LPS) and studied up to 4 days. Subgroups of TLR4(WT) mice were additionally intravenously injected with exogenous recombinant IL-6 (rmIL-6) or murine soluble IL-6 receptor blocking antibody (anti-sIL-6R mAB). KEY RESULTS Hemopoietic TLR4 signaling independently mediated UP-LPS-induced ileus at 24 h, but chemotactic muscularis neutrophil extravasation was not causatively involved and mice lacking CSF-1-dependent macrophages died prematurely. Synergy of hemopoietic and non-hemopoietic cells determined ileus severity and mortality which correlated with synergistic cell lineage specific transcription of inflammatory mediators like IL-6 within the intestinal muscularis. Circulating IL-6 levels were LPS dose dependent, but exogenous rmIL-6 did not spark off a self-perpetuating inflammatory response triggering ileus. Sustained therapeutic inhibition of functional IL-6 signaling efficiently ameliorated late ileus while preemptive antibody-mediated IL-6R blockade was marginally effective in mitigating ileus. However, IL-6R blockade did not prevent endotoxin-associated mortality nor did it alter circulating IL-6 levels. CONCLUSIONS & INFERENCES A time-delayed bone marrow-driven mechanism of murine endotoxin-induced ileus exists, and hemopoietic cells synergize with non-hemopoietic cells thereby prolonging ileus and fueling intestinal inflammation. Importantly, IL-6 signaling via IL-6R/gp130 drives late ileus, yet it did not regulate mortality in endotoxic shock.
Collapse
Affiliation(s)
- Bettina M. Buchholz
- Department of Medicine/Gastroenterology, University of Pittsburgh, Pittsburgh, PA,Department of Surgery, University of Bonn, Bonn, Germany
| | | | | | - Anthony J. Bauer
- Department of Medicine/Gastroenterology, University of Pittsburgh, Pittsburgh, PA
| |
Collapse
|
34
|
Paladugu M, Thakur A, Lum LG, Mittal S, Parajuli P. Generation and immunologic functions of Th17 cells in malignant gliomas. Cancer Immunol Immunother 2012; 62:75-86. [PMID: 22752645 DOI: 10.1007/s00262-012-1312-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2012] [Accepted: 06/15/2012] [Indexed: 12/22/2022]
Abstract
Th17 cells, a recently discovered inflammatory T cell subtype, have been implicated with autoimmune disorders. However, mechanism of generation or functions of intratumoral Th17 cells are still unclear. We have been investigating the mechanism of induction and role of Th17 cells in malignant gliomas using primary tumor as well as cell lines. We report here that: (1) a higher frequency of Th17 cells in gliomas were associated with higher number of myeloid (CD11b) cells as well as the expression of TGF-β1 or IL-6; (2) conditioned medium from glioma cells (Gl CM) induced Th17 cell differentiation, which was inhibited by anti-TGF-β1 and anti-IL-6; (3) glioma-associated monocytes secreted Th17-promoting cytokines IL-1β and IL-23; (4) CM from glioma and monocyte co-culture (Gl+Mo CM) induced high frequency of Th17 cells in naïve T cell culture, which was abrogated by anti-IL-1β and anti-IL-23 antibodies; (5) In vitro Gl+Mo CM-mediated Th17 generation was associated with a decrease in IFN-γ and a concomitant increase in IL-10 secretion. Anti-TGF-β1, but not anti-IL-6, significantly reversed this cytokine profile. These results demonstrate prevalence of Th17 cells in gliomas and implicate the cytokines derived from the tumor as well as infiltrating myeloid cells in the induction of Th17 cells in glioma microenvironment. Moreover, the data also suggest that glioma-associated Th17 cells may contribute to immune-suppression via TGF-β1-induced IL-10 secretion. Further studies on the mechanism of tumor-infiltration, developmental pathways, and pro-/anti-tumor functions of Th17 cells will provide rationale for developing novel adjuvant immunotherapeutic strategies for malignant gliomas.
Collapse
Affiliation(s)
- Manjeera Paladugu
- Department of Neurosurgery, Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201, USA
| | | | | | | | | |
Collapse
|
35
|
Alpayci M, Bozan N, Erdem S, Gunes M, Erden M. The possible underlying pathophysiological mechanisms for development of multiple sclerosis in familial Mediterranean fever. Med Hypotheses 2012; 78:717-20. [DOI: 10.1016/j.mehy.2012.02.017] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2011] [Revised: 02/09/2012] [Accepted: 02/09/2012] [Indexed: 10/28/2022]
|
36
|
Katz Sand IB, Krieger S. Emerging strategies for the treatment of multiple sclerosis. FUTURE NEUROLOGY 2012. [DOI: 10.2217/fnl.12.9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Despite extraordinary advances in the field of neuroimmunology, ideal treatment for patients with multiple sclerosis remains an unmet need. Existing treatments are only partially effective in preventing multiple sclerosis relapses, have a limited impact on the accrual of disability, have not been effective in progressive forms of the disease, and treatment remains preventive rather than restorative. This review provides an overview of emerging therapies and targets, and incorporates strategies for two different approaches to multiple sclerosis: prevention, through immune modulation; and repair, through neuroprotection and remyelination. Agents at all stages of development, from late-stage clinical trials of BG-12, teriflunomide, alemtuzumab, daclizumab and anti-CD20 agents, to novel approaches in preclinical testing, are discussed.
Collapse
Affiliation(s)
- Ilana B Katz Sand
- Corinne Goldsmith Dickinson Center for Multiple Sclerosis, Mount Sinai School of Medicine, 5 East 98th Street, Box 1138, New York, NY, 10029, USA
| | - Stephen Krieger
- Corinne Goldsmith Dickinson Center for Multiple Sclerosis, Mount Sinai School of Medicine, 5 East 98th Street, Box 1138, New York, NY, 10029, USA
| |
Collapse
|
37
|
Th17 cell cytokine secretion profile in host defense and autoimmunity. Inflamm Res 2011; 61:87-96. [DOI: 10.1007/s00011-011-0419-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2011] [Revised: 11/10/2011] [Accepted: 12/10/2011] [Indexed: 01/13/2023] Open
|