1
|
Sánchez‐García S, Povo‐Retana A, Marin S, Madurga S, Fariñas M, Aleixandre N, Castrillo A, de la Rosa JV, Alvarez‐Lucena C, Landauro‐Vera R, Prieto P, Cascante M, Boscá L. Immunometabolic Effect of Nitric Oxide on Human Macrophages Challenged With the SARS-CoV2-Induced Cytokine Storm. A Fluxomic Approach. Adv Healthc Mater 2025; 14:e2401688. [PMID: 39502019 PMCID: PMC11694080 DOI: 10.1002/adhm.202401688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 10/04/2024] [Indexed: 01/03/2025]
Abstract
The cytokine storm associated with SARS-CoV-2 infection is one of the most distinctive pathological signatures in COVID-19 patients. Macrophages respond to this pro-inflammatory challenge by reprogramming their functional and metabolic phenotypes. Interestingly, human macrophages fail to express the inducible form of the NO synthase (NOS2) in response to pro-inflammatory activation and, therefore, NO is not synthesized by these cells. The contribution of exogenously added NO, via a chemical NO-donor, on the immunometabolic changes associated with the cytokine storm is investigated. By using metabolic, transcriptomic, and functional assays the effect of NO in human macrophages is evaluated and found specific responses. Moreover, through integrative fluxomic analysis, pathways modified by NO that contribute to the expression of a particular phenotype in human macrophages are identified, which includes a decrease in mitochondrial respiration and TCA with a slight increase in the glycolytic flux. A significant ROS increase and preserved cell viability are observed in the presence of NO, which may ease the inflammatory response and host defense. Also, NO reverses the cytokine storm-induced itaconate accumulation. These changes offer additional clues to understanding the potential crosstalk between NO and the COVID-19 cytokine storm-dependent signaling pathways.
Collapse
Affiliation(s)
- Sergio Sánchez‐García
- Instituto de Investigaciones Biomédicas Sols‐Morreale, Consejo Superior de Investigaciones Científicas‐Universidad Autónoma de MadridArturo Duperier 4Madrid28029Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV)Av. Monforte de Lemos 3–5, P‐11Madrid28029Spain
| | - Adrián Povo‐Retana
- Instituto de Investigaciones Biomédicas Sols‐Morreale, Consejo Superior de Investigaciones Científicas‐Universidad Autónoma de MadridArturo Duperier 4Madrid28029Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV)Av. Monforte de Lemos 3–5, P‐11Madrid28029Spain
| | - Silvia Marin
- Department of Biochemistry and Molecular Biomedicine‐Institute of Biomedicine (IBUB), Faculty of BiologyUniversitat de BarcelonaBarcelona08028Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD)Av. Monforte de Lemos 3–5, P‐11Madrid28029Spain
| | - Sergio Madurga
- Department of Material Science and Physical Chemistry & Research Institute of Theoretical and Computational Chemistry (IQTCUB)University of BarcelonaBarcelona08028Spain
| | - Marco Fariñas
- Department of Biochemistry and Molecular Biomedicine‐Institute of Biomedicine (IBUB), Faculty of BiologyUniversitat de BarcelonaBarcelona08028Spain
| | - Nuria Aleixandre
- Department of Biochemistry and Molecular Biomedicine‐Institute of Biomedicine (IBUB), Faculty of BiologyUniversitat de BarcelonaBarcelona08028Spain
- Department of Material Science and Physical Chemistry & Research Institute of Theoretical and Computational Chemistry (IQTCUB)University of BarcelonaBarcelona08028Spain
| | - Antonio Castrillo
- Instituto de Investigaciones Biomédicas Sols‐Morreale, Consejo Superior de Investigaciones Científicas‐Universidad Autónoma de MadridArturo Duperier 4Madrid28029Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV)Av. Monforte de Lemos 3–5, P‐11Madrid28029Spain
- Unidad de Biomedicina (Unidad Asociada al CSIC) de la Universidad de Las Palmas de Gran CanariaLas Palmas35016Spain
- Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS) de la Universidad de Las Palmas de Gran CanariaLas Palmas35016Spain
| | - Juan V. de la Rosa
- Unidad de Biomedicina (Unidad Asociada al CSIC) de la Universidad de Las Palmas de Gran CanariaLas Palmas35016Spain
- Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS) de la Universidad de Las Palmas de Gran CanariaLas Palmas35016Spain
| | - Carlota Alvarez‐Lucena
- Instituto de Investigaciones Biomédicas Sols‐Morreale, Consejo Superior de Investigaciones Científicas‐Universidad Autónoma de MadridArturo Duperier 4Madrid28029Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV)Av. Monforte de Lemos 3–5, P‐11Madrid28029Spain
| | - Rodrigo Landauro‐Vera
- Instituto de Investigaciones Biomédicas Sols‐Morreale, Consejo Superior de Investigaciones Científicas‐Universidad Autónoma de MadridArturo Duperier 4Madrid28029Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV)Av. Monforte de Lemos 3–5, P‐11Madrid28029Spain
| | - Patricia Prieto
- Instituto de Investigaciones Biomédicas Sols‐Morreale, Consejo Superior de Investigaciones Científicas‐Universidad Autónoma de MadridArturo Duperier 4Madrid28029Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV)Av. Monforte de Lemos 3–5, P‐11Madrid28029Spain
- Departamento de Farmacología, Farmacognosia y BotánicaFacultad de Farmacia, Universidad Complutense de MadridMadrid28040Spain
| | - Marta Cascante
- Department of Biochemistry and Molecular Biomedicine‐Institute of Biomedicine (IBUB), Faculty of BiologyUniversitat de BarcelonaBarcelona08028Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD)Av. Monforte de Lemos 3–5, P‐11Madrid28029Spain
| | - Lisardo Boscá
- Instituto de Investigaciones Biomédicas Sols‐Morreale, Consejo Superior de Investigaciones Científicas‐Universidad Autónoma de MadridArturo Duperier 4Madrid28029Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV)Av. Monforte de Lemos 3–5, P‐11Madrid28029Spain
- Unidad de Biomedicina (Unidad Asociada al CSIC) de la Universidad de Las Palmas de Gran CanariaLas Palmas35016Spain
| |
Collapse
|
2
|
Ju S, Singh MK, Han S, Ranbhise J, Ha J, Choe W, Yoon KS, Yeo SG, Kim SS, Kang I. Oxidative Stress and Cancer Therapy: Controlling Cancer Cells Using Reactive Oxygen Species. Int J Mol Sci 2024; 25:12387. [PMID: 39596452 PMCID: PMC11595237 DOI: 10.3390/ijms252212387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 10/31/2024] [Accepted: 11/13/2024] [Indexed: 11/28/2024] Open
Abstract
Cancer is a multifaceted disease influenced by various mechanisms, including the generation of reactive oxygen species (ROS), which have a paradoxical role in both promoting cancer progression and serving as targets for therapeutic interventions. At low concentrations, ROS serve as signaling agents that enhance cancer cell proliferation, migration, and resistance to drugs. However, at elevated levels, ROS induce oxidative stress, causing damage to biomolecules and leading to cell death. Cancer cells have developed mechanisms to manage ROS levels, including activating pathways such as NRF2, NF-κB, and PI3K/Akt. This review explores the relationship between ROS and cancer, focusing on cell death mechanisms like apoptosis, ferroptosis, and autophagy, highlighting the potential therapeutic strategies that exploit ROS to target cancer cells.
Collapse
Affiliation(s)
- Songhyun Ju
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.); (M.K.S.); (S.H.); (J.R.); (J.H.); (W.C.); (K.-S.Y.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Manish Kumar Singh
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.); (M.K.S.); (S.H.); (J.R.); (J.H.); (W.C.); (K.-S.Y.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Sunhee Han
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.); (M.K.S.); (S.H.); (J.R.); (J.H.); (W.C.); (K.-S.Y.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Jyotsna Ranbhise
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.); (M.K.S.); (S.H.); (J.R.); (J.H.); (W.C.); (K.-S.Y.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Joohun Ha
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.); (M.K.S.); (S.H.); (J.R.); (J.H.); (W.C.); (K.-S.Y.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Wonchae Choe
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.); (M.K.S.); (S.H.); (J.R.); (J.H.); (W.C.); (K.-S.Y.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Kyung-Sik Yoon
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.); (M.K.S.); (S.H.); (J.R.); (J.H.); (W.C.); (K.-S.Y.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Seung Geun Yeo
- Department of Otorhinolaryngology—Head and Neck Surgery, College of Medicine, Kyung Hee University Medical Center, Kyung Hee University, Seoul 02453, Republic of Korea;
| | - Sung Soo Kim
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.); (M.K.S.); (S.H.); (J.R.); (J.H.); (W.C.); (K.-S.Y.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Insug Kang
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.); (M.K.S.); (S.H.); (J.R.); (J.H.); (W.C.); (K.-S.Y.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
3
|
Baicalin attenuates lipopolysaccharide induced inflammation and apoptosis of cow mammary epithelial cells by regulating NF-κB and HSP72. Int Immunopharmacol 2016; 40:139-145. [PMID: 27588914 DOI: 10.1016/j.intimp.2016.08.032] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Revised: 08/25/2016] [Accepted: 08/25/2016] [Indexed: 01/06/2023]
Abstract
Baicalin is the main ingredient of traditional Chinese herbal medicine, Scutellaria baicalensis, which has been widely used clinically as an anti-inflammatory agent. However, molecular mechanism of action of this drug is not yet clear. In the present study, the protective mechanism of baicalin against lipopolysaccharide (LPS) induced inflammatory injury in cow mammary epithelial cells (CMECs) was explored. For this purpose, in vitro cultured CMECs were treated with baicalin (10μg/mL) and LPS (10μg/mL) for 24 and 12h, respectively, and the cell viability was measured by using cell counting kit-8 (CCK-8). The results revealed that LPS induced inflammatory responses, as p-p65/p65 and p-IκBα/IκBα ratios and TNF-α and IL-1β production was increased in the CMECs. Both Bcl-2/Bax ratio and cell viability were decreased and caspase-3 cleaved following LPS treatment, indicating apoptosis of CMECs. Moreover, both LPS and baicalin increased HSP72 expression of the CMECs. However, cellular inflammatory responses and apoptosis were significantly reduced in baicalin treated CMECs. In conclusion, baicalin ameliorated inflammation and apoptosis of the CMECs induced by LPS via inhibiting NF-κB activation and up regulation of HSP72.
Collapse
|
4
|
Kim SJ, Lee JH, Kim BS, So HS, Park R, Myung NY, Um JY, Hong SH. (-)-Epigallocatechin-3-gallate protects against NO-induced ototoxicity through the regulation of caspase- 1, caspase-3, and NF-κB activation. PLoS One 2012; 7:e43967. [PMID: 23028481 PMCID: PMC3461011 DOI: 10.1371/journal.pone.0043967] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2011] [Accepted: 07/30/2012] [Indexed: 12/16/2022] Open
Abstract
Excessive nitric oxide (NO) production is toxic to the cochlea and induces hearing loss. However, the mechanism through which NO induces ototoxicity has not been completely understood. The aim of this study was to gain further insight into the mechanism mediating NO-induced toxicity in auditory HEI-OC1 cells and in ex vivo analysis. We also elucidated whether and how epigallocatechin-3-gallate (EGCG), the main component of green tea polyphenols, regulates NO-induced auditory cell damage. To investigate NO-mediated ototoxicity, S-nitroso-N-acetylpenicillamine (SNAP) was used as an NO donor. SNAP was cytotoxic, generating reactive oxygen species, releasing cytochrome c, and activating caspase-3 in auditory cells. NO-induced ototoxicity also mediated the nuclear factor (NF)-κB/caspase-1 pathway. Furthermore, SNAP destroyed the orderly arrangement of the 3 outer rows of hair cells in the basal, middle, and apical turns of the organ of Corti from the cochlea of Sprague-Dawley rats at postnatal day 2. However, EGCG counteracted this ototoxicity by suppressing the activation of caspase-3/NF-κB and preventing the destruction of hair cell arrays in the organ of Corti. These findings may lead to the development of a model for pharmacological mechanism of EGCG and potential therapies against ototoxicity.
Collapse
Affiliation(s)
- Su-Jin Kim
- Department of Cosmeceutical Science, Daegu Hanny University, Kyungsan, Gyeoungbuk, Republic of Korea
| | - Jeong-Han Lee
- Center for Metabolic Function Regulation, Wonkwang University, Iksan, Republic of Korea
| | - Beom-Su Kim
- Wonkwang Bone Regeneration Research Institute, Wonkwang University, Iksan, Jeonbuk, Republic of Korea
| | - Hong-Seob So
- Center for Metabolic Function Regulation, Wonkwang University, Iksan, Republic of Korea
| | - Raekil Park
- Center for Metabolic Function Regulation, Wonkwang University, Iksan, Republic of Korea
| | - Noh-Yil Myung
- Center for Metabolic Function Regulation, Wonkwang University, Iksan, Republic of Korea
| | - Jae-Young Um
- College of Oriental Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Seung-Heon Hong
- Center for Metabolic Function Regulation, Wonkwang University, Iksan, Republic of Korea
| |
Collapse
|
5
|
Affiliation(s)
- Cristina E Carnovale
- Instituto de Fisiología Experimental-CONICET, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Suipacha 570-2000 Rosario, Argentina.
| | | |
Collapse
|
6
|
Nitrosative stress induces osteoblast apoptosis through downregulating MAPK-mediated NFκB/AP-1 activation and subsequent Bcl-XL expression. Chem Biol Interact 2010; 184:359-65. [DOI: 10.1016/j.cbi.2010.01.040] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2009] [Revised: 01/18/2010] [Accepted: 01/25/2010] [Indexed: 01/14/2023]
|
7
|
NF-kappaB blockade upregulates Bax, TSP-1, and TSP-2 expression in rat granulation tissue. J Mol Med (Berl) 2009; 87:481-92. [PMID: 19189070 DOI: 10.1007/s00109-009-0443-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2008] [Revised: 12/22/2008] [Accepted: 01/07/2009] [Indexed: 12/12/2022]
Abstract
Several diseases are characterized by chronic inflammation, a condition frequently associated with angiogenesis and fibrogenesis that account for the development of granulation tissue. Nuclear Factor kappa-light-chain-enhancer of activated B cells (NF-kappaB) is a crucial modulator of intracellular prosurvival signaling pathways and is implicated in the pathogenesis of inflammatory process. In this study, we have investigated the role of NF-kappaB in the angiogenic and fibrogenic response induced by lambda-carrageenin in a rat model of chronic inflammation at 1, 3, and 5 days. The subcutaneous implant of lambda-carrageenin-soaked sponges in rat induced a time-related increase of granulation tissue formation accompanied by intense neovascularization. These lambda-carrageenin-induced changes were significantly reduced by coinjection of wild-type oligodeoxynucleotide (WT ODN) decoy to NF-kappaB. Molecular, morphological, and ultrastructural analysis performed on whole granulation tissue demonstrated: (1) inhibition of NF-kappaB/DNA binding activity; (2) downregulation of cyclooxygenase-2, matrix metalloproteinase-9, tumor necrosis factor-alpha, and vascular endothelial growth factor; (3) upregulation of thrombospondin (TSP)-1 at 1 day and TSP-2 at 5 days; and (4) increase in Bax to Bcl-2 ratio. Our findings show that the blockade of NF-kappaB activation by WT ODN decoy prevents the development of granulation tissue induced by lambda-carrageenin-soaked sponge implant upregulating Bax as well as TSP-1 and TSP-2 expression.
Collapse
|
8
|
Lovastatin induces apoptosis of k-ras-transformed thyroid cells via inhibition of ras farnesylation and by modulating redox state. J Mol Med (Berl) 2008; 86:1341-51. [DOI: 10.1007/s00109-008-0396-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2008] [Revised: 08/01/2008] [Accepted: 08/04/2008] [Indexed: 01/29/2023]
|
9
|
Radke JR, Siddiqui ZK, Miura TA, Routes JM, Cook JL. E1A oncogene enhancement of caspase-2-mediated mitochondrial injury sensitizes cells to macrophage nitric oxide-induced apoptosis. THE JOURNAL OF IMMUNOLOGY 2008; 180:8272-9. [PMID: 18523293 DOI: 10.4049/jimmunol.180.12.8272] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The adenovirus E1A oncogene induces innate immune rejection of tumors by sensitizing tumor cells to apoptosis in response to injuries, such as those inflicted by macrophage-produced TNF alpha and NO. E1A sensitizes cells to TNF by repressing its activation of NF-kappaB-dependent, antiapoptotic defenses. This suggested the hypothesis that E1A blockade of the NF-kappaB activation response might be the central mechanism of E1A induced cellular sensitivity to other proapoptotic injuries, such as macrophage-produced NO. However, creation of E1A-positive NIH-3T3 mouse cell variants with high-level, NF-kappaB-dependent resistance to TNF did not coselect for resistance to apoptosis induced by either macrophage-NO or chemical-NO, as the hypothesis would predict. E1A expression did block cellular recovery from NO-induced mitochondrial injury and converted the reversible, NO-induced cytostasis response of cells to an apoptotic response. This viral oncogene-induced phenotypic conversion of the cellular injury response of mouse and human cells was mediated by an E1A-related increase in NO-induced activation of caspase-2, an apical initiator of intrinsic apoptosis. Blocking caspase-2 activation or expression eliminated the NO-induced apoptotic response of E1A-positive cells. These results define an NF-kappaB-independent pathway through which the E1A gene of human adenovirus sensitizes mouse and human cells to apoptosis by enhancement of caspase-2-mediated mitochondrial injury.
Collapse
Affiliation(s)
- Jay R Radke
- Section of Infectious Diseases, Immunology, and International Medicine, Department of Medicine and Microbiology-Immunology and the Cancer Center, University of Illinois, Chicago, IL 60612, USA
| | | | | | | | | |
Collapse
|
10
|
Rishi L, Dhiman R, Raje M, Majumdar S. Nitric oxide induces apoptosis in cutaneous T cell lymphoma (HuT-78) by downregulating constitutive NF-κB. Biochim Biophys Acta Gen Subj 2007; 1770:1230-9. [PMID: 17555878 DOI: 10.1016/j.bbagen.2007.04.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2007] [Revised: 04/18/2007] [Accepted: 04/25/2007] [Indexed: 01/27/2023]
Abstract
Constitutive active NF-kappaB have been shown to protect cutaneous T cell lymphoma (CTCL) cells from apoptosis. In the present study, we have studied the cytotoxic potential of nitric oxide generating compound, sodium nitroprusside (SNP) on CTCL cell line, HuT-78. Treatment of cells with SNP resulted in decrease in mitochondrial membrane potential, cytochrome c release, activation of caspase-3 and poly (ADP ribose) polymerase cleavage. SNP treatment inhibited activation of NF-kappaB in a concentration dependent manner. SNP increased the expression of IkappaBalpha without affecting the phosphorylation of IkappaBalpha. Downregulation of NF-kappaB by SNP decreased p65 nuclear translocation as evident by confocal fluorescence microscopy. Further it was found that SNP treatment caused downregulation of Bcl-2 family member (Bcl-xl) in HuT-78 cells. Thus, we have provided evidence that SNP induces apoptosis in CTCL cell line, HuT-78 by downregulating constitutive NF-kappaB and thereby Bcl-xl expression.
Collapse
Affiliation(s)
- Loveena Rishi
- Division of Cell Biology and Immunology, Institute of Microbial Technology, Chandigarh-160 036, India
| | | | | | | |
Collapse
|
11
|
De Stefano D, Maiuri MC, Simeon V, Grassia G, Soscia A, Cinelli MP, Carnuccio R. Lycopene, quercetin and tyrosol prevent macrophage activation induced by gliadin and IFN-gamma. Eur J Pharmacol 2007; 566:192-9. [PMID: 17477920 DOI: 10.1016/j.ejphar.2007.03.051] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2007] [Revised: 03/23/2007] [Accepted: 03/26/2007] [Indexed: 01/13/2023]
Abstract
Oxidative stress plays an important role in inflammatory process of celiac disease. We have studied the effect of the lycopene, quercetin and tyrosol natural antioxidants on the inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2) gene expression in RAW 264.7 macrophages stimulated by gliadin in association with IFN-gamma. The IFN-gamma plus gliadin combination treatment was capable of enhancing iNOS and COX-2 gene expression and nuclear factor-kappaB (NF-kappaB), interferon regulatory factor-1 (IRF-1) and signal transducer and activator of transcription-1alpha (STAT-1alpha) activation induced by reactive oxygen species generation at 24 h. Lycopene, quercetin and tyrosol inhibited all these effects. The results here reported suggest that these compounds may represent non toxic agents for the control of pro-inflammatory genes involved in celiac disease.
Collapse
Affiliation(s)
- Daniela De Stefano
- Dipartimento di Farmacologia Sperimentale, Via D. Montesano, 49, Università degli Studi di Napoli Federico II, 80131 Naples, Italy.
| | | | | | | | | | | | | |
Collapse
|
12
|
Lin SK, Kok SH, Lin LD, Wang CC, Kuo MYP, Lin CT, Hsiao M, Hong CY. Nitric oxide promotes the progression of periapical lesion via inducing macrophage and osteoblast apoptosis. ACTA ACUST UNITED AC 2007; 22:24-9. [PMID: 17241167 DOI: 10.1111/j.1399-302x.2007.00316.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
This study aimed to elucidate the modulation by nitric oxide (NO) of the apoptosis of macrophages and osteoblasts, the essential cellular components in the development of periapical lesions. Lipopolysaccharide (LPS) induced prominent nitrite synthesis in J774 mouse macrophage cell lines. Exposure to LPS induced obvious apoptosis in J774 cells, whereas transient transfection with murine inducible nitric oxide synthase (iNOS), small interfering RNA (siRNA) diminished this effect. Tumor necrosis factor-alpha (TNF-alpha) and S-nitroso-N-acetyl-DL-penicillamine (SNAP) (a NO donor) triggered apoptosis in UMR-106 rat osteoblastic cell lines and a synergistic effect was noted when TNF-alpha and SNAP were added to the medium together. Administration of siRNAs for c-Fos and c-Jun: components of activator protein-1 (AP-1) and transforming growth factor-beta1 attenuated the combined effect markedly. Terminal deoxynucleotidyl transferase-mediated nick end-labeling (TUNEL) stain in a rat model of induced periapical lesion showed positive apoptotic signals in macrophages and osteoblasts. Administration of N(G)-monomethyl-l-arginine markedly diminished the extent of bone loss and the amounts of apoptotic macrophages and osteoblasts. In conclusion, NO mediates LPS-stimulated apoptosis of macrophages. It also induces osteoblast apoptosis and augments the pro-apoptotic effect of cytokines. Inhibition of NO synthesis in vivo attenuates apoptosis and the size of periapical lesions. Taken together, these results suggest that NO may promote the progression of periapical lesion by inducing the apoptosis of macrophages and osteoblasts.
Collapse
Affiliation(s)
- S-K Lin
- Department of Dentistry, National Taiwan University Hospital, Taipei, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
13
|
Kafoury RM, Madden MC. Diesel exhaust particles induce the over expression of tumor necrosis factor-alpha (TNF-alpha) gene in alveolar macrophages and failed to induce apoptosis through activation of nuclear factor-kappaB (NF-kappaB). INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2006; 2:107-13. [PMID: 16705808 PMCID: PMC3814704 DOI: 10.3390/ijerph2005010107] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Exposure to particulate matter (PM2.5-10), including diesel exhaust particles (DEP) has been reported to induce lung injury and exacerbation of asthma and chronic obstructive pulmonary disease. Alveolar macrophages play a major role in the lung's response to inhaled particles and therefore, are a primary target for PM2.5-10 effect. The molecular and cellular events underlying DEP-induced toxicity in the lung, however, remain unclear. To determine the effect of DEP on alveolar macrophages, RAW 264.7 cells were grown in RPMI 1640 with supplements until confluency. RAW 264.7 cultures were exposed to Hank's buffered saline solution (vehicle), vehicle containing an NF-kappaB inhibitor, BAY11-7082 (25 microM with 11/2 hr pre-incubation), or vehicle containing DEP (250 microg/ml) in the presence or absence of BAY11-7082 (25 microM with 11/2 hr pre-incubation) for 4 hr and TNF-alpha release was determined by enzyme-linked immunosorbent assay and confirmed by western blots. RAW 264.7 apoptotic response was determined by DNA fragmentation assays. U937 cells treated with campothecin (4 microg/ml x 3 hr), an apoptosis-inducing agent, were used as positive control. We report that exposure to the carbonaceous core of DEP induces significant release of TNF-alpha in a concentration-dependent fashion (31 +/- 4 pg/ml, n = 4, p = 0.08; 162 +/- 23 pg/ml, n = 4, p < 0.05; 313 +/- 31 pg/ml, n = 4, p < 0.05 at 25, 100, and 250 microg/ml, respectively). DEP exposure, however, failed to induce any apoptotic response in RAW 264.7 cells. Moreover, inhibition of NF-kappaB binding activity has resulted in DEP-induced apoptotic response in alveolar macrophages, as demonstrated by the NF-kappaB inhibitor, BAY11-7082 studies. The results of the present study indicate that DEP induce the release of TNF-alpha in alveolar macrophages, a primary target for inhaled particles effect. DEP-induced TNF-alpha gene expression is regulated at the transcriptional level by NF-kappaB. Furthermore, DEP-induced increase in NF-kappaB-DNA binding activity appears to protect against apoptosis.
Collapse
Affiliation(s)
- Ramzi M Kafoury
- Center of Environmental Health and Molecular Toxicology Laboratory, Jackson State University, Jackson, Mississippi, USA.
| | | |
Collapse
|
14
|
De Stefano D, Maiuri MC, Iovine B, Ialenti A, Bevilacqua MA, Carnuccio R. The role of NF-κB, IRF-1, and STAT-1α transcription factors in the iNOS gene induction by gliadin and IFN-γ in RAW 264.7 macrophages. J Mol Med (Berl) 2005; 84:65-74. [PMID: 16284791 DOI: 10.1007/s00109-005-0713-x] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2005] [Accepted: 08/05/2005] [Indexed: 01/01/2023]
Abstract
Nitric oxide (NO) plays an important role in the pathogenesis of celiac disease. We have examined the involvement of nuclear factor-kappaB (NF-kappaB), interferon regulatory factor-1 (IRF-1), and signal transducer and activator of transcription-1alpha (STAT-1alpha) on the synergistic induction of inducible nitric oxide synthase (iNOS) gene expression by gliadin (G) in association with interferon-gamma (IFN-gamma) in RAW 264.7 macrophages. We found that IFN-gamma was efficient in enhancing the basal transcription of the iNOS promoter at 1, 6, and 24 h, whereas G had no effect. The G plus IFN-gamma association caused an increase in iNOS promoter activity which was inhibited by pyrrolidine dithiocarbammate (PDTC) at 6 and 24 h as well as by genistein (Gen) and tyrphostine B42 (TB42) at 1 h, inhibitors of NF-kappaB, IRF-1, and STAT-1alpha activation, respectively. Similarly, the IFN-gamma and G combination treatment led to a higher increase in iNOS mRNA levels at 1, 6, and 24 h compared with IFN-gamma alone. Gen and TB42 inhibited iNOS mRNA levels at 1 h, whereas PDTC inhibited iNOS mRNA levels at 6 and 24 h. In addition, the synergistic induction of iNOS gene expression by G plus IFN-gamma correlated with the induction of NF-kappaB, IRF-1, and STAT-1alpha/DNA binding activity and mRNA expression. In conclusion, our study, which provides evidence that the effect of G on iNOS gene transcription in IFN-gamma-stimulated RAW 264.7 cells can be ascribed to all three transcription factors, may contribute to lead to new insights into the molecular mechanisms governing the inflammatory process in celiac disease.
Collapse
Affiliation(s)
- Daniela De Stefano
- Dipartimento di Farmacologia Sperimentale, Via D. Montesano 49, University of Naples Federico II, 80131 Naples, Italy
| | | | | | | | | | | |
Collapse
|
15
|
De Rosa G, Maiuri MC, Ungaro F, De Stefano D, Quaglia F, La Rotonda MI, Carnuccio R. Enhanced intracellular uptake and inhibition of NF-kappaB activation by decoy oligonucleotide released from PLGA microspheres. J Gene Med 2005; 7:771-81. [PMID: 15702489 DOI: 10.1002/jgm.724] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Nuclear factor-kappaB (NF-kappaB) transcription factor regulates the expression of genes involved in immune response and inflammation. NF-kappaB activity can be efficiently inhibited by double-stranded oligodeoxynucleotides (ODNs). In the present study, we investigated the potential of poly(DL-lactic-co-glycolic acid) (PLGA) microspheres as delivery system for an ODN against NF-kappaB in RAW 264.7 macrophages stimulated with lipopolysaccharide (LPS). METHODS Microspheres encapsulating ODN were prepared by the multiple emulsion/solvent evaporation technique and characterised in terms of size, morphology, encapsulation efficiency and in vitro release profile. In vitro uptake after 4 h and activity of ODN released from microspheres were evaluated in RAW 264.7 macrophages stimulated with LPS for 24, 48 and 72 h. RESULTS We prepared microspheres with a high encapsulation efficiency showing a very slow and almost constant in vitro release of ODN for up to 1 month. ODN slowly released from microspheres translocated better into LPS-stimulated cells as compared with naked ODN. Incubation of cells with ODN-encapsulating microspheres resulted in a decrease of tumor necrosis factor-alpha (TNF-alpha) and nitrite production, inducible nitric oxide synthase (iNOS) protein expression, as well as NF-kappaB/DNA-binding activity. Similar results were obtained with naked ODN only at about 80 times higher concentrations. CONCLUSIONS Our results suggest that PLGA microspheres could be a useful tool to improve pharmacokinetics of a ODN decoy to NF-kappaB and may represent a promising strategy to effectively inhibit the transcriptional activity of NF-kappaB in inflammatory process.
Collapse
Affiliation(s)
- Giuseppe De Rosa
- Dipartimento di Chimica Farmaceutica e Tossicologica, Università degli Studi di Napoli Federico II, Via Montesano 49, 80131 Naples, Italy
| | | | | | | | | | | | | |
Collapse
|
16
|
Maiuri MC, De Stefano D, Di Meglio P, Irace C, Savarese M, Sacchi R, Cinelli MP, Carnuccio R. Hydroxytyrosol, a phenolic compound from virgin olive oil, prevents macrophage activation. Naunyn Schmiedebergs Arch Pharmacol 2005; 371:457-65. [PMID: 16025269 DOI: 10.1007/s00210-005-1078-y] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2005] [Accepted: 06/09/2005] [Indexed: 01/04/2023]
Abstract
We investigated the effect of hydroxytyrosol (HT), a phenolic compound from virgin olive oil, on inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2) expression in J774 murine macrophages stimulated with lipopolysaccharide (LPS). Incubation of cells with LPS caused an increase in iNOS and COX-2 mRNA and protein level as well as ROS generation, which was prevented by HT. In addition, HT blocked the activation of nuclear factor-kappaB (NF-kappaB), signal transducer and activator of transcription-1alpha (STAT-1alpha) and interferon regulatory factor-1 (IRF-1). These results, showing that HT down-regulates iNOS and COX-2 gene expression by preventing NF-kappaB, STAT-1alpha and IRF-1 activation mediated through LPS-induced ROS generation, suggest that it may represent a non-toxic agent for the control of pro-inflammatory genes.
Collapse
Affiliation(s)
- Maria Chiara Maiuri
- Dipartimento di Farmacologia Sperimentale, Università degli Studi di Napoli Federico II, Via D. Montesano n. 49, 80131 Naples, Italy
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Abstract
Low NO concentrations synthesized by constitutively expressed NO synthases act on several signaling pathways activating transcription factors (TF), such as NF-kappaB or AP-1, and thereby influence gene expression. In contrast, during inflammatory reactions the inducible NO synthase produces NO for prolonged periods of time. The resulting nitrosative stress directly affects redox-sensitive TF like NF-kappaB, AP-1, Oct-1, c-Myb, or zinc finger-containing TF, but also additional mechanisms have been identified. Nitrosative stress in some cases induces expression of TF (AP-1, p53), indirectly modulates activity or stability of TF (HIF-1, p53) or their inhibitors (NF-kappaB), or modulates accessibility of promoters via increased DNA methylation or histone deacetylation. Depending on the promoter the result is induced, increased, decreased or even totally inhibited expression of various target genes. In unstimulated cells nitrosative stress increases NF-kappaB- or AP-1-dependent transcription, while in activated cells nitrosative stress rather abolishes NF-kappaB- or AP-1-dependent transcription. Sometimes the oxygen concentration also is of prime importance, since under normoxic conditions nitrosative stress activates HIF-1-dependent transcription, while under hypoxic conditions nitrosative stress leads to inhibition of HIF-1-dependent transcription. This review summarizes what is known about effects of physiological NO levels as well as of nitrosative stress on transcription.
Collapse
Affiliation(s)
- Klaus-Dietrich Kröncke
- Institute of Molecular Medicine, Research Group Immunobiology, Medical Department, Heinrich-Heine-University Düsseldorf, D-20225 Düsseldorf, Germany
| |
Collapse
|
18
|
Maiuri MC, Tajana G, Iuvone T, De Stefano D, Mele G, Ribecco MT, Cinelli MP, Romano MF, Turco MC, Carnuccio R. Nuclear factor-kappaB regulates inflammatory cell apoptosis and phagocytosis in rat carrageenin-sponge implant model. THE AMERICAN JOURNAL OF PATHOLOGY 2004; 165:115-26. [PMID: 15215167 PMCID: PMC1618533 DOI: 10.1016/s0002-9440(10)63280-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 03/15/2004] [Indexed: 10/18/2022]
Abstract
In the present study we investigated whether apoptosis and phagocytosis are regulated by nuclear factor (NF)-kappaB in a model of chronic inflammation. The subcutaneous implant of lambda-carrageenin-soaked sponges elicited an inflammatory response, characterized by a time-related increase of leukocyte infiltration into the sponge and tissue formation, which was inhibited by simultaneous injection of wild-type oligodeoxynucleotide decoy to NF-kappaB. Molecular and morphological analysis performed on infiltrated cells demonstrated: 1) an inhibition of NF-kappaB/DNA binding activity; 2) an increase of polymorphonuclear leukocyte apoptosis correlated either to an increase of p53 or Bax and decrease of Bcl-2 protein expression; and 3) an increase of phagocytosis of apoptotic polymorphonuclear leukocytes by macrophages associated with an increase of transforming growth factor-beta1 and decrease of tumor necrosis factor-alpha as well as nitrite/nitrate production. Our results, showing that blockade of NF-kappaB by oligodeoxynucleotide decoy increases inflammatory cell apoptosis and phagocytosis, may contribute to lead to new insights into the mechanisms governing the inflammatory process.
Collapse
Affiliation(s)
- Maria Chiara Maiuri
- Dipartimento di Farmacologia Sperimentale, Scienze Biomorfologiche, and Biochimica e Biotecnologie Mediche, Federico II University of Naples, Via D. Montesano 49, 80131 Naples, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Differential expression of genes involved in metabolism between tumorigenitic human leukemia cell lines K562 and K562-n. Chin J Cancer Res 2003. [DOI: 10.1007/bf02974910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
20
|
Franek WR, Chowdary YC, Lin X, Hu M, Miller EJ, Kazzaz JA, Razzano P, Romashko J, Davis JM, Narula P, Horowitz S, Scott W, Mantell LL. Suppression of nuclear factor-kappa B activity by nitric oxide and hyperoxia in oxygen-resistant cells. J Biol Chem 2002; 277:42694-700. [PMID: 12215428 DOI: 10.1074/jbc.m202623200] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Inhaled nitric oxide (iNO) is used clinically to treat pulmonary hypertension in newborns, often in conjunction with hyperoxia (NO/O2). Prolonged exposure to NO/O2 causes synergistic lung injury and death of lung epithelial cells. To explore the mechanisms involved, oxygen-resistant HeLa-80 cells were exposed to NO +/- O2. Exposure to NO and O2 induced a synergistic cytotoxicity, accompanied with apoptotic characteristics, including elevated caspase-3-like activity, Annexin V incorporation, and nuclear condensation. This apoptosis was associated with a synergistic suppression of NF-kappaB activity. Cells lacking functional NF-kappaB p65 subunit were more sensitive to NO/O2 than their wild type counterparts. This injury was partially rescued by transfection with a p65 expression construct, suggesting an inverse relationship between NF-kappaB and susceptibility to the cytotoxicity of NO/O2. Despite the reduced NF-kappaB activity in cells exposed to NO +/- O2, IkappaBalpha was degraded, suggesting that pathways regulating the steady-state levels of IkappaB were not involved. However, exposure to NO/O2 caused a marked reduction in nuclear localization and an increase in protein carbonyl formation of NF-kappaB p65 subunit. These results suggest that NO/O2-induced apoptosis occurs by suppressing NF-kappaB activity.
Collapse
Affiliation(s)
- William R Franek
- Department of Thoracic Cardiovascular Surgery, Winthrop University Hospital, State University of New York/Stony Brook School of Medicine, Mineola, New York 11501, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Abstract
Cells are constantly generating reactive oxygen species (ROS) during aerobic metabolism. As a consequence, each cell is equipped with an extensive antioxidant defence system to combat excessive production of ROS. Oxidative stress occurs in cells when the generation of ROS overwhelms the cell's natural antioxidant defences. There is a growing consensus that oxidative stress and the redox state of a cell plays a pivotal role in regulating apoptosis, a tightly controlled form of cell death in which a cell partakes in its own demise. More recently, a role for reactive nitrogen species (RNI) as both positive and negative regulators of cell death has been established. This review describes the major sources of ROS and RNI in a cell, the control of cell death by these species and the role of antioxidants as regulators of oxidative stress and apoptosis. Finally, the various methods that can be employed in establishing a role for both ROS and RNI in apoptosis will be discussed with particular emphasis on their intracellular detection.
Collapse
Affiliation(s)
- James F Curtin
- Tumour Biology Laboratory, Department of Biochemistry, Lee Maltings, University College Cork, Ireland
| | | | | |
Collapse
|