1
|
Khokhar M, Dey S, Tomo S, Jaremko M, Emwas AH, Pandey RK. Unveiling Novel Drug Targets and Emerging Therapies for Rheumatoid Arthritis: A Comprehensive Review. ACS Pharmacol Transl Sci 2024; 7:1664-1693. [PMID: 38898941 PMCID: PMC11184612 DOI: 10.1021/acsptsci.4c00067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 05/09/2024] [Accepted: 05/14/2024] [Indexed: 06/21/2024]
Abstract
Rheumatoid arthritis (RA) is a chronic debilitating autoimmune disease, that causes joint damage, deformities, and decreased functionality. In addition, RA can also impact organs like the skin, lungs, eyes, and blood vessels. This autoimmune condition arises when the immune system erroneously targets the joint synovial membrane, resulting in synovitis, pannus formation, and cartilage damage. RA treatment is often holistic, integrating medication, physical therapy, and lifestyle modifications. Its main objective is to achieve remission or low disease activity by utilizing a "treat-to-target" approach that optimizes drug usage and dose adjustments based on clinical response and disease activity markers. The primary RA treatment uses disease-modifying antirheumatic drugs (DMARDs) that help to interrupt the inflammatory process. When there is an inadequate response, a combination of biologicals and DMARDs is recommended. Biological therapies target inflammatory pathways and have shown promising results in managing RA symptoms. Close monitoring for adverse effects and disease progression is critical to ensure optimal treatment outcomes. A deeper understanding of the pathways and mechanisms will allow new treatment strategies that minimize adverse effects and maintain quality of life. This review discusses the potential targets that can be used for designing and implementing precision medicine in RA treatment, spotlighting the latest breakthroughs in biologics, JAK inhibitors, IL-6 receptor antagonists, TNF blockers, and disease-modifying noncoding RNAs.
Collapse
Affiliation(s)
- Manoj Khokhar
- Department
of Biochemistry, All India Institute of
Medical Sciences, Jodhpur, 342005 Rajasthan, India
| | - Sangita Dey
- CSO
Department, Cellworks Research India Pvt
Ltd, Bengaluru, 560066 Karnataka, India
| | - Sojit Tomo
- Department
of Biochemistry, All India Institute of
Medical Sciences, Jodhpur, 342005 Rajasthan, India
| | - Mariusz Jaremko
- Smart-Health
Initiative (SHI) and Red Sea Research Center (RSRC), Division of Biological
and Environmental Sciences and Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955 Jeddah, Saudi Arabia
| | - Abdul-Hamid Emwas
- Core
Laboratories, King Abdullah University of
Science and Technology (KAUST), Thuwal 23955-6900, Kingdom of Saudi Arabia
| | - Rajan Kumar Pandey
- Department
of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm 17177, Sweden
| |
Collapse
|
2
|
Minichino D, Lv K, Chu N, Tong W, Behrens EM. BRAF-V600E utilizes posttranscriptional mechanisms to amplify LPS-induced TNFα production in dendritic cells in a mouse model of Langerhans cell histiocytosis. J Leukoc Biol 2022; 112:1089-1104. [PMID: 35648675 PMCID: PMC9939017 DOI: 10.1002/jlb.3a0122-075rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 04/25/2022] [Indexed: 12/24/2022] Open
Abstract
Langerhans cell histiocytosis (LCH) is an inflammatory disease characterized by abnormal dendritic cells (DCs) with hyperactive ERK signaling, called "LCH cells." Since DCs rely on ERK signaling to produce inflammatory molecules in response to pathogenic cues, we hypothesized that hyperactive ERK enhances DCs inflammatory responses. We specifically investigated TLR4-induced TNFα production in LCH cells by utilizing the BRAF-V600Efl/+ :CD11c-Cre mouse model of LCH, which hyperactivates ERK in DCs. We measured LPS-induced TNFα production both in vivo and in vitro using splenic CD11c+ cells and bone marrow-derived DCs with or without pharmacologic BRAFV600E inhibition. We observed a reversible increase in secreted TNFα and a partially reversible increase in TNFα protein per cell, despite a decrease in TLR4 signaling and Tnfa transcripts compared with controls. We examined ERK-driven, posttranscriptional mechanisms that contribute to TNFα production and secretion using biochemical and cellular assays. We identified a reversible increase in TACE activation, the enzyme required for TNFα secretion, and most strikingly, an increase in protein translation, including TNFα. Defining the translatome through polysome-bound RNA sequencing revealed up-regulated translation of the LPS-response program. These data suggest hyperactive ERK signaling utilizes multiple posttranscriptional mechanisms to amplify inflammatory responses in DCs, advancing our understanding of LCH and basic DC biology.
Collapse
Affiliation(s)
- Danielle Minichino
- Cell and Molecular Biology Graduate Group, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Division of Pediatric Rheumatology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Kaosheng Lv
- Division of Hematology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Niansheng Chu
- Division of Pediatric Rheumatology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Wei Tong
- Division of Hematology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Edward M Behrens
- Cell and Molecular Biology Graduate Group, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Division of Pediatric Rheumatology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| |
Collapse
|
3
|
Millet P, Martin T, Opiekun M, Beauchamp GK, Kimball BA. Differing Alterations of Odor Volatiles Among Pathogenic Stimuli. Chem Senses 2021; 46:6300881. [PMID: 34133735 DOI: 10.1093/chemse/bjab030] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Alterations of the volatile metabolome (the collection of volatiles present in secretions and other emanations) that occur in response to inflammation can be detected by conspecifics and chemometric analyses. Using a model system where mouse urinary metabolites are altered by treatment with lipopolysaccharide (found in the outer cell membrane of gram-negative bacteria), we hypothesized that alteration of body odor volatiles will vary according to the pathogen responsible for inducing the inflammation. We tested this hypothesis by treating mice with different immunogens that engage different immune signaling pathways. Results suggest that alterations of body odor volatiles resulting from inflammation do contain detailed information about the type of pathogen that instigated the inflammation and these differences are not merely dependent on the severity of the inflammatory event. These results are encouraging for the future of differential medical diagnosis of febrile diseases by analysis of the volatile metabolome. In particular, our data support the possibility that bacterial infections can be differentiated from viral infections such that antibiotic drug stewardship could be drastically improved by reducing unneeded treatments with antibiotics.
Collapse
Affiliation(s)
- Patrick Millet
- Monell Chemical Senses Center, 3500 Market Street, Philadelphia, PA, USA
| | - Talia Martin
- Monell Chemical Senses Center, 3500 Market Street, Philadelphia, PA, USA
| | - Maryanne Opiekun
- Monell Chemical Senses Center, 3500 Market Street, Philadelphia, PA, USA
| | - Gary K Beauchamp
- Monell Chemical Senses Center, 3500 Market Street, Philadelphia, PA, USA
| | - Bruce A Kimball
- Monell Chemical Senses Center, 3500 Market Street, Philadelphia, PA, USA
| |
Collapse
|
4
|
ElTanbouly MA, Schaafsma E, Smits NC, Shah P, Cheng C, Burns C, Blazar BR, Noelle RJ, Mabaera R. VISTA Re-programs Macrophage Biology Through the Combined Regulation of Tolerance and Anti-inflammatory Pathways. Front Immunol 2020; 11:580187. [PMID: 33178206 PMCID: PMC7593571 DOI: 10.3389/fimmu.2020.580187] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Accepted: 09/08/2020] [Indexed: 12/20/2022] Open
Abstract
We present the novel finding that V-domain Ig suppressor of T cell activation (VISTA) negatively regulates innate inflammation through the transcriptional and epigenetic re-programming of macrophages. Representative of VISTA re-programming is the ability of VISTA agonistic antibodies to augment LPS tolerance and reduce septic shock lethality in mice. This anti-inflammatory effect of anti-VISTA was mimicked in vitro demonstrating that anti-VISTA treatment caused a significant reduction in LPS-induced IL-12p40, IL-6, CXCL2, and TNF; all hallmark pro-inflammatory mediators of endotoxin shock. Even under conditions that typically "break" LPS tolerance, VISTA agonists sustained a macrophage anti-inflammatory profile. Analysis of the proteomic and transcriptional changes imposed by anti-VISTA show that macrophage re-programming was mediated by a composite profile of mediators involved in both macrophage tolerance induction (IRG1, miR221, A20, IL-10) as well as transcription factors central to driving an anti-inflammatory profile (e.g., IRF5, IRF8, NFKB1). These findings underscore a novel and new activity of VISTA as a negative checkpoint regulator that induces both tolerance and anti-inflammatory programs in macrophages and controls the magnitude of innate inflammation in vivo.
Collapse
Affiliation(s)
- Mohamed A. ElTanbouly
- Department of Microbiology and Immunology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
| | - Evelien Schaafsma
- Department of Biomedical Data Science, Geisel School of Medicine at Dartmouth, Hanover, NH, United States
| | - Nicole C. Smits
- Department of Microbiology and Immunology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
| | - Parth Shah
- Department of Medicine, Dartmouth Hitchcock Medical Center, Lebanon, NH, United States
| | - Chao Cheng
- Department of Medicine, Baylor College of Medicine, Houston, TX, United States
| | - Christopher Burns
- Department of Medicine, Dartmouth Hitchcock Medical Center, Lebanon, NH, United States
| | - Bruce R. Blazar
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, United States
| | - Randolph J. Noelle
- Department of Microbiology and Immunology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
| | - Rodwell Mabaera
- Department of Medicine, Dartmouth Hitchcock Medical Center, Lebanon, NH, United States
| |
Collapse
|
5
|
Jeong† M, Kim† JH, Lee JS, Kang SD, Shim S, Jung MY, Yang H, Byun S, Lee KW. Heat-Killed Lactobacillus brevis Enhances Phagocytic Activity and Generates Immune-Stimulatory Effects through Activating the TAK1 Pathway. J Microbiol Biotechnol 2020; 30:1395-1403. [PMID: 32627755 PMCID: PMC9728231 DOI: 10.4014/jmb.2002.02004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 05/26/2020] [Accepted: 06/17/2020] [Indexed: 12/15/2022]
Abstract
There is an increasing interest in using inactivated probiotics to modulate the host immune system and protect against pathogens. As the immunomodulatory function of heat-killed Lactobacillus brevis KCTC 12777BP (LBB) and its mechanism is unclear, we investigated the effect of LBB on immune response based on the hypothesis that LBB might exert stimulatory effects on immunity. In the current study, we demonstrate that administration of LBB can exert immune-stimulatory effects and promote clearance of foreign matters through enhancing phagocytosis. Treatment with LBB induced the production of TNF-α, IL-6, and nitric oxide in macrophages. Importantly, LBB directly increased the phagocytic activity of macrophages against bacterial particles. LBB was able to promote the production of TNF-α in bone marrow-derived macrophages and splenocytes and also increase the proliferation rate of splenocytes, suggesting that the immune-stimulating activity of LBB can be observed in primary immune cells. Investigation into the molecular mechanism responsible revealed that LBB upregulates TAK1 activity and its downstream ERK, p38, and JNK signaling pathways. To further confirm the immunomodulatory capability of LBB in vivo, we orally administered LBB to mice and assessed the effect on primary splenocytes. Splenocytes isolated from LBB-treated mice exhibited higher TNF-α expression and proliferative capacity. These results show that heat-killed L. brevis, a wildly consumed probiotic, may provide protection against pathogens through enhancing host immunity.
Collapse
Affiliation(s)
- Minju Jeong†
- Department of Agricultural Biotechnology, Seoul National University, Seoul 08826, Republic of Korea
| | - Jae Hwan Kim†
- Department of Agricultural Biotechnology, Seoul National University, Seoul 08826, Republic of Korea
| | - Ji Su Lee
- Division of Bioengineering, Incheon National University, Incheon 22012, Republic of Korea
| | - Shin Dal Kang
- Research Institute of Food and Biotechnology, SPC Group, Seoul 151742, Republic of Korea
| | - Sangmin Shim
- Research Institute of Food and Biotechnology, SPC Group, Seoul 151742, Republic of Korea
| | - Moon Young Jung
- Research Institute of Food and Biotechnology, SPC Group, Seoul 151742, Republic of Korea
| | - Hee Yang
- Advanced Institutes of Convergence Technology, Seoul National University, Suwon 16229, Republic of Korea,Center for Food and Bioconvergence, Seoul National University, Seoul 08826, Republic of Korea
| | - Sanguine Byun
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea,Corresponding authors K.W.L. Phone: +82-2-880-4662 E-mail:
| | - Ki Won Lee
- Department of Agricultural Biotechnology, Seoul National University, Seoul 08826, Republic of Korea,Advanced Institutes of Convergence Technology, Seoul National University, Suwon 16229, Republic of Korea,Center for Food and Bioconvergence, Seoul National University, Seoul 08826, Republic of Korea,Corresponding authors K.W.L. Phone: +82-2-880-4662 E-mail:
| |
Collapse
|
6
|
Isoprenylcysteine carboxyl methyltransferase inhibitors exerts anti-inflammatory activity. Biochem Pharmacol 2020; 182:114219. [PMID: 32931773 DOI: 10.1016/j.bcp.2020.114219] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 09/09/2020] [Accepted: 09/10/2020] [Indexed: 11/20/2022]
Abstract
Isoprenylcysteine carboxylmethyltransferase (ICMT) has been reported to regulate the inflammatory response through the Ras/MAPK/AP-1 pathway. Nevertheless, the potential of ICMT inhibitors as therapeutic agents against inflammatory diseases has not been examined. Therefore, in this study, we investigated the anti-inflammatory properties of two ICMT inhibitors, cysmethynil (CyM) and 3-methoxy-N-[2-2,2,6,6-tetramethyl-4-phenyltetrahydropyran-4-yl)ethyl]aniline (MTPA), using in vitro analyses and in vivo analyses (lipopolysaccharide (LPS)/D-GalN-triggered hepatitis and DSS-induced colitis mouse models). CyM and MTPA inhibited the production of nitric oxide (NO) and prostaglandin E (PGE)2 and the expression of cyclooxygenase (COX)-2, tumor necrosis factor (TNF)-α and interleukin (IL)-1β in LPS-induced RAW264.7 cells and peritoneal macrophages without cytotoxicity. CyM also reduced AP-1-mediated luciferase activity in LPS-stimulated RAW264.7 cells and MyD88- and TRIF-expressing HEK293 cells. In addition, CyM and MTPA suppressed the translocation of Ras to the cell membrane and ER as well as phosphorylation of Ras-dependent AP-1 signaling molecules including Raf, MEK1/2, ERK p38, and JNK. Consistent with these results, CyM diminished the expression of inflammatory genes (COX-2, TNF-α, IL-1β, and IL-6), AP-1-Luc activity, and phosphorylation of Ras-mediated signaling enzymes in Ras-overexpressing HEK 293 cells. Moreover, CyM and MTPA ameliorated symptoms of hepatitis and colitis in mice and restrained the ICMT/Ras-dependent AP-1 pathway in inflammatory lesions of the mouse model systems. Taken together, our results indicate that CyM and MTPA alleviate the LPS-induced ICMT/Ras/AP-1 signaling pathway, thereby inhibiting the inflammatory response as promising anti-inflammatory drugs.
Collapse
|
7
|
Faro J, Romero R, Schwenkel G, Garcia-Flores V, Arenas-Hernandez M, Leng Y, Xu Y, Miller D, Hassan SS, Gomez-Lopez N. Intra-amniotic inflammation induces preterm birth by activating the NLRP3 inflammasome†. Biol Reprod 2020; 100:1290-1305. [PMID: 30590393 DOI: 10.1093/biolre/ioy261] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 11/12/2018] [Accepted: 12/22/2018] [Indexed: 01/23/2023] Open
Abstract
Intra-amniotic inflammation is strongly associated with spontaneous preterm labor and birth, the leading cause of perinatal mortality and morbidity worldwide. Previous studies have suggested a role for the NLRP3 (NLR family pyrin domain-containing protein 3) inflammasome in the mechanisms that lead to preterm labor and birth. However, a causal link between the NLRP3 inflammasome and preterm labor/birth induced by intra-amniotic inflammation has not been established. Herein, using an animal model of lipopolysaccharide-induced intra-amniotic inflammation (IAI), we demonstrated that there was priming of the NLRP3 inflammasome (1) at the transcriptional level, indicated by enhanced mRNA expression of inflammasome-related genes (Nlrp3, Casp1, Il1b); and (2) at the protein level, indicated by greater protein concentrations of NLRP3, in both the fetal membranes and decidua basalis prior to preterm birth. Additionally, we showed that there was canonical activation of the NLRP3 inflammasome in the fetal membranes, but not in the decidua basalis, prior to IAI-induced preterm birth as evidenced by increased protein levels of active caspase-1. Protein concentrations of released IL1β were also increased in both the fetal membranes and decidua basalis, as well as in the amniotic fluid, prior to IAI-induced preterm birth. Finally, using the specific NLRP3 inhibitor, MCC950, we showed that in vivo inhibition of the NLRP3 inflammasome reduced IAI-induced preterm birth and neonatal mortality. Collectively, these results provide a causal link between NLRP3 inflammasome activation and spontaneous preterm labor and birth in the context of intra-amniotic inflammation. We also showed that, by targeting the NLRP3 inflammasome, adverse pregnancy and neonatal outcomes can be significantly reduced.
Collapse
Affiliation(s)
- Jonathan Faro
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Roberto Romero
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA.,Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan, USA.,Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, Michigan, USA.,Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan, USA
| | - George Schwenkel
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Valeria Garcia-Flores
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Marcia Arenas-Hernandez
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Yaozhu Leng
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Yi Xu
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Derek Miller
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Sonia S Hassan
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA.,Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Nardhy Gomez-Lopez
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA.,Department of Immunology, Microbiology and Biochemistry, Wayne State University School of Medicine, Detroit, Michigan, USA
| |
Collapse
|
8
|
Škandík M, Mrvová N, Bezek Š, Račková L. Semisynthetic quercetin-quinone mitigates BV-2 microglia activation through modulation of Nrf2 pathway. Free Radic Biol Med 2020; 152:18-32. [PMID: 32142880 DOI: 10.1016/j.freeradbiomed.2020.02.030] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 02/26/2020] [Accepted: 02/27/2020] [Indexed: 12/12/2022]
Abstract
During brain ageing, microglia, the resident immune cells of the CNS, are immunologically activated and contribute to neuroinflammation, a vicious cycle that supports development of neurological disorders. Therapeutic approaches focus mainly on downregulation of their pro-inflammatory activated state that is associated with health benefits. Electrophilic compounds, such as natural quinones and their reduced pro-electrophilic precursors, flavonoids, represent a wide group of diverse substances with important biological effects. They can cause considerable cytotoxicity when used at higher dosages, but on the other hand, they have versatile health benefits at lower dosages. In this study, we investigated the cytotoxicity and prooxidant profile of synthetic conjugate of two electrophilic compounds, quercetin and 1,4-naphthoquinone, 4'-O-(2-chloro-1,4-naphthoquinone-3-yloxy) quercetin (CHNQ), and its attenuation of inflammatory responses and modulation of Nrf2 pathway in BV-2 microglial cells. CHNQ showed higher cytotoxicity than its precursors, accompanied by promotion of production of reactive oxygen species along with G2/M cell cycle arrest at higher concentrations tested. Nevertheless, at a lower non-toxic concentration, CHNQ, more significantly than did its precursors, downregulated LPS-stimulated microglia cells as documented by decreased iNOS, COX-2 and TNFα protein levels. Moreover, CHNQ most effectively upregulated expression of phase II antioxidant enzyme HO-1 and β5 subunit of constitutive proteasome. The enhanced anti-inflammatory effect of CHNQ was accompanied by prominent increase in cytosolic expression of Nrf2 and c-Jun, however, induction effect on nuclear Nrf2 translocation was comparable to QUER. Moreover, a conditioned medium from activated BV-2 cells co-treated with quercetin and CHNQ maintained viability of neuron-like PC12 cells. The compounds tested did not show any disturbance of phagocytosis of live or dead PC12 cells. The present experimental data predict a preventive and therapeutic potential of semisynthetic derivative CHNQ in ageing and related pathologies, mediated by activation of proteins of the antioxidant response.
Collapse
Affiliation(s)
- Martin Škandík
- Centre of Experimental Medicine, Institute of Experimental Pharmacology and Toxicology, Slovak Academy of Sciences, Dubravská cesta 9, 841 04, Bratislava, Slovak Republic
| | - Nataša Mrvová
- Centre of Experimental Medicine, Institute of Experimental Pharmacology and Toxicology, Slovak Academy of Sciences, Dubravská cesta 9, 841 04, Bratislava, Slovak Republic
| | - Štefan Bezek
- Centre of Experimental Medicine, Institute of Experimental Pharmacology and Toxicology, Slovak Academy of Sciences, Dubravská cesta 9, 841 04, Bratislava, Slovak Republic
| | - Lucia Račková
- Centre of Experimental Medicine, Institute of Experimental Pharmacology and Toxicology, Slovak Academy of Sciences, Dubravská cesta 9, 841 04, Bratislava, Slovak Republic.
| |
Collapse
|
9
|
Isoprenylcysteine Carboxyl Methyltransferase and Its Substrate Ras Are Critical Players Regulating TLR-Mediated Inflammatory Responses. Cells 2020; 9:cells9051216. [PMID: 32422978 PMCID: PMC7291029 DOI: 10.3390/cells9051216] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 05/03/2020] [Accepted: 05/11/2020] [Indexed: 12/12/2022] Open
Abstract
In this study, we investigated the functional role of isoprenylcysteine carboxyl methyltransferase (ICMT) and its methylatable substrate Ras in Toll-like receptor (TLR)-activated macrophages and in mouse inflammatory disease conditions. ICMT and RAS expressions were strongly increased in macrophages under the activation conditions of TLRs by lipopolysaccharide (LPS, a TLR4 ligand), pam3CSK (TLR2), or poly(I:C) (TLR3) and in the colons, stomachs, and livers of mice with colitis, gastritis, and hepatitis. The inhibition and activation of ICMT and Ras through genetic and pharmacological approaches significantly affected the activation of interleukin-1 receptor-associated kinase (IRAK)s, tumor necrosis factor receptor associated factor 6 (TRAF6), transforming growth factor-β-activated kinase 1 (TAK1), mitogen-activated protein kinase (MAPK), and MAPK kinases (MAPKKs); translocation of the AP-1 family; and the expressions of inflammation-related genes that depend on both MyD88 and TRIF. Interestingly, the Ras/ICMT-mediated inflammatory reaction critically depends on the TIR domains of myeloid differentiation primary response 88 (MyD88) and TIR-domain-containing adapter-inducing interferon-β (TRIF). Taken together, these results suggest that ICMT and its methylated Ras play important roles in the regulation of inflammatory responses through cooperation with the TIR domain of adaptor molecules.
Collapse
|
10
|
Jain A, Irizarry-Caro RA, McDaniel MM, Chawla AS, Carroll KR, Overcast GR, Philip NH, Oberst A, Chervonsky AV, Katz JD, Pasare C. T cells instruct myeloid cells to produce inflammasome-independent IL-1β and cause autoimmunity. Nat Immunol 2020; 21:65-74. [PMID: 31848486 PMCID: PMC6927526 DOI: 10.1038/s41590-019-0559-y] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 11/12/2019] [Indexed: 12/30/2022]
Abstract
The cytokine interleukin (IL)-1β is a key mediator of antimicrobial immunity as well as autoimmune inflammation. Production of IL-1β requires transcription by innate immune receptor signaling and maturational cleavage by inflammasomes. Whether this mechanism applies to IL-1β production seen in T cell-driven autoimmune diseases remains unclear. Here, we describe an inflammasome-independent pathway of IL-1β production that was triggered upon cognate interactions between effector CD4+ T cells and mononuclear phagocytes (MPs). The cytokine TNF produced by activated CD4+ T cells engaged its receptor TNFR on MPs, leading to pro-IL-1β synthesis. Membrane-bound FasL, expressed by CD4+ T cells, activated death receptor Fas signaling in MPs, resulting in caspase-8-dependent pro-IL-1β cleavage. The T cell-instructed IL-1β resulted in systemic inflammation, whereas absence of TNFR or Fas signaling protected mice from CD4+ T cell-driven autoimmunity. The TNFR-Fas-caspase-8-dependent pathway provides a mechanistic explanation for IL-1β production and its consequences in CD4+ T cell-driven autoimmune pathology.
Collapse
Affiliation(s)
- Aakanksha Jain
- Immunology Graduate Program, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
| | - Ricardo A Irizarry-Caro
- Immunology Graduate Program, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Margaret M McDaniel
- Immunology Graduate Program, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Amanpreet Singh Chawla
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Kaitlin R Carroll
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Garrett R Overcast
- Immunology Graduate Program, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Naomi H Philip
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Andrew Oberst
- Department of Immunology, University of Washington, Seattle, WA, USA
| | | | - Jonathan D Katz
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati, College of Medicine, Cincinnati, OH, USA
| | - Chandrashekhar Pasare
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
- Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
- Department of Pediatrics, University of Cincinnati, College of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
11
|
Shimoyama S, Furukawa T, Ogata Y, Nikaido Y, Koga K, Sakamoto Y, Ueno S, Nakamura K. Lipopolysaccharide induces mouse translocator protein (18 kDa) expression via the AP-1 complex in the microglial cell line, BV-2. PLoS One 2019; 14:e0222861. [PMID: 31536603 PMCID: PMC6752844 DOI: 10.1371/journal.pone.0222861] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Accepted: 09/09/2019] [Indexed: 02/06/2023] Open
Abstract
It has been reported that neuroinflammation occurs in the central nervous system (CNS) in patients with neuropathic pain, Alzheimer’s disease and autism spectrum disorder. The 18-kDa translocator protein TSPO is used as an imaging target in positron emission tomography to detect neuroinflammation, and its expression is correlated with microglial activation. However, the mechanism underlying the transcriptional regulation of Tspo induced by inflammation is not clear. Here, we revealed that lipopolysaccharide (LPS) -induced Tspo expression was activated by the AP-1 complex in a mouse microglial cell line, BV-2. Knockdown of c-Fos and c-Jun, the components of AP-1, reduced LPS-induced Tspo expression. Furthermore, the enrichment of Sp1 in the proximal promoter region of Tspo was increased in the presence of LPS. In addition, the binding of histone deacetylase 1 (HDAC1) to the enhancer region, which contains the AP-1 site, was decreased by LPS treatment, but there were no significant differences in HDAC1 binding to the proximal promoter region with or without LPS. These results indicated that HDAC1 is involved not in the proximal promoter region but in the enhancer region. Our study revealed that inflammatory signals induce the recruitment of AP-1 to the enhancer region and Sp1 to the proximal promoter region of the Tspo gene and that Sp1 may regulate the basal expression of Tspo.
Collapse
Affiliation(s)
- Shuji Shimoyama
- Research Center for Child Mental Development, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
- Department of Neurophysiology, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Tomonori Furukawa
- Department of Neurophysiology, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Yoshiki Ogata
- Department of Neurophysiology, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Yoshikazu Nikaido
- Department of Neurophysiology, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Kohei Koga
- Department of Neurophysiology, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Yui Sakamoto
- Department of Neuropsychiatry, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Shinya Ueno
- Research Center for Child Mental Development, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
- Department of Neurophysiology, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Kazuhiko Nakamura
- Research Center for Child Mental Development, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
- Department of Neuropsychiatry, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
- * E-mail:
| |
Collapse
|
12
|
Yu L, O'Brien VP, Livny J, Dorsey D, Bandyopadhyay N, Colonna M, Caparon MG, Roberson EDO, Hultgren SJ, Hannan TJ. Mucosal infection rewires TNFɑ signaling dynamics to skew susceptibility to recurrence. eLife 2019; 8:46677. [PMID: 31429405 PMCID: PMC6701943 DOI: 10.7554/elife.46677] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 07/20/2019] [Indexed: 12/20/2022] Open
Abstract
A mucosal infectious disease episode can render the host either more or less susceptible to recurrent infection, but the specific mechanisms that tip the balance remain unclear. We investigated this question in a mouse model of recurrent urinary tract infection and found that a prior bladder infection resulted in an earlier onset of tumor necrosis factor-alpha (TNFɑ)-mediated bladder inflammation upon subsequent bacterial challenge, relative to age-matched naive mice. However, the duration of TNFɑ signaling activation differed according to whether the first infection was chronic (Sensitized) or self-limiting (Resolved). TNFɑ depletion studies revealed that transient early-phase TNFɑ signaling in Resolved mice promoted clearance of bladder-colonizing bacteria via rapid recruitment of neutrophils and subsequent exfoliation of infected bladder cells. In contrast, sustained TNFɑ signaling in Sensitized mice prolonged damaging inflammation, worsening infection. This work reveals how TNFɑ signaling dynamics can be rewired by a prior infection to shape diverse susceptibilities to future mucosal infections.
Collapse
Affiliation(s)
- Lu Yu
- Department of Molecular Microbiology and Center for Women's Infectious Disease Research, Washington University School of Medicine, St Louis, United States
| | - Valerie P O'Brien
- Department of Molecular Microbiology and Center for Women's Infectious Disease Research, Washington University School of Medicine, St Louis, United States
| | - Jonathan Livny
- The Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, United States
| | - Denise Dorsey
- Department of Molecular Microbiology and Center for Women's Infectious Disease Research, Washington University School of Medicine, St Louis, United States
| | - Nirmalya Bandyopadhyay
- The Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, United States
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, United States
| | - Michael G Caparon
- Department of Molecular Microbiology and Center for Women's Infectious Disease Research, Washington University School of Medicine, St Louis, United States
| | - Elisha DO Roberson
- Department of Medicine, Division of Rheumatology, Washington University School of Medicine, St Louis, United States.,Department of Genetics, Washington University School of Medicine, St Louis, United States
| | - Scott J Hultgren
- Department of Molecular Microbiology and Center for Women's Infectious Disease Research, Washington University School of Medicine, St Louis, United States
| | - Thomas J Hannan
- Department of Molecular Microbiology and Center for Women's Infectious Disease Research, Washington University School of Medicine, St Louis, United States.,Department of Pathology and Immunology, Washington University School of Medicine, St Louis, United States
| |
Collapse
|
13
|
Signaling mechanisms inducing hyporesponsiveness of phagocytes during systemic inflammation. Blood 2019; 134:134-146. [PMID: 31076441 DOI: 10.1182/blood.2019000320] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 04/30/2019] [Indexed: 12/17/2022] Open
Abstract
The inflammatory responsiveness of phagocytes to exogenous and endogenous stimuli is tightly regulated. This regulation plays an important role in systemic inflammatory response syndromes (SIRSs). In SIRSs, phagocytes initially develop a hyperinflammatory response, followed by a secondary state of hyporesponsiveness, a so-called "tolerance." This hyporesponsiveness can be induced by endotoxin stimulation of Toll-like receptor 4 (TLR4), resulting in an ameliorated response after subsequent restimulation. This modification of inflammatory response patterns has been described as innate immune memory. Interestingly, tolerance can also be triggered by endogenous TLR4 ligands, such as the alarmins myeloid-related protein 8 (MRP8, S100A8) and MRP14 (S100A9), under sterile conditions. However, signaling pathways that trigger hyporesponsiveness of phagocytes in clinically relevant diseases are only barely understood. Through our work, we have now identified 2 main signaling cascades that are activated during MRP-induced tolerance of phagocytes. We demonstrate that the phosphatidylinositol 3-kinase/AKT/GSK-3β pathway interferes with NF-κB-driven gene expression and that inhibition of GSK-3β mimics tolerance in vivo. Moreover, we identified interleukin-10-triggered activation of transcription factors STAT3 and BCL-3 as master regulators of MRP-induced tolerance. Accordingly, patients with dominant-negative STAT3 mutations show no tolerance development. In a clinically relevant condition of systemic sterile stress, cardiopulmonary bypass surgery, we confirmed the initial induction of MRP expression and the tolerance induction of monocytes associated with nuclear translocation of STAT3 and BCL-3 as relevant mechanisms. Our data indicate that the use of pharmacological JAK-STAT inhibitors may be promising targets for future therapeutic approaches to prevent complications associated with secondary hyporesponsiveness during SIRS.
Collapse
|
14
|
Fuller EA, Sominsky L, Sutherland JM, Redgrove KA, Harms L, McLaughlin EA, Hodgson DM. Neonatal immune activation depletes the ovarian follicle reserve and alters ovarian acute inflammatory mediators in neonatal rats. Biol Reprod 2018; 97:719-730. [PMID: 29040417 DOI: 10.1093/biolre/iox123] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 10/07/2017] [Indexed: 12/30/2022] Open
Abstract
Normal ovarian development is crucial for female reproductive success and longevity. Interruptions to the delicate process of initial folliculogenesis may lead to ovarian dysfunction. We have previously demonstrated that an early life immune challenge in the rat, induced by administration of lipopolysaccharide (LPS) on postnatal day (PND) 3 and 5, depletes ovarian follicle reserve long term. Here, we hypothesized that this neonatal immune challenge leads to an increase in peripheral and ovarian inflammatory signaling, contributing to an acute depletion of ovarian follicles. Morphological analysis of neonatal ovaries indicated that LPS administration significantly depleted PND 5 primordial follicle populations and accelerated follicle maturation. LPS exposure upregulated circulating interleukin 6, tumor necrosis factor alpha (TNFa), and C-reactive protein on PND 5, and upregulated ovarian mRNA expression of Tnfa, mitogen-activated protein kinase 8 (Mapk8/Jnk1), and growth differentiation factor 9 (Gdf9) (P < 0.05). Mass spectrometry and cell signaling pathway analysis indicated upregulation of cellular pathways associated with acute phase signaling, and cellular survival and assembly. Apoptosis assessed by terminal deoxynucleotidyl transferase dUTP nick end labeling indicated significantly increased positive staining in the ovaries of LPS-treated neonates. These findings suggest that increased proinflammatory signaling within the neonatal ovary may be responsible for the LPS-induced depletion of the primordial follicle pool. These findings also have implications for female reproductive health, as the ovarian reserve is a major determinate of female reproductive longevity.
Collapse
Affiliation(s)
- Erin A Fuller
- Laboratory of Neuroimmunology, Priority Research Centre for Brain and Mental Health Research, School of Psychology, University of Newcastle, Callaghan, New South Wales, Australia
| | - Luba Sominsky
- School of Health and Biomedical Sciences, Royal Melbourne Institute of Technology University, Melbourne, Victoria, Australia
| | - Jessie M Sutherland
- School of Environmental and Life Sciences, Priority Research Centre in Chemical Biology, University of Newcastle, Callaghan, New South Wales, Australia
| | - Kate A Redgrove
- School of Environmental and Life Sciences, Priority Research Centre in Chemical Biology, University of Newcastle, Callaghan, New South Wales, Australia
| | - Lauren Harms
- Laboratory of Neuroimmunology, Priority Research Centre for Brain and Mental Health Research, School of Psychology, University of Newcastle, Callaghan, New South Wales, Australia
| | - Eileen A McLaughlin
- School of Environmental and Life Sciences, Priority Research Centre in Chemical Biology, University of Newcastle, Callaghan, New South Wales, Australia.,School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | - Deborah M Hodgson
- Laboratory of Neuroimmunology, Priority Research Centre for Brain and Mental Health Research, School of Psychology, University of Newcastle, Callaghan, New South Wales, Australia
| |
Collapse
|
15
|
Shelly A, Banerjee C, Saurav GK, Ray A, Rana VS, Raman R, Mazumder S. Aeromonas hydrophila-induced alterations in cytosolic calcium activate pro-apoptotic cPKC-MEK1/2-TNFα axis in infected headkidney macrophages of Clarias gariepinus. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2017; 76:392-402. [PMID: 28713009 DOI: 10.1016/j.dci.2017.07.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2017] [Revised: 07/12/2017] [Accepted: 07/12/2017] [Indexed: 06/07/2023]
Abstract
Alterations in intracellular-calcium (Ca2+)i homeostasis is critical to Aeromonas hydrophila-induced headkidney macrophages (HKM) apoptosis of Clarias gariepinus, though the implications are poorly understood. Here, we describe the role of intermediate molecules of Ca2+-signaling pathway that are involved in HKM apoptosis. We observed phosphoinositide-3-kinase/phospholipase C is critical for (Ca2+)i release in infected HKM. Heightened protein kinase-C (PKC) activity and phosphorylation of MEK1/2-ERK1/2 was noted which declined in presence of 2-APB, Go6976 and PD98059, inhibitors to IP3-receptor, conventional PKC isoforms (cPKC) and MEK1/2 respectively implicating Ca2+/cPKC/MEK-ERK1/2 axis imperative in A. hydrophila-induced HKM apoptosis. Significant tumor necrosis factor-α (TNFα) production and its subsequent reduction in presence of MEK-ERK1/2 inhibitor U0126 suggested TNFα production downstream to cPKC-mediated signaling via MEK1/2-ERK1/2 pathway. RNAi and inhibitor studies established the role of TNFα in inducing caspase-8-mediated apoptosis of infected HKM. We conclude, alterations in A. hydrophila-induced (Ca2+)i alterations activate cPKC-MEK1/2-ERK1/2-TNFα signaling cascade triggering HKM apoptosis.
Collapse
Affiliation(s)
- Asha Shelly
- Immunobiology Laboratory, Department of Zoology, University of Delhi, Delhi 110 007, India
| | - Chaitali Banerjee
- Immunobiology Laboratory, Department of Zoology, University of Delhi, Delhi 110 007, India
| | - Gunjan Kumar Saurav
- Gut Biology Laboratory, Department of Zoology, University of Delhi, Delhi 110 007, India
| | - Atish Ray
- Immunobiology Laboratory, Department of Zoology, University of Delhi, Delhi 110 007, India
| | - Vipin Singh Rana
- Gut Biology Laboratory, Department of Zoology, University of Delhi, Delhi 110 007, India
| | - Rajagopal Raman
- Gut Biology Laboratory, Department of Zoology, University of Delhi, Delhi 110 007, India
| | - Shibnath Mazumder
- Immunobiology Laboratory, Department of Zoology, University of Delhi, Delhi 110 007, India.
| |
Collapse
|
16
|
Park J, Kwak CH, Ha SH, Kwon KM, Abekura F, Cho SH, Chang YC, Lee YC, Ha KT, Chung TW, Kim CH. Ganglioside GM3 suppresses lipopolysaccharide-induced inflammatory responses in rAW 264.7 macrophage cells through NF-κB, AP-1, and MAPKs signaling. J Cell Biochem 2017; 119:1173-1182. [PMID: 28708322 DOI: 10.1002/jcb.26287] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 07/13/2017] [Indexed: 02/06/2023]
Abstract
Gangliosides are known to specifically inhibit vascular leukocyte recruitment and consequent interaction with the injured endothelium, the basic inflammatory process. In this study, we have found that the production of nitric oxide (NO), a main regulator of inflammation, is suppressed by GM3 on murine macrophage RAW 264.7 cells, when induced by LPS. In addition, GM3 attenuated the increase in cyclooxyenase-2 (COX-2) protein and mRNA levels in lipopolysaccharide (LPS)-activated RAW 264.7 cells in a dose-dependent manner. Moreover, GM3 inhibited the expression and release of pro-inflammatory cytokines of tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), and interleukin-1β (IL-1β) in RAW 264.7 macrophages. At the intracellular level, GM3 inhibited LPS-induced nuclear translocation of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) and activator protein (AP)-1 in RAW 264.7 macrophages. We, therefore, investigated whether GM3 affects mitogen-activated protein kinase (MAPK) phosphorylation, a process known as the upstream signaling regulator. GM3 dramatically reduced the expression levels of the phosphorylated forms of ERK, JNK, and p38 in LPS-activated RAW 264.7 cells. These results indicate that GM3 is a promising suppressor of the vascular inflammatory responses and ganglioside GM3 suppresses the LPS-induced inflammatory response in RAW 264.7 macrophages by suppression of NF-κB, AP-1, and MAPKs signaling. Accordingly, GM3 is suggested as a beneficial agent for the treatment of diseases that are associated with inflammation.
Collapse
Affiliation(s)
- Junyoung Park
- Molecular and Cellular Glycobiology Unit, Department of Biological Sciences, Sungkyunkwan University, Seoburo, Jangan-Gu, Suwon, Gyunggi-Do, Republic of Korea
| | - Choong-Hwan Kwak
- Molecular and Cellular Glycobiology Unit, Department of Biological Sciences, Sungkyunkwan University, Seoburo, Jangan-Gu, Suwon, Gyunggi-Do, Republic of Korea.,Division of Applied Medicine, School of Korean Medicine, Pusan National University, Yangsan City, Gyeongsangnam-Do, Republic of Korea
| | - Sun-Hyung Ha
- Molecular and Cellular Glycobiology Unit, Department of Biological Sciences, Sungkyunkwan University, Seoburo, Jangan-Gu, Suwon, Gyunggi-Do, Republic of Korea
| | - Kyung-Min Kwon
- Molecular and Cellular Glycobiology Unit, Department of Biological Sciences, Sungkyunkwan University, Seoburo, Jangan-Gu, Suwon, Gyunggi-Do, Republic of Korea.,Research Institute, Davinch-K Co., Ltd, Geumcheon-Gu, Seoul, Republic of Korea
| | - Fukushi Abekura
- Molecular and Cellular Glycobiology Unit, Department of Biological Sciences, Sungkyunkwan University, Seoburo, Jangan-Gu, Suwon, Gyunggi-Do, Republic of Korea
| | - Seung-Hak Cho
- Division of Enteric Diseases, Center for Infectious Diseases Research, Korea National Institute of Health, Heungdeok-gu, Cheongju, Republic of Korea
| | - Young-Chae Chang
- Research Institute of Biomedical Engineering and Department of Medicine, Catholic University of Daegu School of Medicine, Daegu, Republic of Korea
| | - Young-Choon Lee
- Faculty of Medicinal Biotechnology, Dong-A University, Saha-Gu, Busan, Republic of Korea
| | - Ki-Tae Ha
- Division of Applied Medicine, School of Korean Medicine, Pusan National University, Yangsan City, Gyeongsangnam-Do, Republic of Korea
| | - Tae-Wook Chung
- Division of Applied Medicine, School of Korean Medicine, Pusan National University, Yangsan City, Gyeongsangnam-Do, Republic of Korea
| | - Cheorl-Ho Kim
- Molecular and Cellular Glycobiology Unit, Department of Biological Sciences, Sungkyunkwan University, Seoburo, Jangan-Gu, Suwon, Gyunggi-Do, Republic of Korea
| |
Collapse
|
17
|
Balkan İA, İlter Akülke AZ, Bağatur Y, Telci D, Gören AC, Kırmızıbekmez H, Yesilada E. Sambulin A and B, non-glycosidic iridoids from Sambucus ebulus, exert significant in vitro anti-inflammatory activity in LPS-induced RAW 264.7 macrophages via inhibition of MAPKs's phosphorylation. JOURNAL OF ETHNOPHARMACOLOGY 2017; 206:347-352. [PMID: 28606808 DOI: 10.1016/j.jep.2017.06.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 05/24/2017] [Accepted: 06/02/2017] [Indexed: 06/07/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The leaves of Sambucus ebulus L. (Adoxaceae) are widely used in Turkish folk medicine particularly against inflammatory disorders. The fresh leaves after wilted over fire or the poultices prepared are directly applied externally to heal burns, edema, eczema, urticarial and abscess. Two iridoids were recently isolated (sambulin A, sambulin B) from the leaves of S. ebulus. AIM OF THE STUDY This study aims to investigate the in vitro anti-inflammatory activities of these iridoids on LPS-induced RAW 264.7 macrophages. MATERIALS AND METHODS Raw 264.7 macrophages were treated with 12.5, 25 and 50µg/ml Sambulin A and 6.25, 12.5 and 25µg/ml Sambulin B and induced with 1µg/ml lipopolysaccaharides (LPS). Effect of the compounds on nitric oxide (NO) production and cytokines (TNFα, IL-6) were determined by Griess and ELISA assays respectively. iNOS and the phosphorylation levels of MAPKs (ERK, JNK) were examined by Western Blot. RESULTS Sambulin A and sambulin B inhibited 52.82% and 72.88% of NO production at 50 and 25µg/ml concentrations respectively. The levels of iNOS were significantly decreased by both molecules, sambulin B at 25µg/ml almost completely decreased iNOS levels (97.53%). Both molecules significantly inhibited TNFα productions. However, only sambulin B inhibited IL-6 production. Consequently, it was shown that sambulin B exerted its effect through the inhibition of ERK and JNK phosphorylations. CONCLUSION The prominent bioactivities exerted by two iridoids will contribute to explanation of the usage of S. ebulus in traditional medicine against rheumatoid diseases.
Collapse
Affiliation(s)
- İrem Atay Balkan
- Yeditepe University, Faculty of Pharmacy, Department of Pharmacognosy and Phytotherapy, 34755 Ataşehir, İstanbul, Turkey.
| | - Ayca Zeynep İlter Akülke
- Yeditepe University, Faculty of Engineering, Department of Genetics and Bioengineering, 34755 Ataşehir, İstanbul, Turkey.
| | - Yeşim Bağatur
- Yeditepe University, Faculty of Engineering, Department of Genetics and Bioengineering, 34755 Ataşehir, İstanbul, Turkey.
| | - Dilek Telci
- Yeditepe University, Faculty of Engineering, Department of Genetics and Bioengineering, 34755 Ataşehir, İstanbul, Turkey.
| | - Ahmet Ceyhan Gören
- TUBITAK National Metrology Institute, Chemistry Group Laboratories, 41470 Gebze, Kocaeli, Turkey.
| | - Hasan Kırmızıbekmez
- Yeditepe University, Faculty of Pharmacy, Department of Pharmacognosy and Phytotherapy, 34755 Ataşehir, İstanbul, Turkey
| | - Erdem Yesilada
- Yeditepe University, Faculty of Pharmacy, Department of Pharmacognosy and Phytotherapy, 34755 Ataşehir, İstanbul, Turkey.
| |
Collapse
|
18
|
Huang X, Kong G, Li Y, Zhu W, Xu H, Zhang X, Li J, Wang L, Zhang Z, Wu Y, Liu X, Wang X. Decitabine and 5-azacitidine both alleviate LPS induced ARDS through anti-inflammatory/antioxidant activity and protection of glycocalyx and inhibition of MAPK pathways in mice. Biomed Pharmacother 2016; 84:447-453. [DOI: 10.1016/j.biopha.2016.09.072] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Revised: 09/19/2016] [Accepted: 09/19/2016] [Indexed: 11/25/2022] Open
|
19
|
Vodovotz Y, Shubing Liu, McCloskey C, Shapiro R, Green A, Billiar TR. The hepatocyte as a microbial product-responsive cell. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/09680519010070050401] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Much research has focused on the responses to microbial products of immune cells such as monocytes, macrophages, and neutrophils. Although the liver is a primary response organ in various infections, relatively little is known about the antimicrobial responses of its major cell type, the hepatocyte. It is now known that the recognition of bacteria occurs via cell-surface proteins that are members of the Toll-like receptor (TLR) family. In addition, lipopolysaccharide (LPS) is bound by circulating LPS-binding protein (LBP) and presented to cell-surface CD14, which in turn interacts with TLR and transduces an intracellular signal. We investigated the CD14 and TLR2 responses of whole liver and isolated hepatocytes, and demonstrated that these cells can be induced to express the molecules necessary for responses to both Gram-positive and Gram-negative bacteria. Our findings may have clinical implications for pathological states such as sepsis.
Collapse
Affiliation(s)
- Yoram Vodovotz
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Shubing Liu
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Carol McCloskey
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Richard Shapiro
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Angela Green
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Timothy R. Billiar
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA,
| |
Collapse
|
20
|
Guyton K, Bond R, Romeo C, Southern R, Cochran J, Teti G, Cook JA. Endotoxin-induced cross-tolerance to Gram-positive sepsis. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/09680519990050030501] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The manifestations of Gram-positive sepsis and Gram-negative sepsis share some common clinical features suggesting common pathways of activation. The goal of this study was to assess whether lipopolysaccharide (LPS) can produce cross-tolerance to Gram-positive sepsis induced by group B streptococcus (GBS). Thromboxane (TxB2), tumor necrosis factor (TNFα), and nitric oxide (NO) production by in vitro LPS- and heat killed GBS-stimulated rat peritoneal macrophages were measured. Since our previous studies have demonstrated altered macrophage activation of extracellular signal-regulated kinases 1 and 2 (ERK 1/2) in tolerance, we also examined the effect of LPS and killed GBS on ERK 1/2 activation in normal and LPS tolerant macrophages. Tolerance was induced in rats by intraperitoneal injection of Salmonella enteritidis LPS or vehicle for two consecutive days at doses of 0.1 and 0.5 mg/kg body weight. Three days after the second LPS dose, rats were injected intravenously with viable GBS (5 x l09 cfu/kg) and D-galactosamine (1 g/kg). LPS tolerance significantly prolonged ( P <0.05) mean survival time to severe GBS sepsis in D-galactosamine sensitized rats from 12.9 ± 1.7 h in control rats to 44.0 ± 8.9 h in tolerant rats. Peritoneal macrophages from LPS tolerant rats exhibited suppressed LPS induced in vitro TxB2 and TNFα production ( P <0.05). Tolerance also decreased in vitro heat killed GBS-induced TNFα production, but did not significantly affect TxB2 production. NO production stimulated by LPS (10 µg/ml was not impaired in LPS tolerance; however at lower doses (0.02—1.25 µg/ml), NO production was significantly decreased ( P <0.05). NO production was augmented ( P <0.05) in response to stimulation with GBS (10 µg/ml) and unaltered at lower doses (0.02—1.25 µg/ml) in tolerant cells. LPS activated ERK 1/2 in control macrophages, but activation of ERK 1/2 was suppressed in LPS tolerance. GBS did not significantly affect ERK 1/2 activity in control or tolerant macrophages. Nevertheless, the selective mitogen-activated kinase (MAPK)/ERK kinase (MEK) inhibitor, PD 98059 blocked ( P <0.05) both GBS- and LPS-induced TNFα and TxB2 production, but not NO production. Thus, some level of ERK 1/2 activity appears essential for GBS- and LPS-induced macrophage activation. In conclusion, LPS tolerance induces partial cross-tolerance to Gram-positive sepsis induced lethality, and alters LPS- and GBS-induced in vitro peritoneal macrophage mediator production. This suggests common pathways of cellular activation for GBS and LPS that are altered by LPS tolerance.
Collapse
Affiliation(s)
- Kelly Guyton
- Departments of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Robert Bond
- Department of Physiology, University of South Carolina School of Medicine, Columbia, South Carolina, USA
| | - Cristina Romeo
- Department of Microbiology, Medical University of Messina, Messina, Italy
| | - Rodney Southern
- Department of Physiology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Joel Cochran
- Department of Pediatrics, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Giuseppe Teti
- Department of Microbiology, Medical University of Messina, Messina, Italy
| | - James A. Cook
- Department of Physiology, Medical University of South Carolina, Charleston, South Carolina, USA
| |
Collapse
|
21
|
Amoureux MC, Hegyi E, Le D, Grandics P, Tong H, Szathmary S. A new method for removing endotoxin from plasma using hemocompatible affinity chromatography technology, applicable for extracorporeal treatment of septic patients. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/09680519040100020401] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The pathogenesis of sepsis begins with the proliferation of micro-organisms at a site of infection, followed by invasion of the bloodstream and other organs. Gram-negative bacteria account for a large part of sepsis cases. The structural component of Gram-negative bacteria, endotoxin or lipopolysaccharide (LPS), induces the synthesis and release of endogenous mediators of sepsis. A growing number of investigations of the molecular mechanisms occurring in sepsis, point to endotoxin as a central mediator leading to multi-organ failure and death. In numerous clinical trials, attempts to target molecules downstream of endotoxin have been made, but have not been associated with improved survival. We describe an affinity-based system for the selective removal of endotoxin from plasma. The small-scale device, a 1.5 ml cartridge, contains beads that bind endotoxin with high specificity and efficiency. In addition, evidence is presented that this device does not affect plasma hemostasis, nor does it activate the complement system. Taken together, these results represent a proof of principle for endotoxin removal from plasma, which may be of clinical value to treat sepsis by extracorporeal circulation of the blood through a scaled-up version of this endotoxin-removing device.
Collapse
Affiliation(s)
| | - Edit Hegyi
- Clarigen Inc., Carlsbad, California, USA
| | - Dzung Le
- Clarigen Inc., Carlsbad, California, USA
| | | | - Hung Tong
- Clarigen Inc., Carlsbad, California, USA
| | | |
Collapse
|
22
|
Lappas M. RAF1 is increased in labouring myometrium and modulates inflammation-induced pro-labour mediators. Reproduction 2016; 151:411-20. [DOI: 10.1530/rep-15-0607] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 01/25/2016] [Indexed: 01/25/2023]
Abstract
Inflammation plays a central role in the terminal process of human labour and delivery, including myometrial contractions. RAF1 proto-oncogene serine/threonine-protein kinase (RAF1) can activate ERK (official gene symbolMAPK1) and/or nuclear factor-kappa B (NF-κB) to regulate genes involved in inflammation. There are, however, no studies on the role of RAF1 in the processes of human labour and delivery. Thus, the aims of this study were to determine the effect of i) human labour and pro-inflammatory cytokines interleukin 1 beta (IL1B) and tumour necrosis factor (TNF) alpha on RAF1 protein expression in myometrium and ii) siRNA knockdown ofRAF1on pro-inflammatory and pro-labour mediators in human myometrial primary cells. Term labour was associated with an increase in RAF1 protein expression. Furthermore, RAF1 protein expression was increased in myometrial cells treated with IL1B and TNF, two likely factors contributing to preterm birth. Knockdown ofRAF1by siRNA in primary myometrial cells significantly decreased IL1B- and TNF-inducedIL1A, IL1B, IL6,(C-X-C motif) ligand 8 (CXCL8)and chemokine (C-C motif) ligand 2 (CCL2) mRNA abundance and IL6, IL8 and CCL2; prostaglandin-endoperoxide synthase 2 (PTGS2) mRNA levels and prostaglandin PGF2αrelease; and NF-κB activation. Furthermore,RAF1knockdown was associated with decreased activation of ERK in the presence of IL1B but not TNF. Concordantly, the ERK inhibitor U0126 significantly decreased IL1B-inducedIL6,CXCL8,CCL2andPTGS2mRNA abundance; IL6, CXCL8, CCL2 and PGF2αrelease; and NF-κB activation. In conclusion, IL1B induces the expression and secretion of pro-labour mediators through the RAF1–MAPK1–NF-κB signalling pathway. TNF, on the other hand, regulates pro-labour mediators through the RAF1–NF-κB signalling pathway via an MAPK1-independent mechanism.
Collapse
|
23
|
Tang L, Okamoto S, Shiuchi T, Toda C, Takagi K, Sato T, Saito K, Yokota S, Minokoshi Y. Sympathetic Nerve Activity Maintains an Anti-Inflammatory State in Adipose Tissue in Male Mice by Inhibiting TNF-α Gene Expression in Macrophages. Endocrinology 2015; 156:3680-94. [PMID: 26132918 DOI: 10.1210/en.2015-1096] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Adipose tissue macrophages (ATMs) play an important role in the inflammatory response in obese animals. How ATMs are regulated in lean animals has remained elusive, however. We now show that the sympathetic nervous system (SNS) is necessary to maintain the abundance of the mRNA for the proinflammatory cytokine TNF-α at a low level in ATMs of lean mice. Intracerebroventricular injection of agouti-related neuropeptide increased the amount of TNF-α mRNA in epididymal (epi) white adipose tissue (WAT), but not in interscapular brown adipose tissue (BAT), through inhibition of sympathetic nerve activity in epiWAT. The surgical denervation and β-adrenergic antagonist propranolol up-regulated TNF-α mRNA in both epiWAT and BAT in vivo. Signaling by the β2-adrenergic receptor (AR) and protein kinase A down-regulated TNF-α mRNA in epiWAT explants and suppressed lipopolysaccharide-induced up-regulation of TNF-α mRNA in the stromal vascular fraction of this tissue. β-AR-deficient (β-less) mice manifested an increased plasma TNF-α concentration and increased TNF-α mRNA abundance in epiWAT and BAT. TNF-α mRNA abundance was greater in ATMs (CD11b(+) cells of the stromal vascular fraction) from epiWAT or BAT of wild-type mice than in corresponding CD11b(-) cells, and β2-AR mRNA abundance was greater in ATMs than in CD11b(-) cells of epiWAT. Our results show that the SNS and β2-AR-protein kinase A pathway maintain an anti-inflammatory state in ATMs of lean mice in vivo, and that the brain melanocortin pathway plays a role in maintaining this state in WAT of lean mice via the SNS.
Collapse
MESH Headings
- Adipose Tissue, Brown/drug effects
- Adipose Tissue, Brown/innervation
- Adipose Tissue, Brown/metabolism
- Adipose Tissue, White/drug effects
- Adipose Tissue, White/innervation
- Adipose Tissue, White/metabolism
- Adrenergic beta-Antagonists/pharmacology
- Agouti-Related Protein/administration & dosage
- Animals
- Cell Line
- Epididymis/drug effects
- Epididymis/metabolism
- Gene Expression/drug effects
- Immunoblotting
- Inflammation Mediators/metabolism
- Injections, Intraventricular
- Macrophages/metabolism
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Peptide Fragments/administration & dosage
- Propranolol/pharmacology
- Receptors, Adrenergic, beta/genetics
- Receptors, Adrenergic, beta/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Sympathectomy
- Sympathetic Nervous System/metabolism
- Tumor Necrosis Factor-alpha/genetics
- Tumor Necrosis Factor-alpha/metabolism
Collapse
Affiliation(s)
- Lijun Tang
- Division of Endocrinology and Metabolism (L.T., S.O., K.T., T.Sa., K.S., S.Y., Y.M.), Department of Developmental Physiology, National Institute for Physiological Sciences, Okazaki, Aichi 444-8585, Japan; Department of Physiological Sciences (S.O., T.Sa., Y.M.), Sokendai (The Graduate University for Advanced Studies), Hayama, Kanagawa 240-0193, Japan; Department of Integrative Physiology (T.Sh.), Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima City, Tokushima 770-8503, Japan; and Section of Comparative Medicine (C.T.), Yale University School of Medicine, New Haven, Connecticut 06520-8016
| | - Shiki Okamoto
- Division of Endocrinology and Metabolism (L.T., S.O., K.T., T.Sa., K.S., S.Y., Y.M.), Department of Developmental Physiology, National Institute for Physiological Sciences, Okazaki, Aichi 444-8585, Japan; Department of Physiological Sciences (S.O., T.Sa., Y.M.), Sokendai (The Graduate University for Advanced Studies), Hayama, Kanagawa 240-0193, Japan; Department of Integrative Physiology (T.Sh.), Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima City, Tokushima 770-8503, Japan; and Section of Comparative Medicine (C.T.), Yale University School of Medicine, New Haven, Connecticut 06520-8016
| | - Tetsuya Shiuchi
- Division of Endocrinology and Metabolism (L.T., S.O., K.T., T.Sa., K.S., S.Y., Y.M.), Department of Developmental Physiology, National Institute for Physiological Sciences, Okazaki, Aichi 444-8585, Japan; Department of Physiological Sciences (S.O., T.Sa., Y.M.), Sokendai (The Graduate University for Advanced Studies), Hayama, Kanagawa 240-0193, Japan; Department of Integrative Physiology (T.Sh.), Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima City, Tokushima 770-8503, Japan; and Section of Comparative Medicine (C.T.), Yale University School of Medicine, New Haven, Connecticut 06520-8016
| | - Chitoku Toda
- Division of Endocrinology and Metabolism (L.T., S.O., K.T., T.Sa., K.S., S.Y., Y.M.), Department of Developmental Physiology, National Institute for Physiological Sciences, Okazaki, Aichi 444-8585, Japan; Department of Physiological Sciences (S.O., T.Sa., Y.M.), Sokendai (The Graduate University for Advanced Studies), Hayama, Kanagawa 240-0193, Japan; Department of Integrative Physiology (T.Sh.), Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima City, Tokushima 770-8503, Japan; and Section of Comparative Medicine (C.T.), Yale University School of Medicine, New Haven, Connecticut 06520-8016
| | - Kazuyo Takagi
- Division of Endocrinology and Metabolism (L.T., S.O., K.T., T.Sa., K.S., S.Y., Y.M.), Department of Developmental Physiology, National Institute for Physiological Sciences, Okazaki, Aichi 444-8585, Japan; Department of Physiological Sciences (S.O., T.Sa., Y.M.), Sokendai (The Graduate University for Advanced Studies), Hayama, Kanagawa 240-0193, Japan; Department of Integrative Physiology (T.Sh.), Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima City, Tokushima 770-8503, Japan; and Section of Comparative Medicine (C.T.), Yale University School of Medicine, New Haven, Connecticut 06520-8016
| | - Tatsuya Sato
- Division of Endocrinology and Metabolism (L.T., S.O., K.T., T.Sa., K.S., S.Y., Y.M.), Department of Developmental Physiology, National Institute for Physiological Sciences, Okazaki, Aichi 444-8585, Japan; Department of Physiological Sciences (S.O., T.Sa., Y.M.), Sokendai (The Graduate University for Advanced Studies), Hayama, Kanagawa 240-0193, Japan; Department of Integrative Physiology (T.Sh.), Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima City, Tokushima 770-8503, Japan; and Section of Comparative Medicine (C.T.), Yale University School of Medicine, New Haven, Connecticut 06520-8016
| | - Kumiko Saito
- Division of Endocrinology and Metabolism (L.T., S.O., K.T., T.Sa., K.S., S.Y., Y.M.), Department of Developmental Physiology, National Institute for Physiological Sciences, Okazaki, Aichi 444-8585, Japan; Department of Physiological Sciences (S.O., T.Sa., Y.M.), Sokendai (The Graduate University for Advanced Studies), Hayama, Kanagawa 240-0193, Japan; Department of Integrative Physiology (T.Sh.), Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima City, Tokushima 770-8503, Japan; and Section of Comparative Medicine (C.T.), Yale University School of Medicine, New Haven, Connecticut 06520-8016
| | - Shigefumi Yokota
- Division of Endocrinology and Metabolism (L.T., S.O., K.T., T.Sa., K.S., S.Y., Y.M.), Department of Developmental Physiology, National Institute for Physiological Sciences, Okazaki, Aichi 444-8585, Japan; Department of Physiological Sciences (S.O., T.Sa., Y.M.), Sokendai (The Graduate University for Advanced Studies), Hayama, Kanagawa 240-0193, Japan; Department of Integrative Physiology (T.Sh.), Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima City, Tokushima 770-8503, Japan; and Section of Comparative Medicine (C.T.), Yale University School of Medicine, New Haven, Connecticut 06520-8016
| | - Yasuhiko Minokoshi
- Division of Endocrinology and Metabolism (L.T., S.O., K.T., T.Sa., K.S., S.Y., Y.M.), Department of Developmental Physiology, National Institute for Physiological Sciences, Okazaki, Aichi 444-8585, Japan; Department of Physiological Sciences (S.O., T.Sa., Y.M.), Sokendai (The Graduate University for Advanced Studies), Hayama, Kanagawa 240-0193, Japan; Department of Integrative Physiology (T.Sh.), Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima City, Tokushima 770-8503, Japan; and Section of Comparative Medicine (C.T.), Yale University School of Medicine, New Haven, Connecticut 06520-8016
| |
Collapse
|
24
|
Li Y, Zhang H, Kosturakis AK, Cassidy RM, Zhang H, Kennamer-Chapman RM, Jawad AB, Colomand CM, Harrison DS, Dougherty PM. MAPK signaling downstream to TLR4 contributes to paclitaxel-induced peripheral neuropathy. Brain Behav Immun 2015; 49:255-66. [PMID: 26065826 PMCID: PMC4567501 DOI: 10.1016/j.bbi.2015.06.003] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Revised: 06/02/2015] [Accepted: 06/02/2015] [Indexed: 01/07/2023] Open
Abstract
Toll-like receptor 4 (TLR4) has been implicated as a locus for initiation of paclitaxel related chemotherapy induced peripheral neuropathy (CIPN). This project explores the involvement of the immediate down-stream signal molecules in inducing paclitaxel CIPN. Mitogen-activated protein kinases (MAPKs) and nuclear factor-κB (NFκB) were measured in dorsal root ganglia (DRG) and the spinal cord over time using Western blot and immunohistochemistry in a rat model of paclitaxel CIPN. The effects of MAPK inhibitors in preventing and reversing behavioral signs of CIPN were also measured (group sizes 4-9). Extracellular signal related kinase (ERK1/2) and P38 but not c-Jun N terminal kinase (JNK) or PI3K-Akt signaling expression was increased in DRG. Phospho-ERK1/2 staining was co-localized to small CGRP-positive DRG neurons in cell profiles surrounding large DRG neurons consistent with satellite glial cells. The expression of phospho-P38 was co-localized to small IB4-positive and CGRP-positive DRG neurons. The TLR4 antagonist LPS derived from Rhodobacter sphaeroides (LPS-RS) inhibited paclitaxel-induced phosphorylation of ERK1/2 and P38. The MAPK inhibitors PD98059 (MEK1/2), U0126 (MEK1/2) and SB203580 (P38) prevented but did not reverse paclitaxel-induced behavioral hypersensitivity. Paclitaxel treatment resulted in phosphorylation of Inhibitor α of NFκB (IκBα) in DRG resulting in an apparent release of NFκB from the IκBα-NFκB complex as increased expression of nuclear NFκB was also observed. LPS-RS inhibited paclitaxel-induced translocation of NFκB in DRG. No change was observed in spinal NFκB. These results implicate TLR4 signaling via MAP kinases and NFκB in the induction and maintenance of paclitaxel-related CIPN.
Collapse
Affiliation(s)
- Yan Li
- Department of Anesthesia and Pain Medicine Research, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030
| | - Hongmei Zhang
- Department of Anesthesia and Pain Medicine Research, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030
| | - Alyssa K. Kosturakis
- Department of Anesthesia and Pain Medicine Research, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030,The University of Texas Health Science Center, San Antonio, Texas 78229
| | - Ryan M. Cassidy
- The University of Texas Health Science Center, Houston, Texas 77030
| | - Haijun Zhang
- Department of Anesthesia and Pain Medicine Research, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030,Department of Anesthesiology, The University of Texas Medical School at Houston, Houston, Texas 77030
| | | | | | | | | | - Patrick M. Dougherty
- Department of Anesthesia and Pain Medicine Research, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030
| |
Collapse
|
25
|
Tang S, Chen T, Yang M, Wang L, Yu Z, Xie B, Qian C, Xu S, Li N, Cao X, Wang J. Extracellular calcium elicits feedforward regulation of the Toll-like receptor-triggered innate immune response. Cell Mol Immunol 2015; 14:180-191. [PMID: 26277896 DOI: 10.1038/cmi.2015.59] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2015] [Revised: 05/30/2015] [Accepted: 05/31/2015] [Indexed: 12/13/2022] Open
Abstract
Despite the expanding knowledge on feedback regulation of Toll-like receptor (TLR) signaling, the feedforward regulation of TLR signaling for the proper innate response to invading microbes is not fully understood. Here, we report that extracellular calcium can coordinate the activation of the small GTPases Ras and Ras-proximate-1 (Rap1) upon TLR stimulation which favors activation of macrophages through a feedforward mechanism. We show that different doses of TLR agonists can trigger different levels of cytokine production, which can be potentiated by extracellular calcium but are impaired by the chelating reagent ethylene glycol tetraacetic acid (EGTA) or by knockdown of stromal interaction molecule 1 (STIM1). Upon TLR engagement, GTP-bound Ras levels are increased and GTP-bound Rap1 is decreased, which can be reversed by EGTA-mediated removal of extracellular calcium. Furthermore, we demonstrate that Rap1 knockdown rescues the inhibitory effects of EGTA on the TLR-triggered innate response. Examination of the TLR signaling pathway reveals that extracellular calcium may regulate the TLR response via feedforward activation of the extracellular signal-regulated kinase signaling pathway. Our data suggest that an influx of extracellular calcium, mediated by STIM1-operated calcium channels, may transmit the information about the intensity of extracellular TLR stimuli to initiate innate responses at an appropriate level. Our study may provide mechanistic insight into the feedforward regulation of the TLR-triggered innate immune response.
Collapse
Affiliation(s)
- Songqing Tang
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou 310058, China.,National Key Laboratory of Medical Immunology & Institute of Immunology, Second Military Medical University, Shanghai 200433, China
| | - Taoyong Chen
- National Key Laboratory of Medical Immunology & Institute of Immunology, Second Military Medical University, Shanghai 200433, China
| | - Mingjin Yang
- National Key Laboratory of Medical Immunology & Institute of Immunology, Second Military Medical University, Shanghai 200433, China.,National Key Laboratory of Medical Molecular Biology & Department of Immunology, Chinese Academy of Medical Sciences, Beijing 100005, China
| | - Lei Wang
- National Key Laboratory of Medical Molecular Biology & Department of Immunology, Chinese Academy of Medical Sciences, Beijing 100005, China
| | - Zhou Yu
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Bin Xie
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou 310058, China.,National Key Laboratory of Medical Immunology & Institute of Immunology, Second Military Medical University, Shanghai 200433, China
| | - Cheng Qian
- National Key Laboratory of Medical Immunology & Institute of Immunology, Second Military Medical University, Shanghai 200433, China
| | - Sheng Xu
- National Key Laboratory of Medical Immunology & Institute of Immunology, Second Military Medical University, Shanghai 200433, China
| | - Nan Li
- National Key Laboratory of Medical Immunology & Institute of Immunology, Second Military Medical University, Shanghai 200433, China
| | - Xuetao Cao
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou 310058, China.,National Key Laboratory of Medical Immunology & Institute of Immunology, Second Military Medical University, Shanghai 200433, China.,National Key Laboratory of Medical Molecular Biology & Department of Immunology, Chinese Academy of Medical Sciences, Beijing 100005, China
| | - Jianli Wang
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou 310058, China
| |
Collapse
|
26
|
Shi-Lin D, Yuan X, Zhan S, Luo-Jia T, Chao-Yang T. Trametinib, a novel MEK kinase inhibitor, suppresses lipopolysaccharide-induced tumor necrosis factor (TNF)-α production and endotoxin shock. Biochem Biophys Res Commun 2015; 458:667-673. [PMID: 25684183 DOI: 10.1016/j.bbrc.2015.01.160] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 01/27/2015] [Indexed: 12/19/2022]
Abstract
Lipopolysaccharide (LPS), one of the most prominent pathogen-associated molecular patterns (PAMPs), activates macrophages, causing release of toxic cytokines (i.e. tumor necrosis factor (TNF)-α) that may provoke inflammation and endotoxin shock. Here, we tested the potential role of trametinib, a novel and highly potent MAPK/ERK kinase (MEK) inhibitor, against LPS-induced TNF-α response in monocytes, and analyzed the underlying mechanisms. We showed that trametinib, at nM concentrations, dramatically inhibited LPS-induced TNF-α mRNA expression and protein secretion in transformed (RAW 264.7 cells) and primary murine macrophages. In ex-vivo cultured human peripheral blood mononuclear cells (PBMCs), this MEK inhibitor similarly suppressed TNF-α production by LPS. For the mechanism study, we found that trametinib blocked LPS-induced MEK-ERK activation in above monocytes, which accounted for the defective TNF-α response. Macrophages or PBMCs treated with a traditional MEK inhibitor PD98059 or infected with MEK1/2-shRNA lentivirus exhibited a similar defect as trametinib, and nullified the activity of trametinib. On the other hand, introducing a constitutively-active (CA) ERK1 restored TNF-α production by LPS in the presence of trametinib. In vivo, mice administrated with trametinib produced low levels of TNF-α after LPS stimulation, and these mice were protected from LPS-induced endotoxin shock. Together, these results show that trametinib inhibits LPS-induced TNF-α expression and endotoxin shock probably through blocking MEK-ERK signaling.
Collapse
Affiliation(s)
- Du Shi-Lin
- Department of Emergency, Zhongshan Hospital, Shanghai Medical School, Fudan University, Shanghai, China
| | - Xue Yuan
- Department of Emergency, Zhongshan Hospital, Shanghai Medical School, Fudan University, Shanghai, China
| | - Sun Zhan
- Department of Emergency, Zhongshan Hospital, Shanghai Medical School, Fudan University, Shanghai, China
| | - Tang Luo-Jia
- Department of Emergency, Zhongshan Hospital, Shanghai Medical School, Fudan University, Shanghai, China
| | - Tong Chao-Yang
- Department of Emergency, Zhongshan Hospital, Shanghai Medical School, Fudan University, Shanghai, China.
| |
Collapse
|
27
|
Huang BP, Lin CH, Chen HM, Lin JT, Cheng YF, Kao SH. AMPK Activation Inhibits Expression of Proinflammatory Mediators Through Downregulation of PI3K/p38 MAPK and NF-κB Signaling in Murine Macrophages. DNA Cell Biol 2015; 34:133-41. [DOI: 10.1089/dna.2014.2630] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Bee-Piao Huang
- Department of Pathology, Tungs' Taichung MetroHarbor Hospital, Taichung, Taiwan
| | - Chun-Hsiang Lin
- Institute of Biochemistry and Biotechnology, Chung Shan Medical University, Taichung, Taiwan
| | - Han-Min Chen
- Department of Life Science, Catholic Fu-Jen University, New Taipei City, Taiwan
- Institute of Applied Science and Engineering, Catholic Fu-Jen University, New Taipei City, Taiwan
- Energenesis Biomedical Co. Ltd., New Taipei City, Taiwan
| | - Jiun-Tsai Lin
- Energenesis Biomedical Co. Ltd., New Taipei City, Taiwan
| | - Yi-Fang Cheng
- Energenesis Biomedical Co. Ltd., New Taipei City, Taiwan
| | - Shao-Hsuan Kao
- Institute of Biochemistry and Biotechnology, Chung Shan Medical University, Taichung, Taiwan
- Clinical Laboratory, Chung Shan Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
28
|
Simma N, Bose T, Kahlfuss S, Mankiewicz J, Lowinus T, Lühder F, Schüler T, Schraven B, Heine M, Bommhardt U. NMDA-receptor antagonists block B-cell function but foster IL-10 production in BCR/CD40-activated B cells. Cell Commun Signal 2014; 12:75. [PMID: 25477292 PMCID: PMC4269920 DOI: 10.1186/s12964-014-0075-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Accepted: 11/12/2014] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND B cells are important effectors and regulators of adaptive and innate immune responses, inflammation and autoimmunity, for instance in anti-NMDA-receptor (NMDAR) encephalitis. Thus, pharmacological modulation of B-cell function could be an effective regimen in therapeutic strategies. Since the non-competitive NMDAR antagonist memantine is clinically applied to treat advanced Alzheimer`s disease and ketamine is supposed to improve the course of resistant depression, it is important to know how these drugs affect B-cell function. RESULTS Non-competitive NMDAR antagonists impaired B-cell receptor (BCR)- and lipopolysaccharide (LPS)-induced B-cell proliferation, reduced B-cell migration towards the chemokines SDF-1α and CCL21 and downregulated IgM and IgG secretion. Mechanistically, these effects were mediated through a blockade of Kv1.3 and KCa3.1 potassium channels and resulted in an attenuated Ca(2+)-flux and activation of Erk1/2, Akt and NFATc1. Interestingly, NMDAR antagonist treatment increased the frequency of IL-10 producing B cells after BCR/CD40 stimulation. CONCLUSIONS Non-competitive NMDAR antagonists attenuate BCR and Toll-like receptor 4 (TLR4) B-cell signaling and effector function and can foster IL-10 production. Consequently, NMDAR antagonists may be useful to target B cells in autoimmune diseases or pathological systemic inflammation. The drugs' additional side effects on B cells should be considered in treatments of neuronal disorders with NMDAR antagonists.
Collapse
Affiliation(s)
- Narasimhulu Simma
- Institute of Molecular and Clinical Immunology, Otto-von-Guericke-University Magdeburg, Leipziger Str. 44, 39120, Magdeburg, Germany.
| | - Tanima Bose
- RG Molecular Physiology, Leibniz Institute of Neurobiology, Brenneckestr. 6, 39118, Magdeburg, Germany.
| | - Sascha Kahlfuss
- Institute of Molecular and Clinical Immunology, Otto-von-Guericke-University Magdeburg, Leipziger Str. 44, 39120, Magdeburg, Germany.
| | - Judith Mankiewicz
- Institute of Molecular and Clinical Immunology, Otto-von-Guericke-University Magdeburg, Leipziger Str. 44, 39120, Magdeburg, Germany.
| | - Theresa Lowinus
- Institute of Molecular and Clinical Immunology, Otto-von-Guericke-University Magdeburg, Leipziger Str. 44, 39120, Magdeburg, Germany.
| | - Fred Lühder
- Department of Neuroimmunology, Institute for Multiple Sclerosis Research and The Hertie Foundation, Waldweg 33, 37073, Göttingen, Germany.
| | - Thomas Schüler
- Institute of Molecular and Clinical Immunology, Otto-von-Guericke-University Magdeburg, Leipziger Str. 44, 39120, Magdeburg, Germany.
| | - Burkhart Schraven
- Institute of Molecular and Clinical Immunology, Otto-von-Guericke-University Magdeburg, Leipziger Str. 44, 39120, Magdeburg, Germany. .,Department of Immune Control, Helmholtz Centre for Infection Research, Inhoffenstr. 7, 38124, Braunschweig, Germany.
| | - Martin Heine
- RG Molecular Physiology, Leibniz Institute of Neurobiology, Brenneckestr. 6, 39118, Magdeburg, Germany.
| | - Ursula Bommhardt
- Institute of Molecular and Clinical Immunology, Otto-von-Guericke-University Magdeburg, Leipziger Str. 44, 39120, Magdeburg, Germany.
| |
Collapse
|
29
|
Hull M, Lee E, Lee T, Anand N, LaLone V, Parameswaran N. Lithium chloride induces TNFα in mouse macrophages via MEK-ERK-dependent pathway. J Cell Biochem 2014; 115:71-80. [PMID: 23904208 DOI: 10.1002/jcb.24634] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Accepted: 07/19/2013] [Indexed: 12/12/2022]
Abstract
Lithium (Li) is one of the currently prescribed drugs for bipolar disorders (BPDs) and has many neuro-regulatory and immune-modulating properties. Because many neuro-pathological diseases including BPDs have been associated with some level of inflammation, Li's effect on inflammation may have some crucial consequences. Even though Li has been shown to have pro- and anti-inflammatory activities in different cell models, mechanisms involved in these effects are not well understood. Moreover, Li's effect on inflammation in the presence of activators of Toll-like receptors (TLRs), especially TLR-2 (that activates MyD88-dependent pathway) and TLR-3 (that activates TRIF-dependent pathway) is not known. Here we tested the role of Li in the presence and absence of TLR2, and TLR3 on MAPK and NFκB pathways and the consequent production of tumor necrosis factor-α (TNFα) in Raw264.7 macrophages. Our results indicate that Li enhances TNFα production both in the absence and presence of TLR stimulation. Interestingly, Li differentially modulates MAPK and NFκB pathways in the absence and presence of TLR2/3 ligands. Our results further indicate that the effect of Li on TNFα occurs at the post-transcriptional level. Together, these studies demonstrate that Li induces TNFα production in macrophages and that it modulates signaling at different levels depending on the presence or absence of TLR2/3 stimulation.
Collapse
Affiliation(s)
- Megan Hull
- Department of Physiology and Division of Pathology, Michigan State University, East Lansing, Michigan, USA
| | - Eunhee Lee
- Department of Physiology and Division of Pathology, Michigan State University, East Lansing, Michigan, USA
| | - Taehyung Lee
- Department of Physiology and Division of Pathology, Michigan State University, East Lansing, Michigan, USA
| | - Nandita Anand
- Department of Physiology and Division of Pathology, Michigan State University, East Lansing, Michigan, USA
| | - Vernon LaLone
- Department of Physiology and Division of Pathology, Michigan State University, East Lansing, Michigan, USA
| | - Narayanan Parameswaran
- Department of Physiology and Division of Pathology, Michigan State University, East Lansing, Michigan, USA
| |
Collapse
|
30
|
Huang BP, Lin CH, Chen YC, Kao SH. Anti-inflammatory effects of Perilla frutescens leaf extract on lipopolysaccharide-stimulated RAW264.7 cells. Mol Med Rep 2014; 10:1077-83. [PMID: 24898576 DOI: 10.3892/mmr.2014.2298] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Accepted: 03/28/2014] [Indexed: 11/05/2022] Open
Abstract
Perilla leaves are widely used in Chinese herbal medicine and in Japanese herbal agents used to treat respiratory diseases. This study aimed to investigate the anti‑inflammatory effects and the underlying mechanisms of Perilla frutescens leaf extract (PLE). Murine macrophage RAW264.7 cells were used as a model. Cell viability and morphological changes were studied by the MTT assay and microscopy. mRNA expression of pro‑inflammatory mediators was assessed by both semi‑quantitative reverse transcription‑polymerase chain reaction (RT‑PCR) and quantitative (q) RT‑PCR. Nitric oxide (NO) and prostaglandin E2 (PGE2) production were analyzed by the Griess test and sandwich enzyme‑linked immunosorbent assay (ELISA), respectively. The activation of kinase cascades was studied by immunoblotting. Our findings showed that PLE slightly affects cell viability, but alleviates LPS‑induced activation of RAW264.7 cells. Furthermore, PLE significantly reduced the LPS‑induced mRNA expression of the interleukin (IL)‑6, IL‑8, tumor necrosis factor‑α (TNF‑α), cyclooxygenase‑2 (COX‑2) and inducible nitric oxide synthase (iNOS), genes in a dose‑dependent manner. In addition, PLE reduced NO production and PGE2 secretion induced by LPS. PLE also inhibited activation of mitogen‑activated protein kinases (MAPKs), increased the cytosolic IκBα level, and reduced the level of nuclear factor (NF)‑κB. Taken together, these findings indicate that PLE significantly decreases the mRNA expression and protein production of pro‑inflammatory mediators, via the inhibition of extracellular‑signal‑regulated kinase (ERK)1/2, c‑Jun N‑terminal kinase (JNK), p38, as well as NF‑κB signaling in RAW264.7 cells stimulated with LPS.
Collapse
Affiliation(s)
- Bee-Piao Huang
- Department of Pathology, Tungs' Taichung MetroHarbor Hospital, Taichung, Taiwan, R.O.C
| | - Chun-Hsiang Lin
- Institute of Biochemistry and Biotechnology, Chung Shan Medical University Hospital, Taichung, Taiwan, R.O.C
| | - Yi-Ching Chen
- Institute of Biochemistry and Biotechnology, Chung Shan Medical University Hospital, Taichung, Taiwan, R.O.C
| | - Shao-Hsuan Kao
- Institute of Biochemistry and Biotechnology, Chung Shan Medical University Hospital, Taichung, Taiwan, R.O.C
| |
Collapse
|
31
|
Poulsen KL, Albee RP, Ganey PE, Roth RA. Trovafloxacin potentiation of lipopolysaccharide-induced tumor necrosis factor release from RAW 264.7 cells requires extracellular signal-regulated kinase and c-Jun N-Terminal Kinase. J Pharmacol Exp Ther 2014; 349:185-91. [PMID: 24525298 DOI: 10.1124/jpet.113.211276] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Trovafloxacin (TVX) is a fluoroquinolone antibiotic known to cause idiosyncratic, drug-induced liver injury (IDILI) in humans. The mechanism underlying this toxicity remains unknown. Previously, an animal model of IDILI in mice revealed that TVX synergizes with inflammatory stress from bacterial lipopolysaccharide (LPS) to produce a hepatotoxic interaction. The liver injury required prolongation of the appearance of tumor necrosis factor-α (TNF) in the plasma. The results presented here describe a model of TVX/LPS coexposure in RAW 264.7 cells acting as a surrogate for TNF-releasing cells in vivo. Pretreating cells with TVX for 2 hours before LPS addition led to increased TNF protein release into culture medium in a concentration- and time-dependent manner relative to cells treated with LPS or TVX alone. During the pretreatment period, TVX increased TNF mRNA, but this was less apparent when cells were exposed to TVX after LPS addition, suggesting that the pivotal signaling events that increase TNF expression occurred during the TVX pretreatment period. Indeed, TVX exposure increased activation of extracellular signal-regulated kinase (ERK), c-Jun N-terminal kinase (JNK), and p38 mitogen-activated protein kinase. Inhibition of either ERK or JNK decreased the TVX-mediated increase in TNF mRNA and LPS-induced TNF protein release, but p38 inhibition did not. These results demonstrated that the increased TNF appearance from TVX-LPS interaction in vivo can be reproduced in vitro and occurs in an ERK- and JNK-dependent manner.
Collapse
Affiliation(s)
- Kyle L Poulsen
- Department of Pharmacology & Toxicology, Center for Integrative Toxicology, Michigan State University, East Lansing, Michigan
| | | | | | | |
Collapse
|
32
|
Kim HS, Park JW, Kwon OK, Kim JH, Oh SR, Lee HK, Bach TT, Quang BH, Ahn KS. Anti-inflammatory activity of a methanol extract from Ardisia tinctoria on mouse macrophages and paw edema. Mol Med Rep 2014; 9:1388-94. [PMID: 24534870 DOI: 10.3892/mmr.2014.1941] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2013] [Accepted: 01/22/2014] [Indexed: 11/06/2022] Open
Abstract
Ardisia tinctoria (AT) is a plant of the Myrsinaceae family. No studies on its anti-inflammatory effects have yet been reported. This study investigated the anti-inflammatory activity of AT. A non-cytotoxic methanol extract of AT inhibited the expression of inducible NO synthase (iNOS) and cyclooxygenase-2 (COX-2), leading to significantly reduced levels of nitric oxide (NO) and prostaglandin E2 (PGE2) and of two proteins regulated by these, interleukin-1β (IL-1β) and IL-6, in lipopolysaccharide (LPS)-stimulated RAW 264.7 macrophage cells. The thickness of paw edema induced in vivo in mice by carrageenan administration was effectively reduced by the AT extract. Translocation of the nuclear factor-κB (NF-κB) subunit 65 (p65) into the nucleus and phosphorylation of mitogen-activated protein kinase kinase (MEK) and extracellular signal-related kinase (ERK) were inhibited by the AT extract. Our results indicated that a methanol extract of AT downregulates the inflammatory response by blocking phosphorylation of MEK and ERK and activation of NF-κB. To the best of our knowledge, this is the first study of anti-inflammatory effects of an AT extract, and demonstrates its potential in the treatment of inflammatory diseases.
Collapse
Affiliation(s)
- Hui-Seong Kim
- Biomolecular Science, University of Science and Technology, Daejeon 305‑806, Republic of Korea
| | - Ji-Won Park
- Department of Biotechnology, Korea University, Seoul 136‑713, Republic of Korea
| | - Ok-Kyoung Kwon
- Department of Toxicology, College of Pharmacy, Chungnam National University, Daejeon 305-764, Republic of Korea
| | - Jea-Hong Kim
- Department of Biotechnology, Korea University, Seoul 136‑713, Republic of Korea
| | - Sei-Ryang Oh
- Biomolecular Science, University of Science and Technology, Daejeon 305‑806, Republic of Korea
| | - Hyeong-Kyu Lee
- Biomolecular Science, University of Science and Technology, Daejeon 305‑806, Republic of Korea
| | - Tran The Bach
- Department of Botany, Institute of Ecology and Biological Resources,Vietnam Academy of Science and Technology, Hanoi 10307, Vietnam
| | - Bui Hong Quang
- Department of Botany, Institute of Ecology and Biological Resources,Vietnam Academy of Science and Technology, Hanoi 10307, Vietnam
| | - Kyung-Seop Ahn
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk 363-883, Republic of Korea
| |
Collapse
|
33
|
Wu CL, Tsai HC, Chen ZW, Wu CM, Li TM, Fong YC, Tang CH. Ras activation mediates WISP-1-induced increases in cell motility and matrix metalloproteinase expression in human osteosarcoma. Cell Signal 2013; 25:2812-22. [DOI: 10.1016/j.cellsig.2013.09.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Revised: 09/02/2013] [Accepted: 09/02/2013] [Indexed: 12/30/2022]
|
34
|
Lachance C, Wojewodka G, Skinner TAA, Guilbault C, De Sanctis JB, Radzioch D. Fenretinide corrects the imbalance between omega-6 to omega-3 polyunsaturated fatty acids and inhibits macrophage inflammatory mediators via the ERK pathway. PLoS One 2013; 8:e74875. [PMID: 24069363 PMCID: PMC3771966 DOI: 10.1371/journal.pone.0074875] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Accepted: 08/09/2013] [Indexed: 12/24/2022] Open
Abstract
We previously identified Fragile X-related protein 1 (FXR1) as an RNA-binding protein involved in the post-transcriptional control of TNF and other cytokines in macrophages. Macrophages derived from FXR1-KO mice overexpress several inflammatory cytokines including TNF. Recently, we showed that fenretinide (4HPR) is able to inhibit several inflammatory cytokines in the lungs of cystic fibrosis mice, which also have abnormal immune responses. Therefore, we hypothesized that 4HPR might also be able to downregulate excessive inflammation even in macrophages with ablated FXR1. Indeed, our results demonstrate that 4HPR inhibited the excessive production of inflammatory mediators, including TNF, IL-6, CCL2 and CCL-5 in LPS-stimulated FXR1-KO macrophages, by selectively inhibiting phosphorylation of ERK1/2, which is naturally more phosphorylated in FXR1-KO cells. We also found that LPS stimulation of FXR1-KO macrophages led to significantly higher ratio of arachidonic acid/docosahexaenoic acid than observed in FXR1-WT macrophages. Interestingly, treatment with 4HPR was associated with the normalization of arachidonic acid/docosahexaenoic acid ratio in macrophages, which we found to impact phosphorylation of ERK1/2. Overall, this study shows for the first time that 4HPR modulates inflammatory cytokine expression in macrophages by correcting a phospholipid-bound fatty acid imbalance that impacts the phosphorylation of ERK1/2.
Collapse
Affiliation(s)
- Claude Lachance
- McGill University, Department of Medicine and Department of Human Genetics, McGill University Health Center Research Institute, Montreal, Quebec, Canada
| | - Gabriella Wojewodka
- McGill University, Department of Medicine and Department of Human Genetics, McGill University Health Center Research Institute, Montreal, Quebec, Canada
| | - Tom A. A. Skinner
- McGill University, Department of Medicine and Department of Human Genetics, McGill University Health Center Research Institute, Montreal, Quebec, Canada
| | - Claudine Guilbault
- McGill University, Department of Medicine and Department of Human Genetics, McGill University Health Center Research Institute, Montreal, Quebec, Canada
| | - Juan B. De Sanctis
- Central University of Venezuela, Institute of Immunology, Caracas, Venezuela
| | - Danuta Radzioch
- McGill University, Department of Medicine and Department of Human Genetics, McGill University Health Center Research Institute, Montreal, Quebec, Canada
- * E-mail:
| |
Collapse
|
35
|
Morphine mediates a proinflammatory phenotype via μ-opioid receptor–PKCɛ–Akt–ERK1/2 signaling pathway in activated microglial cells. Biochem Pharmacol 2013; 86:487-96. [DOI: 10.1016/j.bcp.2013.05.027] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Revised: 05/28/2013] [Accepted: 05/31/2013] [Indexed: 11/18/2022]
|
36
|
|
37
|
Kim SM, Jang H, Son Y, Lee SA, Bae SS, Park YC, Eo SK, Kim K. 27-Hydroxycholesterol induces production of tumor necrosis factor-alpha from macrophages. Biochem Biophys Res Commun 2013; 430:454-9. [DOI: 10.1016/j.bbrc.2012.12.021] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Accepted: 12/05/2012] [Indexed: 10/27/2022]
|
38
|
Novel aminobenzyl-acetamidine derivative modulate the differential regulation of NOSs in LPS induced inflammatory response: Role of PI3K/Akt pathway. Biochim Biophys Acta Gen Subj 2012; 1820:2095-104. [DOI: 10.1016/j.bbagen.2012.08.014] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2012] [Revised: 08/02/2012] [Accepted: 08/16/2012] [Indexed: 12/13/2022]
|
39
|
Huang JL, Zhang YL, Wang CC, Zhou JR, Ma Q, Wang X, Shen XH, Jiang CL. Enhanced phosphorylation of MAPKs by NE promotes TNF-α production by macrophage through α adrenergic receptor. Inflammation 2012; 35:527-34. [PMID: 21590324 DOI: 10.1007/s10753-011-9342-4] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The aim of this study was to investigate whether norepinephrine (NE) could regulate macrophage production of tumor necrosis factor alpha (TNF-α) by influencing the phosphorylation of mitogen-activated protein kinases (MAPKs). Primary macrophages from male BALB/c mice were applied to explore the mechanism by which NE influences the the secretion of TNF-α when macrophages were activated by lipopolysaccharides (LPS). We found that NE could increase crophage production of TNF-α when macrophages were activated by LPS, and this effect could be inhibited by α adrenergic antagonist phentolamine. Also, NE could increase the phosphorylation of c-Jun N-terminal kinase (JNK), extracellular signal-regulated kinases (ERK), and p38, through α receptor. Furthermore, JNK inhibitor SP600125, ERK inhibitor U0126, and p38 inhibitor SB203580 could all partially counteract NE's effect on the phosphorylation of MAPKs, as well as TNF-α production by macrophages. This study revealed that as macrophages were activated by LPS, NE promoted the secretion of inflammatory factors by increasing the phosphorylation of MAPKs through an α receptor-dependent pathway. Our results provide the evidence of a relationship between stress and diseases, as well as the mechanism by which stress induces or affects the inflammation-related diseases.
Collapse
Affiliation(s)
- Jun-Long Huang
- Department of Military Nautical Medicine, Laboratory of Stress Medicine, Faculty of Naval Medicine, Second Military Medical University, Xiangyin Road 800, Shanghai 200433, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Kang H, Jeong HD, Choi HY. The Chloroform Fraction of Solanum nigrum Suppresses Nitric Oxide and Tumor Necrosis Factor-α in LPS-Stimulated Mouse Peritoneal Macrophages Through Inhibition of p38, JNK and ERK1/2. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2012; 39:1261-73. [DOI: 10.1142/s0192415x11009548] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Solanum nigrum L., commonly known as black nightshade, is used worldwide for the treatment of skin and mucosal ulcers, liver cirrhosis and edema. We aimed to determine the anti-inflammatory active fraction of S. nigrum by serial extractions. S. nigrum was first extracted with methanol, then fractionated with chloroform and water. The effects of S. nigrum fractions, diosgenin and α-solanine on LPS/interferon-gamma-induced nitric oxide (NO) and inducible NO synthase (iNOS), or LPS-induced tumor necrosis factor-α (TNF-α) and interleukin (IL)-6, in mouse peritoneal macrophages were determined. Western blotting analysis was used to detect LPS-induced phosphorylation of p38, JNK and ERK1/2. The chloroform fraction of S. nigrum was cytotoxic in a time and concentration dependent manner; however, the methanol and water fractions were not. The chloroform fraction reduced NO through inhibition of iNOS synthesis and inhibited TNF-α and IL-6 at the level of protein secretion; the methanol and water fractions showed a weak or no effect. The chloroform fraction also suppressed p38, JNK and ERK1/2. Diosgenin and α-solanine were cytotoxic at a high concentration. In particular, diosgenin was able to inhibit TNF-α and IL-6, but both compounds did not affect LPS-induced iNOS expression. These results indicate that the anti-inflammatory compounds of S. nigrum exist preferentially in the nonpolar fraction, ruling out the possibility that diosgenin and α-solanine are the likely candidates. The inhibition of iNOS, TNF-α and IL-6 by the chloroform fraction may be partly due to the suppression of p38, JNK and ERK1/2. Further study is required to identify the active compounds of S. nigrum.
Collapse
Affiliation(s)
- Hee Kang
- Department of East-West Medical Science, Graduate School of East-West Medical Science, Kyung Hee University, Yongin-si, Gyeonggi-do 446-701, South Korea
| | - Ha-Deok Jeong
- Department of Herbology, College of Oriental Medicine, Kyung Hee University, Seoul 130-701, South Korea
| | - Ho-Young Choi
- Department of Herbology, College of Oriental Medicine, Kyung Hee University, Seoul 130-701, South Korea
| |
Collapse
|
41
|
Park HC, Bae HB, Jeong CW, Lee SH, Jeung HJ, Kwak SH. Effect of manassantin B, a lignan isolated from Saururus chinensis, on lipopolysaccharide-induced interleukin-1β in RAW 264.7 cells. Korean J Anesthesiol 2012; 62:161-5. [PMID: 22379572 PMCID: PMC3284739 DOI: 10.4097/kjae.2012.62.2.161] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2011] [Revised: 10/05/2011] [Accepted: 10/05/2011] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Elevated systemic levels of pro-inflammatory cytokines cause hypotension during septic shock and induce capillary leakage in acute lung injury. Manassantin B has anti-inflammatory and anti-plasmoidal properties. This study examined the effects of manassantin B on lipopolysaccharide (LPS)-induced inflammatory response in murine macrophages. METHODS RAW 264.7 macrophage cells were incubated without or with (1, 3 and 10 µM) manassantin B and without or with (100 ng/ml) LPS. Manassantin B dissolved in phosphate buffered saline was added to the medium 1 h prior to the addition of LPS. The degree of activation of mitogen-activated protein kinase (MAPK) including extracellular signal-regulated kinases 1 and 2 (ERK1/2), c-Jun amino terminal kinases (JNK) and p38 MAPK, and the level of interleukin (IL)-1β were determined 30 min and 24 h after the addition of LPS respectively. RESULTS Manassantin B inhibited the production of IL-1β and attenuated the phosphorylations of ERK1/2 and p38 MAPK, but not that of JNK, in RAW 264.7 cells treated with LPS. CONCLUSIONS Manassantin B reduces LPS-induced IL-1β expression through effects on ERK1/2- and p38 MAPK-mediated pathways. Manassantin B has potential as a potent anti-inflammatory drug for use in pathological processes such as sepsis or acute lung injury.
Collapse
Affiliation(s)
- Hwan Chul Park
- Department of Anesthesiology and Pain Medicine, Chonnam Nationanl University Hospital, Gwangju, Korea
| | | | | | | | | | | |
Collapse
|
42
|
Tang CH, Tsai CC. CCL2 increases MMP-9 expression and cell motility in human chondrosarcoma cells via the Ras/Raf/MEK/ERK/NF-κB signaling pathway. Biochem Pharmacol 2012; 83:335-44. [DOI: 10.1016/j.bcp.2011.11.013] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2011] [Revised: 11/15/2011] [Accepted: 11/16/2011] [Indexed: 12/24/2022]
|
43
|
Hung HF, Hou CW, Chen YL, Lin CC, Fu HW, Wang JS, Jeng KC. Elephantopus scaber inhibits lipopolysaccharide-induced liver injury by suppression of signaling pathways in rats. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2011; 39:705-17. [PMID: 21721151 DOI: 10.1142/s0192415x11009147] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Elephantopus scaber (ES, Teng-Khia-U) has been traditionally used for the treatment of nephritis, pain, and fever; however, the direct evidence is lacking. We investigated the effect of ES on lipopolysaccharide (LPS) induced inflammation of BV-2 microglial cells and acute liver injury in Sprague-Dawley (SD) rats. Our results showed that ES reduced LPS-induced nitric oxide (NO), interleukin (IL)-1, IL-6, reactive oxygen species (ROS), and prostaglandin (PGE(2)) production in BV-2 cells. ES significantly decreased serum aspartate aminotransferase (AST) and alanine aminotransferase (ALT) levels in LPS-treated rats. Furthermore, the water extract, but not the ethanol extract, of ES dose-dependently inhibited LPS-induced JNK, p38 mitogen-activated protein kinases (MAPK), and slightly inhibited cyclooxygenase (COX-2) in BV-2 cells but decreased p38 MAPK and COX-2 expressions in the liver of LPS-treated rats. Taken together, these results indicate that the protective mechanism of ES involves an antioxidant effect and inhibition of p38 MAP kinase and COX-2 expressions in LPS-stressed acute hepatic injury in SD rats.
Collapse
Affiliation(s)
- Hsiao-Fang Hung
- Institute of Molecular and Cellular Biology, National Tsinghua University, Hsinchu, Taiwan
| | | | | | | | | | | | | |
Collapse
|
44
|
Chen DP, Wong CK, Leung PC, Fung KP, Lau CBS, Lau CP, Li EKM, Tam LS, Lam CWK. Anti-inflammatory activities of Chinese herbal medicine sinomenine and Liang Miao San on tumor necrosis factor-α-activated human fibroblast-like synoviocytes in rheumatoid arthritis. JOURNAL OF ETHNOPHARMACOLOGY 2011; 137:457-68. [PMID: 21679760 DOI: 10.1016/j.jep.2011.05.048] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2010] [Revised: 05/09/2011] [Accepted: 05/10/2011] [Indexed: 05/20/2023]
Abstract
AIM OF THE STUDY Sinomenine, an alkaloid isolated from the root of Sinomenium acutum, has been used to alleviate the symptoms of rheumatic diseases. Liang Miao San (LMS), composed of the herbs Rhizoma Atractylodis (Cangzhu) and Cotex Phellodendri (Huangbai), is another traditional Chinese medicine formula for rheumatoid arthritis (RA) treatment. Although numerous studies have demonstrated the potential anti-inflammatory activities of sinomenine and LMS, the underlying intracellular mechanisms regulating the anti-inflammatory activities of sinomenine and LMS on human primary fibroblast-like synoviocytes (FLS) from RA patients and normal control subjects have not been elucidated. MATERIALS AND METHODS We investigated the in vitro anti-inflammatory activity of sinomenine and LMS on inflammatory cytokine tumor necrosis factor (TNF)-α-mediated activation of human normal and RA-FLS. The underlying intracellular signaling molecules were analyzed quantitatively using flow cytometry. RESULTS Sinomenine was found to significantly inhibit TNF-α induced cell surface expression of vascular cell adhesion molecule (VCAM)-1 and release of inflammatory cytokine and chemokine IL-6, CCL2 and CXCL8 from both normal and RA-FLS (all p<0.05). Moreover, the suppression of sinomenine on TNF-α induced VCAM-1 expression and IL-6 release of RA-FLS was significantly higher than that of normal FLS (p<0.05). LMS significantly inhibited TNF-α-induced inflammatory chemokines CXCL10 and CCL5 release from both normal and RA-FLS, with significantly higher suppression on CXCL10 secretion in RA-FLS than that of normal FLS (all p<0.05). Further investigations showed that sinomenine and LMS could significantly suppress TNF-α-induced phosphorylation of inhibitor κBα and extracellular signal-regulated protein kinase, the central signaling molecules mediating TNF-α-induced VCAM-1 expression and chemokine production. CONCLUSION Our results therefore provide a new insight into the differential anti-inflammatory activities of sinomenine and LMS through the suppression of TNF-α-activated FLS by modulating distinct intracellular signaling pathways in RA.
Collapse
Affiliation(s)
- Da-Peng Chen
- Department of Chemical Pathology, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, N.T., Hong Kong
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
CARHSP1 is required for effective tumor necrosis factor alpha mRNA stabilization and localizes to processing bodies and exosomes. Mol Cell Biol 2010; 31:277-86. [PMID: 21078874 DOI: 10.1128/mcb.00775-10] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Tumor necrosis factor alpha (TNF-α) is a critical mediator of inflammation, and its production is tightly regulated, with control points operating at nearly every step of its biosynthesis. We sought to identify uncharacterized TNF-α 3' untranslated region (3'UTR)-interacting proteins utilizing a novel screen, termed the RNA capture assay. We identified CARHSP1, a cold-shock domain-containing protein. Knockdown of CARHSP1 inhibits TNF-α protein production in lipopolysaccharide (LPS)-stimulated cells and reduces the level of TNF-α mRNA in both resting and LPS-stimulated cells. mRNA stability assays demonstrate that CARHSP1 knockdown decreases TNF-α mRNA stability from a half-life (t(1/2)) of 49 min to a t(1/2) of 22 min in LPS-stimulated cells and from a t(1/2) of 29 min to a t(1/2) of 24 min in resting cells. Transfecting CARHSP1 into RAW264.7 cells results in an increase in TNF-α 3'UTR luciferase expression in resting cells and CARHSP1 knockdown LPS-stimulated cells. We examined the functional effect of inhibiting Akt, calcineurin, and protein phosphatase 2A and established that inhibition of Akt or calcineurin but not PP2A inhibits CARHSP1 function. Subcellular analysis establishes CARHSP1 as a cytoplasmic protein localizing to processing bodies and exosomes but not on translating mRNAs. We conclude CARHSP1 is a TNF-α mRNA stability enhancer required for effective TNF-α production, demonstrating the importance of both stabilization and destabilization pathways in regulating the TNF-α mRNA half-life.
Collapse
|
46
|
Kang H, Han SW, Hong JW, Sohn NW. Suppression of tumour necrosis factor-alpha by Schizonepeta tenuifolia water extract via inhibition of IkappaBalpha degradation and Jun N-terminal kinase/stress-activated protein kinase activation. J Pharm Pharmacol 2010; 62:1069-76. [PMID: 20663042 DOI: 10.1111/j.2042-7158.2010.01126.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
OBJECTIVES The anti-inflammatory effects of an aqueous extract of Schizonepeta tenuifolia on lipopolysaccharide (LPS)-induced tumour necrosis factor-alpha (TNF-alpha) and interleukin-6 (IL-6) in vivo and in vitro have been investigated. METHODS C57BL/6 mice were orally administered phosphate-buffered saline (control) or S. tenuifolia water extract (50, 200, 500 or 1000 mg/kg) for 10 days before intraperitoneal administration of LPS (1.3 mg/kg). Blood samples were obtained 1 h after LPS challenge, followed by determination of TNF-alpha and IL-6 levels. Peritoneal macrophages from thioglycollate-injected mice were obtained and stimulated with LPS and S. tenuifolia water extract for viability assay, cytokine analysis, real-time RT PCR and Western blotting. KEY FINDINGS Oral administration of S. tenuifolia water extract to mice significantly reduced LPS-induced serum levels of TNF-alpha, but not IL-6. When peritoneal macrophages were treated in vitro with S. tenuifolia water extract, the inhibition of LPS-induced TNF-alpha was more pronounced than that of IL-6 at the level of secreted protein and mRNA. S. tenuifolia water extract reduced the degradation of IkappaBalpha and the nuclear relocation of p65 NF-kappaB, but the phosphorylation of IkappaBalpha was not affected. Inhibition of c-Jun N-terminal kinase/stress-activated protein kinase (JNK/SAPK) by S. tenuifolia water extract led secondarily to the inhibition of phospho-c-Jun and phospho-ATF-2. CONCLUSIONS These results indicated that the downregulation of TNF-alpha by S. tenuifolia water extract may have involved the inhibition of both IkappaBalpha degradation and activation of c-Jun and ATF-2 involving suppression of JNK/SAPK.
Collapse
Affiliation(s)
- Hee Kang
- Department of East-West Medical Science, Graduate School of East-West Medical Science, Kyung Hee University, Yongin-si, Gyeonggi-do, South Korea.
| | | | | | | |
Collapse
|
47
|
Bae HB, Li M, Son JK, Seo CS, Chung SH, Kim SJ, Jeong CW, Lee HG, Kim W, Park HC, Kwak SH. Sauchinone, a lignan from Saururus chinensis, reduces tumor necrosis factor-α production through the inhibition of c-raf/MEK1/2/ERK 1/2 pathway activation. Int Immunopharmacol 2010; 10:1022-8. [DOI: 10.1016/j.intimp.2010.06.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2009] [Revised: 05/07/2010] [Accepted: 06/01/2010] [Indexed: 10/19/2022]
|
48
|
Hammarfjord O, Wallin RPA. Dendritic cell function at low physiological temperature. J Leukoc Biol 2010; 88:747-56. [PMID: 20651299 DOI: 10.1189/jlb.0310155] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Compared with the stable core temperature, the skin temperature is lower and varies depending on ambient temperature and convection conditions. The function of DCs, which are plentiful in the skin at lower physiological temperatures, has not been reported. We show that DC performed some functions normally at 28°C, including phagocytosis and macropinocytosis. TLR-4 signaling via MAPK pathways was delayed at 28°C but reached normal levels, which may explain the observed slower kinetics of stimulated macropinocytosis and TNF production. TLR-4-induced NO production was compromised severely at 28°C. Collagen degradation and migration through matrigel-coated transwell inserts were decreased, but no effect on podosome number or DC migration through noncoated transwell filters was seen. Lowering the temperature differentially regulated functions associated with the role of DCs in adaptive immunity. LPS-induced up-regulation of CD86 was normal; however, CD40 up-regulation was suppressed after TLR-4 stimulation at 28°C. Nonactivated DC processed and presented antigen on MHC class II equally well at 28°C and 37°C. However, DCs that were loaded with antigens and stimulated with TLR ligand at 28°C were poor at activating T cells at 37°C compared with DCs that were activated and loaded with antigen at 37°C.
Collapse
Affiliation(s)
- Oscar Hammarfjord
- Center for Infectious Medicine, F59, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Huddinge, Stockholm, Sweden
| | | |
Collapse
|
49
|
Kwon OK, Lee MY, Yuk JE, Oh SR, Chin YW, Lee HK, Ahn KS. Anti-inflammatory effects of methanol extracts of the root of Lilium lancifolium on LPS-stimulated Raw264.7 cells. JOURNAL OF ETHNOPHARMACOLOGY 2010; 130:28-34. [PMID: 20412846 DOI: 10.1016/j.jep.2010.04.002] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2010] [Revised: 03/12/2010] [Accepted: 04/08/2010] [Indexed: 05/29/2023]
Abstract
AIM OF THE STUDY Lilium lancifolium is commonly used to treat bronchitis, pneumonia, etc. In this study, we investigated the anti-inflammatory effects of methanol extracts of the root of Lilium lancifolium (LL extracts) in LPS-stimulated Raw264.7 cells. MATERIAL AND METHODS Levels of NO, PGE(2) and pro-inflammatory cytokines (IL-6 and TNF-alpha) in the supernatant fraction were determined using sandwich ELISA. Expression of COX-2 and iNOS, phosphorylation of MAPK subgroups (ERK and JNK), and NF-kappaB activation in extracts were detected via Western blot and immunocytochemistry assays. RESULTS The LL extract significantly inhibited NO, PGE(2), IL-6 and TNF-alpha production in LPS-stimulated cells, and suppressed iNOS and COX-2 expression. A mechanism-based study showed that phosphorylation of ERK1/2 and JNK and translocation of the NF-kappaB p65 subunit into nuclei were inhibited by the LL extract. Furthermore, interleukin-4 and interleukin-13 production in Con A-induced splenocytes was suppressed. CONCLUSION These results indicate that anti-inflammatory effects of methanol extracts from Lilium lancifolium are due to downregulation of iNOS and COX-2 via suppression of NF-kappaB activation and nuclear translocation as well as blocking of ERK and JNK signaling in LPS-stimulated Raw264.7 cells.
Collapse
Affiliation(s)
- Ok-Kyoung Kwon
- Immune Modulator Research Center, Korea Research Institute of Bioscience and Biotechnology, 685-1 Yangchung-ri, Ochang-eup, Cheongwon-gun, Chungbuk, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
50
|
Murugan V, Peck MJ. Signal transduction pathways linking the activation of alveolar macrophages with the recruitment of neutrophils to lungs in chronic obstructive pulmonary disease. Exp Lung Res 2010; 35:439-85. [PMID: 19842832 DOI: 10.1080/01902140902759290] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is a major and increasing global health problem. It is predicted by the World Health Organization to become the third most common cause of death and the fifth most common cause of disability in the world by 2020. COPD is a complex inflammatory disease involving several types of inflammatory cells and multiple inflammatory mediators. Although abnormal numbers of inflammatory cells such as macrophages, dendritic cells, neutrophils, and T lymphocytes have been documented in COPD, the relationship between these cell types and the sequence of their appearance and persistence is largely unknown. Alveolar macrophages have been identified as one of the major cell types that plays a key role in orchestrating the inflammatory events associated with the pathophysiology of COPD. One of the major functions of macrophages is the secretion of chemotactic factors and this function is markedly increased on exposure to cigarette smoke (CS). This enhanced release of chemoattractants results in increased lung neutrophil infiltration, which is thought to be a key event in the development of COPD. The molecular basis for this amplified inflammatory response is not very clear, but it could be due to an alteration in signal transduction pathways within the macrophage. Based on existing literature, an attempt has been made to create a comprehensive review of the signal transduction pathways that link the activation of macrophages with the increased recruitment of neutrophils into the airways. Some of the major stimuli that activate macrophages and cause them to secrete chemotactic factors have been identified as CS, wood smoke, ozone, bacterial endotoxin, and proinflammatory cytokines such as interleukin (IL)-1beta and tumor necrosis factor (TNF)-alpha. These stimuli seem to activate mainly redox-sensitive transcription factors such as nuclear factor (NF)-kappa B and activator protein (AP)-1, both of which play a major role in the synthesis and secretion of chemotactic factors such as IL-8 and leukotriene B(4) (LTB(4)). The pathways involved in the synthesis and secretion of other factors such as macrophage chemotactic protein-1 (MCP-1) and growth-related oncogene-alpha (Gro-alpha) have also been reviewed.
Collapse
|