1
|
Di Giorgio E, Cortolezzis Y, Gualandi N, Agostini F, Rapozzi V, Xodo LE. NRF2 interacts with distal enhancer and inhibits nitric oxide synthase 2 expression in KRAS-driven pancreatic cancer cells. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119606. [PMID: 37852325 DOI: 10.1016/j.bbamcr.2023.119606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 10/07/2023] [Accepted: 10/10/2023] [Indexed: 10/20/2023]
Abstract
Nitric oxide is a pleiotropic free radical produced by three nitric oxide synthases (NOS1-3), of which inducible NOS2 is involved in tumor initiation and progression. In this study, RNA-seq, ChIP-seq and qRT-PCR experiments combined with bioinformatic analyses showed that NRF2 is a repressor of NOS2 gene by maintaining a distal enhancer located 22 kb downstream of TSS in an inactive state. Deletion of NRF2 leads to activation of the enhancer, which exerts a pioneering function before it is fully activated. Specifically, NRF2 controls the expression of NOS2 in response to intracellular oxidative stress and extracellular oxygen pressure. We found that abrogation of NOS2 expression by siRNAs partially reduced the ability of WT Panc-1 cells to form 3D spheroids, but strongly reduced the formation of 3D spheroids by NRF2-depleted Panc-1 cells. Mechanistically, this effect correlates with the finding that NOS2 and nitric oxide stimulate epithelial-to-mesenchymal transition in NRF2-depleted Panc-1 and MIA PaCa-2 cells. We also found that knockdown of NOS2 leads to blockade of 3D matrigel invasion of NRF2-depleted PDAC cells, demonstrating that a short-circuit in the reciprocal regulation of NOS2 and NRF2 attenuates the malignancy of PDAC cells. In summary, we show for the first time that: (i) NRF2 is a suppressor of NOS2 in pancreatic cancer cells; (ii) NRF2 binds to and inactivates an enhancer located 22 kb downstream of TSS of the NOS2 gene; (iii) activation of NOS2 requires suppression of NRF2; (iv) NOS2 is required for NRF2-depleted Panc-1 cells to maintain their malignancy and invasiveness.
Collapse
Affiliation(s)
- Eros Di Giorgio
- Department of Medicine, Laboratory of Biochemistry, P.le Kolbe 4, 33100 Udine, Italy.
| | - Ylenia Cortolezzis
- Department of Medicine, Laboratory of Biochemistry, P.le Kolbe 4, 33100 Udine, Italy
| | - Nicolò Gualandi
- Department of Medicine, Laboratory of Biochemistry, P.le Kolbe 4, 33100 Udine, Italy
| | - Francesca Agostini
- Department of Medicine, Laboratory of Biochemistry, P.le Kolbe 4, 33100 Udine, Italy
| | - Valentina Rapozzi
- Department of Medicine, Laboratory of Biochemistry, P.le Kolbe 4, 33100 Udine, Italy
| | - Luigi E Xodo
- Department of Medicine, Laboratory of Biochemistry, P.le Kolbe 4, 33100 Udine, Italy.
| |
Collapse
|
2
|
Cheng RYS, Ridnour LA, Wink AL, Gonzalez AL, Femino EL, Rittscher H, Somasundaram V, Heinz WF, Coutinho L, Rangel MC, Edmondson EF, Butcher D, Kinders RJ, Li X, Wong STC, McVicar DW, Anderson SK, Pore M, Hewitt SM, Billiar TR, Glynn SA, Chang JC, Lockett SJ, Ambs S, Wink DA. Interferon-gamma is quintessential for NOS2 and COX2 expression in ER - breast tumors that lead to poor outcome. Cell Death Dis 2023; 14:319. [PMID: 37169743 PMCID: PMC10175544 DOI: 10.1038/s41419-023-05834-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 04/20/2023] [Accepted: 04/24/2023] [Indexed: 05/13/2023]
Abstract
A strong correlation between NOS2 and COX2 tumor expression and poor clinical outcomes in ER breast cancer has been established. However, the mechanisms of tumor induction of these enzymes are unclear. Analysis of The Cancer Genome Atlas (TCGA) revealed correlations between NOS2 and COX2 expression and Th1 cytokines. Herein, single-cell RNAseq analysis of TNBC cells shows potent NOS2 and COX2 induction by IFNγ combined with IL1β or TNFα. Given that IFNγ is secreted by cytolytic lymphocytes, which improve clinical outcomes, this role of IFNγ presents a dichotomy. To explore this conundrum, tumor NOS2, COX2, and CD8+ T cells were spatially analyzed in aggressive ER-, TNBC, and HER2 + breast tumors. High expression and clustering of NOS2-expressing tumor cells occurred at the tumor/stroma interface in the presence of stroma-restricted CD8+ T cells. High expression and clustering of COX2-expressing tumor cells extended into immune desert regions in the tumor core where CD8+ T cell penetration was limited or absent. Moreover, high NOS2-expressing tumor cells were proximal to areas with increased satellitosis, suggestive of cell clusters with a higher metastatic potential. Further in vitro experiments revealed that IFNγ + IL1β/TNFα increased the elongation and migration of treated tumor cells. This spatial analysis of the tumor microenvironment provides important insight into distinct neighborhoods where stroma-restricted CD8+ T cells exist proximal to NOS2-expressing tumor niches that could have increased metastatic potential.
Collapse
Affiliation(s)
- Robert Y S Cheng
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| | - Lisa A Ridnour
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| | - Adelaide L Wink
- Optical Microscopy and Analysis Laboratory, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc. for the National Cancer Institute, Frederick, MD, USA
| | - Ana L Gonzalez
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| | - Elise L Femino
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| | - Helene Rittscher
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| | - Veena Somasundaram
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| | - William F Heinz
- Optical Microscopy and Analysis Laboratory, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc. for the National Cancer Institute, Frederick, MD, USA
| | - Leandro Coutinho
- Center for Translational Research in Oncology, ICESP/HC, Faculdade de Medicina da Universidade de São Paulo; and Comprehensive Center for Precision Oncology, Universidade de São Paulo, São Paulo, SP, Brazil
| | - M Cristina Rangel
- Center for Translational Research in Oncology, ICESP/HC, Faculdade de Medicina da Universidade de São Paulo; and Comprehensive Center for Precision Oncology, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Elijah F Edmondson
- Molecular Histopathology Laboratories, Leidos Biomedical Research Inc. for NCI, Frederick, MD, USA
| | - Donna Butcher
- Molecular Histopathology Laboratories, Leidos Biomedical Research Inc. for NCI, Frederick, MD, USA
| | - Robert J Kinders
- Office of the Director, Division of Cancer Treatment and Diagnosis, NCI, Frederick, MD, USA
| | - Xiaoxian Li
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA, USA
| | - Stephen T C Wong
- Systems Medicine and Bioengineering Department, Houston Methodist Neal Cancer Center, Houston Methodist Hospital and Weill Cornell Medicine, Houston, TX, USA
| | - Daniel W McVicar
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| | - Stephen K Anderson
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| | - Milind Pore
- Imaging Mass Cytometry Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | | | - Timothy R Billiar
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA
| | - Sharon A Glynn
- Discipline of Pathology, Lambe Institute for Translational Research, School of Medicine, University of Galway, Galway, Ireland
| | - Jenny C Chang
- Mary and Ron Neal Cancer Center, Houston Methodist Hospital and Weill Cornell Medicine, Houston, TX, USA
| | - Stephen J Lockett
- Optical Microscopy and Analysis Laboratory, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc. for the National Cancer Institute, Frederick, MD, USA
| | - Stefan Ambs
- Laboratory of Human Carcinogenesis, CCR, NCI, NIH, Bethesda, MD, USA
| | - David A Wink
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA.
| |
Collapse
|
3
|
Cheng RY, Ridnour LA, Wink AL, Gonzalez AL, Femino EL, Rittscher H, Somasundarum V, Heinz WF, Coutinho L, Cristina Rangel M, Edmondson EF, Butcher D, Kinders RJ, Li X, Wong STC, McVicar DW, Anderson SK, Pore M, Hewitt SM, Billiar TR, Glynn S, Chang JC, Lockett SJ, Ambs S, Wink DA. Interferon-gamma is Quintessential for NOS2 and COX2 Expression in ER - Breast Tumors that Lead to Poor Outcome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.06.535916. [PMID: 37066331 PMCID: PMC10104135 DOI: 10.1101/2023.04.06.535916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
A strong correlation between NOS2 and COX2 tumor expression and poor clinical outcomes in ER-breast cancer has been established. However, mechanisms of tumor induction of these enzymes are unclear. Analysis of The Cancer Genome Atlas (TCGA) revealed correlations between NOS2 and COX2 expression and Th1 cytokines. Herein, single cell RNAseq analysis of TNBC cells shows potent NOS2 and COX2 induction by IFNγ combined with IL1β or TNFα. Given that IFNγ is secreted by cytolytic lymphocytes, which improve clinical outcomes, this role of IFNγpresents a dichotomy. To explore this conundrum, tumor NOS2, COX2, and CD8 + T cells were spatially analyzed in aggressive ER-, TNBC, and HER2+ breast tumors. High expression and clustering of NOS2-expressing tumor cells occurred at the tumor/stroma interface in the presence of stroma-restricted CD8 + T cells. High expression and clustering of COX2-expressing tumor cells extended into immune desert regions in the tumor core where CD8 + T cell penetration was limited or absent. Moreover, high NOS2-expressing tumor cells were proximal to areas with increased satellitosis suggestive of cell clusters with a higher metastatic potential. Further in vitro experiments revealed that IFNγ+IL1β/TNFα increased elongation and migration of treated tumor cells. This spatial analysis of the tumor microenvironment provides important insight of distinct neighborhoods where stroma-restricted CD8 + T cells exist proximal to NOS2-expressing tumor niches that could have increased metastatic potential.
Collapse
|
4
|
Russell TM, Richardson DR. The good Samaritan glutathione-S-transferase P1: An evolving relationship in nitric oxide metabolism mediated by the direct interactions between multiple effector molecules. Redox Biol 2023; 59:102568. [PMID: 36563536 PMCID: PMC9800640 DOI: 10.1016/j.redox.2022.102568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 11/22/2022] [Accepted: 12/01/2022] [Indexed: 12/23/2022] Open
Abstract
Glutathione-S-transferases (GSTs) are phase II detoxification isozymes that conjugate glutathione (GSH) to xenobiotics and also suppress redox stress. It was suggested that GSTs have evolved not to enhance their GSH affinity, but to better interact with and metabolize cytotoxic nitric oxide (NO). The interactions between NO and GSTs involve their ability to bind and store NO as dinitrosyl-dithiol iron complexes (DNICs) within cells. Additionally, the association of GSTP1 with inducible nitric oxide synthase (iNOS) results in its inhibition. The function of NO in vasodilation together with studies associating GSTM1 or GSTT1 null genotypes with preeclampsia, additionally suggests an intriguing connection between NO and GSTs. Furthermore, suppression of c-Jun N-terminal kinase (JNK) activity occurs upon increased levels of GSTP1 or NO that decreases transcription of JNK target genes such as c-Jun and c-Fos, which inhibit apoptosis. This latter effect is mediated by the direct association of GSTs with MAPK proteins. GSTP1 can also inhibit nuclear factor kappa B (NF-κB) signaling through its interactions with IKKβ and Iκα, resulting in decreased iNOS expression and the stimulation of apoptosis. It can be suggested that the inhibitory activity of GSTP1 within the JNK and NF-κB pathways may be involved in crosstalk between survival and apoptosis pathways and modulating NO-mediated ROS generation. These studies highlight an innovative role of GSTs in NO metabolism through their interaction with multiple effector proteins, with GSTP1 functioning as a "good Samaritan" within each pathway to promote favorable cellular conditions and NO levels.
Collapse
Affiliation(s)
- Tiffany M Russell
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - Des R Richardson
- Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University, Nathan, 4111, Australia.
| |
Collapse
|
5
|
Song MY, Lee DY, Park SY, Seo SA, Hwang JS, Heo SH, Kim EH. Steamed Ginger Extract Exerts Anti-inflammatory Effects in Helicobacter pylori-infected Gastric Epithelial Cells through Inhibition of NF-κB. J Cancer Prev 2021; 26:289-297. [PMID: 35047455 PMCID: PMC8749313 DOI: 10.15430/jcp.2021.26.4.289] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 12/22/2021] [Accepted: 12/23/2021] [Indexed: 12/13/2022] Open
Abstract
Ginger (Zingiber officinale) has traditionally been used as a treatment for inflammatory diseases in the Asian region. Recently, anti-inflammatory effects of steamed ginger extract (GGE03) have been reported, but its association with Helicobacter pylori (H. pylori)-induced gastritis has not been investigated. The purpose of this study was to assess the anti-inflammatory activity of GGE03 in H. pylori-infected gastric epithelial cells. Our studies revealed that the GGE03 suppressed the growth of H. pylori. GGE03 markedly reduced the expression of the H. pylori-induced pro-inflammatory cytokines including interleukin (IL)-8, TNF-α, IL-6, inducible NOS (iNOS) and IFN-γ. We also demonstrated that GGE03 treatment inhibited the H. pylori-activated NF-κB signaling pathway. In addition, the treatment with GGE03 significantly attenuated nitric oxide production and myeloperoxidase activity in H. pylori-infected gastric epithelial cells. These anti-inflammatory effects of GGE03 were more effective than ginger extract. Finally, we investigated the minimum effective concentration of GGE03 to inhibit H. pylori-induced inflammation. Our findings suggest that GGE03 not only inhibits the growth of H. pylori, but also attenuates H. pylori-induced inflammation.
Collapse
Affiliation(s)
- Moon-Young Song
- Institute of Pharmaceutical Sciences, College of Pharmacy, CHA University, Seongnam, Korea
| | - Da-Young Lee
- Institute of Pharmaceutical Sciences, College of Pharmacy, CHA University, Seongnam, Korea
| | | | - Seul-A Seo
- SD Biotechnologies Co., Ltd., Seoul, Korea
| | | | | | - Eun-Hee Kim
- Institute of Pharmaceutical Sciences, College of Pharmacy, CHA University, Seongnam, Korea
| |
Collapse
|
6
|
The Relationship of Glutathione- S-Transferase and Multi-Drug Resistance-Related Protein 1 in Nitric Oxide (NO) Transport and Storage. Molecules 2021; 26:molecules26195784. [PMID: 34641326 PMCID: PMC8510172 DOI: 10.3390/molecules26195784] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/21/2021] [Accepted: 09/21/2021] [Indexed: 12/18/2022] Open
Abstract
Nitric oxide is a diatomic gas that has traditionally been viewed, particularly in the context of chemical fields, as a toxic, pungent gas that is the product of ammonia oxidation. However, nitric oxide has been associated with many biological roles including cell signaling, macrophage cytotoxicity, and vasodilation. More recently, a model for nitric oxide trafficking has been proposed where nitric oxide is regulated in the form of dinitrosyl-dithiol-iron-complexes, which are much less toxic and have a significantly greater half-life than free nitric oxide. Our laboratory has previously examined this hypothesis in tumor cells and has demonstrated that dinitrosyl-dithiol-iron-complexes are transported and stored by multi-drug resistance-related protein 1 and glutathione-S-transferase P1. A crystal structure of a dinitrosyl-dithiol-iron complex with glutathione-S-transferase P1 has been solved that demonstrates that a tyrosine residue in glutathione-S-transferase P1 is responsible for binding dinitrosyl-dithiol-iron-complexes. Considering the roles of nitric oxide in vasodilation and many other processes, a physiological model of nitric oxide transport and storage would be valuable in understanding nitric oxide physiology and pathophysiology.
Collapse
|
7
|
Martí I Líndez AA, Reith W. Arginine-dependent immune responses. Cell Mol Life Sci 2021; 78:5303-5324. [PMID: 34037806 PMCID: PMC8257534 DOI: 10.1007/s00018-021-03828-4] [Citation(s) in RCA: 121] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 03/23/2021] [Accepted: 03/29/2021] [Indexed: 02/07/2023]
Abstract
A growing body of evidence indicates that, over the course of evolution of the immune system, arginine has been selected as a node for the regulation of immune responses. An appropriate supply of arginine has long been associated with the improvement of immune responses. In addition to being a building block for protein synthesis, arginine serves as a substrate for distinct metabolic pathways that profoundly affect immune cell biology; especially macrophage, dendritic cell and T cell immunobiology. Arginine availability, synthesis, and catabolism are highly interrelated aspects of immune responses and their fine-tuning can dictate divergent pro-inflammatory or anti-inflammatory immune outcomes. Here, we review the organismal pathways of arginine metabolism in humans and rodents, as essential modulators of the availability of this semi-essential amino acid for immune cells. We subsequently review well-established and novel findings on the functional impact of arginine biosynthetic and catabolic pathways on the main immune cell lineages. Finally, as arginine has emerged as a molecule impacting on a plethora of immune functions, we integrate key notions on how the disruption or perversion of arginine metabolism is implicated in pathologies ranging from infectious diseases to autoimmunity and cancer.
Collapse
Affiliation(s)
| | - Walter Reith
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
8
|
Kim H, Yang WS, Htwe KM, Lee MN, Kim YD, Yoon KD, Lee BH, Lee S, Cho JY. Dipterocarpus tuberculatus Roxb. Ethanol Extract Has Anti-Inflammatory and Hepatoprotective Effects In Vitro and In Vivo by Targeting the IRAK1/AP-1 Pathway. Molecules 2021; 26:molecules26092529. [PMID: 33926126 PMCID: PMC8123704 DOI: 10.3390/molecules26092529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 04/22/2021] [Accepted: 04/23/2021] [Indexed: 11/28/2022] Open
Abstract
Dipterocarpus tuberculatus Roxb. has been used traditionally as a remedy for many diseases, especially inflammation. Therefore, we analyzed and explored the mechanism of the anti-inflammatory effect of a Dipterocarpus tuberculatus Roxb. ethanol extract (Dt-EE). Dt-EE clearly and dose-dependently inhibited the expression of pro-inflammatory cytokines such as IL-6, TNF-α, and IL-1β in lipopolysaccharide (LPS)-treated RAW264.7 cells. Also, Dt-EE suppressed the activation of the MyD88/TRIF-mediated AP-1 pathway and the AP-1 pathway related proteins JNK2, MKK4/7, and TAK1, which occurred as a result of inhibiting the kinase activity of IRAK1 and IRAK4, the most upstream factors of the AP-1 pathway. Finally, Dt-EE displayed hepatoprotective activity in a mouse model of hepatitis induced with LPS/D-galactosamine (D-GalN) through decreasing the serum levels of alanine aminotransferase and suppressing the activation of JNK and IRAK1. Therefore, our results strongly suggest that Dt-EE could be a candidate anti-inflammatory herbal medicine with IRAK1/AP-1 inhibitory and hepatoprotective properties.
Collapse
Affiliation(s)
- Haeyeop Kim
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Korea; (H.K.); (W.S.Y.)
| | - Woo Seok Yang
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Korea; (H.K.); (W.S.Y.)
| | - Khin Myo Htwe
- Popa Mountain Park, Forest Department, Kyaukpadaung Township, Mandalay Division, Kyaukpadaung 05241, Myanmar;
| | - Mi-Nam Lee
- Department of Hospitality and Culinary, Ansan University, Ansan 15318, Korea;
| | - Young-Dong Kim
- Department of Life Science, Hallym University, Chuncheon 200-702, Korea;
| | - Ki Dong Yoon
- College of Pharmacy, The Catholic University of Korea, Bucheon 420-743, Korea;
| | - Byoung-Hee Lee
- National Institute of Biological Resources, Environmental Research Complex, Incheon 22689, Korea;
| | - Sarah Lee
- National Institute of Biological Resources, Environmental Research Complex, Incheon 22689, Korea;
- Correspondence: (S.L.); (J.Y.C.); Tel.: +82-32-590-7265 (S.L.); +82-31-290-7868 (J.Y.C.); Fax: +82-32-590-7472 (S.L.); +82-31-290-7870 (J.Y.C.)
| | - Jae Youl Cho
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Korea; (H.K.); (W.S.Y.)
- Correspondence: (S.L.); (J.Y.C.); Tel.: +82-32-590-7265 (S.L.); +82-31-290-7868 (J.Y.C.); Fax: +82-32-590-7472 (S.L.); +82-31-290-7870 (J.Y.C.)
| |
Collapse
|
9
|
Riyadi PH, Wahyudi D, Alexander Tanod W. Effects of dichloromethane Sarcophyton spp. extract on the lipopolysaccharide-induced expression of nuclear factor-kappa B and inducible nitric oxide synthase in mice. Vet World 2019; 12:1897-1902. [PMID: 32095038 PMCID: PMC6989332 DOI: 10.14202/vetworld.2019.1897-1902] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 10/29/2019] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND AND AIM The soft coral genus Sarcophyton is a source of cembraneterpen. Sarcophyton is reported to have anti-inflammatory properties, with the ability to reduce the expression of inducible nitric oxide synthase (iNOS) and inhibit nuclear factor-kappa B (NF-κB) activation. This study aimed to investigate the efficacy of dichloromethane (DCM) extracts of soft coral Sarcophyton spp. to inhibit the expression of NF-κB and iNOS induced by lipopolysaccharide (LPS). MATERIALS AND METHODS Crude extracts of Sarcophyton spp. were macerated with DCM (1:3 v/v) for 24 h. Thirty-six Balb/c mice were divided into six treatment groups, namely, normal control (without LPS induction), negative control (LPS induction 4 mg/mL), comparative control (LPS+Dexamethasone 6 mg/kg), and 3 concentration groups extract (LPS+50, 125, and 250 mg/kg). The expression of NF-κB and iNOS was measured in each treatment group. RESULTS Flow cytometry analysis showed that the relative number of NF-κB+ cells increased (18.38±1.24%) in LPS-induced mice compared with normal mice (13.24±1.15%). The Sarcophyton spp. DCM extracts decreased the relative number of NF-κB+ cells (125 mg/kg: 13.96±0.84%). Immunohistochemical analysis with ImmunoMembrane showed that LPS induction in mice increased iNOS expression when compared to normal mice. The Sarcophyton spp. DCM extracts reduced iNOS expression (especially at 125 mg/kg). CONCLUSION DCM extracts of Sarcophyton spp. inhibited the activation of NF-κB, resulting in suppressed iNOS expression, which directly inhibits NO production.
Collapse
Affiliation(s)
- Putut Har Riyadi
- Postgraduate Program, Faculty of Fisheries and Marine Science, Brawijaya University, Malang 65145, East Java, Indonesia
- Department of Fisheries Post Harvest Technology, Faculty of Fisheries and Marine Science, Diponegoro University, Semarang 50275, Central Java, Indonesia
| | - Didik Wahyudi
- Department of Biology, Faculty of Science and Technology, State Islamic University of Maulana Malik Ibrahim Malang, Malang 65144, East Java, Indonesia
| | - Wendy Alexander Tanod
- Department of Fisheries Product Technology, Institute of Fisheries and Marine (Sekolah Tinggi Perikanan dan Kelautan), Palu 94118, Central Sulawesi, Indonesia
| |
Collapse
|
10
|
Stachon T, Latta L, Kolev K, Seitz B, Langenbucher A, Szentmáry N. Erhöhte NF-κB- und iNOS-Expression in Keratozyten von Keratokonuspatienten – Hinweise auf eine entzündliche Komponente? Klin Monbl Augenheilkd 2019; 238:1010-1017. [DOI: 10.1055/a-1002-0100] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Zusammenfassung
Hintergrund In den letzten Jahren mehren sich Hinweise auf eine entzündliche Komponente beim Keratokonus (KC). Ein Schlüsselgen bei entzündlichen Prozessen ist der Nuclear Factor Kappa B (NF-κB). NF-κB ist ein Transkriptionsfaktor, der unter anderem das Enzym Nitric Oxide Synthase (NOS), das mit dem konkurrierenden Enzym Arginase (Arg) bei entzündlichen Prozessen involviert ist, aktiviert. Ziel dieser Studie war es, die Isotypen von NOS und Arginase zu analysieren, die Expression NF-κB, NOS und Arginase sowie den regulativen Mechanismus von NOS und Arginase in Keratozyten von Keratokonuspatienten mithilfe des Inhibitors 1400W in vitro zu untersuchen.
Methoden Primäre humane Keratozyten wurden durch enzymatische Behandlung mit Kollagenase A aus humanen Korneoskleralscheiben (n = 8) und von Explantaten von geplanten perforierenden Keratoplastiken (KC-Patienten) isoliert (n = 8) und in DMEM/F12-Kulturmedium, versetzt mit 5% fetalem Kälberserum, kultiviert. Die Expression von NF-κB, NOS und Arginase wurden mit quantitativer PCR (qPCR) und Westernblot-Analyse (WB) untersucht. Nitrit- und Ureakonzentrationen im Zellkulturüberstand wurden nach Zugabe des NOS-Inhibitors 1400W (0 – 40 µM) analysiert.
Ergebnisse In den Keratozyten wurden ausschließlich die Isotypen iNOS (induzierbare NO-Synthase) und Arg-II nachgewiesen. Die mRNA-Expression von NF-κB und iNOS waren in KC-Keratozyten höher als in normalen Zellen (p = 0,0135 und p = 0,0001), während in der Arg-II-Expression keine Unterschiede messbar waren. Im WB war bei NF-κB eine höhere Bandenintensität messbar (p = 0,0012), bei iNOS konnten keine Unterschiede in der Bandenintensität nachgewiesen werden. Im Überstand der KC-Keratozyten wurden geringere Konzentrationen von Nitrit und Urea nach Zugabe des Inhibitors 1400W gemessen (p = ≤ 0,014), nicht jedoch bei normalen Zellen (p ≥ 0,178).
Schlussfolgerung Aufgrund der erhöhten Expression von NF-κB und iNOS muss von einer inflammatorischen Komponente beim Keratokonus ausgegangen werden. Die unterschiedliche Regulation der KC-Keratozyten durch den iNOS-Inhibitor 1400W legt eine veränderte metabolische Aktivität nahe, die durch entzündliche Prozesse hervorgerufen werden kann.
Collapse
Affiliation(s)
- Tanja Stachon
- Klinik für Augenheilkunde, Universitätsklinikum des Saarlandes UKS, Homburg/Saar
| | - Lorenz Latta
- Klinik für Augenheilkunde, Universitätsklinikum des Saarlandes UKS, Homburg/Saar
| | - Krasimir Kolev
- Department of Medical Biochemistry, Semmelweis University, Budapest, Hungary
| | - Berthold Seitz
- Klinik für Augenheilkunde, Universitätsklinikum des Saarlandes UKS, Homburg/Saar
| | - Achim Langenbucher
- Institut für Experimentelle Ophthalmologie, Universität des Saarlandes, Homburg/Saar
| | - Nóra Szentmáry
- Klinik für Augenheilkunde, Universitätsklinikum des Saarlandes UKS, Homburg/Saar
| |
Collapse
|
11
|
Tsuyama H, Fujishiro H, Himeno S, Sumi D. Arsenite suppresses NO production evoked by lipopolysaccharide and poly(I:C) via the suppression of interferon-β expression in RAW264.7 cells. J Toxicol Sci 2019; 44:83-92. [PMID: 30726814 DOI: 10.2131/jts.44.83] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Immunological functions are disturbed in humans who have been chronically exposed to arsenic via contaminated groundwater. Little is known about the specific mechanisms underlying the impairment of immunological defense system caused by arsenic. The activation of macrophage cells upon infection with bacteria and viruses plays important roles in the defense against these pathogens. Here we show that exposure to arsenite (As(III)) suppresses nitric oxide (NO) production in murine RAW264.7 macrophage cells stimulated with lipopolysaccharide (LPS) and poly(I:C), the compounds mimicking bacterial and viral infection, respectively. As(III) suppressed the LPS- or poly(I:C)-evoked induction of inducible NO synthase (iNOS) without affecting the transactivation of NF-κB. As the interferon (IFN)-β/STAT1 pathway is also involved in the induction of iNOS in addition to NF-κB, we examined the effects of As(III) on the expression and secretion of IFN-β, the expression of the components of IFN-α/β receptor, the phosphorylation of STAT1, and the levels of cytokines involved in STAT1 activation. The results showed that the expression and secretion of IFN-β were specifically suppressed by As(III) treatment in RAW264.7 cells stimulated with LPS or poly(I:C). These results suggest that As(III) suppresses the expression and secretion of IFN-β, leading to the reduced STAT1 activation and consequently the reduced iNOS induction in macrophage cells. Our data suggest an important role of the arsenic-induced suppression of IFN-β on the disturbances in immunological defense against both bacteria and viruses.
Collapse
Affiliation(s)
- Hiromasa Tsuyama
- Laboratory of Molecular Nutrition and Toxicology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University
| | - Hitomi Fujishiro
- Laboratory of Molecular Nutrition and Toxicology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University
| | - Seiichiro Himeno
- Laboratory of Molecular Nutrition and Toxicology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University
| | - Daigo Sumi
- Laboratory of Molecular Nutrition and Toxicology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University
| |
Collapse
|
12
|
Ghanbari R. Review on the Bioactive Peptides from Marine Sources: Indication for Health Effects. Int J Pept Res Ther 2018. [DOI: 10.1007/s10989-018-9766-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
13
|
Lee M, Rey K, Besler K, Wang C, Choy J. Immunobiology of Nitric Oxide and Regulation of Inducible Nitric Oxide Synthase. Results Probl Cell Differ 2017; 62:181-207. [PMID: 28455710 DOI: 10.1007/978-3-319-54090-0_8] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Nitric oxide (NO) is a bioactive gas that has multiple roles in innate and adaptive immune responses. In macrophages, nitric oxide is produced by inducible nitric oxide synthase upon microbial and cytokine stimulation. It is needed for host defense against pathogens and for immune regulation. This review will summarize the role of NO and iNOS in inflammatory and immune responses and will discuss the regulatory mechanisms that control inducible nitric oxide synthase expression and activity.
Collapse
Affiliation(s)
- Martin Lee
- Department of Molecular Biology and Biochemistry, Simon Fraser University, 8888 University Drive, Burnaby, BC, V5A 1S6, Canada
| | - Kevin Rey
- Department of Molecular Biology and Biochemistry, Simon Fraser University, 8888 University Drive, Burnaby, BC, V5A 1S6, Canada
| | - Katrina Besler
- Department of Molecular Biology and Biochemistry, Simon Fraser University, 8888 University Drive, Burnaby, BC, V5A 1S6, Canada
| | - Christine Wang
- Department of Molecular Biology and Biochemistry, Simon Fraser University, 8888 University Drive, Burnaby, BC, V5A 1S6, Canada
| | - Jonathan Choy
- Department of Molecular Biology and Biochemistry, Simon Fraser University, 8888 University Drive, Burnaby, BC, V5A 1S6, Canada.
| |
Collapse
|
14
|
Buzzo CDL, Medina T, Branco LM, Lage SL, Ferreira LCDS, Amarante-Mendes GP, Hottiger MO, De Carvalho DD, Bortoluci KR. Epigenetic regulation of nitric oxide synthase 2, inducible (Nos2) by NLRC4 inflammasomes involves PARP1 cleavage. Sci Rep 2017; 7:41686. [PMID: 28150715 PMCID: PMC5288713 DOI: 10.1038/srep41686] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 12/21/2016] [Indexed: 12/11/2022] Open
Abstract
Nitric oxide synthase 2, inducible (Nos2) expression is necessary for the microbicidal activity of macrophages. However, NOS2 over-activation causes multiple inflammatory disorders, suggesting a tight gene regulation is necessary. Using cytosolic flagellin as a model for inflammasome-dependent NOS2 activation, we discovered a surprising new role for NLRC4/caspase-1 axis in regulating chromatin accessibility of the Nos2 promoter. We found that activation of two independent mechanisms is necessary for NOS2 expression by cytosolic flagellin: caspase-1 and NF-κB activation. NF-κB activation was necessary, but not sufficient, for NOS2 expression. Conversely, caspase-1 was necessary for NOS2 expression, but dispensable for NF-κB activation, indicating that this protease acts downstream NF-κB activation. We demonstrated that epigenetic regulation of Nos2 by caspase-1 involves cleavage of the chromatin regulator PARP1 (also known as ARTD1) and chromatin accessibility of the NF-κB binding sites located at the Nos2 promoter. Remarkably, caspase-1-mediated Nos2 transcription and NO production contribute to the resistance of macrophages to Salmonella typhimurium infection. Our results uncover the molecular mechanism behind the constricted regulation of Nos2 expression and open new therapeutic opportunities based on epigenetic activities of caspase-1 against infectious and inflammatory diseases.
Collapse
Affiliation(s)
- Carina de Lima Buzzo
- Centro de Terapia Celular e Molecular (CTC-Mol) e Departamento de Ciências Biológicas - Universidade Federal de São Paulo, São Paulo, Brazil.,Departamento de Microbiologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Tiago Medina
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G 2M9, Canada
| | - Laura M Branco
- Centro de Terapia Celular e Molecular (CTC-Mol) e Departamento de Ciências Biológicas - Universidade Federal de São Paulo, São Paulo, Brazil.,Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo and Instituto de Investigação em Imunologia, Instituto Nacional de Ciência e Tecnologia (INCT-iii), Brazil
| | - Silvia L Lage
- Centro de Terapia Celular e Molecular (CTC-Mol) e Departamento de Ciências Biológicas - Universidade Federal de São Paulo, São Paulo, Brazil.,Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo and Instituto de Investigação em Imunologia, Instituto Nacional de Ciência e Tecnologia (INCT-iii), Brazil
| | | | - Gustavo P Amarante-Mendes
- Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo and Instituto de Investigação em Imunologia, Instituto Nacional de Ciência e Tecnologia (INCT-iii), Brazil
| | - Michael O Hottiger
- Department of Molecular Mechanisms of Disease, University of Zurich, Zurich, Switzerland
| | - Daniel D De Carvalho
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G 2M9, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, ON, M5G 2M9, Canada
| | - Karina R Bortoluci
- Centro de Terapia Celular e Molecular (CTC-Mol) e Departamento de Ciências Biológicas - Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
15
|
Mya Mya Mu, Chakravortty D, Sugiyama T, Koide N, Takahashi K, Mori I, Yoshida T, Yokochi T. The inhibitory action of quercetin on lipopolysaccharide-induced nitric oxide production in RAW 264.7 macrophage cells. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/09680519010070060601] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The effect of quercetin on lipopolysaccharide (LPS)-induced nitric oxide (NO) production was studied. Quercetin pretreatment significantly inhibited NO production in an LPS-stimulated RAW 264.7 murine macrophage cell line. Post-treatment with quercetin partially inhibited NO production. The inhibitory action of quercetin was due to neither the cytotoxic action nor altered LPS binding. The expression of inducible-type NO synthase (iNOS) was markedly down-regulated by quercetin. Quercetin suppressed the release of free nuclear factor (NF)-κB by preventing degradation of IκB-α and IκB-β. Moreover, quercetin blocked the phosphorylation of extracellular signal regulated kinase 1/2 (Erk1/2), p38, and c-Jun NH 2-terminal kinase/stress-activated protein kinase (JNK/SAPK) and, further, the activity of tyrosine kinases in LPS-stimulated RAW cells. Quercetin also inhibited interferon (IFN)-γ-induced NO production. Taken together, these results indicate that the inhibitory action of quercetin on NO production in LPS- and/or IFN-γ-stimulated macrophages might be due to abrogation of iNOS protein induction by impairment of a series of intracellular signal pathways.
Collapse
Affiliation(s)
- Mya Mya Mu
- Department of Microbiology and Immunology and Division of Bacterial Toxin, Research Center for Infectious Disease, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
| | - Dipshikha Chakravortty
- Department of Microbiology and Immunology and Division of Bacterial Toxin, Research Center for Infectious Disease, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
| | - Tsuyoshi Sugiyama
- Department of Microbiology and Immunology and Division of Bacterial Toxin, Research Center for Infectious Disease, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
| | - Naoki Koide
- Department of Microbiology and Immunology and Division of Bacterial Toxin, Research Center for Infectious Disease, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
| | - Kazuko Takahashi
- Department of Microbiology and Immunology and Division of Bacterial Toxin, Research Center for Infectious Disease, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
| | - Isamu Mori
- Department of Microbiology and Immunology and Division of Bacterial Toxin, Research Center for Infectious Disease, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
| | - Tomoaki Yoshida
- Department of Microbiology and Immunology and Division of Bacterial Toxin, Research Center for Infectious Disease, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
| | - Takashi Yokochi
- Department of Microbiology and Immunology and Division of Bacterial Toxin, Research Center for Infectious Disease, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan, -med-u.ac.jp
| |
Collapse
|
16
|
AP-1 Inhibition by SR 11302 Protects Human Hepatoma HepG2 Cells from Bile Acid-Induced Cytotoxicity by Restoring the NOS-3 Expression. PLoS One 2016; 11:e0160525. [PMID: 27490694 PMCID: PMC4973998 DOI: 10.1371/journal.pone.0160525] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 07/20/2016] [Indexed: 12/27/2022] Open
Abstract
The harmful effects of bile acid accumulation occurring during cholestatic liver diseases have been associated with oxidative stress increase and endothelial nitric oxide synthase (NOS-3) expression decrease in liver cells. We have previously reported that glycochenodeoxycholic acid (GCDCA) down-regulates gene expression by increasing SP1 binding to the NOS-3 promoter in an oxidative stress dependent manner. In the present study, we aimed to investigate the role of transcription factor (TF) AP-1 on the NOS-3 deregulation during GCDCA-induced cholestasis. The cytotoxic response to GCDCA was characterized by 1) the increased expression and activation of TFs cJun and c-Fos; 2) a higher binding capability of these at position -666 of the NOS-3 promoter; 3) a decrease of the transcriptional activity of the promoter and the expression and activity of NOS-3; and 4) the expression increase of cyclin D1. Specific inhibition of AP-1 by the retinoid SR 11302 counteracted the cytotoxic effects induced by GCDCA while promoting NOS-3 expression recovery and cyclin D1 reduction. NOS activity inhibition by L-NAME inhibited the protective effect of SR 11302. Inducible NOS isoform was no detected in this experimental model of cholestasis. Our data provide direct evidence for the involvement of AP-1 in the NOS-3 expression regulation during cholestasis and define a critical role for NOS-3 in regulating the expression of cyclin D1 during the cell damage induced by bile acids. AP-1 appears as a potential therapeutic target in cholestatic liver diseases given its role as a transcriptional repressor of NOS-3.
Collapse
|
17
|
Type I interferons exert anti-tumor effect via reversing immunosuppression mediated by mesenchymal stromal cells. Oncogene 2016; 35:5953-5962. [PMID: 27109100 PMCID: PMC5079855 DOI: 10.1038/onc.2016.128] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2015] [Revised: 03/06/2016] [Accepted: 03/15/2016] [Indexed: 12/13/2022]
Abstract
Mesenchymal stromal cells (MSCs) are strongly immunosuppressive via producing nitric oxide (NO) and known to migrate into tumor sites to promote tumor growth, but the underlying mechanisms remain largely elusive. Here, we found that interferon alpha (IFNα)-secreting MSCs showed more dramatic inhibition effect on tumor progression than that of IFNα alone. Interestingly, IFNα-primed MSCs could also effectively suppress tumor growth. Mechanistically, we demonstrated that both IFNα and IFNβ (type I IFNs) reversed the immunosuppressive effect of MSCs on splenocyte proliferation. This effect of type I IFNs was exerted through inhibiting inducible NO synthase (iNOS) expression in IFNγ and TNFα-stimulated MSCs. Notably, only NO production was inhibited by IFNα production of other cytokines or chemokines tested was not suppressed. Furthermore, IFNα promoted the switch from signal transducer and activator of transcription 1 (Stat1) homodimers to Stat1-Stat2 heterodimers. Studies using the luciferase reporter system and chromatin immunoprecipitation assay revealed that IFNα suppressed iNOS transcription through inhibiting the binding of Stat1 to iNOS promoter. Therefore, the synergistic anti-tumor effects of type I IFNs and MSCs were achieved by inhibiting NO production. This study provides essential information for understanding the mechanisms of MSC-mediated immunosuppression and for the development of better clinical strategies using IFNs and MSCs for cancer immunotherapy.
Collapse
|
18
|
Barani R, Motalleb G, Maghsoudi H. Evaluation of iNOS Expression in Esophageal Cancer Patients. Gastrointest Tumors 2016; 3:44-58. [PMID: 27722156 DOI: 10.1159/000443976] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 01/13/2016] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Esophageal cancer is a public health concern around the world; this cancer is the sixth leading cause of death of cancer in the world with about 386,000 deaths per year. Its risk factors include environmental factors such as tobacco smoke, gastroesophageal reflux and genetic changes. iNOS is stated by the effect of various inflammatory factors and is thus called inducible NOS. Investigating iNOS expression is a powerful tool for understanding effective molecular parameters at tissue and cellular responses to external factors. In this research work, iNOS expression in patients with esophageal cancer was studied in Iran. MATERIALS AND METHODS 15 formalin-fixed and paraffin-embedded (FFPE) esophageal cancer tissue samples and 15 normal FFPE samples were collected from various medical centers (Zabol, Zahedan, Kashan) to measure iNOS expression by real-time reverse transcriptase polymerase chain reaction (real-time RT-PCR). All PCR reactions were conducted by three replicates for iNOS and internal control (β-actin) by 2-ΔΔCT (Livak) method. Differences were measured in target gene expression in patients and control group using the t test. All statistical analyses were done using the SPSS software. RESULTS The results showed that there was no significant difference between iNOS expression in the case and control groups (p > 0.05); however, there was an increase in iNOS expression in the case group. On the other hand, there was a significant difference between iNOS expression in males and females in the two groups of healthy subjects and patients, and it was higher in women than in men. CONCLUSION Further studies need to be conducted with larger sample sizes and in other populations to validate these findings.
Collapse
Affiliation(s)
- Romina Barani
- Department of Biotechnology, Faculty of Science, Payame Noor University, Tehran
| | | | - Hossein Maghsoudi
- Department of Biotechnology, Faculty of Science, Payame Noor University, Tehran
| |
Collapse
|
19
|
Vannini F, Kashfi K, Nath N. The dual role of iNOS in cancer. Redox Biol 2015; 6:334-343. [PMID: 26335399 PMCID: PMC4565017 DOI: 10.1016/j.redox.2015.08.009] [Citation(s) in RCA: 354] [Impact Index Per Article: 39.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Revised: 08/07/2015] [Accepted: 08/10/2015] [Indexed: 01/02/2023] Open
Abstract
Nitric oxide (NO) is one of the 10 smallest molecules found in nature. It is a simple gaseous free radical whose predominant functions is that of a messenger through cGMP. In mammals, NO is synthesized by the enzyme nitric oxide synthase (NOS) of which there are three isoforms. Neuronal (nNOS, NOS1) and endothelial (eNOS, NOS3) are constitutive calcium-dependent forms of the enzyme that regulate neural and vascular function respectively. The third isoform (iNOS, NOS2), is calcium-independent and is inducible. In many tumors, iNOS expression is high, however, the role of iNOS during tumor development is very complex and quite perplexing, with both promoting and inhibiting actions having been described. This review will aim to summarize the dual actions of iNOS-derived NO showing that the microenvironment of the tumor is a contributing factor to these observations and ultimately to cellular outcomes. NO is pro- and anti-tumorigenic. High concentrations of NO maybe anti-tumorigenic. iNOS produces high concentrations of NO and relates to tumor growth or its inhibition. iNOS is associated with cytotoxicity, apoptosis and bystander anti-tumor effects. Tumor- and stromal-iNOS, and the ‘cell situation’ contribute to anti or pro-tumor effects. Dual role of iNOS is influenced by the cell situation and is environment dependent.
Collapse
Affiliation(s)
- Federica Vannini
- Department of Physiology, Pharmacology and Neuroscience, Sophie Davis School of Biomedical Education, City University of New York Medical School, New York, NY 10031, United States
| | - Khosrow Kashfi
- Department of Physiology, Pharmacology and Neuroscience, Sophie Davis School of Biomedical Education, City University of New York Medical School, New York, NY 10031, United States
| | - Niharika Nath
- Department of Life Sciences, New York Institute of Technology, NY 10023, United States.
| |
Collapse
|
20
|
Ma S, Tan S, Fang D, Zhang R, Zhou S, Wu W, Zheng K. Probing the binding mechanism of novel dual NF-κB/AP-1 inhibitors by 3D-QSAR, docking and molecular dynamics simulations. RSC Adv 2015. [DOI: 10.1039/c5ra10831d] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Potent dual NF-κB/AP-1 inhibitors could effectively treat immunoinflammatory diseases. An integrated computational study was carried out to identify the most favourable binding sites, the structural features and the interaction mechanisms.
Collapse
Affiliation(s)
- Shaojie Ma
- Department of Physical Chemistry
- College of Pharmacy
- Guangdong Pharmaceutical University
- Guangzhou 510006
- PR China
| | - Shepei Tan
- Department of Physical Chemistry
- College of Pharmacy
- Guangdong Pharmaceutical University
- Guangzhou 510006
- PR China
| | - Danqing Fang
- Department of Cardiothoracic Surgery
- Affiliated Second Hospital of Guangzhou Medical University
- Guangzhou 510260
- PR China
| | - Rong Zhang
- Department of Physical Chemistry
- College of Pharmacy
- Guangdong Pharmaceutical University
- Guangzhou 510006
- PR China
| | - Shengfu Zhou
- Department of Physical Chemistry
- College of Pharmacy
- Guangdong Pharmaceutical University
- Guangzhou 510006
- PR China
| | - Wenjuan Wu
- Department of Physical Chemistry
- College of Pharmacy
- Guangdong Pharmaceutical University
- Guangzhou 510006
- PR China
| | - Kangcheng Zheng
- School of Chemistry and Chemical Engineering
- SunYat-Sen University
- Guangzhou 510275
- PR China
| |
Collapse
|
21
|
Hoos MD, Vitek MP, Ridnour LA, Wilson J, Jansen M, Everhart A, Wink DA, Colton CA. The impact of human and mouse differences in NOS2 gene expression on the brain's redox and immune environment. Mol Neurodegener 2014; 9:50. [PMID: 25403885 PMCID: PMC4247207 DOI: 10.1186/1750-1326-9-50] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Accepted: 10/10/2014] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Mouse models are used in the study of human disease. Despite well-known homologies, the difference in immune response between mice and humans impacts the application of data derived from mice to human disease outcomes. Nitric oxide synthase-2 (NOS2) is a key gene that displays species-specific outcomes via altered regulation of the gene promoter and via post-transcriptional mechanisms in humans that are not found in mice. The resulting levels of NO produced by activation of human NOS2 are different from the levels of NO produced by mouse Nos2. Since both tissue redox environment and immune responsiveness are regulated by the level of NO and its interactions, we investigated the significance of mouse and human differences on brain oxidative stress and on immune activation in HuNOS2tg/mNos2-/- mice that express the entire human NOS2 gene and that lack a functional mNos2 compared to wild type (WT) mice that express normal mNos2. METHODS/RESULTS Similarly to human, brain tissue from HuNOS2tg/mNos2-/- mice showed the presence of a NOS2 gene 3'UTR binding site. We also identified miRNA-939, the binding partner for this site, in mouse brain lysates and further demonstrated reduced levels of nitric oxide (NO) typical of the human immune response on injection with lipopolysaccharide (LPS). HuNOS2tg/mNos2-/- brain samples were probed for characteristic differences in redox and immune gene profiles compared to WT mice using gene arrays. Selected genes were also compared against mNos2-/- brain lysates. Reconstitution of the human NOS2 gene significantly altered genes that encode multiple anti-oxidant proteins, oxidases, DNA repair, mitochondrial proteins and redox regulated immune proteins. Expression levels of typical pro-inflammatory, anti-inflammatory and chemokine genes were not significantly different with the exception of increased TNFα and Ccr1 mRNA expression in the HuNOS2tg/mNos2-/- mice compared to WT or mNos2-/- mice. CONCLUSIONS NO is a principle factor in establishing the tissue redox environment and changes in NO levels impact oxidative stress and immunity, both of which are primary characteristics of neurodegenerative diseases. The HuNOS2tg/mNos2-/- mice provide a potentially useful mechanism to address critical species- specific immune differences that can impact the study of human diseases.
Collapse
Affiliation(s)
- Michael D Hoos
- />Department of Neurosurgery, Stonybrook Health Sciences, Stony Brook, NY 11794 USA
| | - Michael P Vitek
- />Department of Neurology, Duke University Medical Center, Durham, NC 27710 USA
| | - Lisa A Ridnour
- />Radiation Biology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892 USA
| | - Joan Wilson
- />Department of Neurology, Duke University Medical Center, Durham, NC 27710 USA
| | - Marilyn Jansen
- />Department of Neurology, Duke University Medical Center, Durham, NC 27710 USA
| | - Angela Everhart
- />Department of Neurology, Duke University Medical Center, Durham, NC 27710 USA
| | - David A Wink
- />Radiation Biology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892 USA
| | - Carol A Colton
- />Department of Neurology, Duke University Medical Center, Durham, NC 27710 USA
| |
Collapse
|
22
|
Differential signaling of inducible nitric oxide synthase induction in Mycobacterium tuberculosis infected alveolar epithelial cell line A549 in response to cytokines IFN-γ, TNF-α and IL-1β. Int J Mycobacteriol 2014; 3:17-24. [DOI: 10.1016/j.ijmyco.2014.01.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Revised: 01/13/2014] [Accepted: 01/16/2014] [Indexed: 11/18/2022] Open
|
23
|
Borner T, Pinkernell S, Lutz TA, Riediger T. Lipopolysaccharide inhibits ghrelin-excited neurons of the arcuate nucleus and reduces food intake via central nitric oxide signaling. Brain Behav Immun 2012; 26:867-79. [PMID: 22465682 DOI: 10.1016/j.bbi.2012.03.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2012] [Revised: 03/14/2012] [Accepted: 03/19/2012] [Indexed: 12/30/2022] Open
Abstract
Lipopolysaccharide (LPS) induces anorexia and expression of inducible nitric oxide synthase (iNOS) in the hypothalamic arcuate nucleus (Arc). Peripheral administration of the iNOS inhibitor 1400 W counteracts the anorectic effects of LPS. Here we investigated the role of central NO signaling in LPS anorexia. In electrophysiological studies we tested whether 1400 W counteracts the iNOS-dependent inhibition of Arc neurons triggered by in vivo or in vitro stimulation with LPS. We used the hormone ghrelin as a functional reference stimulus because ghrelin is known to activate orexigenic Arc neurons. Further, we investigated whether in vitro LPS stimulation induces an iNOS-mediated formation of the second messenger cGMP. Since the STAT1 pathway contributes to the regulation of iNOS expression we investigated whether LPS treatment induces STAT1 phosphorylation in the Arc. Finally we tested the effect of intracerebroventricular injection of 1400 W on LPS-induced anorexia. Superfusion with 1400 W (10(-4) M) increased neuronal activity in 37% of neurons in Arc slices from LPS treated (100 μg/kg ip) but not from saline treated rats. Similarly, 1400 W excited 45% of Arc neurons after in vitro stimulation with LPS (100 ng/ml). In both approaches, a considerable percentage of 1400 W sensitive neurons were excited by ghrelin (10(-8)M; 50% and 75%, respectively). In vitro stimulation with LPS induced cGMP formation in the Arc, which was blocked by co-incubation with 1400 W. LPS treatment elicited a pSTAT1 response in the Arc of mice. Central 1400 W injection (4 μg/rat) attenuated LPS-induced anorexia and counteracted the LPS-dependent decrease in respiratory quotient and energy expenditure. In conclusion, the current findings substantiate a role of central iNOS dependent NO formation in LPS-induced effects on eating and energy homeostasis. A pharmacological blockade of NO formation might be a therapeutic approach to ameliorate disease-related anorexia.
Collapse
Affiliation(s)
- Tito Borner
- Institute of Veterinary Physiology and Centre of Integrative Human Physiology, University of Zurich, 8057 Zurich, Switzerland
| | | | | | | |
Collapse
|
24
|
McAdam E, Haboubi HN, Forrester G, Eltahir Z, Spencer-Harty S, Davies C, Griffiths AP, Baxter JN, Jenkins GJS. Inducible nitric oxide synthase (iNOS) and nitric oxide (NO) are important mediators of reflux-induced cell signalling in esophageal cells. Carcinogenesis 2012; 33:2035-43. [PMID: 22826608 DOI: 10.1093/carcin/bgs241] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Nitric oxide (NO) produced by inducible nitric oxide synthase (iNOS) has been implicated in both DNA damage induction and aberrant cell signalling in various tissue and cell backgrounds. We investigated here the role of iNOS and NO in DNA damage induction and nuclear factor-kappa B (NF-κB) signalling in esophageal cells in vitro. As esophageal adenocarcinoma develops in a background of Barrett's esophagus secondary to reflux disease, it is possible that inflammatory mediators like NO may be important in esophageal cancer development. We show that reflux components like stomach acid and bile acids [deoxycholic acid (DCA)] can induce iNOS gene and protein expression and produce NO generation in esophageal cells, using real-time PCR, western blotting and NO sensitive fluorescent probes, respectively. This up-regulation of iNOS expression was not dependent on NF-κB activity. DCA-induced DNA damage was independent of NF-κB and only partially dependent on iNOS and NO, as measured by the micronucleus assay. These same reflux constituents also activated the oncogenic transcription factor NF-κB, as measured by transcription factor enzyme-linked immunosorbent assay and gene expression studies with NF-κB linked genes (e.g. interleukin-8). Importantly, we show here for the first time that basal levels of NF-κB activity (and possibly acid and DCA-induced NF-κB) are dependent on iNOS/NO and this may lead to a positive feedback loop whereby induced iNOS is upstream of NF-κB, hence prolonging and potentially amplifying this signalling, presumably through NO activation of NF-κB. Furthermore, we confirm increased protein levels of iNOS in esophageal adenocarcinoma and, therefore, in neoplastic development in the esophagus.
Collapse
Affiliation(s)
- E McAdam
- Institute of Life Science, School of Medicine, Swansea University Swansea, SA28PP, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Khasnavis S, Jana A, Roy A, Mazumder M, Bhushan B, Wood T, Ghosh S, Watson R, Pahan K. Suppression of nuclear factor-κB activation and inflammation in microglia by physically modified saline. J Biol Chem 2012; 287:29529-42. [PMID: 22753407 DOI: 10.1074/jbc.m111.338012] [Citation(s) in RCA: 89] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Chronic inflammation involving activated microglia and astroglia is becoming a hallmark of many human diseases, including neurodegenerative disorders. Although NF-κB is a multifunctional transcription factor, it is an important target for controlling inflammation as the transcription of many proinflammatory molecules depends on the activation of NF-κB. Here, we have undertaken a novel approach to attenuate NF-κB activation and associated inflammation in activated glial cells. RNS60 is a 0.9% saline solution containing charge-stabilized nanostructures that are generated by subjecting normal saline to Taylor-Couette-Poiseuille (TCP) flow under elevated oxygen pressure. RNS60, but not normal saline, RNS10.3 (TCP-modified saline without excess oxygen), and PNS60 (saline containing excess oxygen without TCP modification) were found to inhibit the production of nitric oxide (NO) and the expression of inducible NO synthase in activated microglia. Similarly, RNS60 also inhibited the expression of inducible NO synthase in activated astroglia. Inhibition of NF-κB activation by RNS60 suggests that RNS60 exerts its anti-inflammatory effect through the inhibition of NF-κB. Interestingly, RNS60 induced the activation of type IA phosphatidylinositol (PI) 3-kinase and Akt and rapidly up-regulated IκBα, a specific endogenous inhibitor of NF-κB. Inhibition of PI 3-kinase and Akt by either chemical inhibitors or dominant-negative mutants abrogated the RNS60-mediated up-regulation of IκBα. Furthermore, we demonstrate that RNS60 induced the activation of cAMP-response element-binding protein (CREB) via the PI 3-kinase-Akt pathway and that RNS60 up-regulated IκBα via CREB. These results describe a novel anti-inflammatory property of RNS60 via type IA PI 3-kinase-Akt-CREB-mediated up-regulation of IκBα, which may be of therapeutic benefit in neurodegenerative disorders.
Collapse
Affiliation(s)
- Saurabh Khasnavis
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL 60612, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Lin CC, Shih CH, Yang YL, Bien MY, Lin CH, Yu MC, Sureshbabu M, Chen BC. Thrombin induces inducible nitric oxide synthase expression via the MAPK, MSK1, and NF-κB signaling pathways in alveolar macrophages. Eur J Pharmacol 2011; 672:180-7. [DOI: 10.1016/j.ejphar.2011.10.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2011] [Revised: 09/28/2011] [Accepted: 10/02/2011] [Indexed: 12/17/2022]
|
27
|
The JAK-STAT pathway controls Plasmodium vivax load in early stages of Anopheles aquasalis infection. PLoS Negl Trop Dis 2011; 5:e1317. [PMID: 22069502 PMCID: PMC3206008 DOI: 10.1371/journal.pntd.0001317] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2010] [Accepted: 07/28/2011] [Indexed: 12/24/2022] Open
Abstract
Malaria affects 300 million people worldwide every year and 450,000 in Brazil. In coastal areas of Brazil, the main malaria vector is Anopheles aquasalis, and Plasmodium vivax is responsible for the majority of malaria cases in the Americas. Insects possess a powerful immune system to combat infections. Three pathways control the insect immune response: Toll, IMD, and JAK-STAT. Here we analyze the immune role of the A. aquasalis JAK-STAT pathway after P. vivax infection. Three genes, the transcription factor Signal Transducers and Activators of Transcription (STAT), the regulatory Protein Inhibitors of Activated STAT (PIAS) and the Nitric Oxide Synthase enzyme (NOS) were characterized. Expression of STAT and PIAS was higher in males than females and in eggs and first instar larvae when compared to larvae and pupae. RNA levels for STAT and PIAS increased 24 and 36 hours (h) after P. vivax challenge. NOS transcription increased 36 h post infection (hpi) while this protein was already detected in some midgut epithelial cells 24 hpi. Imunocytochemistry experiments using specific antibodies showed that in non-infected insects STAT and PIAS were found mostly in the fat body, while in infected mosquitoes the proteins were found in other body tissues. The knockdown of STAT by RNAi increased the number of oocysts in the midgut of A. aquasalis. This is the first clear evidence for the involvement of a specific immune pathway in the interaction of the Brazilian malaria vector A. aquasalis with P. vivax, delineating a potential target for the future development of disease controlling strategies.
Collapse
|
28
|
Dual specificity phosphatase 1 regulates human inducible nitric oxide synthase expression by p38 MAP kinase. Mediators Inflamm 2011; 2011:127587. [PMID: 21547253 PMCID: PMC3086212 DOI: 10.1155/2011/127587] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2010] [Accepted: 02/15/2011] [Indexed: 01/12/2023] Open
Abstract
The role of dual specificity phosphatase 1 (DUSP1) in inducible nitric oxide synthase (iNOS) expression in A549 human pulmonary epithelial cells, J774 mouse macrophages and primary mouse bone marrow-derived macrophages (BMMs) was investigated. iNOS expression was induced by a cytokine mixture (TNF, IFNγ and IL-1β) in A549 cells and by LPS in J774 cells, and it was inhibited by p38 MAPK inhibitors SB202190 and BIRB 796. Stimulation with cytokine mixture or LPS enhanced also DUSP1 expression. Down-regulation of DUSP1 by siRNA increased p38 MAPK phosphorylation and iNOS expression in A549 and J774 cells. In addition, LPS-induced iNOS expression was enhanced in BMMs from DUSP1(−/−) mice as compared to that in BMMs from wild-type mice. The results indicate that DUSP1 suppresses iNOS expression by limiting p38 MAPK activity in human and mouse cells. Compounds that enhance DUSP1 expression or modulate its function may be beneficial in diseases complicated with increased iNOS-mediated NO production.
Collapse
|
29
|
Kai JI, Huang WC, Tsai CC, Chang WT, Chen CL, Lin CF. Glycogen synthase kinase-3β indirectly facilitates interferon-γ-induced nuclear factor-κB activation and nitric oxide biosynthesis. J Cell Biochem 2011; 111:1522-30. [PMID: 20872791 DOI: 10.1002/jcb.22881] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Either glycogen synthase kinase (GSK)-3β or nuclear factor (NF)-κB regulates interferon (IFN)-γ-induced nitric oxide (NO) biosynthesis; however, the inter-regulation between GSK-3β and NF-κB is unknown. We have previously shown that IFN-γ-activated GSK-3β negatively regulates Src homology-2 domain-containing phosphatase (SHP) 2 to facilitate Janus kinase (Jak) 2-signal transducer and activator of transcription (STAT) 1 activation. Because Jaks-IFN-inducible dsRNA-activated serine-threonine protein kinase (PKR) axis signaling is essential for IFN-γ-activation of NF-κB, in this study we investigate the potential mechanism for GSK-3β-facilitated NF-κB activation in IFN-γ-stimulated RAW264.7 murine macrophages. Pharmacological inhibitors of GSK-3β or NF-κB signaling, such as the inhibitor of κB (IκB) kinase β (IKKβ) and IκBα, inhibited IFN-γ-induced inducible NO synthase (iNOS) and thus NO biosynthesis. Inhibiting GSK-3β decreased IFN-γ-induced NF-κB phosphorylation (Ser536) and activation. The upstream regulators for GSK-3β activation, including okadaic acid-sensitive protein phosphatase and proline-rich tyrosine kinase 2, were also important for IFN-γ-induced IκBα phosphorylation (Ser32) and degradation. Under IFN-γ stimulation, Jak2-PKR axis signaling induced IκBα inactivation as well as iNOS/NO biosynthesis. It is notable that inhibiting GSK-3β caused SHP2-mediated dephosphorylation of PKR (Thr446), IKKβ (Ser180), and NF-κB (Ser536). Taken together, we provide the first evidence to demonstrate that GSK-3β indirectly facilitates IFN-γ-induced NF-κB activation by inhibiting SHP2, in turn activating the PKR-IKKβ-IκBα axis signaling pathway.
Collapse
Affiliation(s)
- Jui-In Kai
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | | | | | | | | | | |
Collapse
|
30
|
Ramasamy K, Dwyer-Nield LD, Serkova NJ, Hasebroock KM, Tyagi A, Raina K, Singh RP, Malkinson AM, Agarwal R. Silibinin prevents lung tumorigenesis in wild-type but not in iNOS-/- mice: potential of real-time micro-CT in lung cancer chemoprevention studies. Clin Cancer Res 2011; 17:753-61. [PMID: 21148748 PMCID: PMC3066181 DOI: 10.1158/1078-0432.ccr-10-2290] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
PURPOSE Sustained nitric oxide (NO) generation positively correlates with lung cancer development and progression. Herein, we genetically confirmed this role of iNOS and evaluated the chemopreventive efficacy of silibinin in carcinogen-treated B6/129 wild-type (WT) and iNOS(-/-) mice. EXPERIMENTAL DESIGN Male B6/129-Nos2(tm1Lau) (iNOS(-/-)) and B6/129PF2 WT mice were injected i.p. with 1 mg/g body weight urethane once weekly for 7 consecutive weeks, followed by silibinin gavage (742 mg/kg body weight) for 5 d/wk for 18 weeks. RESULTS Quantification of micro-CT data in real-time showed that silibinin significantly decreases urethane-induced tumor number and size in WT mice, consistent with measurements made ex vivo at study termination. Genetic ablation of iNOS decreased urethane-induced tumor multiplicity by 87% (P < 0.001) compared to WT mice. Silibinin decreased tumor multiplicity by 71% (P < 0.01) in WT mice, but did not show any such considerable effect in iNOS(-/-) mice. Tumors from WT mice expressed more iNOS (P < 0.01) but almost similar eNOS and nNOS than those in silibinin-treated mice. In these tumors, silibinin moderately (P < 0.01) inhibited cell proliferation but strongly (P < 0.01) reduced the number of newly formed nestin-positive microvessels. Silibinin decreased VEGFR2 level, and STAT3 and NF-κB activation in tumors. CONCLUSIONS The lack of effect of silibinin in iNOS(-/-) mice suggests that silibinin exerts most of its chemopreventive and angiopreventive effects through its inhibition of iNOS expression in lung tumors. Our results support iNOS as a potential target for controlling lung cancer, and demonstrate the value of real-time noninvasive micro-CT imaging modality for evaluating the efficacy of lung cancer chemopreventive agents.
Collapse
Affiliation(s)
| | - Lori D. Dwyer-Nield
- Department of Pharmaceutical Sciences, University of Colorado Denver, Aurora, Colorado, USA
| | - Natalie J. Serkova
- Departments of Anesthesiology and Radiology, University of Colorado Denver, Aurora, Colorado, USA
- University of Colorado Cancer Center, University of Colorado Denver, Aurora, Colorado, USA
| | - Kendra M. Hasebroock
- Departments of Anesthesiology and Radiology, University of Colorado Denver, Aurora, Colorado, USA
| | - Alpna Tyagi
- Department of Pharmaceutical Sciences, University of Colorado Denver, Aurora, Colorado, USA
| | - Komal Raina
- Department of Pharmaceutical Sciences, University of Colorado Denver, Aurora, Colorado, USA
| | - Rana P. Singh
- Department of Pharmaceutical Sciences, University of Colorado Denver, Aurora, Colorado, USA
- Cancer Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Alvin M. Malkinson
- Department of Pharmaceutical Sciences, University of Colorado Denver, Aurora, Colorado, USA
- University of Colorado Cancer Center, University of Colorado Denver, Aurora, Colorado, USA
| | - Rajesh Agarwal
- Department of Pharmaceutical Sciences, University of Colorado Denver, Aurora, Colorado, USA
- University of Colorado Cancer Center, University of Colorado Denver, Aurora, Colorado, USA
| |
Collapse
|
31
|
Li W, Liu H, Fu L, Li D, Zhao Y. Identification of Yin Yang 1-interacting partners at −1026C/A in the human iNOS promoter. Arch Biochem Biophys 2010; 498:119-26. [DOI: 10.1016/j.abb.2010.04.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2009] [Revised: 04/22/2010] [Accepted: 04/23/2010] [Indexed: 11/27/2022]
|
32
|
Pautz A, Art J, Hahn S, Nowag S, Voss C, Kleinert H. Regulation of the expression of inducible nitric oxide synthase. Nitric Oxide 2010; 23:75-93. [PMID: 20438856 DOI: 10.1016/j.niox.2010.04.007] [Citation(s) in RCA: 383] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2010] [Accepted: 04/23/2010] [Indexed: 12/13/2022]
Abstract
Nitric oxide (NO) generated by the inducible isoform of nitric oxide synthase (iNOS) is involved in complex immunomodulatory and antitumoral mechanisms and has been described to have multiple beneficial microbicidal, antiviral and antiparasital effects. However, dysfunctional induction of iNOS expression seems to be involved in the pathophysiology of several human diseases. Therefore iNOS has to be regulated very tightly. Modulation of expression, on both the transcriptional and post-transcriptional level, is the major regulation mechanism for iNOS. Pathways resulting in the induction of iNOS expression vary in different cells or species. Activation of the transcription factors NF-kappaB and STAT-1alpha and thereby activation of the iNOS promoter seems to be an essential step for the iNOS induction in most human cells. However, at least in the human system, also post-transcriptional mechanisms involving a complex network of RNA-binding proteins build up by AUF1, HuR, KSRP, PTB and TTP is critically involved in the regulation of iNOS expression. Recent data also implicate regulation of iNOS expression by non-coding RNAs (ncRNAs).
Collapse
Affiliation(s)
- Andrea Pautz
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg University Mainz, D-55101 Mainz, Germany
| | | | | | | | | | | |
Collapse
|
33
|
Wang SX, Hu LM, Gao XM, Guo H, Fan GW. Anti-inflammatory activity of salvianolic acid B in microglia contributes to its neuroprotective effect. Neurochem Res 2010; 35:1029-37. [PMID: 20238162 DOI: 10.1007/s11064-010-0151-1] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2009] [Accepted: 03/06/2010] [Indexed: 01/04/2023]
Abstract
This study examined whether Salvianolic acid B (Sal B), a major active component of Chinese herb Radix Salviae Miltiorrhizae, may exert an anti-inflammatory effect in microglia and may be neuroprotective by regulating microglial activation. Our results showed that Sal B significantly reduced the production of nitric oxide (NO), tumor necrosis factor-alpha (TNF-alpha), interleukin-1beta (IL-1beta) and reactive oxygen species (ROS) induced by lipopolysaccharide (LPS) treatment in rat primary microglia in a dose-dependent manner. Sal B had no effects on ATP-dependent IL-1beta release and interferon (IFN)-gamma-induced NO production. Sal B also suppressed LPS-induced inducible nitric oxide synthase (iNOS), TNF-alpha, and IL-1beta mRNA expression, which was accompanied by inhibiting transcription factor NF-kappaB activation. Sal B could protect neurons through inhibition of microglial activation in a microglia-neuron coculture system. In conclusion, these data demonstrate that anti-inflammatory activity of Sal B in microglia contributes to its neuroprotective effect and suggest that it may be useful for preventing microglia-mediated neuroinflammation.
Collapse
Affiliation(s)
- Shao-Xia Wang
- Tianjin Key Laboratory of Traditional Chinese Medicine Pharmacology, Tianjin University of Traditional Chinese Medicine, 312 Anshanxi Road, 300193, Nankai District, Tianjin, China
| | | | | | | | | |
Collapse
|
34
|
|
35
|
Schmidt N, Pautz A, Art J, Rauschkolb P, Jung M, Erkel G, Goldring MB, Kleinert H. Transcriptional and post-transcriptional regulation of iNOS expression in human chondrocytes. Biochem Pharmacol 2009; 79:722-32. [PMID: 19854161 DOI: 10.1016/j.bcp.2009.10.012] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2009] [Revised: 10/13/2009] [Accepted: 10/13/2009] [Indexed: 11/18/2022]
Abstract
Chondrocytes are important for the development and maintenance of articular cartilage. However, both in osteoarthritis (OA) and rheumatoid arthritis (RA) chondrocytes are involved in the process of cartilage degradation and synthesize important immunomodulatory mediators, including nitric oxide (NO) generated by the inducible NO synthase (iNOS). To uncover the role of iNOS in the pathomechanisms of OA and RA, we analyzed the regulation of iNOS expression using immortalized human chondrocytes as a reproducible model. In C-28/I2 chondrocytes, iNOS expression was associated with the expression of the chondrocyte phenotype. Peak induction by a cytokine cocktail occurred between 6 and 8h and declined by 24h. Inhibition of p38MAPK, NF-kappaB and the JAK2-STAT-1alpha pathways resulted in a reduction of iNOS expression. In contrast to other cell types, the cytokine-mediated induction of the human iNOS promoter paralleled the induction rate of the iNOS mRNA expression in C-28/I2 chondrocytes. However, in addition post-transcriptional regulation of iNOS expression by the RNA binding protein KSRP seems to operate in these cells. As seen in other chondrocyte models, glucocorticoids were not able to inhibit cytokine-induced iNOS expression in C-28/I2 cells, due to the lack of the glucocorticoid receptor mRNA expression. In this model of glucocorticoid-resistance, the new fungal anti-inflammatory compound S-curvularin was able to inhibit cytokine-induced iNOS expression and iNOS-dependent NO-production. In summary, we demonstrate for the first time that differentiated human immortalized C-28/I2 chondrocytes are a representative cell culture model to investigate iNOS gene expression in human joint diseases.
Collapse
MESH Headings
- Anti-Inflammatory Agents/pharmacology
- Cartilage, Articular/drug effects
- Cartilage, Articular/enzymology
- Cell Line, Transformed
- Chondrocytes/drug effects
- Chondrocytes/enzymology
- Cytokines/pharmacology
- Enzyme Induction
- Gene Expression Regulation, Enzymologic/drug effects
- Gene Expression Regulation, Enzymologic/genetics
- Humans
- Interferon-Stimulated Gene Factor 3/antagonists & inhibitors
- Janus Kinase 2/antagonists & inhibitors
- NF-kappa B p50 Subunit/antagonists & inhibitors
- Nitric Oxide Synthase Type II/biosynthesis
- Nitric Oxide Synthase Type II/genetics
- RNA Processing, Post-Transcriptional/drug effects
- RNA Processing, Post-Transcriptional/genetics
- RNA, Messenger/metabolism
- RNA-Binding Proteins/genetics
- RNA-Binding Proteins/metabolism
- Receptors, Glucocorticoid/genetics
- Receptors, Glucocorticoid/metabolism
- Trans-Activators/genetics
- Trans-Activators/metabolism
- Zearalenone/analogs & derivatives
- Zearalenone/pharmacology
- p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
Collapse
Affiliation(s)
- Nadine Schmidt
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg University Mainz, Obere Zahlbacher Strasse 67, D-55131 Mainz, Germany
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Brahmachari S, Jana A, Pahan K. Sodium benzoate, a metabolite of cinnamon and a food additive, reduces microglial and astroglial inflammatory responses. THE JOURNAL OF IMMUNOLOGY 2009; 183:5917-27. [PMID: 19812204 DOI: 10.4049/jimmunol.0803336] [Citation(s) in RCA: 108] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Upon activation, microglia and astrocytes produce a number of proinflammatory molecules that participate in the pathophysiology of several neurodegenerative disorders. This study explores the anti-inflammatory property of cinnamon metabolite sodium benzoate (NaB) in microglia and astrocytes. NaB, but not sodium formate, was found to inhibit LPS-induced expression of inducible NO synthase (iNOS), proinflammatory cytokines (TNF-alpha and IL-1beta) and surface markers (CD11b, CD11c, and CD68) in mouse microglia. Similarly, NaB also inhibited fibrillar amyloid beta (Abeta)-, prion peptide-, double-stranded RNA (polyinosinic-polycytidylic acid)-, HIV-1 Tat-, 1-methyl-4-phenylpyridinium(+)-, IL-1beta-, and IL-12 p40(2)-induced microglial expression of iNOS. In addition to microglia, NaB also suppressed the expression of iNOS in mouse peritoneal macrophages and primary human astrocytes. Inhibition of NF-kappaB activation by NaB suggests that NaB exerts its anti-inflammatory effect through the inhibition of NF-kappaB. Although NaB reduced the level of cholesterol in vivo in mice, reversal of the inhibitory effect of NaB on iNOS expression, and NF-kappaB activation by hydroxymethylglutaryl-CoA, mevalonate, and farnesyl pyrophosphate, but not cholesterol and ubiquinone, suggests that depletion of intermediates, but not end products, of the mevalonate pathway is involved in the anti-inflammatory effect of NaB. Furthermore, we demonstrate that an inhibitor of p21(ras) farnesyl protein transferase suppressed the expression of iNOS, that activation of p21(ras) alone was sufficient to induce the expression of iNOS, and that NaB suppressed the activation of p21(ras) in microglia. These results highlight a novel anti-inflammatory role of NaB via modulation of the mevalonate pathway and p21(ras).
Collapse
Affiliation(s)
- Saurav Brahmachari
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL 60612, USA
| | | | | |
Collapse
|
37
|
Marigo I, Dolcetti L, Serafini P, Zanovello P, Bronte V. Tumor-induced tolerance and immune suppression by myeloid derived suppressor cells. Immunol Rev 2009; 222:162-79. [PMID: 18364001 DOI: 10.1111/j.1600-065x.2008.00602.x] [Citation(s) in RCA: 489] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Emerging evidence indicates that the Achilles' heel of cancer immunotherapies is often the complex interplay of tumor-derived factors and deviant host properties, which involve a wide range of immune elements in the lymphoid and myeloid compartments. Regulatory lymphocytes, tumor-conditioned myeloid-derived suppressor cells (MDSCs), tumor-associated macrophages, and dysfunctional and immature dendritic cells take part in a complex immunoregulatory network. Despite the fact that some mechanisms governing tumor-induced immune tolerance and suppression are starting to be better understood and their complexity dissected, little is known about the diachronic picture of immune tolerance. Based on observations of MDSCs, we present a time-structured and topologically consistent idea of tumor-dependent tolerance progression in tumor-bearing hosts.
Collapse
Affiliation(s)
- Ilaria Marigo
- Department of Oncology and Surgical Sciences, Padova University, Padova, Italy, and Venetian Institute for Molecular Medicine, Padova, Italy
| | | | | | | | | |
Collapse
|
38
|
Roles of NF-kappaB activation and peroxisome proliferator-activated receptor gamma inhibition in the effect of rifampin on inducible nitric oxide synthase transcription in human lung epithelial cells. Antimicrob Agents Chemother 2008; 53:1539-45. [PMID: 19114679 DOI: 10.1128/aac.00961-08] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Rifampin (rifampicin), an important antibiotic agent and a major drug used for the treatment of tuberculosis, exerts immunomodulatory effects. Previous studies have found that rifampin increases inducible nitric oxide (NO) synthase (iNOS) expression and NO production. The present study investigated the potential mechanism(s) underlying these actions. The incubation of human lung epithelial A549 cells with a cytokine mix (interleukin-1beta, tumor necrosis factor alpha, and gamma interferon) induced the expression of iNOS mRNA. The addition of rifampin increased the iNOS level by 1.9 +/- 0.3-fold at a dose of 10 microg/ml (P < 0.01) and by 4.0 +/- 0.3-fold at a dose of 50 microg/ml (P < 0.001). Rifampin treatment also affected the transcription factors that regulate iNOS mRNA: there was an increased and prolonged degradation of the inhibitory subunit of NF-kappaB, a corresponding increase in the level of cytokine-induced DNA binding of NF-kappaB (2.1 +/- 0.2-fold), and a decrease in the level of expression of peroxisome proliferator-activated receptor gamma (PPARgamma). Specifically, the level of PPARgamma expression dropped by 15% in response to cytokine stimulation and by an additional 40% when rifampin was added (P < 0.001). Rifampin had no effect on the activation of mitogen-activated protein kinases or the signal transducer and transcription activator (STAT-1). In conclusion, rifampin augments NO production by upregulating iNOS mRNA. It also increases the level of NF-kappaB activation and decreases the level of PPARgamma expression. The increases in the levels of NF-kappaB activation and NO production probably contribute to the therapeutic effects of rifampin. However, given the role of NF-kappaB in upregulating many inflammatory genes and the roles of PPARgamma in downregulating inflammatory genes and in lipid and glucose metabolism, these findings have implications for potential adverse effects of rifampin in patients with chronic inflammatory diseases and glucose or lipid disorders.
Collapse
|
39
|
Bachmann M, Paulukat J, Pfeilschifter J, Mühl H. Molecular mechanisms of IL-18BP regulation in DLD-1 cells: pivotal direct action of the STAT1/GAS axis on the promoter level. J Cell Mol Med 2008; 13:1987-1994. [PMID: 19046253 DOI: 10.1111/j.1582-4934.2008.00604.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Interleukin (IL)-18, formerly known as interferon (IFN)-gamma-inducing factor, is a crucial mediator of host defence and inflammation. Control of IL-18 bioactivity by its endogenous antagonist IL-18 binding protein (IL-18BP) is a major objective of immunoregulation. IL-18BP is strongly up-regulated by IFN-gamma, thereby establishing a negative feedback mechanism detectable in cell culture and in vivo. Here we sought to investigate in D.L. Dexter (DLD) colon carcinoma cells molecular mechanisms of IL-18BP induction under the influence of IFN-gamma. Mutational analysis revealed that a proximal gamma-activated sequence (GAS) at the IL-18BP promoter is of pivotal importance for expression by IFN-gamma-activated cells. Use of siRNA underscored the essential role of the signal transducer and activator of transcription (STAT)-1 in this process. Indeed, electrophoretic mobility shift assay and chromatin immunoprecipitation analysis proved STAT1 binding to this particular GAS site. Maximal expression of IL-18BP was dependent on de novo protein synthesis but unaffected by silencing of interferon regulatory factor-1. Altogether, data presented herein indicate that direct action of STAT1 on the IL-18BP promoter at the proximal GAS element is key to IL-18BP expression by IFN-gamma-stimulated DLD-1 colon carcinoma cells.
Collapse
Affiliation(s)
- Malte Bachmann
- Pharmazentrum Frankfurt/ZAFES, University Hospital Goethe-University Frankfurt, Frankfurt am Main, Germany
| | - Jens Paulukat
- Pharmazentrum Frankfurt/ZAFES, University Hospital Goethe-University Frankfurt, Frankfurt am Main, Germany
| | - Josef Pfeilschifter
- Pharmazentrum Frankfurt/ZAFES, University Hospital Goethe-University Frankfurt, Frankfurt am Main, Germany
| | - Heiko Mühl
- Pharmazentrum Frankfurt/ZAFES, University Hospital Goethe-University Frankfurt, Frankfurt am Main, Germany
| |
Collapse
|
40
|
Juan CC, Lien CC, Chang CL, Huang YH, Ho LT. Involvement of iNOS and NO in TNF-alpha-downregulated resistin gene expression in 3T3-L1 adipocytes. Obesity (Silver Spring) 2008; 16:1219-25. [PMID: 18369347 DOI: 10.1038/oby.2008.200] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVE In order to characterize the regulation of resistin gene expression, we explore the effect of tumornecrosis factor-alpha (TNF-alpha) on resistin mRNA expression and its underlying mechanism in 3T3-L1 adipocytes. METHODS AND PROCEDURES Differentiated 3T3-L1 adipocytes were treated for 24 h with 0-10 ng/ml of TNF-alpha or with 2.5 ng/ml of TNF-alpha for 0-24 h, and then resistin mRNA levels were measured by northern blotting. To further explore the involvement of nitric oxide (NO) in TNF-alpha-regulated resistin expression, the effect of the NO donor, sodium nitroprusside (SNP), on resistin mRNA levels in adipocytes and the effect of the nitric oxide synthase (NOS) inhibitors, N(G)-nitro-L-arginine methyl ester (L-NAME), and S,S'-1,3-phenylene-bis(1,2-ethanediyl)-bis-isothiourea.2HBr (PBITU), on the TNF-alpha effect in adipocytes were examined. The effects of TNF-alpha on inducible NOS (iNOS) protein expression in adipocytes were also measured by western blotting. RESULTS Our results showed that TNF-alpha caused a dose-dependent reduction in resistin mRNA levels. This effect seemed to be associated with the TNF-alpha-induced expression of iNOS. The results showed that TNF-alpha induced iNOS expression and release of NO after 24-h treatment of differentiated 3T3-L1 adipocytes. Pretreatment with L-NAME and PBITU significantly reversed the TNF-alpha-induced downregulation of resistin expression, while treatment with SNP mimicked the inhibitory effect of TNF-alpha on resistin expression. In addition, pretreatment with protein tyrosine kinase (PTK) inhibitors, genistein and AG-1288, prevented TNF-alpha-induced iNOS expression and subsequent resistin downregulation. DISCUSSION Our data suggest that TNF-alpha suppresses resistin expression by inducing iNOS expression, thus causing overproduction of NO, which downregulates resistin gene expression.
Collapse
Affiliation(s)
- Chi-Chang Juan
- Department of Physiology, School of Medicine, National Yang-Ming University, Taipei, Taiwan.
| | | | | | | | | |
Collapse
|
41
|
Vareille M, Rannou F, Thélier N, Glasser AL, de Sablet T, Martin C, Gobert AP. Heme Oxygenase-1 Is a Critical Regulator of Nitric Oxide Production in EnterohemorrhagicEscherichia coli-Infected Human Enterocytes. THE JOURNAL OF IMMUNOLOGY 2008; 180:5720-6. [DOI: 10.4049/jimmunol.180.8.5720] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
42
|
Jana M, Jana A, Liu X, Ghosh S, Pahan K. Involvement of phosphatidylinositol 3-kinase-mediated up-regulation of I kappa B alpha in anti-inflammatory effect of gemfibrozil in microglia. THE JOURNAL OF IMMUNOLOGY 2007; 179:4142-52. [PMID: 17785853 PMCID: PMC2604815 DOI: 10.4049/jimmunol.179.6.4142] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The present study underlines the importance of PI3K in mediating the anti-inflammatory effect of gemfibrozil, a prescribed lipid-lowering drug for humans, in mouse microglia. Gemfibrozil inhibited LPS-induced expression of inducible NO synthase (iNOS) and proinflammatory cytokines in mouse BV-2 microglial cells and primary microglia. By overexpressing wild-type and dominant-negative constructs of peroxisome proliferator-activated receptor-alpha (PPAR-alpha) in microglial cells and isolating primary microglia from PPAR-alpha-/- mice, we have demonstrated that gemfibrozil inhibits the activation of microglia independent of PPAR-alpha. Interestingly, gemfibrozil induced the activation of p85alpha-associated PI3K (p110beta but not p110alpha) and inhibition of that PI3K by either chemical inhibitors or dominant-negative mutants abrogated the inhibitory effect of gemfibrozil. Conversely, overexpression of the constitutively active mutant of p110 enhanced the inhibitory effect of gemfibrozil on LPS-induced expression of proinflammatory molecules. Similarly, gemfibrozil also inhibited fibrillar amyloid beta (Abeta)-, prion peptide (PrP)-, dsRNA (poly IC)-, HIV-1 Tat-, and 1-methyl-4-phenylpyridinium (MPP+)-, but not IFN-gamma-, induced microglial expression of iNOS. Inhibition of PI3K also abolished the inhibitory effect of gemfibrozil on Abeta-, PrP-, poly IC-, Tat-, and MPP+-induced microglial expression of iNOS. Involvement of NF-kappaB activation in LPS-, Abeta-, PrP-, poly IC-, Tat-, and MPP+-, but not IFN-gamma-, induced microglial expression of iNOS and stimulation of IkappaBalpha expression and inhibition of NF-kappaB activation by gemfibrozil via the PI3K pathway suggests that gemfibrozil inhibits the activation of NF-kappaB and the expression of proinflammatory molecules in microglia via PI3K-mediated up-regulation of IkappaBalpha.
Collapse
Affiliation(s)
- Malabendu Jana
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL 60612
| | - Arundhati Jana
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL 60612
| | - Xiaojuan Liu
- Section of Neuroscience, Department of Oral Biology, College of Dentistry, University of Nebraska Medical Center, Lincoln, NE 68583
| | - Sankar Ghosh
- Section of Immunobiology and Department of Molecular Biophysics and Biochemistry, School of Medicine, Yale University, New Haven, CT 06536
| | - Kalipada Pahan
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL 60612
- Section of Neuroscience, Department of Oral Biology, College of Dentistry, University of Nebraska Medical Center, Lincoln, NE 68583
- Address correspondence and reprint requests to Dr. Kalipada Pahan, Department of Neurological Sciences, Rush University Medical Center, 1735 West Harrison Street, Suite 320, Chicago, IL 60612. E-mail address:
| |
Collapse
|
43
|
Won JS, Singh AK, Singh I. Lactosylceramide: a lipid second messenger in neuroinflammatory disease. J Neurochem 2007; 103 Suppl 1:180-91. [DOI: 10.1111/j.1471-4159.2007.04822.x] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
44
|
Rubovitch V, Gershnabel S, Kalina M. Lung Epithelial Cells Modulate the Inflammatory Response of Alveolar Macrophages. Inflammation 2007; 30:236-43. [PMID: 17851743 DOI: 10.1007/s10753-007-9042-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2007] [Accepted: 07/17/2007] [Indexed: 11/25/2022]
Abstract
The goal of this study was to examine the effect of alveolar epithelial cells on inflammatory responses in macrophages. Lung epithelial cells (either rat RLE-6TN or human A549 cells) reduced LPS-induced NO production in alveolar macrophages (AM) in a contact-independent mechanism. The inhibitory effect of the epithelial cells was present already at the transcriptional level: LPS-induced inducible NO synthase (iNOS) expression was significantly smaller. Surfactant protein A (SP-A)-induced NO production by alveolar macrophages was also reduced in the presence of A549 cells, though, by a different kinetics. LPS-induced interleukin-6 (IL-6) production (another inflammatory pathway) by alveolar macrophages was also reduced in the presence of RLE-6TN cells. These data suggest a role for lung epithelial cells in the complicated modulation of inflammatory processes, and provide an insight into the mechanism underlying.
Collapse
Affiliation(s)
- Vardit Rubovitch
- Department of Cell and Developmental Biology, Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel.
| | | | | |
Collapse
|
45
|
Guo L, Guo H, Gao C, Mi Z, Russell WB, Kuo PC. Stat1 acetylation inhibits inducible nitric oxide synthase expression in interferon-gamma-treated RAW264.7 murine macrophages. Surgery 2007; 142:156-62. [PMID: 17689680 PMCID: PMC2034510 DOI: 10.1016/j.surg.2007.02.016] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2007] [Revised: 02/22/2007] [Accepted: 02/25/2007] [Indexed: 01/14/2023]
Abstract
BACKGROUND We hypothesized that acetylation of the Stat1 regulates interferon-gamma (IFN-gamma) mediated macrophage expression of inducible nitric oxide synthase (iNOS). METHODS RAW 264.7 iNOS expression was induced with IFN-gamma. Deacetylase inhibitors trichostatin A (TSA) or valproic acid (VPA) were added. Stat1 and iNOS mRNA and protein were measured. Acetylated Stat1 was determined by immunoprecipitation. Chromatin immunoprecipitation assessed in vivo binding of Stat1 to the iNOS promoter. RESULTS IFN-gamma significantly increased nitrite, iNOS protein and iNOS mRNA, and iNOS promoter activation. (P < .01 vs control for nitrite, protein, and mRNA). TSA-mediated acetylation decreased these to levels that were not different from controls. IFN-gamma increased acetylated Stat1 by 5-fold (P < .02 vs control); TSA + IFN-gamma caused an additional 4-fold increase in acetylated Stat1 (P < .05 vs IFN alone). Stat1 binding to the iNOS promoter increased 8-fold with IFN-gamma (P < .01 vs control). In TSA + IFN-gamma, Stat1 binding was not different from controls. Although less potent than TSA, VPA also significantly decreased nitrite, iNOS protein, iNOS mRNA, Stat1 acetylation, and Stat1 binding. CONCLUSIONS Acetylation of Stat1 protein correlates with decreased Stat1 binding to the iNOS promoter with resultant inhibition of IFN-gamma-mediated iNOS expression. Acetylation of the Stat1 protein may downregulate iNOS expression in proinflammatory states.
Collapse
Affiliation(s)
- Lucie Guo
- Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | | | | | | | | | | |
Collapse
|
46
|
Ziesché E, Bachmann M, Kleinert H, Pfeilschifter J, Mühl H. The interleukin-22/STAT3 pathway potentiates expression of inducible nitric-oxide synthase in human colon carcinoma cells. J Biol Chem 2007; 282:16006-15. [PMID: 17438334 DOI: 10.1074/jbc.m611040200] [Citation(s) in RCA: 99] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Inducible nitric-oxide synthase (iNOS) has been identified as a marker and mediator of disease in human colonic inflammation and carcinogenesis. Accordingly, identification of mediators that trigger iNOS in colon carcinoma/epithelial cells is an important topic of current research. Here we demonstrate that interleukin (IL)-22, a newly described member of the IL-10 cytokine family, potently synergizes with interferon (IFN)-gamma for iNOS expression in human DLD-1 colon carcinoma cells. Detection of both IL-22 receptor chains and STAT3 phosphorylation proved robust IL-22 responsiveness of these cells. Short interfering RNA technology identified STAT3 as being crucial for up-regulation of iNOS. Compared with IFNgamma, STAT1 phosphorylation by IL-22 was insufficient. IL-22 did not stabilize IL-1beta/tumor necrosis factor-alpha/IFNgamma-induced iNOS mRNA. IL-22 also failed to amplify expression of the prototypic IFNgamma-inducible parameters IL-18-binding protein and CXCL-10, indicating that IL-22 is not a general amplifier of IFNgamma functions. This assumption is furthermore supported by the observation that IL-22 was unable to enhance cellular activation of the pro-inflammatory transcription factor nuclear factor-kappaB. In contrast, IL-22 increased iNOS promoter activation as detected by using DLD-1 cells stably transfected with a corresponding 16-kb promoter construct (pNOS2(16)-Luc). IL-22 likewise enhanced iNOS in Caco-2 colon carcinoma cells. With IL-22 we introduce a novel potent determinant of iNOS expression in human colon carcinoma/epithelial cells. Considering the eminent functions of STAT3 and iNOS in inflammation and carcinogenesis, IL-22 may represent a novel target for immunotherapeutic intervention.
Collapse
Affiliation(s)
- Elisabeth Ziesché
- Pharmazentrum Frankfurt/ZAFES, Klinikum der Johann Wolfgang Goethe-Universität Frankfurt am Main, 60590 Frankfurt am Main, Germany
| | | | | | | | | |
Collapse
|
47
|
Weinberg JB, Fermor B, Guilak F. Nitric oxide synthase and cyclooxygenase interactions in cartilage and meniscus: relationships to joint physiology, arthritis, and tissue repair. Subcell Biochem 2007; 42:31-62. [PMID: 17612045 DOI: 10.1007/1-4020-5688-5_2] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Rheumatoid arthritis and osteoarthritis are painful and debilitating diseases with complex pathophysiology. There is growing evidence that pro-inflammatory cytokines (e.g., interleukin-1 and tumor necrosis factor alpha) and mediators (e.g., prostaglandins, leukotrienes, and nitric oxide) play critical roles in the development and perpetuation of tissue inflammation and damage in joint tissues such as articular cartilage and meniscus. While earlier studies have generally focused on cells of the synovium (especially macrophages), there is increasing evidence that chondrocytes and meniscal cells actively contribute to inflammatory processes. In particular, it is now apparent that mechanical forces engendered by joint loading are transduced to biological signals at the cellular level and that these signals modulate gene expression and biochemical processes. Here we give an overview of the interplay of cytokines and mechanical stress in the production of cyclooxygenases and prostaglandins; lipoxygenases and leukotrienes; and nitric oxide synthases and nitric oxide in arthritis, with particular focus on the interactions of these pathways in articular cartilage and meniscus.
Collapse
MESH Headings
- Animals
- Arthritis, Rheumatoid/enzymology
- Arthritis, Rheumatoid/physiopathology
- Cartilage, Articular/enzymology
- Cartilage, Articular/pathology
- Cartilage, Articular/physiopathology
- Chondrocytes/enzymology
- Chondrocytes/pathology
- Gene Expression Regulation, Enzymologic
- Humans
- Inflammation/enzymology
- Inflammation/pathology
- Inflammation Mediators/metabolism
- Macrophages/enzymology
- Macrophages/pathology
- Menisci, Tibial/enzymology
- Menisci, Tibial/pathology
- Menisci, Tibial/physiopathology
- Nitric Oxide Synthase/biosynthesis
- Osteoarthritis, Knee/enzymology
- Osteoarthritis, Knee/pathology
- Osteoarthritis, Knee/physiopathology
- Prostaglandin-Endoperoxide Synthases/biosynthesis
- Regeneration
- Signal Transduction
- Stress, Mechanical
- Synovial Membrane/enzymology
- Synovial Membrane/pathology
Collapse
Affiliation(s)
- J Brice Weinberg
- Department of Medicine, Division of Hematology-Oncology, VA and Duke University Medical Centers, Durham, North Carolina 27705, USA.
| | | | | |
Collapse
|
48
|
Eberhardt W, Doller A, Akool ES, Pfeilschifter J. Modulation of mRNA stability as a novel therapeutic approach. Pharmacol Ther 2007; 114:56-73. [PMID: 17320967 DOI: 10.1016/j.pharmthera.2007.01.002] [Citation(s) in RCA: 129] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2007] [Revised: 01/10/2007] [Accepted: 01/10/2007] [Indexed: 12/21/2022]
Abstract
During the last decade evidence has accumulated that modulation of mRNA stability plays a central role in cellular homeostasis, including cell differentiation, proliferation and adaptation to external stimuli. The functional relevance of posttranscriptional gene regulation is highlighted by many pathologies, wherein occurrence tightly correlates with a dysregulation in mRNA stability, including chronic inflammation, cardiovascular diseases and cancer. Most commonly, the cis-regulatory elements of mRNA decay are represented by the adenylate- and uridylate (AU)-rich elements (ARE) which are specifically bound by trans-acting RNA binding proteins, which finally determine whether mRNA decay is delayed or facilitated. Regulation of mRNA decay by RNA stabilizing and RNA destabilizing factors is furthermore controlled by different intrinsic and environmental stimuli. The modulation of mRNA binding proteins, therefore, illuminates a promising approach for the pharmacotherapy of those key pathologies mentioned above and characterized by a posttranscriptional dysregulation. Most promisingly, intracellular trafficking of many of the mRNA stability regulating factors is, in turn, regulated by some major signaling pathways, including the mitogen-activated protein kinase (MAPK) cascade, the AMP-activated kinase (AMPK) and the protein kinase (PK) C (PKC) family. In this review, we present timely examples of genes regulated by mRNA stability with a special focus on signaling pathways involved in the ARE-dependent mRNA decay. A better understanding of these processes may form the basis for the development of novel therapeutics to treat major human diseases.
Collapse
Affiliation(s)
- Wolfgang Eberhardt
- Pharmazentrum frankfurt/ZAFES, Klinikum der Johann Wolfgang Goethe-Universität, Frankfurt am Main, Germany.
| | | | | | | |
Collapse
|
49
|
Kang KW, Wagley Y, Kim HW, Pokharel YR, Chung YY, Chang IY, Kim JJ, Moon JS, Kim YK, Nah SY, Kang HS, Oh JW. Novel role of IL-6/SIL-6R signaling in the expression of inducible nitric oxide synthase (iNOS) in murine B16, metastatic melanoma clone F10.9, cells. Free Radic Biol Med 2007; 42:215-27. [PMID: 17189827 DOI: 10.1016/j.freeradbiomed.2006.10.034] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2006] [Revised: 09/15/2006] [Accepted: 10/08/2006] [Indexed: 12/27/2022]
Abstract
Inducible nitric oxide synthase (iNOS) has been shown to be frequently expressed in melanomas; up-regulation of this enzyme is though to be associated with tumor progression. In this study, we investigated whether diverse cytokines such as: IL-6, TNF-alpha, IL-1beta, IFN-gamma and IL6RIL6 (a highly active fusion protein of the soluble form of the IL-6R (sIL-6R) and IL-6) enhance the iNOS gene expression in B16/F10.9 murine metastatic melanoma cells. An increase at iNOS expression and NO production was observed with the co-treatment of IL6RIL6 plus TNF-alpha. Gel shift and reporter gene analyses revealed that IL6RIL6 selectively activated AP-1; while TNF-alpha increased the activities of both NF-kappaB and AP-1. Persistent activation of AP-1 was also seen in cells treated with IL6RIL6 plus TNF-alpha. Stimulation of cells with IL6RIL6/TNF-alpha resulted in the activation of mitogen-activated protein kinases (MAPK) such as c-Jun N-terminal kinase (JNK) and p38, and the abrogation by pretreatment with JNK or p38 MAPK inhibitor. IL6RIL6 or IL6RIL6/TNFalpha-inducible AP-1 binding increase was supershifted by anti-c-Jun or c-Fos antibodies, and the activation of c-Jun and c-Fos was dependent on JNK and p38, respectively. These results suggest that IL-6/sIL-6R/gp130 complex signaling has an unexpected positive effect on iNOS gene expression through JNK/p38 MAPK mediated-AP-1 activation in melanoma cells.
Collapse
Affiliation(s)
- Keon Wook Kang
- College of Pharmacy, Chosun University, Gwangju 501-759, Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Cronauer MV, Ince Y, Engers R, Rinnab L, Weidemann W, Suschek CV, Burchardt M, Kleinert H, Wiedenmann J, Sies H, Ackermann R, Kröncke KD. Nitric oxide-mediated inhibition of androgen receptor activity: possible implications for prostate cancer progression. Oncogene 2006; 26:1875-84. [PMID: 16983333 DOI: 10.1038/sj.onc.1209984] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Chronic inflammation increases the risk of cancer and many cancers, including prostate cancer, arise at sites of chronic inflammation. Inducible nitric oxide synthase (iNOS) is an enzyme dominantly expressed during inflammatory reactions. Although synthesis of high amounts of nitric oxide (NO) by iNOS has been demonstrated in pathophysiological processes, such as acute or chronic inflammation, autoimmune diseases or tumorigenesis, the role of iNOS activity in most of these diseases is poorly understood. Analysing prostate cancer biopsies by immunohistochemistry we found iNOS protein expression in tumor cells strongly paralleled by nitrotyrosine suggesting that iNOS is fully active. In vitro, NO inhibits androgen receptor-dependent promoter activity and prostate specific antigen production as well as DNA-binding activity of the androgen receptor (AR) in a concentration-dependent manner. Inhibition of the activity of androgen receptor-dependent reporter constructs is neither owing to diminished AR protein levels nor owing to an inhibition of its nuclear import. In addition, NO inhibits the proliferation of androgen receptor-positive prostate cancer cells significantly more efficiently than proliferation of androgen receptor-negative prostate cancer cells. In summary, our findings suggest that intratumoral iNOS activity favors development of prostate cancer cells that are able to proliferate androgen receptor-independently, thereby promoting prostate tumor progression.
Collapse
Affiliation(s)
- M V Cronauer
- Department of Urology, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|