1
|
Reddy-Vari H, Kim Y, Rajput C, Sajjan US. Increased expression of miR146a dysregulates TLR2-induced HBD2 in airway epithelial cells from patients with COPD. ERJ Open Res 2023; 9:00694-2022. [PMID: 37228294 PMCID: PMC10204848 DOI: 10.1183/23120541.00694-2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 02/23/2023] [Indexed: 05/27/2023] Open
Abstract
Background Airway epithelial cells from patients with COPD show suboptimal innate immune responses to nontypeable Haemophilus influenzae (NTHi) and Toll-like receptor (TLR)2 ligands despite expressing TLR2 similar to normal airway epithelial cells, but the underlying mechanisms are poorly understood. Methods Normal or COPD mucociliary-differentiated airway epithelial cells were treated with TLR2 agonists or infected with NTHi and expression of β-defensin (HBD)2 was examined. Interleukin-1 receptor-associated kinase (IRAK)-1 and microRNA (miR)146a were genetically inhibited in normal and COPD airway epithelial cell cultures, respectively, and HBD2 responses to TLR2 ligands were determined. IRAK-1 expression in lung sections was determined by immunofluorescence microscopy. Results Compared to normal, COPD airway epithelial cell cultures showed impaired expression of HBD2 in response to TLR2 agonists or NTHi infection. Apical secretions from TLR2 agonist-treated normal, but not COPD, airway epithelial cells efficiently killed NTHi. Knockdown of HBD2 significantly reduced NTHi killing by apical secretions of normal airway epithelial cells. Compared to normal, COPD cells showed significantly reduced expression of IRAK-1 and this was associated with increased expression of miR146a. Inhibition of miR146a increased the expression of IRAK-1, improved the expression of HBD2 in response to TLR2 agonists in COPD cells and enhanced the killing of bacteria by apical secretions obtained from TLR2 agonist-treated COPD cells. Bronchial epithelium of COPD patients showed reduced expression of IRAK-1. Conclusions These results suggest that reduced levels of IRAK-1 due to increased expression of miR146a may contribute to impaired expression of TLR2-induced HBD2 in COPD airway epithelial cells.
Collapse
Affiliation(s)
- Hymavathi Reddy-Vari
- Department of Microbiology Immunology and Inflammation, Lewis Katz Medical School, Temple University, Philadelphia, PA, USA
| | - Yerin Kim
- Department of Microbiology Immunology and Inflammation, Lewis Katz Medical School, Temple University, Philadelphia, PA, USA
| | - Charu Rajput
- Department of Microbiology Immunology and Inflammation, Lewis Katz Medical School, Temple University, Philadelphia, PA, USA
| | - Umadevi S. Sajjan
- Department of Microbiology Immunology and Inflammation, Lewis Katz Medical School, Temple University, Philadelphia, PA, USA
- Department of Thoracic Surgery and Medicine, Lewis Katz Medical School, Temple University, Philadelphia, PA, USA
| |
Collapse
|
2
|
Campbell GR, Rawat P, Teodorof-Diedrich C, Spector SA. IRAK1 inhibition blocks the HIV-1 RNA mediated pro-inflammatory cytokine response from microglia. J Gen Virol 2023; 104:001858. [PMID: 37256770 PMCID: PMC10336426 DOI: 10.1099/jgv.0.001858] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 05/10/2023] [Indexed: 06/02/2023] Open
Abstract
Human immunodeficiency virus (HIV)-associated neurocognitive disorders (HAND) are a common source of morbidity in people living with HIV (PLWH). Although antiretroviral therapy (ART) has lessened the severity of neurocognitive disorders, cognitive impairment still occurs in PLWH receiving ART. The pathogenesis of HAND is likely multifaceted, but common factors include the persistence of HIV transcription within the central nervous system, higher levels of pro-inflammatory cytokines in the cerebrospinal fluid, and the presence of activated microglia. Toll-like receptor (TLR) 7 and TLR8 are innate pathogen recognition receptors located in microglia and other immune and non-immune cells that can recognise HIV RNA and trigger pro-inflammatory responses. IL-1 receptor-associated kinase (IRAK) 1 is key to these signalling pathways. Here, we show that IRAK1 inhibition inhibits the TLR7 and TLR8-dependent pro-inflammatory response to HIV RNA. Using genetic and pharmacological inhibition, we demonstrate that inhibition of IRAK1 prevents IRAK1 phosphorylation and ubiquitination, and the subsequent recruitment of TRAF6 and the TAK1 complex to IRAK1, resulting in the inhibition of downstream signalling and the suppression of pro-inflammatory cytokine and chemokine release.
Collapse
Affiliation(s)
- Grant R. Campbell
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, USA
| | - Pratima Rawat
- Division of Infectious Diseases, Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
- Present address: Microbiologics Inc, San Diego, CA, USA
| | - Carmen Teodorof-Diedrich
- Division of Infectious Diseases, Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
| | - Stephen A. Spector
- Division of Infectious Diseases, Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
- Rady Children’s Hospital, San Diego, CA, USA
| |
Collapse
|
3
|
Sun H, Wang K, Yao W, Liu J, Lv L, Shi X, Chen H. Inter-Fighting between Influenza A Virus NS1 and β-TrCP: A Novel Mechanism of Anti-Influenza Virus. Viruses 2022; 14:v14112426. [PMID: 36366524 PMCID: PMC9699209 DOI: 10.3390/v14112426] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 10/29/2022] [Indexed: 11/06/2022] Open
Abstract
Influenza A virus (IAV) prevents innate immune signaling during infection. In our previous study, the production of pro-inflammatory cytokines was associated with Cullin-1 RING ligase (CRL1), which was related to NF-κB activation. However, the underlying mechanism is unclear. Here, an E3 ligase, β-transducin repeat-containing protein (β-TrCP), was significantly downregulated during IAV infection. Co-IP analysis revealed that non-structural 1 protein (NS1) interacts with β-TrCP. With co-transfection, an increase in NS1 expression led to a reduction in β-TrCP expression, affecting the level of IκBα and then resulting in repression of the activation of the NF-κB pathway during IAV infection. In addition, β-TrCP targets the viral NS1 protein and significantly reduces the replication level of influenza virus. Our results provide a novel mechanism for influenza to modulate its immune response during infection, and β-TrCP may be a novel target for influenza virus antagonism.
Collapse
Affiliation(s)
- Haiwei Sun
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
- Biosafety Research Center, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
- Correspondence: (H.S.); (H.C.)
| | - Kai Wang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Wei Yao
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Jingyi Liu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
- Biosafety Research Center, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Lu Lv
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
- Biosafety Research Center, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Xinjin Shi
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
- Biosafety Research Center, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Hongjun Chen
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
- Biosafety Research Center, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
- Correspondence: (H.S.); (H.C.)
| |
Collapse
|
4
|
Zhou X, Zhang Z, Xu H, Zhu B, Zhang L, Lie L, Huang Y, Du X, Liu H, Li Y, Huang Y, Hu S, Zhou C, Wen Q, Pepplenbosch MP, Ma L. Viperin impairs the innate immune response through the IRAK1-TRAF6-TAK1 axis to promote Mtb infection. Sci Signal 2022; 15:eabe1621. [PMID: 36194648 DOI: 10.1126/scisignal.abe1621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Mycobacterium tuberculosis (Mtb) infection is a long-standing public health threat, and the development of host-directed therapy for eradicating Mtb infection requires better insights into Mtb-host interactions. Viperin [virus-inhibitory protein, endoplasmic reticulum-associated, interferon (IFN) inducible] is an IFN-inducible protein with broad antiviral activities. Here, we demonstrated that Viperin was increased in abundance in patients with lymphatic and pulmonary tuberculosis (TB). Viperin-deficient mice had decreased Mtb bacterial loads and enhanced macrophage responses compared with their wild-type counterparts. Viperin suppressed the formation of a complex containing interleukin-1 receptor-associated kinase 1, TNF receptor-associated factor 6, and transforming growth factor β-activated kinase 1 (TAK1) and inhibited the TAK1-dependent activation of IκB kinase α/β, thereby impairing the production of nitric oxide and proinflammatory cytokines. These results suggest that Viperin promotes Mtb infection by inhibiting host innate immune responses in macrophages, suggesting that Viperin may be a candidate target for adjunct host-directed therapy in patients with TB.
Collapse
Affiliation(s)
- Xinying Zhou
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510515, China
| | - Zelin Zhang
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510515, China
| | - Hui Xu
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510515, China
| | - Bo Zhu
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510515, China
| | - Lijie Zhang
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510515, China
| | - Linmiao Lie
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510515, China
| | - Yingqi Huang
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510515, China
| | - Xialin Du
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510515, China
| | - Honglin Liu
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510515, China
| | - Yanfen Li
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510515, China
| | - Yulan Huang
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510515, China
| | - Shengfeng Hu
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510515, China
| | - Chaoying Zhou
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510515, China
| | - Qian Wen
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510515, China
| | - Mailkel P Pepplenbosch
- Department of Gastroenterology and Hepatology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Li Ma
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
5
|
Minimal structure of IRAK-1 to induce degradation of TRAF6. Immunobiology 2022; 227:152256. [DOI: 10.1016/j.imbio.2022.152256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 06/30/2022] [Accepted: 07/28/2022] [Indexed: 11/20/2022]
|
6
|
Hoyler T, Bannert B, André C, Beck D, Boulay T, Buffet D, Caesar N, Calzascia T, Dawson J, Kyburz D, Hennze R, Huppertz C, Littlewood-Evans A, Loetscher P, Mertz KD, Niwa S, Robert G, Rush JS, Ruzzante G, Sarret S, Stein T, Touil I, Wieczorek G, Zipfel G, Hawtin S, Junt T. Nonhematopoietic IRAK1 drives arthritis via neutrophil chemoattractants. JCI Insight 2022; 7:149825. [PMID: 35801586 PMCID: PMC9310529 DOI: 10.1172/jci.insight.149825] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 06/03/2022] [Indexed: 11/17/2022] Open
Abstract
IL-1 receptor-activated kinase 1 (IRAK1) is involved in signal transduction downstream of many TLRs and the IL-1R. Its potential as a drug target for chronic inflammatory diseases is underappreciated. To study its functional role in joint inflammation, we generated a mouse model expressing a functionally inactive IRAK1 (IRAK1 kinase deficient, IRAK1KD), which also displayed reduced IRAK1 protein expression and cell type–specific deficiencies of TLR signaling. The serum transfer model of arthritis revealed a potentially novel role of IRAK1 for disease development and neutrophil chemoattraction exclusively via its activity in nonhematopoietic cells. Consistently, IRAK1KD synovial fibroblasts showed reduced secretion of neutrophil chemoattractant chemokines following stimulation with IL-1β or human synovial fluids from patients with rheumatoid arthritis (RA) and gout. Together with patients with RA showing prominent IRAK1 expression in fibroblasts of the synovial lining, these data suggest that targeting IRAK1 may be therapeutically beneficial. As pharmacological inhibition of IRAK1 kinase activity had only mild effects on synovial fibroblasts from mice and patients with RA, targeted degradation of IRAK1 may be the preferred pharmacologic modality. Collectively, these data position IRAK1 as a central regulator of the IL-1β–dependent local inflammatory milieu of the joints and a potential therapeutic target for inflammatory arthritis.
Collapse
Affiliation(s)
- Thomas Hoyler
- Department of Autoimmunity Transplantation and Inflammation, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Bettina Bannert
- Department of Rheumatology, University Hospital Basel, Basel, Switzerland
| | - Cédric André
- Department of Autoimmunity Transplantation and Inflammation, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Damian Beck
- Department of Autoimmunity Transplantation and Inflammation, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Thomas Boulay
- Department of Autoimmunity Transplantation and Inflammation, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - David Buffet
- Department of Autoimmunity Transplantation and Inflammation, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Nadja Caesar
- Department of Autoimmunity Transplantation and Inflammation, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Thomas Calzascia
- Department of Autoimmunity Transplantation and Inflammation, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Janet Dawson
- Department of Autoimmunity Transplantation and Inflammation, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Diego Kyburz
- Department of Rheumatology, University Hospital Basel, Basel, Switzerland
| | - Robert Hennze
- Department of Autoimmunity Transplantation and Inflammation, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Christine Huppertz
- Department of Autoimmunity Transplantation and Inflammation, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Amanda Littlewood-Evans
- Department of Autoimmunity Transplantation and Inflammation, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Pius Loetscher
- Department of Autoimmunity Transplantation and Inflammation, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Kirsten D Mertz
- Institute of Pathology, Cantonal Hospital Baselland, Liestal, Switzerland
| | - Satoru Niwa
- Department of Autoimmunity Transplantation and Inflammation, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Gautier Robert
- Department of Autoimmunity Transplantation and Inflammation, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - James S Rush
- Department of Autoimmunity Transplantation and Inflammation, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Giulia Ruzzante
- Department of Autoimmunity Transplantation and Inflammation, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Sophie Sarret
- Department of Autoimmunity Transplantation and Inflammation, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Thomas Stein
- Department of Autoimmunity Transplantation and Inflammation, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Ismahane Touil
- Department of Autoimmunity Transplantation and Inflammation, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Grazyna Wieczorek
- Department of Autoimmunity Transplantation and Inflammation, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Geraldine Zipfel
- Department of Autoimmunity Transplantation and Inflammation, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Stuart Hawtin
- Department of Autoimmunity Transplantation and Inflammation, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Tobias Junt
- Department of Autoimmunity Transplantation and Inflammation, Novartis Institutes for BioMedical Research, Basel, Switzerland
| |
Collapse
|
7
|
Campbell GR, Rawat P, Spector SA. Pacritinib Inhibition of IRAK1 Blocks Aberrant TLR8 Signalling by SARS-CoV-2 and HIV-1-Derived RNA. J Innate Immun 2022; 15:96-106. [PMID: 35785771 PMCID: PMC10643889 DOI: 10.1159/000525292] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 05/27/2022] [Indexed: 11/19/2022] Open
Abstract
Macrophages promote an early host response to infection by releasing pro-inflammatory cytokines such as interleukin (IL) 1β (IL-1β), tumour necrosis factor (TNF), and IL-6. One of the mechanisms through which cells sense pathogenic microorganisms is through Toll-like receptors (TLRs). IL-1 receptor-associated kinase (IRAK) 1, IRAK2, IRAK3, and IRAK4 are integral to TLR and IL-1 receptor signalling pathways. Recent studies suggest a role for aberrant TLR8 and NLRP3 inflammasome activation during both COVID-19 and HIV-1 infection. Here, we show that pacritinib inhibits the TLR8-dependent pro-inflammatory cytokine response elicited by GU-rich single-stranded RNA derived from SARS-CoV-2 and HIV-1. Using genetic and pharmacologic inhibition, we demonstrate that pacritinib inhibits IRAK1 phosphorylation and ubiquitination which then inhibits the recruitment of the TAK1 complex to IRAK1, thus inhibiting the activation of downstream signalling and the production of pro-inflammatory cytokines.
Collapse
Affiliation(s)
- Grant R. Campbell
- Division of Infectious Diseases, Department of Pediatrics, University of California San Diego, La Jolla, California, USA
| | - Pratima Rawat
- Division of Infectious Diseases, Department of Pediatrics, University of California San Diego, La Jolla, California, USA
| | - Stephen A. Spector
- Division of Infectious Diseases, Department of Pediatrics, University of California San Diego, La Jolla, California, USA
- Rady Children's Hospital, San Diego, California, USA
| |
Collapse
|
8
|
Fan W, Liu X, Zhang J, Qin L, Du J, Li X, Qian S, Chen H, Qian P. TRIM67 Suppresses TNFalpha-Triggered NF-kB Activation by Competitively Binding Beta-TrCP to IkBa. Front Immunol 2022; 13:793147. [PMID: 35273593 PMCID: PMC8901487 DOI: 10.3389/fimmu.2022.793147] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 01/31/2022] [Indexed: 12/22/2022] Open
Abstract
The transcription factor NF-κB plays an important role in modulation of inflammatory pathways, which are associated with inflammatory diseases, neurodegeneration, apoptosis, immune responses, and cancer. Increasing evidence indicates that TRIM proteins are crucial role in the regulation of NF-κB signaling pathways. In this study, we identified TRIM67 as a negative regulator of TNFα-triggered NF-κB activation. Ectopic expression of TRIM67 significantly represses TNFα-induced NF-κB activation and the expression of pro-inflammatory cytokines TNFα and IL-6. In contrast, Trim67 depletion promotes TNFα-induced expression of TNFα, IL-6, and Mcp-1 in primary mouse embryonic fibroblasts. Mechanistically, we found that TRIM67 competitively binding β-transducin repeat-containing protein (β-TrCP) to IκBα results inhibition of β-TrCP-mediated degradation of IκBα, which finally caused inhibition of TNFα-triggered NF-κB activation. In summary, our findings revealed that TRIM67 function as a novel negative regulator of NF-κB signaling pathway, implying TRIM67 might exert an important role in regulation of inflammation disease and pathogen infection caused inflammation.
Collapse
Affiliation(s)
- Wenchun Fan
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China.,Division of Animal Infectious Diseases, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Xueyan Liu
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China.,Division of Animal Infectious Diseases, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Jinyan Zhang
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China.,Division of Animal Infectious Diseases, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Liuxing Qin
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China.,Division of Animal Infectious Diseases, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Jian Du
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China.,Division of Animal Infectious Diseases, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Xiangmin Li
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China.,Division of Animal Infectious Diseases, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, China
| | - Suhong Qian
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China.,Division of Animal Infectious Diseases, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Huanchun Chen
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China.,Division of Animal Infectious Diseases, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, China
| | - Ping Qian
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China.,Division of Animal Infectious Diseases, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
9
|
Lee S, Ha J, Park J, Kang E, Jeon SH, Han SB, Ningsih S, Paik JH, Cho S. Antioxidant and Anti-Inflammatory Effects of Bischofia javanica (Blume) Leaf Methanol Extracts through the Regulation of Nrf2 and TAK1. Antioxidants (Basel) 2021; 10:antiox10081295. [PMID: 34439543 PMCID: PMC8389227 DOI: 10.3390/antiox10081295] [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: 07/15/2021] [Revised: 08/12/2021] [Accepted: 08/13/2021] [Indexed: 11/16/2022] Open
Abstract
Bischofia javanica (Blume) has been traditionally used to treat inflammatory diseases such as tonsillitis and ulcers throughout Asia, including China, Indonesia, and the Philippines: however, the molecular mechanisms by which B. javanica exerts its antioxidant and anti-inflammatory properties remain largely unknown. In this study, we analyzed the antioxidant and anti-inflammatory mechanisms of methanol extracts of B. javanica leaves (MBJ) in vitro and in vivo. MBJ decreased nitric oxide (NO) production and the expression of pro-inflammatory cytokines, including interleukin (IL)-1β, IL-6, and tumor necrosis factor-α, in lipopolysaccharide (LPS)-treated RAW 264.7 cells. The observed suppression of inflammatory responses by MBJ was correlated with an inhibition of the nuclear factor-κB (NF-κB) and the mitogen-activated protein kinase (MAPK) pathways. Additionally, MBJ induced nuclear translocation of the nuclear factor erythroid 2-related factor 2 (Nrf2), a transcription factor that upregulates the expression of anti-inflammatory and antioxidant genes. Furthermore, MBJ exhibited antioxidant and anti-inflammatory effects in an acute hepatitis mouse model. In conclusion, our results confirm the medicinal properties of B. javanica, and therefore MBJ could be applied to improve inflammatory and redox imbalances in different types of pathologies.
Collapse
Affiliation(s)
- Sewoong Lee
- Laboratory of Molecular and Pharmacological Cell Biology, College of Pharmacy, Chung-Ang University, Seoul 06974, Korea; (S.L.); (J.H.); (J.P.); (E.K.)
| | - Jain Ha
- Laboratory of Molecular and Pharmacological Cell Biology, College of Pharmacy, Chung-Ang University, Seoul 06974, Korea; (S.L.); (J.H.); (J.P.); (E.K.)
| | - Jiyoung Park
- Laboratory of Molecular and Pharmacological Cell Biology, College of Pharmacy, Chung-Ang University, Seoul 06974, Korea; (S.L.); (J.H.); (J.P.); (E.K.)
| | - Eunjeong Kang
- Laboratory of Molecular and Pharmacological Cell Biology, College of Pharmacy, Chung-Ang University, Seoul 06974, Korea; (S.L.); (J.H.); (J.P.); (E.K.)
| | - Sung-Hyun Jeon
- Biomedical Mass Spectrometry Lab, College of Pharmacy, Chung-Ang University, Seoul 06974, Korea; (S.-H.J.); (S.B.H.)
| | - Sang Beom Han
- Biomedical Mass Spectrometry Lab, College of Pharmacy, Chung-Ang University, Seoul 06974, Korea; (S.-H.J.); (S.B.H.)
| | - Sri Ningsih
- Center for Pharmaceutical and Medical Technology, Deputy for Agroindustrial Technology and Biotechnology, The Agency for the Assessment and Application of Technology (BPPT), Jl. Raya Puspiptek, Kota Tangerang Selatan 15310, Banten, Indonesia;
| | - Jin Hyub Paik
- International Biological Material Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Korea;
| | - Sayeon Cho
- Laboratory of Molecular and Pharmacological Cell Biology, College of Pharmacy, Chung-Ang University, Seoul 06974, Korea; (S.L.); (J.H.); (J.P.); (E.K.)
- Correspondence: ; Tel.: +82-2-820-5595; Fax: +82-2-816-7338
| |
Collapse
|
10
|
Chen Y, Sun D, Yang R, Lim J, Sondey C, Presland J, Rakhilina L, Addona G, Kariv I, Chen H. Establishing and Validating Cellular Functional Target Engagement Assay for Selective IRAK4 Inhibitor Discovery. SLAS DISCOVERY 2021; 26:1040-1054. [PMID: 34130529 DOI: 10.1177/24725552211021074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
One of the main reasons for the lack of drug efficacy in late-stage clinical trials is the lack of specific and selective target engagement. To increase the likelihood of success of new therapeutics, one approach is to conduct proximal target engagement testing during the early phases of preclinical drug discovery. To identify and optimize selective IRAK4 inhibitors, a kinase that has been implicated in multiple inflammatory and autoimmune diseases, we established an electrochemiluminescence (ECL)-based cellular endogenous IRAK1 activation assay as the most proximal functional evaluation of IRAK4 engagement to support structure-activity relationship (SAR) studies. Since IRAK1 activation is dependent on both the IRAK4 scaffolding function in Myddosome formation and IRAK4 kinase activity for signal transduction, this assay potentially captures inhibitors with different mechanisms of action. Data from this IRAK1 assay with compounds representing different structural classes showed statistically significant correlations when compared with results from both IRAK4 biochemical kinase activity and functional peripheral blood mononuclear cell (PBMC)-derived tumor necrosis factor α (TNFα) secretion assays, validating the biological relevancy of the IRAK1 target engagement as a biomarker of the IRAK4 activity. Plate uniformity and potency reproducibility evaluations demonstrated that this assay is amenable to high throughput. Using Bland-Altman assay agreement analysis, we demonstrated that incorporating such proximal pharmacological assessment of cellular target engagement to an in vitro screening funnel for SAR studies can prevent compound optimization toward off-target activity.
Collapse
Affiliation(s)
- Yiping Chen
- Department of Quantitative Bioscience, Merck & Co., Inc., Boston, MA, USA
| | - Dongyu Sun
- Department of Quantitative Bioscience, Merck & Co., Inc., Boston, MA, USA
| | - Ruojing Yang
- Department of Quantitative Bioscience, Merck & Co., Inc., Boston, MA, USA
| | - Jongwon Lim
- Department of Discovery Chemistry, Merck & Co., Inc., Boston, MA, USA
| | - Christopher Sondey
- Department of Quantitative Bioscience, Merck & Co., Inc., Boston, MA, USA
| | - Jeremy Presland
- Department of Quantitative Bioscience, Merck & Co., Inc., Boston, MA, USA
| | - Larissa Rakhilina
- Department of Quantitative Bioscience, Merck & Co., Inc., Boston, MA, USA
| | - George Addona
- Department of Quantitative Bioscience, Merck & Co., Inc., Boston, MA, USA
| | - Ilona Kariv
- Department of Quantitative Bioscience, Merck & Co., Inc., Boston, MA, USA
| | - Hongmin Chen
- Department of Quantitative Bioscience, Merck & Co., Inc., Boston, MA, USA
| |
Collapse
|
11
|
Mulas F, Wang X, Song S, Nishanth G, Yi W, Brunn A, Larsen PK, Isermann B, Kalinke U, Barragan A, Naumann M, Deckert M, Schlüter D. The deubiquitinase OTUB1 augments NF-κB-dependent immune responses in dendritic cells in infection and inflammation by stabilizing UBC13. Cell Mol Immunol 2021; 18:1512-1527. [PMID: 32024978 PMCID: PMC8167118 DOI: 10.1038/s41423-020-0362-6] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 01/01/2020] [Indexed: 01/09/2023] Open
Abstract
Dendritic cells (DCs) are indispensable for defense against pathogens but may also contribute to immunopathology. Activation of DCs upon the sensing of pathogens by Toll-like receptors (TLRs) is largely mediated by pattern recognition receptor/nuclear factor-κB (NF-κB) signaling and depends on the appropriate ubiquitination of the respective signaling molecules. However, the ubiquitinating and deubiquitinating enzymes involved and their interactions are only incompletely understood. Here, we reveal that the deubiquitinase OTU domain, ubiquitin aldehyde binding 1 (OTUB1) is upregulated in DCs upon murine Toxoplasma gondii infection and lipopolysaccharide challenge. Stimulation of DCs with the TLR11/12 ligand T. gondii profilin and the TLR4 ligand lipopolysaccharide induced an increase in NF-κB activation in OTUB1-competent cells, resulting in elevated interleukin-6 (IL-6), IL-12, and tumor necrosis factor (TNF) production, which was also observed upon the specific stimulation of TLR2, TLR3, TLR7, and TLR9. Mechanistically, OTUB1 promoted NF-κB activity in DCs by K48-linked deubiquitination and stabilization of the E2-conjugating enzyme UBC13, resulting in increased K63-linked ubiquitination of IRAK1 (IL-1 receptor-associated kinase 1) and TRAF6 (TNF receptor-associated factor 6). Consequently, DC-specific deletion of OTUB1 impaired the production of cytokines, in particular IL-12, by DCs over the first 2 days of T. gondii infection, resulting in the diminished production of protective interferon-γ (IFN-γ) by natural killer cells, impaired control of parasite replication, and, finally, death from chronic T. encephalitis, all of which could be prevented by low-dose IL-12 treatment in the first 3 days of infection. In contrast, impaired OTUB1-deficient DC activation and cytokine production by OTUB1-deficient DCs protected mice from lipopolysaccharide-induced immunopathology. Collectively, these findings identify OTUB1 as a potent novel regulator of DCs during infectious and inflammatory diseases.
Collapse
Affiliation(s)
- Floriana Mulas
- Institute of Medical Microbiology and Hospital Hygiene, Otto-von-Guericke University Magdeburg, 39120, Magdeburg, Germany
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School, 30625, Hannover, Germany
| | - Xu Wang
- Institute of Medical Microbiology and Hospital Hygiene, Otto-von-Guericke University Magdeburg, 39120, Magdeburg, Germany.
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School, 30625, Hannover, Germany.
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, 325035, Wenzhou, China.
| | - Shanshan Song
- Institute of Medical Microbiology and Hospital Hygiene, Otto-von-Guericke University Magdeburg, 39120, Magdeburg, Germany
| | - Gopala Nishanth
- Institute of Medical Microbiology and Hospital Hygiene, Otto-von-Guericke University Magdeburg, 39120, Magdeburg, Germany
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School, 30625, Hannover, Germany
| | - Wenjing Yi
- Institute of Medical Microbiology and Hospital Hygiene, Otto-von-Guericke University Magdeburg, 39120, Magdeburg, Germany
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School, 30625, Hannover, Germany
| | - Anna Brunn
- Department of Neuropathology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931, Cologne, Germany
| | - Pia-Katharina Larsen
- Institute for Experimental Infection Research, TWINCORE Centre for Experimental and Clinical Infection Research, a joint venture between the Hannover Medical School and the Helmholtz Centre for Infection Research, 30625, Hannover, Germany
| | - Berend Isermann
- Institute for Clinical Chemistry and Pathobiochemistry, Otto-von-Guericke University Magdeburg, 39120, Magdeburg, Germany
| | - Ulrich Kalinke
- Institute for Experimental Infection Research, TWINCORE Centre for Experimental and Clinical Infection Research, a joint venture between the Hannover Medical School and the Helmholtz Centre for Infection Research, 30625, Hannover, Germany
- Cluster of Excellence-Resolving Infection Susceptibility (RESIST), Hannover Medical School, 30625, Hannover, Germany
| | - Antonio Barragan
- Department of Molecular Biosciences, Stockholm University, 10691, Stockholm, Sweden
| | - Michael Naumann
- Institute for Experimental Internal Medicine, Otto-von-Guericke University Magdeburg, 39120, Magdeburg, Germany
| | - Martina Deckert
- Department of Neuropathology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931, Cologne, Germany
| | - Dirk Schlüter
- Institute of Medical Microbiology and Hospital Hygiene, Otto-von-Guericke University Magdeburg, 39120, Magdeburg, Germany.
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School, 30625, Hannover, Germany.
- Cluster of Excellence-Resolving Infection Susceptibility (RESIST), Hannover Medical School, 30625, Hannover, Germany.
| |
Collapse
|
12
|
Sokolova O, Naumann M. Manifold role of ubiquitin in Helicobacter pylori infection and gastric cancer. Cell Mol Life Sci 2021; 78:4765-4783. [PMID: 33825941 PMCID: PMC8195768 DOI: 10.1007/s00018-021-03816-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 02/22/2021] [Accepted: 03/18/2021] [Indexed: 02/07/2023]
Abstract
Infection with H. pylori induces a strong host cellular response represented by induction of a set of molecular signaling pathways, expression of proinflammatory cytokines and changes in proliferation. Chronic infection and inflammation accompanied by secretory dysfunction can result in the development of gastric metaplasia and gastric cancer. Currently, it has been determined that the regulation of many cellular processes involves ubiquitinylation of molecular effectors. The binding of ubiquitin allows the substrate to undergo a change in function, to interact within multimolecular signaling complexes and/or to be degraded. Dysregulation of the ubiquitinylation machinery contributes to several pathologies, including cancer. It is not understood in detail how H. pylori impacts the ubiquitinylation of host substrate proteins. The aim of this review is to summarize the existing literature in this field, with an emphasis on the role of E3 ubiquitin ligases in host cell homeodynamics, gastric pathophysiology and gastric cancer.
Collapse
Affiliation(s)
- Olga Sokolova
- Medical Faculty, Otto Von Guericke University, Institute of Experimental Internal Medicine, 39120 Magdeburg, Germany
| | - Michael Naumann
- Medical Faculty, Otto Von Guericke University, Institute of Experimental Internal Medicine, 39120 Magdeburg, Germany
| |
Collapse
|
13
|
Kim H, Yang WS, Htwe KM, Lee MN, Kim YD, Yoon KD, Lee BH, Lee S, Cho JY. Dipterocarpus tuberculatus Roxb. Ethanol Extract Has Anti-Inflammatory and Hepatoprotective Effects In Vitro and In Vivo by Targeting the IRAK1/AP-1 Pathway. Molecules 2021; 26:molecules26092529. [PMID: 33926126 PMCID: PMC8123704 DOI: 10.3390/molecules26092529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 04/22/2021] [Accepted: 04/23/2021] [Indexed: 11/28/2022] Open
Abstract
Dipterocarpus tuberculatus Roxb. has been used traditionally as a remedy for many diseases, especially inflammation. Therefore, we analyzed and explored the mechanism of the anti-inflammatory effect of a Dipterocarpus tuberculatus Roxb. ethanol extract (Dt-EE). Dt-EE clearly and dose-dependently inhibited the expression of pro-inflammatory cytokines such as IL-6, TNF-α, and IL-1β in lipopolysaccharide (LPS)-treated RAW264.7 cells. Also, Dt-EE suppressed the activation of the MyD88/TRIF-mediated AP-1 pathway and the AP-1 pathway related proteins JNK2, MKK4/7, and TAK1, which occurred as a result of inhibiting the kinase activity of IRAK1 and IRAK4, the most upstream factors of the AP-1 pathway. Finally, Dt-EE displayed hepatoprotective activity in a mouse model of hepatitis induced with LPS/D-galactosamine (D-GalN) through decreasing the serum levels of alanine aminotransferase and suppressing the activation of JNK and IRAK1. Therefore, our results strongly suggest that Dt-EE could be a candidate anti-inflammatory herbal medicine with IRAK1/AP-1 inhibitory and hepatoprotective properties.
Collapse
Affiliation(s)
- Haeyeop Kim
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Korea; (H.K.); (W.S.Y.)
| | - Woo Seok Yang
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Korea; (H.K.); (W.S.Y.)
| | - Khin Myo Htwe
- Popa Mountain Park, Forest Department, Kyaukpadaung Township, Mandalay Division, Kyaukpadaung 05241, Myanmar;
| | - Mi-Nam Lee
- Department of Hospitality and Culinary, Ansan University, Ansan 15318, Korea;
| | - Young-Dong Kim
- Department of Life Science, Hallym University, Chuncheon 200-702, Korea;
| | - Ki Dong Yoon
- College of Pharmacy, The Catholic University of Korea, Bucheon 420-743, Korea;
| | - Byoung-Hee Lee
- National Institute of Biological Resources, Environmental Research Complex, Incheon 22689, Korea;
| | - Sarah Lee
- National Institute of Biological Resources, Environmental Research Complex, Incheon 22689, Korea;
- Correspondence: (S.L.); (J.Y.C.); Tel.: +82-32-590-7265 (S.L.); +82-31-290-7868 (J.Y.C.); Fax: +82-32-590-7472 (S.L.); +82-31-290-7870 (J.Y.C.)
| | - Jae Youl Cho
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Korea; (H.K.); (W.S.Y.)
- Correspondence: (S.L.); (J.Y.C.); Tel.: +82-32-590-7265 (S.L.); +82-31-290-7868 (J.Y.C.); Fax: +82-32-590-7472 (S.L.); +82-31-290-7870 (J.Y.C.)
| |
Collapse
|
14
|
Wang H, Zhou H, Zhang Q, Poulsen KL, Taylor V, McMullen MR, Czarnecki D, Dasarathy D, Yu M, Liao Y, Allende DS, Chen X, Hong L, Zhao J, Yang J, Nagy LE, Li X. Inhibition of IRAK4 kinase activity improves ethanol-induced liver injury in mice. J Hepatol 2020; 73:1470-1481. [PMID: 32682051 PMCID: PMC8007112 DOI: 10.1016/j.jhep.2020.07.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 07/03/2020] [Accepted: 07/07/2020] [Indexed: 02/08/2023]
Abstract
BACKGROUNDS & AIMS Alcohol-related liver disease (ALD) is a major cause of chronic liver disease worldwide with limited therapeutic options. Interleukin-1 receptor associated kinase 4 (IRAK4), the master kinase of Toll-like receptor (TLR)/IL-1R-mediated signalling activation, is considered a novel therapeutic target in inflammatory diseases, but has not been investigated in the context of ALD. METHODS IRAK4 phosphorylation and IRAK1 protein were analysed in liver from alcohol-related hepatitis patients and healthy controls. IRAK4 kinase activity-inactive knock-in (Irak4 KI) mice and bone marrow chimeric mice were exposed to chronic ethanol-induced liver injury. IL-1β-induced IRAK4-mediated signalling and acute phase response were investigated in cultured hepatocytes. IRAK1/4 inhibitor was used to test the therapeutic potential for ethanol-induced liver injury in mice. RESULTS Increased IRAK4 phosphorylation and reduced IRAK1 protein were found in livers of patients with alcoholic hepatitis. In the chronic ethanol-induced liver injury mouse model, hepatic inflammation and hepatocellular damage were attenuated in Irak4 KI mice. IRAK4 kinase activity promotes expression of acute phase proteins in response to ethanol exposure, including C-reactive protein and serum amyloid A1 (SAA1). SAA1 and IL-1β synergistically exacerbate ethanol-induced cell death ex vivo. Pharmacological blockage of IRAK4 kinase abrogated ethanol-induced liver injury, inflammation, steatosis, as well as acute phase gene expression and protein production in mice. CONCLUSIONS Our data elucidate the critical role of IRAK4 kinase activity in the pathogenesis of ethanol-induced liver injury in mice and provide preclinical validation for use of an IRAK1/4 inhibitor as a new potential therapeutic strategy for the treatment of ALD. LAY SUMMARY Herein, we have identified the role of IRAK4 kinase activity in the development of alcohol-induced liver injury in mice. Hepatocyte-specific IRAK4 is associated with an acute phase response and release of proinflammatory cytokines/chemokines, which synergistically exacerbate alcohol-induced hepatocyte cell death ex vivo. Pharmacological inhibition of IRAK4 kinase activity effectively attenuates alcohol-induced liver injury in mice and could have therapeutic implications.
Collapse
Affiliation(s)
- Han Wang
- School of Life Sciences, Lanzhou University, Lanzhou, Gansu 730000, China,Inflammation and Immunity Department, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA,Northern Ohio Alcohol Center, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Hao Zhou
- Inflammation and Immunity Department, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA,Northern Ohio Alcohol Center, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Quanri Zhang
- Inflammation and Immunity Department, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA,Northern Ohio Alcohol Center, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Kyle L. Poulsen
- Inflammation and Immunity Department, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA,Northern Ohio Alcohol Center, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Vanessa Taylor
- Rigel Pharmaceuticals, South San Francisco, CA 94080, USA
| | - Megan R. McMullen
- Inflammation and Immunity Department, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA,Northern Ohio Alcohol Center, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Doug Czarnecki
- Inflammation and Immunity Department, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA,Northern Ohio Alcohol Center, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Dhweeja Dasarathy
- Inflammation and Immunity Department, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA,Harvard University, Massachusetts Hall, Cambridge, MA 02138, USA
| | - Minjia Yu
- Inflammation and Immunity Department, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA,Department of Medicine, Mount Auburn Hospital, Harvard Medical School, Cambridge, MA, 02138, USA
| | - Yun Liao
- Inflammation and Immunity Department, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Daniela S. Allende
- Inflammation and Immunity Department, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA,Pathology Department, Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Xing Chen
- Inflammation and Immunity Department, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Lingzi Hong
- Inflammation and Immunity Department, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Junjie Zhao
- Inflammation and Immunity Department, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Jinbo Yang
- School of Life Sciences, Lanzhou University, Lanzhou, Gansu 730000, China.
| | - Laura E. Nagy
- Inflammation and Immunity Department, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA,Northern Ohio Alcohol Center, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Xiaoxia Li
- Inflammation and Immunity Department, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; Northern Ohio Alcohol Center, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA.
| |
Collapse
|
15
|
Bi Y, Cui D, Xiong X, Zhao Y. The characteristics and roles of β-TrCP1/2 in carcinogenesis. FEBS J 2020; 288:3351-3374. [PMID: 33021036 DOI: 10.1111/febs.15585] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 09/02/2020] [Accepted: 10/01/2020] [Indexed: 12/11/2022]
Abstract
β-transducin repeat-containing protein (β-TrCP), one of the well-characterized F-box proteins, acts as a substrate receptor and constitutes an active SCFβ-TrCP E3 ligase with a scaffold protein CUL1, a RING protein RBX1, and an adaptor protein SKP1. β-TrCP plays a critical role in the regulation of various physiological and pathological processes, including signal transduction, cell cycle progression, cell migration, DNA damage response, and tumorigenesis, by governing burgeoning amounts of key regulators for ubiquitination and proteasomal degradation. Given that a variety of β-TrCP substrates are well-known oncoproteins and tumor suppressors, and dysregulation of β-TrCP is frequently identified in human cancers, β-TrCP plays a vital role in carcinogenesis. In this review, we first briefly introduce the characteristics of β-TrCP1, β-TrCP2, and SCFβ-TrCP ubiquitin ligase, and then discuss SCFβ-TrCP ubiquitin ligase regulated biological processes by targeting its substrates for degradation. Moreover, we summarize the regulation of β-TrCP1 and β-TrCP2 at multiple layers and further discuss the various roles of β-TrCP1 and β-TrCP2 in human cancer, functioning as either an oncoprotein or a tumor suppressor in a manner dependent of cellular context. Finally, we provide novel insights for future perspectives on the potential of targeting β-TrCP1 and β-TrCP2 for cancer therapy.
Collapse
Affiliation(s)
- Yanli Bi
- Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Danrui Cui
- Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiufang Xiong
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China.,Cancer Institute of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yongchao Zhao
- Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
16
|
Morgan EL, Chen Z, Van Waes C. Regulation of NFκB Signalling by Ubiquitination: A Potential Therapeutic Target in Head and Neck Squamous Cell Carcinoma? Cancers (Basel) 2020; 12:E2877. [PMID: 33036368 PMCID: PMC7601648 DOI: 10.3390/cancers12102877] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 09/22/2020] [Accepted: 09/29/2020] [Indexed: 02/08/2023] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is the sixth most common cancer worldwide, with over 600,000 cases per year. The primary causes for HNSCC include smoking and alcohol consumption, with an increasing number of cases attributed to infection with Human Papillomavirus (HPV). The treatment options for HNSCC currently include surgery, radiotherapy, and/or platinum-based chemotherapeutics. Cetuximab (targeting EGFR) and Pembrolizumab (targeting PD-1) have been approved for advanced stage, recurrent, and/or metastatic HNSCC. Despite these advances, whilst HPV+ HNSCC has a 3-year overall survival (OS) rate of around 80%, the 3-year OS for HPV- HNSCC is still around 55%. Aberrant signal activation of transcription factor NFκB plays an important role in the pathogenesis and therapeutic resistance of HNSCC. As an important mediator of inflammatory signalling and the immune response to pathogens, the NFκB pathway is tightly regulated to prevent chronic inflammation, a key driver of tumorigenesis. Here, we discuss how NFκB signalling is regulated by the ubiquitin pathway and how this pathway is deregulated in HNSCC. Finally, we discuss the current strategies available to target the ubiquitin pathway and how this may offer a potential therapeutic benefit in HNSCC.
Collapse
Affiliation(s)
- Ethan L. Morgan
- Tumor Biology Section, Head and Neck Surgery Branch, National Institute of Deafness and Other Communication Disorders, NIH, Bethesda, MD 20892, USA;
| | - Zhong Chen
- Tumor Biology Section, Head and Neck Surgery Branch, National Institute of Deafness and Other Communication Disorders, NIH, Bethesda, MD 20892, USA;
| | | |
Collapse
|
17
|
Singh A, Devkar R, Basu A. Myeloid Differentiation Primary Response 88-Cyclin D1 Signaling in Breast Cancer Cells Regulates Toll-Like Receptor 3-Mediated Cell Proliferation. Front Oncol 2020; 10:1780. [PMID: 33072559 PMCID: PMC7531238 DOI: 10.3389/fonc.2020.01780] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Accepted: 08/11/2020] [Indexed: 11/13/2022] Open
Abstract
Toll-like receptor 3 (TLR3)-mediated apoptotic changes in cancer cells are well-documented, and hence, several synthetic ligands of TLR3 are being used for adjuvant therapy, but there are reports showing a contradictory effect of TLR3 signaling, which include our previous report that had shown cell proliferation following surface localization of TLR 3. However, the underlying mechanism of cell surface localization of TLR3 and subsequent cell proliferation lacks clarity. This study addresses the TLR3 ligand-mediated signaling cascade that regulates a proliferative effect in breast cancer cells (MDA-MB-231 and T47D) challenged with TLR3 ligand in the presence of myeloid differentiation primary response 88 (MyD88) inhibitor. Evidences were obtained using immunoblotting, coimmunoprecipitation, confocal microscopy, immunocytochemistry, ELISA, and flow cytometry. Results had revealed that TLR3 ligand treatment significantly enhanced breast cancer cell proliferation marked by an upregulated expression of cyclinD1, but the same was suppressed by the addition of MyD88 inhibitor. Also, expression of interleukin 1 receptor-associated kinase 1 (IRAK1)-TNF receptor-associated factor 6 (TRAF6)-transforming growth factor beta-activated kinase 1 (TAK1) was altered in the given TLR3-signaling pathway. Inhibition of MyD88 disrupted the downstream adaptor complex and mediated signaling through the TLR3-MyD88-NF-κB (p65)-IL-6-cyclin D1 pathway. TLR3-mediated alternative signaling of the TLR3-MyD88-IRAK1-TRAF6-TAK1-TAB1-NF-κB axis leads to upregulation of IL6 and cyclin D1. This response is hypothesized to be via the MyD88 gateway that culminates in the proliferation of breast cancer cells. Overall, this study provides first comprehensive evidence on the involvement of canonical signaling of TLR3 using MyD88-cyclin D1-mediated breast cancer cell proliferation. The findings elucidated herein will provide valuable insights into understanding the TLR3-mediated adjuvant therapy in cancer.
Collapse
Affiliation(s)
- Aradhana Singh
- Molecular Biology and Human Genetics Laboratory, Department of Zoology, The University of Burdwan, Bardhaman, India
| | - Ranjitsinh Devkar
- Department of Zoology, Faculty of Science, The M.S. University of Baroda, Vadodara, India
| | - Anupam Basu
- Molecular Biology and Human Genetics Laboratory, Department of Zoology, The University of Burdwan, Bardhaman, India
| |
Collapse
|
18
|
Seltzer J, Moorad R, Schifano JM, Landis JT, Dittmer DP. Interleukin-1 Receptor-Associated Kinase (IRAK) Signaling in Kaposi Sarcoma-Associated Herpesvirus-Induced Primary Effusion Lymphoma. J Virol 2020; 94:e02123-19. [PMID: 32161170 PMCID: PMC7199399 DOI: 10.1128/jvi.02123-19] [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: 01/03/2020] [Accepted: 03/05/2020] [Indexed: 12/20/2022] Open
Abstract
Kaposi sarcoma-associated herpesvirus (KSHV) is necessary but not sufficient for primary effusion lymphoma (PEL) development. Alterations in cellular signaling pathways are also a characteristic of PEL. Other B cell lymphomas have acquired an oncogenic mutation in the myeloid differentiation primary response 88 (MYD88) gene. The MYD88 L265P mutant results in the activation of interleukin-1 receptor associated kinase (IRAK). To probe IRAK/MYD88 signaling in PEL, we employed CRISPR/Cas9 technology to generate stable deletion clones in BCBL-1Cas9 and BC-1Cas9 cells. To look for off-target effects, we determined the complete exome of the BCBL-1Cas9 and BC-1Cas9 cells. Deletion of either MYD88, IRAK4, or IRAK1 abolished interleukin-1 beta (IL-1β) signaling; however, we were able to grow stable subclones from each population. Transcriptome sequencing (RNA-seq) analysis of IRAK4 knockout cell lines (IRAK4 KOs) showed that the IRAK pathway induced cellular signals constitutively, independent of IL-1β stimulation, which was abrogated by deletion of IRAK4. Transient complementation with IRAK1 increased NF-κB activity in MYD88 KO, IRAK1 KO, and IRAK4 KO cells even in the absence of IL-1β. IL-10, a hallmark of PEL, was dependent on the IRAK pathway, as IRAK4 KOs showed reduced IL-10 levels. We surmise that, unlike B cell receptor (BCR) signaling, MYD88/IRAK signaling is constitutively active in PEL, but that under cell culture conditions, PEL rapidly became independent of this pathway.IMPORTANCE One hundred percent of primary effusion lymphoma (PEL) cases are associated with Kaposi sarcoma-associated herpesvirus (KSHV). PEL cell lines, such as BCBL-1, are the workhorse for understanding this human oncovirus and the host pathways that KSHV dysregulates. Understanding their function is important for developing new therapies as well as identifying high-risk patient groups. The myeloid differentiation primary response 88 (MYD88)/interleukin-1 receptor associated kinase (IRAK) pathway, which has progrowth functions in other B cell lymphomas, has not been fully explored in PEL. By performing CRISPR/Cas9 knockout (KO) studies targeting the IRAK pathway in PEL, we were able to determine that established PEL cell lines can circumvent the loss of IRAK1, IRAK4, and MYD88; however, the deletion clones are deficient in interleukin-10 (IL-10) production. Since IL-10 suppresses T cell function, this suggests that the IRAK pathway may serve a function in vivo and during early-stage development of PEL.
Collapse
Affiliation(s)
- Jedediah Seltzer
- Department of Microbiology and Immunology, Center for AIDS Research, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Razia Moorad
- Department of Microbiology and Immunology, Center for AIDS Research, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Jason M Schifano
- Department of Microbiology and Immunology, Center for AIDS Research, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Justin T Landis
- Department of Microbiology and Immunology, Center for AIDS Research, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Dirk P Dittmer
- Department of Microbiology and Immunology, Center for AIDS Research, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
19
|
Liu X, Jiao K, Jia CC, Li GX, Yuan Q, Xu JK, Hou Y, Wang B. BAP31 regulates IRAK1-dependent neuroinflammation in microglia. J Neuroinflammation 2019; 16:281. [PMID: 31883536 PMCID: PMC6935200 DOI: 10.1186/s12974-019-1661-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Accepted: 11/26/2019] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Microglia, the mononuclear immune cells of the central nervous system (CNS), are essential for the maintenance of CNS homeostasis. BAP31, a resident and ubiquitously expressed protein of the endoplasmic reticulum, serves as a sorting factor for its client proteins, mediating the subsequent export, retention, and degradation or survival. Recently, BAP31 has been defined as a regulatory molecule in the CNS, but the function of BAP31 in microglia has yet to be determined. In the present study, we investigated whether BAP31 is involved in the inflammatory response of microglia. METHODS This study used the BV2 cell line and BAP31 conditional knockdown mice generated via the Cre/LoxP system. A BAP31 knockdown experiment was performed to elucidate the role of BAP31 in the endogenous inflammatory cytokine production by microglial BV2 cells. A mouse model of lipopolysaccharide (LPS)-induced cognitive impairment was established to evaluate the neuroprotective effect of BAP31 against neuroinflammation-induced memory deficits. Behavioral alterations were assessed with the open field test (OFT), Y maze, and Morris water maze. The activation of microglia in the hippocampus of mice was observed by immunohistochemistry. Western blot, enzyme-linked immunosorbent assay (ELISA), immunofluorescence staining, and reverse transcription quantitative real-time polymerase chain reaction (RT-PCR) were used to clarify the mechanisms. RESULTS BAP31 deficiency upregulates LPS-induced proinflammatory cytokines in BV2 cells and mice by upregulating the protein level of IRAK1, which in turn increases the translocation and transcriptional activity of NF-κB p65 and c-Jun, and moreover, knockdown of IRAK1 or use of an IRAK1 inhibitor reverses these functions. In the cognitive impairment animal model, the BAP31 knockdown mice displayed increased severity in memory deficiency accompanied by an increased expression of proinflammatory factors in the hippocampus. CONCLUSIONS These findings indicate that BAP31 may modulate inflammatory cytokines and cognitive impairment induced by neuroinflammation through IRAK1, which demonstrates that BAP31 plays an essential role in microglial inflammation and prevention of memory deficits caused by neuroinflammation.
Collapse
Affiliation(s)
- Xia Liu
- College of Life and Health Science, Northeastern University, 195 Chuangxin Road, Hunnan District, Shenyang, Liaoning, 110819, People's Republic of China
| | - Kun Jiao
- College of Life and Health Science, Northeastern University, 195 Chuangxin Road, Hunnan District, Shenyang, Liaoning, 110819, People's Republic of China
| | - Cong-Cong Jia
- College of Life and Health Science, Northeastern University, 195 Chuangxin Road, Hunnan District, Shenyang, Liaoning, 110819, People's Republic of China
| | - Guo-Xun Li
- College of Life and Health Science, Northeastern University, 195 Chuangxin Road, Hunnan District, Shenyang, Liaoning, 110819, People's Republic of China
| | - Qing Yuan
- College of Life and Health Science, Northeastern University, 195 Chuangxin Road, Hunnan District, Shenyang, Liaoning, 110819, People's Republic of China
| | - Ji-Kai Xu
- College of Life and Health Science, Northeastern University, 195 Chuangxin Road, Hunnan District, Shenyang, Liaoning, 110819, People's Republic of China
| | - Yue Hou
- College of Life and Health Science, Northeastern University, 195 Chuangxin Road, Hunnan District, Shenyang, Liaoning, 110819, People's Republic of China.
| | - Bing Wang
- College of Life and Health Science, Northeastern University, 195 Chuangxin Road, Hunnan District, Shenyang, Liaoning, 110819, People's Republic of China.
| |
Collapse
|
20
|
Shin KK, Park JG, Hong YH, Aziz N, Park SH, Kim S, Kim E, Cho JY. Anti-Inflammatory Effects of Licania macrocarpa Cuatrec Methanol Extract Target Src- and TAK1-Mediated Pathways. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2019; 2019:4873870. [PMID: 31611922 PMCID: PMC6757254 DOI: 10.1155/2019/4873870] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2019] [Revised: 08/13/2019] [Accepted: 08/17/2019] [Indexed: 12/15/2022]
Abstract
In this study, we investigated the anti-inflammatory effects of Licania macrocarpa Cuatrec methanol extract (Lm-ME) in vitro and in vivo and found pharmacological target proteins of Lm-ME in TLR4-mediated inflammatory signaling. This extract reduced NO production and mRNA expression of inflammatory cytokines such as iNOS, COX-2, IL-6, and IL-1β. In the NF-κB- and AP-1-mediated luciferase reporter gene assay, transcription factor activities decreased under cotransfection with MyD88 or TRIF. Phosphorylated protein levels of Src, PI3K, IKKα/β, and IκBα as well as p50 and p65 in the NF-κB signal pathway were downregulated, and phosphorylation of TAK1, MEK1/2, MKK4/7, and MKK3/6 as well as ERK, JNK, and p38 was decreased in the AP-1 signal pathway. Through overexpression of HA-Src and HA-TAK1, respectively, Lm-ME inhibited autophosphorylation of overexpressed proteins and thereby activated fewer downstream signaling molecules. Lm-ME also attenuated stomach ulcers in an HCl/EtOH-induced acute gastritis model mice, and COX-2 mRNA expression and phosphorylated TAK1 levels in gastric tissues were diminished. The flavonoids kaempferol and quercetin were identified in the HPLC analysis of Lm-ME; both are actively anti-inflammatory. Therefore, these results suggest that Lm-ME can be used for anti-inflammatory remedy by targeting Src and TAK1.
Collapse
Affiliation(s)
- Kon Kuk Shin
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Jae Gwang Park
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Republic of Korea
- Division of Translational Science, Research Institute, National Cancer Center, Goyang 10408, Republic of Korea
| | - Yo Han Hong
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Nur Aziz
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Sang Hee Park
- Department of Biocosmetics, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Sunggyu Kim
- Department of Biocosmetics, Sungkyunkwan University, Suwon 16419, Republic of Korea
- Research and Business Foundation, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Eunji Kim
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Jae Youl Cho
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Republic of Korea
- Department of Biocosmetics, Sungkyunkwan University, Suwon 16419, Republic of Korea
| |
Collapse
|
21
|
Geranylgeraniol Suppresses the Expression of IRAK1 and TRAF6 to Inhibit NFκB Activation in Lipopolysaccharide-Induced Inflammatory Responses in Human Macrophage-Like Cells. Int J Mol Sci 2019; 20:ijms20092320. [PMID: 31083375 PMCID: PMC6540148 DOI: 10.3390/ijms20092320] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Revised: 05/06/2019] [Accepted: 05/08/2019] [Indexed: 12/15/2022] Open
Abstract
Geranylgeraniol (GGOH), a natural isoprenoid found in plants, has anti-inflammatory effects via inhibiting the activation of nuclear factor-kappa B (NFκB). However, its detailed mechanism has not yet been elucidated. Recent studies have revealed that isoprenoids can modulate signaling molecules in innate immune responses. We found that GGOH decreased the expression of lipopolysaccharide (LPS)-induced inflammatory genes in human macrophage-like THP-1 cells. Furthermore, we observed that the suppression of NFκB signaling proteins, in particular interleukin-1 receptor-associated kinase 1 (IRAK1) and tumor necrosis factor receptor-associated factor 6 (TRAF6), occurred in GGOH-treated cells prior to LPS stimulation, suggesting an immunomodulatory effect. These results indicate that GGOH may modulate and help prevent excessive NFκB activation that can lead to numerous diseases.
Collapse
|
22
|
Zhang Y, Guo H, Cheng BCY, Su T, Fu XQ, Li T, Zhu PL, Tse KW, Pan SY, Yu ZL. Dingchuan tang essential oil inhibits the production of inflammatory mediators via suppressing the IRAK/NF-κB, IRAK/AP-1, and TBK1/IRF3 pathways in lipopolysaccharide-stimulated RAW264.7 cells. DRUG DESIGN DEVELOPMENT AND THERAPY 2018; 12:2731-2748. [PMID: 30233137 PMCID: PMC6129014 DOI: 10.2147/dddt.s160645] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Background Dingchuan tang (asthma-relieving decoction), a formula of nine herbs, has been used for treating respiratory inflammatory diseases for >400 years in the People’s Republic of China. However, the mechanisms underlying the anti-inflammatory action of dingchuan tang is not fully understood. This study aims to investigate the effects of Dingchuan tang essential oil (DCEO) on inflammatory mediators and the underlying mechanism of action. Materials and methods DCEO was extracted by steam distillation. Lipopolysaccharide (LPS)-stimulated RAW264.7 macrophages were used as the cell model. Production of nitric oxide (NO) was determined by the Griess test. Protein secretion and mRNA levels of inflammatory mediators were measured by the enzyme-linked immunosorbent assay (ELISA) and quantitative real-time polymerase chain reaction (qRT-PCR), respectively. Protein levels were examined by Western blot. Nuclear localization of nuclear factor-kappa B (NF-κB) was detected using immunofluorescence analyses. Results DCEO significantly reduced LPS-triggered production of NO and prostaglandin E2 (PGE2) and decreased protein and mRNA levels of inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2). LPS induced upregulation of protein and mRNA levels of cytokines (interleukin-1β [IL-1β], interleukin-6 [IL-6], tumor necrosis factor-α [TNF-α]), and chemokines (monocyte chemoattractant protein-1 [MCP-1], chemokine [C-C motif] ligand 5 [CCL-5], and macrophage inflammatory protein [MIP]-1α) were suppressed by DCEO treatment. Phosphorylation and nuclear protein levels of transcription factors (activator protein-1 [AP-1], NF-κB, interferon regulatory factor 3 [IRF3]) were decreased by DCEO. Protein levels of phosphorylated IκB-α, IκB kinase α/β (IKKα/β), phosphatidylinositol 3-kinase (PI3K), protein kinase B (Akt), TGF β-activated kinase 1 (TAK1), TANK-binding kinase 1 (TBK1), extracellular signal-regulated kinase (ERK), p38 mitogen-activated protein kinase (p38), and c-Jun N-terminal kinase (JNK) were lowered by DCEO. Moreover, degradation of interleukin-1 receptor-associated kinase 1 (IRAK1) and IRAK4 induced by LPS was inhibited by DCEO treatment. Conclusion Suppression of the interleukin-1 receptor-associated kinase (IRAK)/NF-κB, IRAK/AP-1 and TBK1/IRF3 pathways was associated with the inhibitory effects of DCEO on inflammatory mediators in LPS-stimulated RAW264.7 macrophages. This study provides a pharmacological justification for the use of dingchuan tang in managing inflammatory disorders.
Collapse
Affiliation(s)
- Yi Zhang
- Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, .,Department of Pharmacology, Beijing University of Chinese Medicine, Beijing, People's Republic of China
| | - Hui Guo
- Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong,
| | - Brian Chi-Yan Cheng
- Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong,
| | - Tao Su
- Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong,
| | - Xiu-Qiong Fu
- Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong,
| | - Ting Li
- Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong,
| | - Pei-Li Zhu
- Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong,
| | - Kai-Wing Tse
- Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong,
| | - Si-Yuan Pan
- Research and Development Centre for Natural Health Products, HKBU Shenzhen Research Institute and Continuing Education, Shenzhen, People's Republic of China,
| | - Zhi-Ling Yu
- Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, .,Research and Development Centre for Natural Health Products, HKBU Shenzhen Research Institute and Continuing Education, Shenzhen, People's Republic of China, .,Consun Chinese Medicines Research Centre for Renal Diseases, Hong Kong Baptist University, Hong Kong, People's Republic of China,
| |
Collapse
|
23
|
Courtois G, Fauvarque MO. The Many Roles of Ubiquitin in NF-κB Signaling. Biomedicines 2018; 6:E43. [PMID: 29642643 PMCID: PMC6027159 DOI: 10.3390/biomedicines6020043] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 03/31/2018] [Accepted: 04/02/2018] [Indexed: 12/24/2022] Open
Abstract
The nuclear factor κB (NF-κB) signaling pathway ubiquitously controls cell growth and survival in basic conditions as well as rapid resetting of cellular functions following environment changes or pathogenic insults. Moreover, its deregulation is frequently observed during cell transformation, chronic inflammation or autoimmunity. Understanding how it is properly regulated therefore is a prerequisite to managing these adverse situations. Over the last years evidence has accumulated showing that ubiquitination is a key process in NF-κB activation and its resolution. Here, we examine the various functions of ubiquitin in NF-κB signaling and more specifically, how it controls signal transduction at the molecular level and impacts in vivo on NF-κB regulated cellular processes.
Collapse
|
24
|
Kong F, Liu Z, Jain VG, Shima K, Suzuki T, Muglia LJ, Starczynowski DT, Pasare C, Bhattacharyya S. Inhibition of IRAK1 Ubiquitination Determines Glucocorticoid Sensitivity for TLR9-Induced Inflammation in Macrophages. THE JOURNAL OF IMMUNOLOGY 2017; 199:3654-3667. [PMID: 29038250 DOI: 10.4049/jimmunol.1700443] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 09/19/2017] [Indexed: 01/26/2023]
Abstract
Inflammatory responses are controlled by signaling mediators that are regulated by various posttranslational modifications. Recently, transcription-independent functions for glucocorticoids (GC) in restraining inflammation have emerged, but the underlying mechanisms are unknown. In this study, we report that GC receptor (GR)-mediated actions of GC acutely suppress TLR9-induced inflammation via inhibition of IL-1R-associated kinase 1 (IRAK1) ubiquitination. β-TrCP-IRAK1 interaction is required for K48-linked ubiquitination of IRAK1 at Lys134 and subsequent membrane-to-cytoplasm trafficking of IRAK1 interacting partners TNFR-associated factor 6 and TAK1 that facilitates NF-κB and MAPK activation. Upon costimulation of macrophages with GC and TLR9-engaging ligand, GR physically interacts with IRAK1 and interferes with protein-protein interactions between β-TrCP and IRAK1. Ablation of GR in macrophages prevents GC-dependent suppression of β-TrCP-IRAK1 interactions. This GC-mediated suppression of IRAK1 activation is unique to TLR9, as GC treatment impairs TLR9 but not TLR4 ligand-induced K48-linked IRAK1 ubiquitination and trafficking of IRAK1 interacting partners. Furthermore, mutations in IRAK1 at Lys134 prevent TLR9 ligand-induced activation of inflammatory signaling mediators and synthesis of proinflammatory cytokines to an extent comparable to GC-mediated inhibition. Collectively, these findings identify a transcription-independent, rapid, and nongenomic GC suppression of TLR9 ligand-mediated IRAK1 ubiquitination as a novel mechanism for restraining acute inflammatory reactions.
Collapse
Affiliation(s)
- Fansheng Kong
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH 45229
| | - Zhiwei Liu
- Neonatal Division, International Peace Maternity and Child Health Hospital of China Welfare Institution, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, People's Republic of China
| | - Viral G Jain
- Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229
| | - Kenjiro Shima
- Division of Pulmonary Biology, Translational Pulmonary Science Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229
| | - Takuji Suzuki
- Division of Pulmonary Biology, Translational Pulmonary Science Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229
| | - Louis J Muglia
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH 45229
| | - Daniel T Starczynowski
- Experimental Hematology and Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229; and
| | - Chandrashekhar Pasare
- Department of Immunology, Southwestern Medical Center, University of Texas, Dallas, TX 75390
| | - Sandip Bhattacharyya
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH 45229;
| |
Collapse
|
25
|
Zhou H, Bulek K, Li X, Herjan T, Yu M, Qian W, Wang H, Zhou G, Chen X, Yang H, Hong L, Zhao J, Qin L, Fukuda K, Flotho A, Gao J, Dongre A, Carman JA, Kang Z, Su B, Kern TS, Smith JD, Hamilton TA, Melchior F, Fox PL, Li X. IRAK2 directs stimulus-dependent nuclear export of inflammatory mRNAs. eLife 2017; 6:29630. [PMID: 28990926 PMCID: PMC5675595 DOI: 10.7554/elife.29630] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 10/06/2017] [Indexed: 12/16/2022] Open
Abstract
Expression of inflammatory genes is determined in part by post-transcriptional regulation of mRNA metabolism but how stimulus- and transcript-dependent nuclear export influence is poorly understood. Here, we report a novel pathway in which LPS/TLR4 engagement promotes nuclear localization of IRAK2 to facilitate nuclear export of a specific subset of inflammation-related mRNAs for translation in murine macrophages. IRAK2 kinase activity is required for LPS-induced RanBP2-mediated IRAK2 sumoylation and subsequent nuclear translocation. Array analysis showed that an SRSF1-binding motif is enriched in mRNAs dependent on IRAK2 for nuclear export. Nuclear IRAK2 phosphorylates SRSF1 to reduce its binding to target mRNAs, which promotes the RNA binding of the nuclear export adaptor ALYREF and nuclear export receptor Nxf1 loading for the export of the mRNAs. In summary, LPS activates a nuclear function of IRAK2 that facilitates the assembly of nuclear export machinery to export selected inflammatory mRNAs to the cytoplasm for translation. The innate immune system is the body’s first line of defense against invading microbes. Some immune cells carry specific receptor proteins called Toll-like receptors that can identify microbes and the signals they emit. As soon as the receptors have detected a threat – for example through sensing oily molecules that make up the cell membranes of microbes – they produce signaling proteins called cytokines and chemokines to alert other immune cells. The DNA in the cell’s nucleus carries the instructions needed to make proteins. To produce proteins, including cytokines and chemokines, the information first has to be transferred into mRNA templates, which carry the instructions to the sites in the cell where the proteins are made. Cytokine and chemokine mRNAs are generally short-lived, but previous studies in 2009 and 2011 have shown that an enzyme called IRAK2 can stabilize them to make them last longer. IRAK enzymes are activated by the Toll-like receptors after a threat has been detected. However, until now it was not known whether IRAK2 also helps to transport the mRNAs of cytokines and chemokines out of the cell nucleus. Using immune cells of mice, Zhou et al. – including some of the researchers involved in the previous studies – discovered that IRAK2 helped to export the mRNAs of cytokines and chemokines from the immune cell nucleus into the surrounding cell fluid. The Toll-like receptors recognized the oily molecules of the microbes and consequently activated IRAK2, which lead to IRAK2 being moved into the cell nucleus. Once activated, IRAK2 helped to assemble the export machinery that moved selected mRNAs out of the nucleus to build the proteins. To do so, IRAK2 stopped a destabilizing protein from binding to the mRNA, so that instead the export machinery could transport the mRNA of the cytokines and chemokines out of the cell nucleus. A next step will be to test whether IRAK2 is required to guide exported mRNA tothe sites in the cell where the proteins are made. This new insight could help to develop new treatments for various diseases. For example, diseases in which the immune system attacks the cells of the body, rather than invaders, can be caused by too many cytokines and chemokines. Since IRAK2 helps to control the availability of cytokines and chemokines it may in future be used as a new drug target.
Collapse
Affiliation(s)
- Hao Zhou
- Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, United States
| | - Katarzyna Bulek
- Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, United States.,Department of Immunology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Xiao Li
- Department of Genetics, Stanford University School of Medicine, Stanford, United States
| | - Tomasz Herjan
- Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, United States
| | - Minjia Yu
- Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, United States.,Department of Medicine, Mount Auburn Hospital, Harvard Medical School, Cambridge, United States
| | - Wen Qian
- Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, United States
| | - Han Wang
- Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, United States
| | - Gao Zhou
- Department of Genetics, Stanford University School of Medicine, Stanford, United States
| | - Xing Chen
- Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, United States
| | - Hui Yang
- Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, United States
| | - Lingzi Hong
- Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, United States
| | - Junjie Zhao
- Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, United States
| | - Luke Qin
- Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, United States
| | - Koichi Fukuda
- Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Cleveland, United States
| | - Annette Flotho
- Zentrum für Molekulare Biologie der Universität Heidelberg, DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Ji Gao
- Discovery Biology, Bristol-Myers Squibb, Princeton, United States
| | - Ashok Dongre
- Discovery Biology, Bristol-Myers Squibb, Princeton, United States
| | - Julie A Carman
- Discovery Biology, Bristol-Myers Squibb, Princeton, United States
| | - Zizhen Kang
- Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, United States.,Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Department of Immunobiology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bing Su
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Department of Immunobiology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Department of Immunobiology, Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, United States
| | - Timothy S Kern
- School of Medicine, Case Western Reserve University, Cleveland, United States.,Stokes Veterans Administration Hospital, Cleveland, United States
| | - Jonathan D Smith
- Department of Cellular and Molecular Medicine, Lerner Research Institute Cleveland Clinic, Cleveland, United States
| | - Thomas A Hamilton
- Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, United States
| | - Frauke Melchior
- Zentrum für Molekulare Biologie der Universität Heidelberg, DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Paul L Fox
- Department of Cellular and Molecular Medicine, Lerner Research Institute Cleveland Clinic, Cleveland, United States
| | - Xiaoxia Li
- Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, United States
| |
Collapse
|
26
|
Hosoki K, Redding D, Itazawa T, Chakraborty A, Tapryal N, Qian S, Qi H, Aguilera-Aguirre L, Brasier AR, Phani VS, Hazra TK, Boldogh I, Sur S. Innate mechanism of pollen- and cat dander-induced oxidative stress and DNA damage in the airways. J Allergy Clin Immunol 2017; 140:1436-1439.e5. [PMID: 28583369 DOI: 10.1016/j.jaci.2017.04.044] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 04/19/2017] [Accepted: 04/26/2017] [Indexed: 12/29/2022]
Affiliation(s)
- Koa Hosoki
- Department of Internal Medicine, Division of Allergy and Immunology, University of Texas Medical Branch, Galveston, Tex
| | - David Redding
- Department of Internal Medicine, Division of Allergy and Immunology, University of Texas Medical Branch, Galveston, Tex
| | - Toshiko Itazawa
- Department of Internal Medicine, Division of Allergy and Immunology, University of Texas Medical Branch, Galveston, Tex
| | - Anirban Chakraborty
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston, Tex
| | - Nisha Tapryal
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston, Tex
| | - Sun Qian
- Department of Internal Medicine, Division of Allergy and Immunology, University of Texas Medical Branch, Galveston, Tex
| | - Huibin Qi
- Department of Internal Medicine, Division of Allergy and Immunology, University of Texas Medical Branch, Galveston, Tex
| | | | - Allan R Brasier
- Department of Internal Medicine, Division of Allergy and Immunology, University of Texas Medical Branch, Galveston, Tex; Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston, Tex
| | - Veeranki Sreenivas Phani
- Department of Preventive Medicine and Community Health, University of Texas Medical Branch, Galveston, Tex
| | - Tapas K Hazra
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston, Tex
| | - Istvan Boldogh
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Tex; Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston, Tex
| | - Sanjiv Sur
- Department of Internal Medicine, Division of Allergy and Immunology, University of Texas Medical Branch, Galveston, Tex; Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston, Tex.
| |
Collapse
|
27
|
Han KA, Yoo L, Sung JY, Chung SA, Um JW, Kim H, Seol W, Chung KC. Leucine-Rich Repeat Kinase 2 (LRRK2) Stimulates IL-1β-Mediated Inflammatory Signaling through Phosphorylation of RCAN1. Front Cell Neurosci 2017; 11:125. [PMID: 28553204 PMCID: PMC5425608 DOI: 10.3389/fncel.2017.00125] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 04/18/2017] [Indexed: 12/17/2022] Open
Abstract
Leucine-rich repeat kinase 2 (LRRK2) is a Ser/Thr kinase having mixed lineage kinase-like and GTPase domains, controlling neurite outgrowth and neuronal cell death. Evidence suggests that LRRK2 is involved in innate immune response signaling, but the underlying mechanism is yet unknown. A novel protein inhibitor of phosphatase 3B, RCAN1, is known to positively regulate inflammatory signaling through modulation of several intracellular targets of interleukins in immune cells. In the present study, we report that LRRK2 phosphorylates RCAN1 (RCAN1-1S) and is markedly up-regulated during interleukin-1β (IL-1β) treatment. During IL-1β treatment, LRRK2-mediated phosphorylation of RCAN1 promoted the formation of protein complexes, including that between Tollip and RCAN1. LRRK2 decreased binding between Tollip and IRAK1, which was accompanied by increased formation of the IRAK1-TRAF6 complex. TAK1 activity was significantly enhanced by LRRK2. Furthermore, LRRK2 enhanced transcriptional activity of NF-κB and cytokine IL-8 production. These findings suggest that LRRK2 might be important in positively modulating IL-1β-mediated signaling through selective phosphorylation of RCAN1.
Collapse
Affiliation(s)
- Kyung A Han
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei UniversitySeoul, South Korea
| | - Lang Yoo
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei UniversitySeoul, South Korea
| | - Jee Y Sung
- Center for Pediatric Oncology, National Cancer CenterGoyang-si, South Korea
| | - Sun A Chung
- Department of Food and Nutrition, College of Human Ecology, Yonsei UniversitySeoul, South Korea
| | - Ji W Um
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST)Daegu, South Korea
| | - Hyeyoung Kim
- Department of Food and Nutrition, College of Human Ecology, Yonsei UniversitySeoul, South Korea
| | - Wongi Seol
- InAm Neuroscience Research Center, Sanbon Medical Center, College of Medicine, Wonkwang UniversityGunpo-si, South Korea
| | - Kwang C Chung
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei UniversitySeoul, South Korea
| |
Collapse
|
28
|
Hossen MJ, Yang WS, Kim D, Aravinthan A, Kim JH, Cho JY. Thymoquinone: An IRAK1 inhibitor with in vivo and in vitro anti-inflammatory activities. Sci Rep 2017; 7:42995. [PMID: 28216638 PMCID: PMC5316937 DOI: 10.1038/srep42995] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Accepted: 01/18/2017] [Indexed: 12/14/2022] Open
Abstract
Thymoquinone (TQ) is a bioactive component of black seed (Nigella sativa) volatile oil and has been shown to have anti-oxidative, anti-inflammatory, and anti-cancer properties. In the present study, we explored the molecular mechanisms that underlie the anti-inflammatory effect of TQ and its target proteins using lipopolysaccharide (LPS)-stimulated murine macrophage-like RAW264.7 and human monocyte-like U937 cells, together with LPS/D-galactosamine (GalN)-induced acute hepatitis and HCl/EtOH-induced gastritis mouse models. TQ strongly inhibited the production of nitric oxide (NO) and repressed NO synthase (iNOS), tumor necrosis factor (TNF)-α, cyclooxygenase (COX)−2, interleukin (IL)−6, and IL-1β expression in LPS-activated RAW264.7 cells. Treatment of LPS/D-GalN–induced hepatitis and EtOH/HCl–induced gastritis mouse models with TQ significantly ameliorated disease symptoms. Using luciferase reporter gene assays, we also showed that the nuclear levels of transcription factors and phosphorylation patterns of signaling proteins, activator protein (AP)−1, and nuclear factor (NF)-κB pathways were all affected by TQ treatment. Finally, we used additional kinase and luciferase validation assays with interleukin-1 receptor-associated kinase 1 (IRAK1) to show that IRAK1 is directly suppressed by TQ treatment. Together, these findings strongly suggest that the anti-inflammatory actions of TQ are caused by suppression of IRAK-linked AP-1/NF-κB pathways.
Collapse
Affiliation(s)
- Muhammad Jahangir Hossen
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea.,Department of Animal Science, Patuakhali Science and Technology University, Dumki, Patuakhali 8602, Bangladesh
| | - Woo Seok Yang
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Daewon Kim
- Laboratory of Bio-informatics, Department of Multimedia Engineering, Dankook University, Cheonan 31116, Republic of Korea
| | - Adithan Aravinthan
- Department of Physiology, College of Veterinary Medicine, Chonbuk National University, Iksan 54596, Republic of Korea
| | - Jong-Hoon Kim
- Department of Physiology, College of Veterinary Medicine, Chonbuk National University, Iksan 54596, Republic of Korea
| | - Jae Youl Cho
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| |
Collapse
|
29
|
Zhou H, Yu M, Zhao J, Martin BN, Roychowdhury S, McMullen MR, Wang E, Fox PL, Yamasaki S, Nagy LE, Li X. IRAKM-Mincle axis links cell death to inflammation: Pathophysiological implications for chronic alcoholic liver disease. Hepatology 2016; 64:1978-1993. [PMID: 27628766 PMCID: PMC5115953 DOI: 10.1002/hep.28811] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 07/20/2016] [Accepted: 08/01/2016] [Indexed: 12/20/2022]
Abstract
UNLABELLED Lipopolysaccharide (LPS)-mediated activation of Toll-like receptors (TLRs) in hepatic macrophages and injury to hepatocytes are major contributors to the pathogenesis of alcoholic liver disease. However, the mechanisms by which TLR-dependent inflammatory responses and alcohol-induced hepatocellular damage coordinately lead to alcoholic liver disease are not completely understood. In this study, we found that mice deficient in interleukin-1 receptor-associated kinase M (IRAKM), a proximal TLR pathway molecule typically associated with inhibition of TLR signaling, were actually protected from chronic ethanol-induced liver injury. In bone marrow-derived macrophages challenged with low concentrations of LPS, which reflect the relevant pathophysiological levels of LPS in both alcoholic patients and ethanol-fed mice, the IRAKM Myddosome was preferentially formed. Further, the IRAKM Myddosome mediated the up-regulation of Mincle, a sensor for cell death. Mincle-deficient mice were also protected from ethanol-induced liver injury. The endogenous Mincle ligand spliceosome-associated protein 130 (SAP130) is a danger signal released by damaged cells; culture of hepatocytes with ethanol increased the release of SAP130. Ex vivo studies in bone marrow-derived macrophages suggested that SAP130 and LPS synergistically activated inflammatory responses, including inflammasome activation. CONCLUSION This study reveals a novel IRAKM-Mincle axis that contributes to the pathogenesis of ethanol-induced liver injury. (Hepatology 2016;64:1978-1993).
Collapse
Affiliation(s)
- Hao Zhou
- Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Minjia Yu
- Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Junjie Zhao
- Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Bradley N. Martin
- Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Sanjoy Roychowdhury
- Center for Liver Disease Research, Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Megan R. McMullen
- Center for Liver Disease Research, Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Emily Wang
- Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Paul L. Fox
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Sho Yamasaki
- Division of Molecular Immunology, Research Center for Infectious Diseases, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi Higashiku, Fukuoka, Japan
| | - Laura E. Nagy
- Center for Liver Disease Research, Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA,Department of Gastroenterology, Cleveland Clinic, Cleveland, Ohio, USA
| | - Xiaoxia Li
- Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
30
|
Kang K, Won M, Yuk JM, Park CY, Byun HS, Park KA, Lee SR, Kang YG, Shen HM, Lee IY, Hur GM. IinQ attenuates systemic inflammatory responses via selectively impairing the Myddosome complex formation upon TLR4 ligation. Biochem Pharmacol 2016; 121:52-66. [PMID: 27664853 DOI: 10.1016/j.bcp.2016.09.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 09/19/2016] [Indexed: 01/21/2023]
Abstract
A specific small-molecule inhibitor of the TLR4 signaling complex upstream of the IKK would likely provide therapeutic benefit for NF-κB-mediated inflammatory disease. We previously identified brazilin as a selective upstream IKK inhibitor targeting the Myddosome complex. In this study, using a cell-based ubiquitination assay for IRAK1 and a chemical library comprising a series of structural analogues of brazilin, a novel small molecule, 2-hydroxy-5,6-dihydroisoindolo[1,2-a]isoquinoline-3,8-dione (IinQ), was identified as a selective and potent inhibitor of IRAK1-dependent NF-κB activation upon TLR4 ligation. In RAW264.7 macrophages, IinQ drastically suppressed activation of upstream IKK signaling events including membrane-bound IRAK1 ubiquitination and IKK phosphorylation by the TLR4 ligand, resulting in reduced expression of proinflammatory mediators including IL-6, TNF-α, and nitric oxide. Interestingly, IinQ did not suppress NF-κB activation via the TLR3 ligand, DNA damaging agents, or a protein kinase C activator, indicating IinQ is specific for TLR4 signaling. Analysis of upstream signaling events further confirmed that IinQ disrupts the MyD88-IRAK1-TRAF6 complex formation induced by LPS treatment, without affecting TLR4 oligomerization. Moreover, intravenous administration of IinQ significantly reduced lethality and attenuated systemic inflammatory responses in an in vivo mouse model of endotoxin shock following LPS challenge. Thus, IinQ represents a novel class of brazilin analogues with improved potency and specificity toward disruption of Myddosome complex formation in TLR4 signaling, indicating that IinQ may be a promising therapeutic candidate for the treatment of systemic inflammatory diseases.
Collapse
Affiliation(s)
- Kidong Kang
- Department of Pharmacology, College of Medicine, Chungnam National University, 266 Munhwa-ro, Daejeon 35015, Republic of Korea; Department of Medical Science, College of Medicine, Chungnam National University, 266 Munhwa-ro, Daejeon 35015, Republic of Korea
| | - Minho Won
- Department of Pharmacology, College of Medicine, Chungnam National University, 266 Munhwa-ro, Daejeon 35015, Republic of Korea
| | - Jae-Min Yuk
- Department of Medical Science, College of Medicine, Chungnam National University, 266 Munhwa-ro, Daejeon 35015, Republic of Korea; Department of Infection Biology, College of Medicine, Chungnam National University, 266 Munhwa-ro, Daejeon 35015, Republic of Korea
| | - Chan-Yong Park
- Eco-Friendly New Materials Research Center, Korea Research Institute of Chemical Technology, 141 Gajeong-ro, Yusung-gu, Daejeon 34114, Republic of Korea
| | - Hee Sun Byun
- Department of Pharmacology, College of Medicine, Chungnam National University, 266 Munhwa-ro, Daejeon 35015, Republic of Korea
| | - Kyeong Ah Park
- Department of Pharmacology, College of Medicine, Chungnam National University, 266 Munhwa-ro, Daejeon 35015, Republic of Korea
| | - So-Ra Lee
- Department of Pharmacology, College of Medicine, Chungnam National University, 266 Munhwa-ro, Daejeon 35015, Republic of Korea; Department of Medical Science, College of Medicine, Chungnam National University, 266 Munhwa-ro, Daejeon 35015, Republic of Korea
| | - Young-Goo Kang
- Eco-Friendly New Materials Research Center, Korea Research Institute of Chemical Technology, 141 Gajeong-ro, Yusung-gu, Daejeon 34114, Republic of Korea
| | - Han-Ming Shen
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Ill Young Lee
- Eco-Friendly New Materials Research Center, Korea Research Institute of Chemical Technology, 141 Gajeong-ro, Yusung-gu, Daejeon 34114, Republic of Korea.
| | - Gang Min Hur
- Department of Pharmacology, College of Medicine, Chungnam National University, 266 Munhwa-ro, Daejeon 35015, Republic of Korea; Department of Medical Science, College of Medicine, Chungnam National University, 266 Munhwa-ro, Daejeon 35015, Republic of Korea.
| |
Collapse
|
31
|
Abstract
"Rotaviruses represent the most important etiological agents of acute, severe gastroenteritis in the young of many animal species, including humans." This statement, variations of which are a common beginning in articles about rotaviruses, reflects the fact that these viruses have evolved efficient strategies for evading the innate immune response of the host and for successfully replicating in the population. In this review, we summarize what is known about the defense mechanisms that host cells employ to prevent rotavirus invasion and the countermeasures that these viruses have successfully developed to surpass cellular defenses. Rotaviruses use at least two viral multifunctional proteins to directly interact with, and prevent the activation of, the interferon system, and they use at least one other protein to halt the protein synthesis machinery and prevent the expression of most of the transcriptional antiviral program of the cell. Characterization of the confrontation between rotaviruses and their host cells has allowed us to learn about the virus-host coevolution that prevents the damaging effects of the innate immune response.
Collapse
Affiliation(s)
- Susana López
- Departamento de Génetica del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos 62210, México;
| | - Liliana Sánchez-Tacuba
- Departamento de Génetica del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos 62210, México;
| | - Joaquin Moreno
- Departamento de Génetica del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos 62210, México;
| | - Carlos F Arias
- Departamento de Génetica del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos 62210, México;
| |
Collapse
|
32
|
Singh AK, Umar S, Riegsecker S, Chourasia M, Ahmed S. Regulation of Transforming Growth Factor β-Activated Kinase Activation by Epigallocatechin-3-Gallate in Rheumatoid Arthritis Synovial Fibroblasts: Suppression of K(63) -Linked Autoubiquitination of Tumor Necrosis Factor Receptor-Associated Factor 6. Arthritis Rheumatol 2016; 68:347-58. [PMID: 26473505 DOI: 10.1002/art.39447] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Accepted: 09/17/2015] [Indexed: 12/19/2022]
Abstract
OBJECTIVE Transforming growth factor β-activated kinase 1 (TAK1) is a key MAPKKK family protein in interleukin-1β (IL-1β), tumor necrosis factor (TNF), and Toll-like receptor signaling. This study was undertaken to examine the posttranslational modification of TAK1 and its therapeutic regulation in rheumatoid arthritis (RA). METHODS The effect of TAK1, IL-1 receptor-associated kinase 1 (IRAK-1), and TNF receptor-associated factor 6 (TRAF6) inhibition was evaluated in IL-1β-stimulated human RA synovial fibroblasts (RASFs). Western blotting, immunoprecipitation, and 20S proteasome assay were used to study the ubiquitination process in RASFs. The efficacy of epigallocatechin-3-gallate (EGCG), a potent antiinflammatory molecule, in regulating these processes in RASFs was evaluated. Molecular docking was performed to examine the interaction of EGCG with human TAK1, IRAK-1, and TRAF6. These findings were confirmed using a rat model of adjuvant-induced arthritis (AIA). RESULTS Inhibition of TAK1, but not IRAK-1 or TRAF6, completely abrogated IL-1β-induced IL-6 and IL-8 synthesis in RASFs. EGCG inhibited TAK1 phosphorylation at Thr(184/187) and occupied the C(174) position, an ATP-binding site, to inhibit its kinase activity. EGCG pretreatment also inhibited K(63) -linked autoubiquitination of TRAF6, a posttranslational modification essential for TAK1 autophosphorylation, by forming a stable H bond at the K(124) position on TRAF6. Furthermore, EGCG enhanced proteasome-associated deubiquitinase expression to rescue proteins from proteasomal degradation. Western blot analyses of joint homogenates from rats with AIA showed a significant increase in K(48) -linked polyubiquitination, TAK1 phosphorylation, and TRAF6 expression when compared to naive rats. Administration of EGCG (50 mg/kg/day) for 10 days ameliorated AIA in rats by reducing TAK1 phosphorylation and K(48) -linked polyubiquitination. CONCLUSION Our findings provide a rationale for targeting TAK1 for the treatment of RA with EGCG.
Collapse
Affiliation(s)
- Anil K Singh
- Washington State University College of Pharmacy, Spokane
| | - Sadiq Umar
- Washington State University College of Pharmacy, Spokane
| | - Sharayah Riegsecker
- University of Toledo College of Pharmacy and Pharmaceutical Sciences, Toledo, Ohio
| | - Mukesh Chourasia
- National Institute of Pharmaceutical Education and Research, Hajipur, India
| | | |
Collapse
|
33
|
Cheng BCY, Yu H, Su T, Fu XQ, Guo H, Li T, Cao HH, Tse AKW, Kwan HY, Yu ZL. A herbal formula comprising Rosae Multiflorae Fructus and Lonicerae Japonicae Flos inhibits the production of inflammatory mediators and the IRAK-1/TAK1 and TBK1/IRF3 pathways in RAW 264.7 and THP-1 cells. JOURNAL OF ETHNOPHARMACOLOGY 2015; 174:195-199. [PMID: 26297845 DOI: 10.1016/j.jep.2015.08.018] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Revised: 08/16/2015] [Accepted: 08/18/2015] [Indexed: 06/04/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE As documented in the Chinese Materia Medica Grand Dictionary (), a herbal formula (RL) consisting of Rosae Multiflorae Fructus (multiflora rose hips) and Lonicerae Japonicae Flos (Japanese honeysuckle flowers) has traditionally been used in treating inflammatory disorders. RL was previously reported to inhibit the expression of various inflammatory mediators regulated by NF-κB and MAPKs that are components of the TLR4 signalling pathways. AIM OF THE STUDY This study aims to provide further justification for clinical application of RL in treating inflammatory disorders by further delineating the involvement of the TLR4 signalling cascades in the effects of RL on inflammatory mediators. MATERIALS AND METHODS RL consisting of Rosae Multiflorae Fructus and Lonicerae Japonicae Flos (in 5:3 ratio) was extracted using absolute ethanol. We investigated the effect of RL on the production of cytokines and chemokines that are regulated by three key transcription factors of the TLR4 signalling pathways AP-1, NF-κB and IRF3 in LPS-stimulated RAW264.7 cells using the multiplex biometric immunoassay. Phosphorylation of AP-1, NF-κB, IRF3, IκB-α, IKKα/β, Akt, TAK1, TBK1, IRAK-1 and IRAK-4 were examined in LPS-stimulated RAW264.7 cells and THP-1 cells using Western blotting. Nuclear localizations of AP-1, NF-κB and IRF3 were also examined using Western blotting. RESULTS RL reduced the secretion of various pro-inflammatory cytokines and chemokines regulated by transcription factors AP-1, NF-κB and IRF3. Phosphorylation and nuclear protein levels of these transcription factors were decreased by RL treatment. Moreover, RL inhibited the activation/phosphorylation of IκB-α, IKKα/β, TAK1, TBK1 and IRAK-1. CONCLUSIONS Suppression of the IRAK-1/TAK1 and TBK1/IRF3 signalling pathways was associated with the effect of RL on inflammatory mediators in LPS-stimulated RAW264.7 and THP-1 cells. This provides further pharmacological basis for the clinical application of RL in the treatment of inflammatory disorders.
Collapse
Affiliation(s)
- Brian Chi Yan Cheng
- Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| | - Hua Yu
- Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| | - Tao Su
- Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| | - Xiu-Qiong Fu
- Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| | - Hui Guo
- Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| | - Ting Li
- Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| | - Hui-Hui Cao
- Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| | - Anfernee Kai-Wing Tse
- Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| | - Hiu-Yee Kwan
- Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| | - Zhi-Ling Yu
- Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong.
| |
Collapse
|
34
|
Murphy MB, Xiong Y, Pattabiraman G, Manavalan TT, Qiu F, Medvedev AE. Pellino-3 promotes endotoxin tolerance and acts as a negative regulator of TLR2 and TLR4 signaling. J Leukoc Biol 2015; 98:963-74. [PMID: 26310831 DOI: 10.1189/jlb.2vma0515-229rr] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 07/30/2015] [Indexed: 12/24/2022] Open
Abstract
Development of endotoxin tolerance in macrophages during sepsis reprograms Toll-like receptor 4 signaling to inhibit proinflammatory cytokines without suppressing anti-inflammatory and antimicrobial mediators and protects the host from excessive inflammation and tissue damage. However, endotoxin tolerance renders septic patients immunocompromised and unable to control secondary infections. Although previous studies have revealed the importance of several negative regulators of Toll-like receptor signaling in endotoxin tolerance, the role of Pellino proteins has not been addressed. The present report shows that the induction of endotoxin tolerance in vivo in mice and in vitro in human monocytes and THP-1 and MonoMac-6 macrophages increases the expression of Pellino-3. Overexpression of Pellino-3 in human embryonic kidney 293/Toll-like receptor 2 or 293/Toll-like receptor 4/myeloid differentiation factor-2 cells inhibited Toll-like receptor 2/4-mediated activation of nuclear factor-κB and induction of CXCL-8 mRNA, and Pellino-3 ablation increased these responses. Pellino-3-deficient THP-1 cells had elevated Toll-like receptor 2/4-driven tumor necrosis factor-α, interleukin-6 mRNA, and Toll-like receptor 4-driven CCL5 gene expression in response to Toll-like receptor agonists and heat-killed Escherichia coli and Staphylococcus aureus, cytokines controlled by the MyD88 and Toll-interleukin-1R domain-containing protein inducing interferon-β-mediated pathways, respectively. In addition, deficiency in Pellino-3 slightly increased phagocytosis of heat-killed bacteria. Transfected Pellino-3 inhibited nuclear factor-κB activation driven by overexpression of MyD88, TIR domain-containing adapter inducing interferon-β, interleukin-1R-associated kinase-1, and tumor necrosis factor receptor activator of nuclear factor-κB-binding kinase-1, TGF-β-activated kinase 1, and tumor necrosis factor receptor-associated factor-6, and inhibited interleukin-1R-associated kinase 1 modifications and tumor necrosis factor receptor activator of nuclear factor-κB-binding kinase 1 phosphorylation. Finally, Pellino-3 ablation in THP-1 decreased the extent of endotoxin tolerization. Thus, Pellino-3 is involved in endotoxin tolerance and functions as a negative regulator of Toll-like receptor 2/4 signaling.
Collapse
Affiliation(s)
- Michael B Murphy
- *Department of Immunology, University of Connecticut Health Center, Farmington, Connecticut, USA; and Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Yanbao Xiong
- *Department of Immunology, University of Connecticut Health Center, Farmington, Connecticut, USA; and Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Goutham Pattabiraman
- *Department of Immunology, University of Connecticut Health Center, Farmington, Connecticut, USA; and Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Tissa T Manavalan
- *Department of Immunology, University of Connecticut Health Center, Farmington, Connecticut, USA; and Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Fu Qiu
- *Department of Immunology, University of Connecticut Health Center, Farmington, Connecticut, USA; and Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Andrei E Medvedev
- *Department of Immunology, University of Connecticut Health Center, Farmington, Connecticut, USA; and Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
35
|
Thai HV, Kim E, Kim SC, Jeong D, Yang S, Baek KS, Kim Y, Ratan ZA, Yoon KD, Kim JH, Cho JY. Boerhavia diffusa L. ethanol extract suppresses inflammatory responses via inhibition of Src/Syk/TRAF6. J Funct Foods 2015. [DOI: 10.1016/j.jff.2015.06.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
|
36
|
Bahia MS, Kaur M, Silakari P, Silakari O. Interleukin-1 receptor associated kinase inhibitors: potential therapeutic agents for inflammatory- and immune-related disorders. Cell Signal 2015; 27:1039-55. [PMID: 25728511 DOI: 10.1016/j.cellsig.2015.02.025] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Revised: 01/30/2015] [Accepted: 02/23/2015] [Indexed: 12/15/2022]
Abstract
The various cells of innate immune system quickly counter-attack invading pathogens, and mount up "first line" defense through their trans-membrane receptors including Toll-like receptors (TLRs) and interleukin receptors (IL-Rs) that result in the secretion of pro-inflammatory cytokines. Albeit such inflammatory responses are beneficial in pathological conditions, their overstimulation may cause severe inflammatory damage; thus, make this defense system a "double edged sword". IRAK-4 has been evaluated as an indispensable element of IL-Rs and TLR pathways that can regulate the abnormal levels of cytokines, and therefore could be employed to manage immune- and inflammation-related disorders. Historically, the identification of selective and potent inhibitors has been challenging; thus, a limited number of small molecule IRAK-4 inhibitors are available in literature. Recently, IRAK-4 achieved great attention, when Ligand® pharmaceutical and Nimbus Discovery® reported the beneficial potentials of IRAK-4 inhibitors in the pre-clinical evaluation for various inflammatory- and immune-related disorders, but not limited to, such as rheumatoid arthritis, inflammatory bowel disease, psoriasis, gout, asthma and cancer.
Collapse
Affiliation(s)
- Malkeet Singh Bahia
- Molecular Modeling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab 147002, India
| | - Maninder Kaur
- Molecular Modeling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab 147002, India
| | - Pragati Silakari
- Molecular Modeling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab 147002, India
| | - Om Silakari
- Molecular Modeling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab 147002, India.
| |
Collapse
|
37
|
Putative E3 ubiquitin ligase of human rotavirus inhibits NF-κB activation by using molecular mimicry to target β-TrCP. mBio 2015; 6:mBio.02490-14. [PMID: 25626907 PMCID: PMC4324316 DOI: 10.1128/mbio.02490-14] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
NF-κB plays a critical role in the induction and maintenance of innate and adaptive immune transcriptional programs. An associated inhibitor of κB protein (IκB) regulates NF-κB activation and contains a degron motif (DSGΦxS) that undergoes phosphorylation following pathogen recognition or other proinflammatory signals. The E3 ubiquitin ligase SCFβ-TrCP recognizes this phosphodegron through its β-transducin repeat-containing protein (β-TrCP) subunit and induces IκB degradation, allowing NF-κB to translocate to the nucleus and modulate gene expression. Rotavirus (RV), a major cause of pediatric gastroenteritis, can block NF-κB activation through the action of its nonstructural protein NSP1, a putative E3 ubiquitin ligase that mediates the degradation of β-TrCP or other immunomodulatory proteins in a virus strain-specific manner. Here, we show that NSP1 targets β-TrCP by mimicking the IκB phosphodegron. The NSP1 proteins of most human and porcine RV strains conserve a C-terminal phosphodegron-like (PDL) motif, DSGΦS. Deletion of this motif or mutation of its serine residues disrupts NSP1-mediated degradation of β-TrCP and inhibition of NF-κB activation. Additionally, a point mutation within the phosphodegron-binding pocket protects β-TrCP from NSP1-mediated turnover. Fusion of the PDL motif to an NSP1 protein known to target other immunomodulatory proteins generates a chimeric NSP1 protein that can induce β-TrCP degradation and block NF-κB activation. Other viral proteins (Epstein-Barr virus LMP1, HIV-1 Vpu, and vaccinia virus A49) also contain a PDL motif and interact with β-TrCP to inhibit NF-κB activation. Taken together, these data suggest that targeting β-TrCP by molecular mimicry may be a common strategy used by human viruses to evade the host immune response. IMPORTANCE The transcription factor NF-κB, a central regulator of the host response to infection, is a frequent target of viral antagonism. Pathogen detection activates NF-κB by inducing the phosphorylation of an associated inhibitor protein (IκB), which targets IκB for degradation by the E3 ubiquitin ligase β-TrCP. Rotavirus, a significant cause of childhood gastroenteritis, antagonizes NF-κB through the activity of its NSP1 protein, a putative E3 ubiquitin ligase that mediates β-TrCP turnover. Here, we show that NSP1 functions by mimicking the IκB phosphodegron recognized by β-TrCP. Nearly all human rotavirus strains conserve this motif at the NSP1 C terminus, and its removal disrupts NSP1 antagonist activity. This sequence conserves the biochemical properties of the IκB phosphodegron and can rescue antagonist activity when fused to an NSP1 protein otherwise inactive against β-TrCP. Other viral proteins also mimic IκB to disrupt NF-κB activation, indicating that this is an important immune evasion strategy. The transcription factor NF-κB, a central regulator of the host response to infection, is a frequent target of viral antagonism. Pathogen detection activates NF-κB by inducing the phosphorylation of an associated inhibitor protein (IκB), which targets IκB for degradation by the E3 ubiquitin ligase β-TrCP. Rotavirus, a significant cause of childhood gastroenteritis, antagonizes NF-κB through the activity of its NSP1 protein, a putative E3 ubiquitin ligase that mediates β-TrCP turnover. Here, we show that NSP1 functions by mimicking the IκB phosphodegron recognized by β-TrCP. Nearly all human rotavirus strains conserve this motif at the NSP1 C terminus, and its removal disrupts NSP1 antagonist activity. This sequence conserves the biochemical properties of the IκB phosphodegron and can rescue antagonist activity when fused to an NSP1 protein otherwise inactive against β-TrCP. Other viral proteins also mimic IκB to disrupt NF-κB activation, indicating that this is an important immune evasion strategy.
Collapse
|
38
|
Huoh YS, Ferguson KM. The pellino e3 ubiquitin ligases recognize specific phosphothreonine motifs and have distinct substrate specificities. Biochemistry 2014; 53:4946-55. [PMID: 25027698 PMCID: PMC4201300 DOI: 10.1021/bi5005156] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
![]()
The
four mammalian Pellinos (Pellinos 1, 2, 3a, and 3b) are E3
ubiquitin ligases that are emerging as critical mediators for a variety
of immune signaling pathways, including those activated by Toll-like
receptors, the T-cell receptor, and NOD2. It is becoming increasingly
clear that each Pellino has a distinct role in facilitating immune
receptor signaling. However, the underlying mechanisms by which these
highly homologous proteins act selectively in these signaling pathways
are not clear. In this study, we investigate whether Pellino substrate
recognition contributes to the divergent functions of Pellinos. Substrate
recognition of each Pellino is mediated by its noncanonical forkhead-associated
(FHA) domain, a well-characterized phosphothreonine-binding module.
Pellino FHA domains share very high sequence identity, so a molecular
basis for differences in substrate recognition is not immediately
apparent. To explore Pellino substrate specificity, we first identify
a high-affinity Pellino2 FHA domain-binding motif in the Pellino substrate,
interleukin-1 receptor-associated kinase 1 (IRAK1). Analysis of binding
of the different Pellinos to a panel of phosphothreonine-containing
peptides derived from the IRAK1-binding motif reveals that each Pellino
has a distinct phosphothreonine peptide binding preference. We observe
a similar binding specificity in the interaction of Pellinos with
a number of known Pellino substrates. These results argue that the
nonredundant roles that Pellinos play in immune signaling are in part
due to their divergent substrate specificities. This new insight into
Pellino substrate recognition could be exploited for pharmacological
advantage in treating inflammatory diseases that have been linked
to the aberrant regulation of Pellinos.
Collapse
Affiliation(s)
- Yu-San Huoh
- Department of Physiology and Graduate Group in Biochemistry and Molecular Biophysics, University of Pennsylvania Perelman School of Medicine , Philadelphia, Pennsylvania 19104, United States
| | | |
Collapse
|
39
|
Jeon J, Lee JH, Park KA, Byun HS, Lee H, Lee Y, Zhang T, Kang K, Seok JH, Kwon HJ, Han MD, Kang SW, Hong JH, Hur GM. Brazilin selectively disrupts proximal IL-1 receptor signaling complex formation by targeting an IKK-upstream signaling components. Biochem Pharmacol 2014; 89:515-25. [DOI: 10.1016/j.bcp.2014.04.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Revised: 04/01/2014] [Accepted: 04/02/2014] [Indexed: 01/15/2023]
|
40
|
Jeong D, Yi YS, Sung GH, Yang WS, Park JG, Yoon K, Yoon DH, Song C, Lee Y, Rhee MH, Kim TW, Kim JH, Cho JY. Anti-inflammatory activities and mechanisms of Artemisia asiatica ethanol extract. JOURNAL OF ETHNOPHARMACOLOGY 2014; 152:487-496. [PMID: 24503036 DOI: 10.1016/j.jep.2014.01.030] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Revised: 01/25/2014] [Accepted: 01/27/2014] [Indexed: 06/03/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Artemisia asiatica Nakai (Compositae) is a representative herbal plant used to treat infection and inflammatory diseases. Although Artemisia asiatica is reported to have immunopharmacological activities, the mechanisms of these activities and the effectiveness of Artemisia asiatica preparations in use are not known. MATERIALS AND METHODS To evaluate the anti-inflammatory activities of Artemisia asiatica ethanol extract (Aa-EE), we assayed nitric oxide (NO), tumor necrosis factor (TNF)-α, and prostaglandin E2 (PGE2) in macrophages and measured the extent of tissue injury in a model of gastric ulcer induced in mice by treatment with HCl in EtOH. Putative enzymatic mediators of Aa-EE activities were identified by nuclear fractionation, reporter gene assay, immunoprecipitation, immunoblotting, and kinase assay. Active compound in Aa-EE was identified using HPLC. RESULTS Treatment of RAW264.7 cells and peritoneal macrophages with Aa-EE suppressed the production of NO, PGE2, and TNF-α in response to lipopolysaccharide (LPS) and induced heme oxygenase-1 expression. The Aa-EE also ameliorated symptoms of gastric ulcer in HCl/EtOH-treated mice. These effects were associated with the inhibition of nuclear translocation of nuclear factor (NF)-κB and activator protein (AP)-1, implying that the anti-inflammatory action of the Aa-EE occurred through transcriptional inhibition. The upstream regulatory signals Syk and Src for translocation of NF-κB and TRAF6 for AP-1 were identified as targets of this effect. Analysis of Aa-EE by HPLC revealed the presence of luteolin, known to inhibit NO and PGE2 activity. CONCLUSION The anti-inflammatory activities attributed to Artemisia asiatica Nakai in traditional medicine may be mediated by luteolin through inhibition of Src/Syk/NF-κB and TRAF6/JNK/AP-1 signaling pathways.
Collapse
Affiliation(s)
- Deok Jeong
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 440-746, Republic of Korea
| | - Young-Su Yi
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 440-746, Republic of Korea
| | - Gi-Ho Sung
- Department of Herbal Crop Research, National Institutes of Horticultural & Herbal Science, Rural Development Administration, Suwon 441-707, Korea
| | - Woo Seok Yang
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 440-746, Republic of Korea
| | - Jae Gwang Park
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 440-746, Republic of Korea
| | - Keejung Yoon
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 440-746, Republic of Korea
| | - Deok Hyo Yoon
- Department of Biochemistry, Kangwon National University, Chuncehon 200-701, Republic of Korea
| | - Changsik Song
- Department of Chemistry, Sungkyunkwan University, Suwon 440-746, Republic of Korea
| | - Yunmi Lee
- Department of Chemistry, Kwangwoon University, Seoul 139-701, Republic of Korea
| | - Man Hee Rhee
- College of Veterinary Medicine, Kyungpook National University, Daegu 702-701, Republic of Korea
| | - Tae Woong Kim
- Department of Biochemistry, Kangwon National University, Chuncehon 200-701, Republic of Korea
| | - Jong-Hoon Kim
- Department of Veterinary Physiology, College of Veterinary Medicine, Biosafety Research Institute, Chonbuk National University, Jeonju 561-756, Republic of Korea.
| | - Jae Youl Cho
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 440-746, Republic of Korea.
| |
Collapse
|
41
|
MEK1 promotes YAP and their interaction is critical for tumorigenesis in liver cancer. FEBS Lett 2013; 587:3921-7. [PMID: 24211253 DOI: 10.1016/j.febslet.2013.10.042] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2013] [Revised: 08/30/2013] [Accepted: 10/27/2013] [Indexed: 01/03/2023]
Abstract
Mitogen-activated protein kinase kinase 1 (MAP2K1/MEK1) as well as Yes-associated protein (YAP), the downstream effector of Hippo signaling pathway, is linked to hepatocarcinogenesis. However, little is known about whether and how MEK1 interacts with YAP. In this study, we find that MEK1-YAP interaction is critical for liver cancer cell proliferation and maintenance of transformed phenotypes both in vitro and in vivo. Moreover, MEK1 and YAP proteins are closely correlated in human liver cancer samples. Mechanistically, inhibition of MEK1 by both PD98059 and U0126 as well as RNAi reduces beta-transducin repeat containing E3 ubiquitin protein ligase (BTRC), which acts as a potential endogenous YAP protector.
Collapse
|
42
|
Yang Y, Yu T, Lee YG, Yang WS, Oh J, Jeong D, Lee S, Kim TW, Park YC, Sung GH, Cho JY. Methanol extract of Hopea odorata suppresses inflammatory responses via the direct inhibition of multiple kinases. JOURNAL OF ETHNOPHARMACOLOGY 2013; 145:598-607. [PMID: 23220195 DOI: 10.1016/j.jep.2012.11.041] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Revised: 11/15/2012] [Accepted: 11/18/2012] [Indexed: 06/01/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Hopea odorata Roxb. (Dipterocarpaceae) is a representative Thai ethnopharmacological herbal plant used in the treatment of various inflammation-related diseases. In spite of its traditional use, systematic studies of its anti-inflammatory action have not been performed. MATERIALS AND METHODS The inhibitory activities of a Hopea odorata methanol extract (Ho-ME) on the production of nitric oxide (NO), tumour necrosis factor (TNF)-α, and prostaglandin E(2) (PGE(2)) in RAW264.7 cells and peritoneal macrophages were investigated. The effects of Ho-ME on the gastritis symptoms induced by HCl/EtOH and on ear oedemas induced by arachidonic acid were also examined. Furthermore, to identify the immunopharmacological targets of this extract, nuclear fractionation, a reporter gene assay, immunoprecipitation, immunoblot analysis, and a kinase assay were employed. RESULTS Ho-ME strongly inhibited the release of NO, PGE(2), and TNF-α in RAW264.7 cells and peritoneal macrophages stimulated by lipopolysaccharide (LPS). Ho-ME also clearly suppressed the gene expression of pro-inflammatory cytokines and chemokines, such as interferon (IFN)-β, interleukin (IL)-12, and monocyte chemotactic protein-1 (MCP-1). By analysing the inhibited target molecules, Syk and Src were found to be suppressed in the inhibition of nuclear factor (NF)-κB pathway. In addition, the observed downregulation of activator protein (AP)-1 and cAMP response element-binding (CREB) was due to the direct inhibition of interleukin-1 receptor-associated kinase (IRAK)1 and IRAK4, which was also linked to the suppression of c-Jun N-terminal kinase (JNK) and p38. In agreement with the in vitro observations, this extract also ameliorated the inflammatory symptoms in EtOH/HCl-induced gastritis and arachidonic acid-induced ear oedemas in mice. CONCLUSION Ho-ME has potential as a functional herbal remedy targeting Syk- and Src-mediated anti-inflammatory mechanisms. Future pre-clinical studies will be needed to investigate this possibility.
Collapse
Affiliation(s)
- Yanyan Yang
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 440-746, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|