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 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] [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 Madrid, Arturo Duperier 4, Madrid, 28029, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Av. Monforte de Lemos 3-5, P-11, Madrid, 28029, Spain
| | - Adrián Povo-Retana
- Instituto de Investigaciones Biomédicas Sols-Morreale, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Arturo Duperier 4, Madrid, 28029, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Av. Monforte de Lemos 3-5, P-11, Madrid, 28029, Spain
| | - Silvia Marin
- Department of Biochemistry and Molecular Biomedicine-Institute of Biomedicine (IBUB), Faculty of Biology, Universitat de Barcelona, Barcelona, 08028, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Av. Monforte de Lemos 3-5, P-11, Madrid, 28029, Spain
| | - Sergio Madurga
- Department of Material Science and Physical Chemistry & Research Institute of Theoretical and Computational Chemistry (IQTCUB), University of Barcelona, Barcelona, 08028, Spain
| | - Marco Fariñas
- Department of Biochemistry and Molecular Biomedicine-Institute of Biomedicine (IBUB), Faculty of Biology, Universitat de Barcelona, Barcelona, 08028, Spain
| | - Nuria Aleixandre
- Department of Biochemistry and Molecular Biomedicine-Institute of Biomedicine (IBUB), Faculty of Biology, Universitat de Barcelona, Barcelona, 08028, Spain
- Department of Material Science and Physical Chemistry & Research Institute of Theoretical and Computational Chemistry (IQTCUB), University of Barcelona, Barcelona, 08028, Spain
| | - Antonio Castrillo
- Instituto de Investigaciones Biomédicas Sols-Morreale, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Arturo Duperier 4, Madrid, 28029, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Av. Monforte de Lemos 3-5, P-11, Madrid, 28029, Spain
- Unidad de Biomedicina (Unidad Asociada al CSIC) de la Universidad de Las Palmas de Gran Canaria, Las Palmas, 35016, Spain
- Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS) de la Universidad de Las Palmas de Gran Canaria, Las Palmas, 35016, Spain
| | - Juan V de la Rosa
- Unidad de Biomedicina (Unidad Asociada al CSIC) de la Universidad de Las Palmas de Gran Canaria, Las Palmas, 35016, Spain
- Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS) de la Universidad de Las Palmas de Gran Canaria, Las Palmas, 35016, Spain
| | - Carlota Alvarez-Lucena
- Instituto de Investigaciones Biomédicas Sols-Morreale, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Arturo Duperier 4, Madrid, 28029, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Av. Monforte de Lemos 3-5, P-11, Madrid, 28029, Spain
| | - Rodrigo Landauro-Vera
- Instituto de Investigaciones Biomédicas Sols-Morreale, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Arturo Duperier 4, Madrid, 28029, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Av. Monforte de Lemos 3-5, P-11, Madrid, 28029, Spain
| | - Patricia Prieto
- Instituto de Investigaciones Biomédicas Sols-Morreale, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Arturo Duperier 4, Madrid, 28029, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Av. Monforte de Lemos 3-5, P-11, Madrid, 28029, Spain
- Departamento de Farmacología, Farmacognosia y Botánica, Facultad de Farmacia, Universidad Complutense de Madrid, Madrid, 28040, Spain
| | - Marta Cascante
- Department of Biochemistry and Molecular Biomedicine-Institute of Biomedicine (IBUB), Faculty of Biology, Universitat de Barcelona, Barcelona, 08028, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Av. Monforte de Lemos 3-5, P-11, Madrid, 28029, Spain
| | - Lisardo Boscá
- Instituto de Investigaciones Biomédicas Sols-Morreale, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Arturo Duperier 4, Madrid, 28029, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Av. Monforte de Lemos 3-5, P-11, Madrid, 28029, Spain
- Unidad de Biomedicina (Unidad Asociada al CSIC) de la Universidad de Las Palmas de Gran Canaria, Las Palmas, 35016, Spain
| |
Collapse
|
2
|
Sánchez-García S, Castrillo A, Boscá L, Prieto P. Potential Beneficial Role of Nitric Oxide in SARS-CoV-2 Infection: Beyond Spike-Binding Inhibition. Antioxidants (Basel) 2024; 13:1301. [PMID: 39594443 PMCID: PMC11591382 DOI: 10.3390/antiox13111301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 10/23/2024] [Accepted: 10/24/2024] [Indexed: 11/28/2024] Open
Abstract
SARS-CoV-2, the causative virus for the COVID-19 disease, uses its spike glycoprotein to bind to human ACE2 as a first step for viral entry into the cell. For this reason, great efforts have been made to find mechanisms that disrupt this interaction, avoiding the infection. Nitric oxide (NO) is a soluble endogenous gas with known antiviral and immunomodulatory properties. In this study, we aimed to test whether NO could inhibit the binding of the viral spike to ACE2 in human cells and its effects on ACE2 enzymatic activity. Our results show that ACE2 activity was decreased by the NO donors DETA-NONOate and GSNO and by the NO byproduct peroxynitrite. Furthermore, we found that DETA-NONOate could break the spike-ACE2 interaction using the spike from two different variants (Alpha and Gamma) and in two different human cell types. Moreover, the same result was obtained when using NO-producing murine macrophages, while no significant changes were observed in ACE2 expression or distribution within the cell. These results support that it is worth considering NO as a therapeutic agent for COVID-19, as previous reports have suggested.
Collapse
Affiliation(s)
- Sergio Sánchez-García
- Instituto de Investigaciones Biomédicas Sols-Morreale, CSIC-UAM, Arturo Duperier 4, 28029 Madrid, Spain;
| | - Antonio Castrillo
- Instituto de Investigaciones Biomédicas Sols-Morreale, CSIC-UAM, Arturo Duperier 4, 28029 Madrid, Spain;
- Unidad de Biomedicina (Unidad Asociada al CSIC), Universidad de Las Palmas de Gran Canaria, 35016 Las Palmas, Spain
| | - Lisardo Boscá
- Instituto de Investigaciones Biomédicas Sols-Morreale, CSIC-UAM, Arturo Duperier 4, 28029 Madrid, Spain;
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Av. Monforte de Lemos 3-5, P-11, 28029 Madrid, Spain
| | - Patricia Prieto
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Av. Monforte de Lemos 3-5, P-11, 28029 Madrid, Spain
- Departamento de Farmacología, Farmacognosia y Botánica, Facultad de Farmacia, Universidad Complutense de Madrid, Plaza Ramón y Cajal, 28040 Madrid, Spain
| |
Collapse
|
3
|
Povo-Retana A, Fariñas M, Landauro-Vera R, Mojena M, Alvarez-Lucena C, Fernández-Moreno MA, Castrillo A, de la Rosa Medina JV, Sánchez-García S, Foguet C, Mas F, Marin S, Cascante M, Boscá L. Immunometabolic actions of trabectedin and lurbinectedin on human macrophages: relevance for their anti-tumor activity. Front Immunol 2023; 14:1211068. [PMID: 37675104 PMCID: PMC10479946 DOI: 10.3389/fimmu.2023.1211068] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 08/03/2023] [Indexed: 09/08/2023] Open
Abstract
In recent years, the central role of cell bioenergetics in regulating immune cell function and fate has been recognized, giving rise to the interest in immunometabolism, an area of research focused on the interaction between metabolic regulation and immune function. Thus, early metabolic changes associated with the polarization of macrophages into pro-inflammatory or pro-resolving cells under different stimuli have been characterized. Tumor-associated macrophages are among the most abundant cells in the tumor microenvironment; however, it exists an unmet need to study the effect of chemotherapeutics on macrophage immunometabolism. Here, we use a systems biology approach that integrates transcriptomics and metabolomics to unveil the immunometabolic effects of trabectedin (TRB) and lurbinectedin (LUR), two DNA-binding agents with proven antitumor activity. Our results show that TRB and LUR activate human macrophages toward a pro-inflammatory phenotype by inducing a specific metabolic rewiring program that includes ROS production, changes in the mitochondrial inner membrane potential, increased pentose phosphate pathway, lactate release, tricarboxylic acids (TCA) cycle, serine and methylglyoxal pathways in human macrophages. Glutamine, aspartate, histidine, and proline intracellular levels are also decreased, whereas oxygen consumption is reduced. The observed immunometabolic changes explain additional antitumor activities of these compounds and open new avenues to design therapeutic interventions that specifically target the immunometabolic landscape in the treatment of cancer.
Collapse
Affiliation(s)
- Adrián Povo-Retana
- Instituto de Investigaciones Biomédicas Alberto Sols, CSIC-UAM, Madrid, Spain
| | - Marco Fariñas
- Department of Biochemistry and Molecular Biomedicine-Institute of Biomedicine (IBUB), Faculty of Biology, Universitat de Barcelona, Barcelona, Spain
| | | | - Marina Mojena
- Instituto de Investigaciones Biomédicas Alberto Sols, CSIC-UAM, Madrid, Spain
| | | | - Miguel A. Fernández-Moreno
- Instituto de Investigaciones Biomédicas Alberto Sols, CSIC-UAM, Madrid, Spain
- Departamento de Bioquímica, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - Antonio Castrillo
- Instituto de Investigaciones Biomédicas Alberto Sols, CSIC-UAM, Madrid, Spain
- Unidad de Biomedicina (Unidad Asociada al CSIC) de la Universidad de Las Palmas de Gran Canaria, Las Palmas, Spain
| | - Juan Vladimir de la Rosa Medina
- Unidad de Biomedicina (Unidad Asociada al CSIC) de la Universidad de Las Palmas de Gran Canaria, Las Palmas, Spain
- Unidad Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS) de la Universidad de Las Palmas de Gran Canaria, Las Palmas, Spain
| | | | - Carles Foguet
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
| | - Francesc Mas
- Department of Material Science and Physical Chemistry & Research Institute of Theoretical and Computational Chemistry (IQTCUB), University of Barcelona, Barcelona, Spain
| | - Silvia Marin
- Department of Biochemistry and Molecular Biomedicine-Institute of Biomedicine (IBUB), Faculty of Biology, Universitat de Barcelona, Barcelona, Spain
- CIBER of Hepatic and Digestive Diseases (CIBEREHD), Institute of Health Carlos III (ISCIII), Madrid, Spain
| | - Marta Cascante
- Department of Biochemistry and Molecular Biomedicine-Institute of Biomedicine (IBUB), Faculty of Biology, Universitat de Barcelona, Barcelona, Spain
- CIBER of Hepatic and Digestive Diseases (CIBEREHD), Institute of Health Carlos III (ISCIII), Madrid, Spain
| | - Lisardo Boscá
- Instituto de Investigaciones Biomédicas Alberto Sols, CSIC-UAM, Madrid, Spain
- Unidad de Biomedicina (Unidad Asociada al CSIC) de la Universidad de Las Palmas de Gran Canaria, Las Palmas, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Av. Monforte de Lemos, Madrid, Spain
| |
Collapse
|
4
|
Functional Crosstalk between PCSK9 Internalization and Pro-Inflammatory Activation in Human Macrophages: Role of Reactive Oxygen Species Release. Int J Mol Sci 2022; 23:ijms23169114. [PMID: 36012389 PMCID: PMC9409451 DOI: 10.3390/ijms23169114] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 08/10/2022] [Accepted: 08/11/2022] [Indexed: 02/06/2023] Open
Abstract
Atherosclerosis is a cardiovascular disease caused mainly by dyslipidemia and is characterized by the formation of an atheroma plaque and chronic inflammation. Proprotein convertase subtilisin/kexin type 9 (PCSK9) is a protease that induces the degradation of the LDL receptor (LDLR), which contributes to increased levels of LDL cholesterol and the progress of atherosclerosis. Given that macrophages are relevant components of the lipidic and inflammatory environment of atherosclerosis, we studied the effects of PCSK9 treatment on human macrophages. Our data show that human macrophages do not express PCSK9 but rapidly incorporate the circulating protein through the LDLR and also activate the pro-inflammatory TLR4 pathway. Both LDLR and TLR4 are internalized after incubation of macrophages with exogenous PCSK9. PCSK9 uptake increases the production of reactive oxygen species and reduces the expression of genes involved in lipid metabolism and cholesterol efflux, while enhancing the production of pro-inflammatory cytokines through a TLR4-dependent mechanism. Under these conditions, the viability of macrophages is compromised, leading to increased cell death. These results provide novel insights into the role of PCSK9 in the crosstalk of lipids and cholesterol metabolism through the LDLR and on the pro-inflammatory activation of macrophages through TLR4 signaling. These pathways are relevant in the outcome of atherosclerosis and highlight the relevance of PCSK9 as a therapeutic target for the treatment of cardiovascular diseases.
Collapse
|
5
|
Glinski A, Lima de Souza T, Zablocki da Luz J, Bezerra Junior AG, Camargo de Oliveira C, de Oliveira Ribeiro CA, Filipak Neto F. Toxicological effects of silver nanoparticles and cadmium chloride in macrophage cell line (RAW 264.7): An in vitro approach. J Trace Elem Med Biol 2021; 68:126854. [PMID: 34488184 DOI: 10.1016/j.jtemb.2021.126854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 08/19/2021] [Accepted: 08/31/2021] [Indexed: 11/16/2022]
Abstract
BACKGROUND Silver nanoparticles (AgNP) are largely used in nanotechnological products, but the real risks for human and environment are still poorly understood if we consider the effects of mixtures of AgNP and environmental contaminants, such as non-essential metals. METHODS The aim of the present study was to investigate the cytotoxicity and toxicological interaction of AgNP (1-4 nm, 0.36 and 3.6 μg mL-1) and cadmium (Cd, 1 and 10 μM) mixtures. The murine macrophage cell line RAW 264.7 was used as a model. RESULTS Effects were observed after a few hours (4 h) on reactive oxygen species (ROS) and became more pronounced after 24 h-exposure. Cell death occurred by apoptosis, and loss of cell viability (24 h-exposure) was preceded by increases of ROS levels and DNA repair foci, but not of NO levels. Co-exposure potentiated some effects (decrease of cell viability and increase of ROS and NO levels), indicating toxicological interaction. CONCLUSION These effects are important findings that must be better investigated, since the interaction of Cd with AgNP from nanoproducts may impair the function of macrophages and represent a health risk for humans.
Collapse
Affiliation(s)
- Andressa Glinski
- Laboratório de Toxicologia Celular, Departamento de Biologia Celular, Universidade Federal do Paraná, CEP 81.531-980, Curitiba, PR, Brazil
| | - Tugstênio Lima de Souza
- Laboratório de Toxicologia Celular, Departamento de Biologia Celular, Universidade Federal do Paraná, CEP 81.531-980, Curitiba, PR, Brazil
| | - Jessica Zablocki da Luz
- Laboratório de Toxicologia Celular, Departamento de Biologia Celular, Universidade Federal do Paraná, CEP 81.531-980, Curitiba, PR, Brazil
| | - Arandi Ginane Bezerra Junior
- Laboratório Fotonanobio, Departamento Acadêmico de Física, Universidade Tecnológica Federal do Paraná, CEP 80.230-901, Curitiba, PR, Brazil
| | - Carolina Camargo de Oliveira
- Laboratório de Células Inflamatórias e Neoplásicas, Departamento de Biologia Celular, Universidade Federal do Paraná, CEP 81.531-980, Curitiba, PR, Brazil
| | - Ciro Alberto de Oliveira Ribeiro
- Laboratório de Toxicologia Celular, Departamento de Biologia Celular, Universidade Federal do Paraná, CEP 81.531-980, Curitiba, PR, Brazil
| | - Francisco Filipak Neto
- Laboratório de Toxicologia Celular, Departamento de Biologia Celular, Universidade Federal do Paraná, CEP 81.531-980, Curitiba, PR, Brazil.
| |
Collapse
|
6
|
Omidkhah N, Ghodsi R. NO-HDAC dual inhibitors. Eur J Med Chem 2021; 227:113934. [PMID: 34700268 DOI: 10.1016/j.ejmech.2021.113934] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 10/08/2021] [Accepted: 10/17/2021] [Indexed: 12/11/2022]
Abstract
HDAC inhibitors and NO donors have both demonstrated independently broad therapeutic potential in a variety of diseases. Borretto et al. presented the topic of NO-HDAC dual inhibitors for the first time in 2013 as an attractive new topic. Here we collected the general structure of all synthesized NO-HDAC dual inhibitors, lead compounds, synthesis methods and biological features of the most potent dual NO-HDAC inhibitor in each category with the intention of assisting in the synthesis and optimization of new drug-like compounds for diverse diseases. Based on studies done so far, NO-HDAC dual inhibitors have displayed satisfactory results against wound healing (3), heart hypertrophy (3), inflammatory, cardiovascular, neuromuscular illnesses (11a-11e) and cancer (6a-6o, 9a-9d, 10a-10d, 16 and 17). NO-HDAC dual inhibitors can have high therapeutic potential for various diseases due to their new properties, NO properties, HDAC inhibitor properties and also due to the effects of NO on HDAC enzymes.
Collapse
Affiliation(s)
- Negar Omidkhah
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Razieh Ghodsi
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
7
|
Ghanavi J, Farnia P, Farnia P, Velayati AA. The role of interferon-gamma and interferon-gamma receptor in tuberculosis and nontuberculous mycobacterial infections. Int J Mycobacteriol 2021; 10:349-357. [PMID: 34916451 DOI: 10.4103/ijmy.ijmy_186_21] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Mycobacterium tuberculosis (Mtb) and nontuberculous mycobacteria (NTM) remain the leading causes of lung disease and mortality worldwide. Interferon-gamma (IFN-γ) and its receptor (IFN-γR) play a key role in mediating immunity against Mtb and NTM. This study was conducted as a systematic review; all information was collected from databases such as: PubMed, Scopus, Medline, SID, and medical databases. Finally, all the collected data were reviewed, and all content was categorized briefly. There is growing evidence that IFN-γ plays an important role in host defense against these two intracellular pathogens by activating macrophages. In addition, IFN-γ has been shown to be an integral part of various antibacterial methods such as granuloma formation and phagosome-lysosome fusion, both of which lead to the death of intracellular Mycobacterium. As a result, its absence is associated with overgrowth of intracellular pathogens and disease caused by Mtb or Mycobacterium nontuberculosis. We also look at the role of IFN-γR in Mtb or NTM because IFN-γ acts through IFN-γR. Finally, we introduce new approaches to the treatment of M. tuberculosis complex (MTC) and NTM disease, such as cell and gene-based therapies that work by modulating IFN-γ and IFN-γR.
Collapse
Affiliation(s)
- Jalaledin Ghanavi
- Mycobacteriology Research Center, National Research Institute of Tuberculosis and Lung Disease, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Poopak Farnia
- Mycobacteriology Research Center, National Research Institute of Tuberculosis and Lung Disease, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Parissa Farnia
- Mycobacteriology Research Center, National Research Institute of Tuberculosis and Lung Disease, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Akbar Velayati
- Mycobacteriology Research Center, National Research Institute of Tuberculosis and Lung Disease, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
8
|
Povo-Retana A, Mojena M, Stremtan AB, Fernández-García VB, Gómez-Sáez A, Nuevo-Tapioles C, Molina-Guijarro JM, Avendaño-Ortiz J, Cuezva JM, López-Collazo E, Martínez-Leal JF, Boscá L. Specific Effects of Trabectedin and Lurbinectedin on Human Macrophage Function and Fate-Novel Insights. Cancers (Basel) 2020; 12:cancers12103060. [PMID: 33092171 PMCID: PMC7590144 DOI: 10.3390/cancers12103060] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 09/28/2020] [Accepted: 10/16/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Tumor-associated macrophages (TAMs) play a crucial role in suppressing the immunosurveillance function of the immune system that prevents tumor growth. Indeed, macrophages can also be targeted by different chemotherapeutic agents improving the action over immune checkpoints to fight cancer. Here we describe the effect of trabectedin and lurbinectedin on human macrophage cell viability and function. METHODS Blood monocytes from healthy donors were differentiated into macrophages and exposed to different stimuli promoting functional polarization and differentiation into tumor-associated macrophages. Cells were challenged with the chemotherapeutic drugs and the effects on cell viability and function were analyzed. RESULTS Human macrophages exhibit at least two different profiles in response to these drugs. One-fourth of the blood donors assayed (164 individuals) were extremely sensitive to trabectedin and lurbinectedin, which promoted apoptotic cell death. Macrophages from other individuals retained viability but responded to the drugs increasing reactive oxygen production and showing a rapid intracellular calcium rise and a loss of mitochondrial oxygen consumption. Cell-membrane exposure of programmed-death ligand 1 (PD-L1) significantly decreased after treatment with therapeutic doses of these drugs, including changes in the gene expression profile of hypoxia-inducible factor 1 alpha (HIF-1α)-dependent genes, among other. CONCLUSIONS The results provide evidence of additional onco-therapeutic actions for these drugs.
Collapse
Affiliation(s)
- Adrián Povo-Retana
- Instituto de Investigaciones Biomédicas Alberto Sols (Centro Mixto CSIC-UAM), 28029 Madrid, Spain; (A.P.-R.); (M.M.); (A.B.S.); (V.B.F.-G.); (A.G.-S.)
| | - Marina Mojena
- Instituto de Investigaciones Biomédicas Alberto Sols (Centro Mixto CSIC-UAM), 28029 Madrid, Spain; (A.P.-R.); (M.M.); (A.B.S.); (V.B.F.-G.); (A.G.-S.)
| | - Adrian B. Stremtan
- Instituto de Investigaciones Biomédicas Alberto Sols (Centro Mixto CSIC-UAM), 28029 Madrid, Spain; (A.P.-R.); (M.M.); (A.B.S.); (V.B.F.-G.); (A.G.-S.)
| | - Victoria B. Fernández-García
- Instituto de Investigaciones Biomédicas Alberto Sols (Centro Mixto CSIC-UAM), 28029 Madrid, Spain; (A.P.-R.); (M.M.); (A.B.S.); (V.B.F.-G.); (A.G.-S.)
| | - Ana Gómez-Sáez
- Instituto de Investigaciones Biomédicas Alberto Sols (Centro Mixto CSIC-UAM), 28029 Madrid, Spain; (A.P.-R.); (M.M.); (A.B.S.); (V.B.F.-G.); (A.G.-S.)
| | - Cristina Nuevo-Tapioles
- Centro de Biología Molecular (Centro Mixto CSIC-UAM), Nicolás Cabrera S/N, Ciudad Universitaria de Cantoblanco, 28049 Madrid, Spain; (C.N.-T.); (J.M.C.)
- Centro de Investigación Biomédica en Red en Enfermedades Raras (CIBERER), 28029 Madrid, Spain
| | | | - José Avendaño-Ortiz
- Instituto de Investigación Sanitaria La Paz (IdiPaz), Hospital Universitario La Paz, 28046 Madrid, Spain; (J.A.-O.); (E.L.-C.)
| | - José M. Cuezva
- Centro de Biología Molecular (Centro Mixto CSIC-UAM), Nicolás Cabrera S/N, Ciudad Universitaria de Cantoblanco, 28049 Madrid, Spain; (C.N.-T.); (J.M.C.)
- Centro de Investigación Biomédica en Red en Enfermedades Raras (CIBERER), 28029 Madrid, Spain
| | - Eduardo López-Collazo
- Instituto de Investigación Sanitaria La Paz (IdiPaz), Hospital Universitario La Paz, 28046 Madrid, Spain; (J.A.-O.); (E.L.-C.)
| | | | - Lisardo Boscá
- Instituto de Investigaciones Biomédicas Alberto Sols (Centro Mixto CSIC-UAM), 28029 Madrid, Spain; (A.P.-R.); (M.M.); (A.B.S.); (V.B.F.-G.); (A.G.-S.)
- Instituto de Investigación Sanitaria La Paz (IdiPaz), Hospital Universitario La Paz, 28046 Madrid, Spain; (J.A.-O.); (E.L.-C.)
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
- Correspondence: ; Tel.: +34-9149-72747
| |
Collapse
|
9
|
Virág L, Jaén RI, Regdon Z, Boscá L, Prieto P. Self-defense of macrophages against oxidative injury: Fighting for their own survival. Redox Biol 2019; 26:101261. [PMID: 31279985 PMCID: PMC6614175 DOI: 10.1016/j.redox.2019.101261] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Revised: 06/17/2019] [Accepted: 06/23/2019] [Indexed: 02/07/2023] Open
Abstract
Activated macrophages play a central role in both the development and resolution of inflammation. These immune cells need to be functional in harmful conditions with high levels of reactive oxygen and nitrogen species that can damage their basic cell components, which may alter their metabolism. An excessive accumulation of these cell alterations drives macrophages inexorably to cell death, which has been associated to the development of several inflammatory diseases and even with aging in a process termed as "immunosenescence". Macrophages, however, exhibit a prolonged survival in this hostile environment because they equip themselves with a complex network of protective mechanisms. Here we provide an overview of these self-defense mechanisms with special attention being paid to bioactive lipid mediators, NRF2 signaling and metabolic reprogramming.
Collapse
Affiliation(s)
- László Virág
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary; MTA-DE Cell Biology and Signaling Research Group, Debrecen, Hungary.
| | - Rafael I Jaén
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM). Arturo Duperier 4, 28029, Madrid, Spain.
| | - Zsolt Regdon
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.
| | - Lisardo Boscá
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM). Arturo Duperier 4, 28029, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), ISCIII, Madrid, Spain.
| | - Patricia Prieto
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM). Arturo Duperier 4, 28029, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), ISCIII, Madrid, Spain.
| |
Collapse
|
10
|
Yadav RK, Tripathi CB, Saraf SA, Ansari MN, Saeedan AS, Aldosary S, Rajinikanth PS, Kaithwas G. Alpha-linolenic acid based nano-suspension protect against lipopolysaccharides induced mastitis by inhibiting NFκBp65, HIF-1α, and mitochondria-mediated apoptotic pathway in albino Wistar rats. Toxicol Appl Pharmacol 2019; 377:114628. [PMID: 31207257 DOI: 10.1016/j.taap.2019.114628] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 03/30/2019] [Accepted: 06/12/2019] [Indexed: 12/25/2022]
|
11
|
Luan T, Fu S, Huang L, Zuo Y, Ding M, Li N, Chen J, Wang H, Wang J. MicroRNA-98 promotes drug resistance and regulates mitochondrial dynamics by targeting LASS2 in bladder cancer cells. Exp Cell Res 2018; 373:188-197. [PMID: 30463687 DOI: 10.1016/j.yexcr.2018.10.013] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2018] [Revised: 10/13/2018] [Accepted: 10/23/2018] [Indexed: 12/12/2022]
Abstract
MicroRNA-98(miR-98) has been shown to be critical for tumorigenesis, however its involvement in bladder cancer are unclear. The present study aims to investigate the expression, biological roles and potential mechanisms of miR-98 in human bladder cancer. We found that miR-98 was upregulated in bladder urothelial carcinoma tissues compared with adjacent normal tissues. In addition, miR-98 expression was higher in bladder cancer cell lines than in uroepithelial cell line SV-HUC-1. Functional studies revealed that miR-98 mimic promoted proliferation of T24 cells while miR-98 inhibitor inhibited proliferation of BIU-87 cells. Moreover, miR-98 mimic increased cisplatin/doxorubicin resistance and inhibited apoptosis in T24 cells, while miR-98 inhibitor decreased chemoresistance and facilitated apoptosis in BIU-87 cells. Further experiments using MitoTracker and JC-1 staining showed that miR-98 could regulate mitochondrial fission/fusion balance and mitochondrial membrane potential. Western blot showed that miR-98 upregulated cyclin D1, p-Drp1 and Drp1. Using luciferase reporter assay, we demonstrated that LASS2 acted as a direct target of miR-98. LASS2 overexpression induced mitochondrial fusion and downregulated mitochondrial potential, with decreased p-Drp1 status. Additionally, LASS2 siRNA abrogated the effects of miR-98 mimic on Drp1phosphorylation and chemoresistance. We also found a negative correlation between LASS2 and miR-98 in bladder cancer tissues. In conclusion, our study demonstrates that miR-98 targets LASS2 and regulates bladder cancer chemoresistance through modulation of mitochondrial function.
Collapse
Affiliation(s)
- Ting Luan
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Yunnan Institute of Urology, Kunming 650101, China
| | - Shi Fu
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Yunnan Institute of Urology, Kunming 650101, China
| | - Lijuan Huang
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Yunnan Institute of Urology, Kunming 650101, China
| | - Yigang Zuo
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Yunnan Institute of Urology, Kunming 650101, China
| | - Mingxia Ding
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Yunnan Institute of Urology, Kunming 650101, China
| | - Ning Li
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Yunnan Institute of Urology, Kunming 650101, China
| | - Jian Chen
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Yunnan Institute of Urology, Kunming 650101, China
| | - Haifeng Wang
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Yunnan Institute of Urology, Kunming 650101, China.
| | - Jiansong Wang
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Yunnan Institute of Urology, Kunming 650101, China.
| |
Collapse
|
12
|
Mojena M, Povo-Retana A, González-Ramos S, Fernández-García V, Regadera J, Zazpe A, Artaiz I, Martín-Sanz P, Ledo F, Boscá L. Benzylamine and Thenylamine Derived Drugs Induce Apoptosis and Reduce Proliferation, Migration and Metastasis Formation in Melanoma Cells. Front Oncol 2018; 8:328. [PMID: 30191142 PMCID: PMC6115490 DOI: 10.3389/fonc.2018.00328] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Accepted: 07/31/2018] [Indexed: 02/05/2023] Open
Abstract
Melanomas are heterogeneous and aggressive tumors, and one of the worse in prognosis. Melanoma subtypes follow distinct pathways until terminal oncogenic transformation. Here, we have evaluated a series of molecules that exhibit potent cytotoxic effects over the murine and human melanoma cell lines B16F10 and MalMe-3M, respectively, both ex vivo and in animals carrying these melanoma cells. Ex vivo mechanistic studies on molecular targets involved in melanoma growth, migration and viability were evaluated in cultured cells treated with these drugs which exhibited potent proapoptotic and cytotoxic effects and reduced cell migration. These drugs altered the Wnt/β-catenin pathway, which is important for the oncogenic phenotype of melanoma cells. In in vivo experiments, male C57BL/6 or nude mice were injected with melanoma cells that rapidly expanded in these animals and, in some cases were able to form metastasis in lungs. Treatment with anti-tumor drugs derived from benzylamine and 2-thiophenemethylamine (F10503LO1 and related compounds) significantly attenuated tumor growth, impaired cell migration, and reduced the metastatic activity. Several protocols of administration were applied, all of them leading to significant reduction in the tumor size and enhanced animal survival. Tumor cells carrying a luciferase transgene allowed a time-dependent study on the progression of the tumor. Molecular analysis of the pathways modified by F10503LO1 and related compounds defined the main relevant targets for tumor regression: the activation of pro-apoptotic and anti-proliferative routes. These data might provide the proof-of-principle and rationale for its further clinical evaluation.
Collapse
Affiliation(s)
- Marina Mojena
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Madrid, Spain
| | - Adrián Povo-Retana
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Madrid, Spain
| | - Silvia González-Ramos
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares y Hepáticas y Digestivas, ISC III, Madrid, Spain
| | | | - Javier Regadera
- Departamento de Anatomía, Histología y Neurociencia, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - Arturo Zazpe
- R&D+i Department Faes-Farma, Avda Autonomía, Leioa, Spain
| | - Inés Artaiz
- R&D+i Department Faes-Farma, Avda Autonomía, Leioa, Spain
| | - Paloma Martín-Sanz
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares y Hepáticas y Digestivas, ISC III, Madrid, Spain
| | - Francisco Ledo
- R&D+i Department Faes-Farma, Avda Autonomía, Leioa, Spain
- *Correspondence: Francisco Ledo
| | - Lisardo Boscá
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares y Hepáticas y Digestivas, ISC III, Madrid, Spain
- Lisardo Boscá
| |
Collapse
|
13
|
Gu X, Huang Z, Ren Z, Tang X, Xue R, Luo X, Peng S, Peng H, Lu B, Tian J, Zhang Y. Potent Inhibition of Nitric Oxide-Releasing Bifendate Derivatives against Drug-Resistant K562/A02 Cells in Vitro and in Vivo. J Med Chem 2017; 60:928-940. [PMID: 28068095 DOI: 10.1021/acs.jmedchem.6b01075] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Multidrug resistance is a major obstacle to successful chemotherapy for leukemia. In this study, a series of nitric oxide (NO)-releasing bifendate derivatives (7a-n) were synthesized. Biological evaluation indicated that the most active compound (7a) produced relatively high levels of NO and significantly inhibited the proliferation of drug-resistant K562/A02 cells in vitro and in vivo. In addition, 7a induced the mitochondrial tyrosine nitration and the intracellular accumulation of rhodamine 123 by inhibiting P-gp activity in K562/A02 cells. Furthermore, 7a remarkably down-regulated AKT, NF-κB, and ERK activation and HIF-1α expression in K562/A02 cells, which are associated with the tumor cell proliferation and drug resistance. Notably, the antitumor effects were dramatically attenuated by an NO scavenger or elimination of the NO-releasing capability of 7a, indicating that NO produced by 7a contributed to, at least partly, its cytotoxicity against drug-resistant K562/A02 cells. Overall, 7a may be a potential agent against drug-resistant myelogenous leukemia.
Collapse
Affiliation(s)
- Xiaoke Gu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University , 24 Tongjiaxiang, Nanjing 210009, People's Republic of China.,Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University , Xuzhou 221004, People's Republic of China
| | - Zhangjian Huang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University , 24 Tongjiaxiang, Nanjing 210009, People's Republic of China
| | - Zhiguang Ren
- Department of Environment and Pharmacy, Tianjin Institute of Health and Environmental Medicine , Tianjin 300050, People's Republic of China
| | - Xiaobo Tang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University , 24 Tongjiaxiang, Nanjing 210009, People's Republic of China
| | - Rongfang Xue
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University , 24 Tongjiaxiang, Nanjing 210009, People's Republic of China
| | - Xiaojun Luo
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University , 24 Tongjiaxiang, Nanjing 210009, People's Republic of China
| | - Sixun Peng
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University , 24 Tongjiaxiang, Nanjing 210009, People's Republic of China
| | - Hui Peng
- Department of Environment and Pharmacy, Tianjin Institute of Health and Environmental Medicine , Tianjin 300050, People's Republic of China
| | - Bin Lu
- Institute of Biophysics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical College , Wenzhou 325035, People's Republic of China
| | - Jide Tian
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles , Los Angeles, California 90095, United States
| | - Yihua Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University , 24 Tongjiaxiang, Nanjing 210009, People's Republic of China
| |
Collapse
|
14
|
Shaki F, Ashari S, Ahangar N. Melatonin can attenuate ciprofloxacin induced nephrotoxicity: Involvement of nitric oxide and TNF-α. Biomed Pharmacother 2016; 84:1172-1178. [PMID: 27780148 DOI: 10.1016/j.biopha.2016.10.053] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2016] [Accepted: 10/17/2016] [Indexed: 12/19/2022] Open
|
15
|
Interplay between oxidant species and energy metabolism. Redox Biol 2015; 8:28-42. [PMID: 26741399 PMCID: PMC4710798 DOI: 10.1016/j.redox.2015.11.010] [Citation(s) in RCA: 217] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Revised: 11/20/2015] [Accepted: 11/25/2015] [Indexed: 02/07/2023] Open
Abstract
It has long been recognized that energy metabolism is linked to the production of reactive oxygen species (ROS) and critical enzymes allied to metabolic pathways can be affected by redox reactions. This interplay between energy metabolism and ROS becomes most apparent during the aging process and in the onset and progression of many age-related diseases (i.e. diabetes, metabolic syndrome, atherosclerosis, neurodegenerative diseases). As such, the capacity to identify metabolic pathways involved in ROS formation, as well as specific targets and oxidative modifications is crucial to our understanding of the molecular basis of age-related diseases and for the design of novel therapeutic strategies. Herein we review oxidant formation associated with the cell's energetic metabolism, key antioxidants involved in ROS detoxification, and the principal targets of oxidant species in metabolic routes and discuss their relevance in cell signaling and age-related diseases. Energy metabolism is both a source and target of oxidant species. Reactive oxygen species are formed in redox reactions in catabolic pathways. Sensitive targets of oxidant species regulate the flux of metabolic pathways. Metabolic pathways and antioxidant systems are regulated coordinately.
Collapse
|
16
|
Nitric Oxide-Mediated Posttranslational Modifications: Impacts at the Synapse. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2016:5681036. [PMID: 26635909 PMCID: PMC4655263 DOI: 10.1155/2016/5681036] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 06/28/2015] [Indexed: 11/18/2022]
Abstract
Nitric oxide (NO) is an important gasotransmitter molecule that is involved in numerous physiological processes throughout the nervous system. In addition to its involvement in physiological plasticity processes (long-term potentiation, LTP; long-term depression, LTD) which can include NMDAR-mediated calcium-dependent activation of neuronal nitric oxide synthase (nNOS), new insights into physiological and pathological consequences of nitrergic signalling have recently emerged. In addition to the canonical cGMP-mediated signalling, NO is also implicated in numerous pathways involving posttranslational modifications. In this review we discuss the multiple effects of S-nitrosylation and 3-nitrotyrosination on proteins with potential modulation of function but limit the analyses to signalling involved in synaptic transmission and vesicular release. Here, crucial proteins which mediate synaptic transmission can undergo posttranslational modifications with either pre- or postsynaptic origin. During normal brain function, both pathways serve as important cellular signalling cascades that modulate a diverse array of physiological processes, including synaptic plasticity, transcriptional activity, and neuronal survival. In contrast, evidence suggests that aging and disease can induce nitrosative stress via excessive NO production. Consequently, uncontrolled S-nitrosylation/3-nitrotyrosination can occur and represent pathological features that contribute to the onset and progression of various neurodegenerative diseases, including Parkinson's, Alzheimer's, and Huntington's.
Collapse
|
17
|
Palanisamy AP, Cheng G, Sutter AG, Liu J, Lewin DN, Chao J, Chavin K. Adenovirus-mediated eNOS expression augments liver injury after ischemia/reperfusion in mice. PLoS One 2014; 9:e93304. [PMID: 24667691 PMCID: PMC3965553 DOI: 10.1371/journal.pone.0093304] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Accepted: 02/28/2014] [Indexed: 01/16/2023] Open
Abstract
Hepatic ischemia/reperfusion (l/R) injury continues to be a critical problem. The role of nitric oxide in liver I/R injury is still controversial. This study examines the effect of endothelial nitric oxide synthase (eNOS) over-expression on hepatic function following I/R. Adenovirus expressing human eNOS (Ad-eNOS) was administered by tail vein injection into C57BL/6 mice. Control mice received either adenovirus expressing LacZ or vehicle only. Sixty minutes of total hepatic ischemia was performed 3 days after adenovirus treatment, and mice were sacrificed after 6 or 24 hrs of reperfusion to assess hepatic injury. eNOS over expression caused increased liver injury as evidenced by elevated AST and ALT levels and decreased hepatic ATP content. While necrosis was not pervasive in any group, TUNEL demonstrated significantly increased apoptosis in Ad-eNOS infected livers. Western blotting demonstrated increased levels of protein nitration and upregulation of the pro-apoptotic proteins bax and p53. Our data suggest that over-expression of eNOS is detrimental in the setting of hepatic I/R.
Collapse
Affiliation(s)
- Arun P. Palanisamy
- Division of Transplant Surgery, Department Of Surgery, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Gang Cheng
- Division of Transplant Surgery, Department Of Surgery, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Alton G. Sutter
- Division of Transplant Surgery, Department Of Surgery, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - John Liu
- Division of Transplant Surgery, Department Of Surgery, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - David N. Lewin
- Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Julie Chao
- Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Kenneth Chavin
- Division of Transplant Surgery, Department Of Surgery, Medical University of South Carolina, Charleston, South Carolina, United States of America
- * E-mail:
| |
Collapse
|
18
|
Angiotensin II induces mitochondrial dysfunction and promotes apoptosis via JNK signalling pathway in primary mouse calvaria osteoblast. Arch Oral Biol 2014; 59:513-23. [PMID: 24632094 DOI: 10.1016/j.archoralbio.2014.02.015] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Revised: 12/20/2013] [Accepted: 02/25/2014] [Indexed: 02/05/2023]
Abstract
OBJECTIVES This present study was designed to investigate the effects of Angiotensin II on mitochondrial functions, ROS generation and c-jun N-terminal kinases (JNK) signalling pathway-mediated cell apoptosis in mouse calvaria osteoblasts. METHODS Calvaria osteoblast were isolated and cultured. The cells were separated into two groups-control and treated groups-where the latter was stimulated with angiotensin II (Ang II). Mitochondrial reactive oxygen species (ROS) and superoxide production were measured. Intracellular ATP levels were also detected. The cell proliferation rate was determined for the two groups. Protein production such as Anti-Bax, Bcl-2, COX IV and activation of c-jun N-terminal kinases signal (JNK) pathway was measured by enzyme-linked immunosorbent assay (ELISA) methods and Western blotting in this study. RESULTS Ang II treated cells showed significantly higher levels of superoxide production compared to the control group (p<0.05). Conversely, Ang II induced inhibitory effects on mitochondrial respiratory enzyme complexes, cause membrane potential dissipation, ATP loss and promote ROS generation, cell apoptosis in cultured osteoblasts. In addition, JNK phosphorylations were involved in activating the mitochondria-dependent apoptotic pathway following Ang II stimulation, as pre-treatment of JNK-specific inhibitor SP600125 could rescue osteoblast cells from apoptosis by enhancing the anti-apoptotic protein Bcl-2 expressions, suppressing the translocation of Bax from cytosol into mitochondria, blocking cytochrome C release and caspase-3 activation. CONCLUSIONS Ang II stimulates osteoblast apoptosis via suppression of the mitochondrial respiratory enzymes, membrane potential and cellular ATP productions. Clinical application with Ang II-stimulated osteoblast could be used for modelling or bone resorption in the oral region.
Collapse
|
19
|
Pan H, Shen K, Wang X, Meng H, Wang C, Jin B. Protective effect of metalloporphyrins against cisplatin-induced kidney injury in mice. PLoS One 2014; 9:e86057. [PMID: 24454954 PMCID: PMC3891880 DOI: 10.1371/journal.pone.0086057] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2013] [Accepted: 12/05/2013] [Indexed: 12/22/2022] Open
Abstract
Oxidative and nitrative stress is a well-known phenomenon in cisplatin-induced nephrotoxicity. The purpose of this work is to study the role of two metalloporphyrins (FeTMPyP and MnTBAP), water soluble complexes, in cisplatin-induced renal damage and their ability to scavenge peroxynitrite. In cisplatin-induced nephropathy study in mice, renal nitrative stress was evident by the increase in protein nitration. Cisplatin-induced nephrotoxicity was also evident by the histological damage from the loss of the proximal tubular brush border, blebbing of apical membranes, tubular epithelial cell detachment from the basement membrane, or intra-luminal aggregation of cells and proteins and by the increase in blood urea nitrogen and serum creatinine. Cisplatin-induced apoptosis and cell death as shown by Caspase 3 assessments, TUNEL staining and DNA fragmentation Cisplatin-induced nitrative stress, apoptosis and nephrotoxicity were attenuated by both metalloporphyrins. Heme oxygenase (HO-1) also plays a critical role in metalloporphyrin-mediated protection of cisplatin-induced nephrotoxicity. It is evident that nitrative stress plays a critical role in cisplatin-induced nephrotoxicity in mice. Our data suggest that peroxynitrite is involved, at least in part, in cisplatin-induced nephrotoxicity and protein nitration and cisplatin-induced nephrotoxicity can be prevented with the use of metalloporphyrins.
Collapse
Affiliation(s)
- Hao Pan
- Department of Urology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Kezhen Shen
- Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, Hangzhou, Zhejiang, China
| | - Xueping Wang
- Department of Urology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Hongzhou Meng
- Department of Urology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Chaojun Wang
- Department of Urology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Baiye Jin
- Department of Urology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- * E-mail:
| |
Collapse
|
20
|
Martino R, Sülsen V, Alonso R, Anesini C. A fraction rich in phenyl propanoids from L. divaricata aqueous extract is capable of inducing apoptosis, in relation to H2O2 modulation, on a murine lymphoma cell line. Leuk Res 2013; 37:1137-43. [DOI: 10.1016/j.leukres.2013.06.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2013] [Revised: 04/24/2013] [Accepted: 06/01/2013] [Indexed: 11/30/2022]
|
21
|
Aslan M, Dogan S, Kucuksayan E. Oxidative stress and potential applications of free radical scavengers in glaucoma. Redox Rep 2013; 18:76-87. [PMID: 23485101 DOI: 10.1179/1351000212y.0000000033] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Glaucoma is the leading cause of irreversible blindness in industrialized countries and comprises a group of diseases characterized by progressive optic nerve degeneration. Glaucoma is commonly associated with elevated intraocular pressure due to impaired outflow of aqueous humor resulting from abnormalities within the drainage system of the anterior chamber angle (open-angle glaucoma) or impaired access of aqueous humor to the drainage system (angle-closure glaucoma). Oxidative injury and altered antioxidant defense mechanisms in glaucoma appear to play a role in the pathophysiology of glaucomatous neurodegeneration that is characterized by death of retinal ganglion cells. Oxidative protein modifications occurring in glaucoma serve as immunostimulatory signals and alter neurosupportive and immunoregulatory functions of glial cells. Initiation of the apoptotic cascade observed in glaucomatous retinopathy can involve oxidant mechanisms and different agents have been shown to be neuroprotective. This review focuses on the molecular mechanisms of oxidant injury and summarizes studies that have investigated novel free radical scavengers in the treatment of glaucomatous neurodegeneration.
Collapse
Affiliation(s)
- Mutay Aslan
- Akdeniz University Medical School, Antalya, Turkey.
| | | | | |
Collapse
|
22
|
Rafikov R, Rafikova O, Aggarwal S, Gross C, Sun X, Desai J, Fulton D, Black SM. Asymmetric dimethylarginine induces endothelial nitric-oxide synthase mitochondrial redistribution through the nitration-mediated activation of Akt1. J Biol Chem 2012; 288:6212-26. [PMID: 23255608 DOI: 10.1074/jbc.m112.423269] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
We have recently demonstrated that asymmetric dimethylarginine (ADMA) induces the translocation of endothelial nitric-oxide synthase (eNOS) to the mitochondrion via a mechanism that requires protein nitration. Thus, the goal of this study was elucidate how eNOS redistributes to mitochondria and to identify the nitrated protein responsible for this event. Our data indicate that exposure of pulmonary arterial endothelial cells to ADMA enhanced eNOS phosphorylation at the Akt1-dependent phosphorylation sites Ser(617) and Ser(1179). Mutation of these serine residues to alanine (S617A and S1179A) inhibited nitration-mediated eNOS translocation to the mitochondria, whereas the phosphormimic mutations (S617D and S1179D) exhibited increased mitochondrial redistribution in the absence of ADMA. The overexpression of a dominant-negative Akt1 also attenuated ADMA-mediated eNOS mitochondrial translocation. Furthermore, ADMA enhanced Akt1 nitration and increased its activity. Mass spectrometry identified a single nitration site in Akt1 located at the tyrosine residue (Tyr(350)) located within the client-binding domain. Replacement of Tyr(350) with phenylalanine abolished peroxynitrite-mediated eNOS translocation to mitochondria. We also found that in the absence of ADMA, eNOS translocation decreased mitochondrial oxygen consumption and superoxide production without altering cellular ATP level. This suggests that under physiologic conditions, eNOS translocation enhances mitochondria coupling. In conclusion, we have identified a new mechanism by which eNOS translocation to mitochondria is regulated by the phosphorylation of eNOS at Ser(617) and Ser(1179) by Akt1 and that this is enhanced when Akt1 becomes nitrated at Tyr(350).
Collapse
Affiliation(s)
- Ruslan Rafikov
- Pulmonary Disease Program, Vascular Biology Center, Georgia Health Sciences University, Augusta, Georgia 30912, USA
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Antico Arciuch VG, Elguero ME, Poderoso JJ, Carreras MC. Mitochondrial regulation of cell cycle and proliferation. Antioxid Redox Signal 2012; 16:1150-80. [PMID: 21967640 PMCID: PMC3315176 DOI: 10.1089/ars.2011.4085] [Citation(s) in RCA: 313] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2011] [Revised: 10/03/2011] [Accepted: 10/03/2011] [Indexed: 01/01/2023]
Abstract
Eukaryotic mitochondria resulted from symbiotic incorporation of α-proteobacteria into ancient archaea species. During evolution, mitochondria lost most of the prokaryotic bacterial genes and only conserved a small fraction including those encoding 13 proteins of the respiratory chain. In this process, many functions were transferred to the host cells, but mitochondria gained a central role in the regulation of cell proliferation and apoptosis, and in the modulation of metabolism; accordingly, defective organelles contribute to cell transformation and cancer, diabetes, and neurodegenerative diseases. Most cell and transcriptional effects of mitochondria depend on the modulation of respiratory rate and on the production of hydrogen peroxide released into the cytosol. The mitochondrial oxidative rate has to remain depressed for cell proliferation; even in the presence of O₂, energy is preferentially obtained from increased glycolysis (Warburg effect). In response to stress signals, traffic of pro- and antiapoptotic mitochondrial proteins in the intermembrane space (B-cell lymphoma-extra large, Bcl-2-associated death promoter, Bcl-2 associated X-protein and cytochrome c) is modulated by the redox condition determined by mitochondrial O₂ utilization and mitochondrial nitric oxide metabolism. In this article, we highlight the traffic of the different canonical signaling pathways to mitochondria and the contributions of organelles to redox regulation of kinases. Finally, we analyze the dynamics of the mitochondrial population in cell cycle and apoptosis.
Collapse
Affiliation(s)
| | - María Eugenia Elguero
- Laboratory of Oxygen Metabolism, University of Buenos Aires, University Hospital, Buenos Aires, Argentina
| | - Juan José Poderoso
- Laboratory of Oxygen Metabolism, University of Buenos Aires, University Hospital, Buenos Aires, Argentina
- Department of Internal Medicine, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
- CONICET, Buenos Aires, Argentina
| | - María Cecilia Carreras
- Laboratory of Oxygen Metabolism, University of Buenos Aires, University Hospital, Buenos Aires, Argentina
- CONICET, Buenos Aires, Argentina
- Department of Clinical Biochemistry, INFIBIOC and School of Pharmacy and Biochemistry, University of Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
24
|
Gabr SA, Al-Ghadir AH. Role of cellular oxidative stress and cytochrome c in the pathogenesis of psoriasis. Arch Dermatol Res 2012; 304:451-7. [PMID: 22421888 DOI: 10.1007/s00403-012-1230-8] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2011] [Revised: 03/03/2012] [Accepted: 03/06/2012] [Indexed: 12/11/2022]
Abstract
Oxidative-free radicals and apoptosis have linked to chronic skin diseases. Higher levels of oxidative radicals and the release of mitochondrial cytochrome c may have a role in the pathogenesis of psoriasis. We investigated the possible role of cellular oxidative stress and release of cytochrome c of mitochondria in the pathogenesis of psoriasis. Disease severity was assessed by psoriasis area severity index score (PASI) of 55 psoriasis patients, they grouped as mild (11), moderate (20) and severe (24), also 20 healthy individuals used as controls. All groups were subjected for serum malondialdehyde (MDA), nitric oxide (NO·), superoxide dismutase (SOD), catalase (CAT), total antioxidant status (TAS) and serum cytochrome c concentrations. We found that, (1) Severity wise increase in MDA and NO·, and decrease in SOD, CAT and TAS levels in all patients with different degrees of psoriasis; (2) PASI showed positive correlation with the increase in MDA and NO·, and negatively with decreased SOD, CAT and TAS levels; (3) significant increase in cytochrome c level was observed among psoriasis patients which showed negative correlation to MDA and NO· levels in mild and positively with moderate and severe groups. The release of mitochondrial cytochrome c indicates the induction of apoptosis mediated via oxidative stress which ultimately plays role in the pathogenesis of psoriasis.
Collapse
Affiliation(s)
- Sami A Gabr
- Department of Anatomy, Faculty of Medicine, Mansoura University, Egypt.
| | | |
Collapse
|
25
|
Martínez-Ruiz A, Cadenas S, Lamas S. Nitric oxide signaling: classical, less classical, and nonclassical mechanisms. Free Radic Biol Med 2011; 51:17-29. [PMID: 21549190 DOI: 10.1016/j.freeradbiomed.2011.04.010] [Citation(s) in RCA: 237] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2011] [Revised: 03/10/2011] [Accepted: 04/04/2011] [Indexed: 12/20/2022]
Abstract
Although nitric oxide (NO) was identified more than 150 years ago and its effects were clinically tested in the form of nitroglycerine, it was not until the decades of 1970-1990 that it was described as a gaseous signal transducer. Since then, a canonical pathway linked to cyclic GMP (cGMP) as its quintessential effector has been established, but other modes of action have emerged and are now part of the common body of knowledge within the field. Classical (or canonical) signaling involves the selective activation of soluble guanylate cyclase, the generation of cGMP, and the activation of specific kinases (cGMP-dependent protein kinases) by this cyclic nucleotide. Nonclassical signaling alludes to the formation of NO-induced posttranslational modifications (PTMs), especially S-nitrosylation, S-glutathionylation, and tyrosine nitration. These PTMs are governed by specific biochemical mechanisms as well as by enzymatic systems. In addition, a less classical but equally important pathway is related to the interaction between NO and mitochondrial cytochrome c oxidase, which might have important implications for cell respiration and intermediary metabolism. Cross talk trespassing these necessarily artificial conceptual boundaries is progressively being identified and hence an integrated systems biology approach to the comprehension of NO function will probably emerge in the near future.
Collapse
Affiliation(s)
- Antonio Martínez-Ruiz
- Servicio de Inmunología, Hospital Universitario de la Princesa, Instituto de Investigación Sanitaria Princesa (IP), Madrid, Spain
| | | | | |
Collapse
|
26
|
Herbst S, Schaible UE, Schneider BE. Interferon gamma activated macrophages kill mycobacteria by nitric oxide induced apoptosis. PLoS One 2011; 6:e19105. [PMID: 21559306 PMCID: PMC3085516 DOI: 10.1371/journal.pone.0019105] [Citation(s) in RCA: 179] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2010] [Accepted: 03/17/2011] [Indexed: 02/03/2023] Open
Abstract
Mycobacterium tuberculosis is an intracellular pathogen of macrophages and escapes the macrophages' bactericidal effectors by interfering with phagosome-lysosome fusion. IFN-γ activation renders the macrophages capable of killing intracellular mycobacteria by overcoming the phagosome maturation block, nutrient deprivation and exposure to microbicidal effectors including nitric oxide (NO). While the importance about NO for the control of mycobacterial infection in murine macrophages is well documented, the underlying mechanism has not been revealed yet. In this study we show that IFN-γ induced apoptosis in mycobacteria-infected macrophages, which was strictly dependent on NO. Subsequently, NO-mediated apoptosis resulted in the killing of intracellular mycobacteria independent of autophagy. In fact, killing of mycobacteria was susceptible to the autophagy inhibitor 3-methyladenine (3-MA). However, 3-MA also suppressed NO production, which is an important off-target effect to be considered in autophagy studies using 3-MA. Inhibition of caspase 3/7 activation, as well as NO production, abolished apoptosis and elimination of mycobacteria by IFN-γ activated macrophages. In line with the finding that drug-induced apoptosis kills intracellular mycobacteria in the absence of NO, we identified NO-mediated apoptosis as a new defense mechanism of activated macrophages against M. tuberculosis.
Collapse
Affiliation(s)
- Susanne Herbst
- Department of Immunology and Infection, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Ulrich E. Schaible
- Department of Immunology and Infection, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, United Kingdom
- Department of Molecular Infection Biology, Research Center Borstel, Borstel, Germany
- * E-mail:
| | - Bianca E. Schneider
- Department of Immunology and Infection, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, United Kingdom
- Department of Molecular Infection Biology, Research Center Borstel, Borstel, Germany
| |
Collapse
|
27
|
Manuele MG, Barreiro Arcos ML, Davicino R, Ferraro G, Cremaschi G, Anesini C. Limonene exerts antiproliferative effects and increases nitric oxide levels on a lymphoma cell line by dual mechanism of the ERK pathway: relationship with oxidative stress. Cancer Invest 2010; 28:135-45. [PMID: 19968502 DOI: 10.3109/07357900903179583] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
D-limonene is a common monoterpene found in nature. Previously, it has been demonstrated that it has antiproliferative effects on a lymphoma cell line by increasing the nitric oxide (NO) level. In the present work this mechanism is analyzed by evaluating the participation of MAP38 and ERK pathways. Limonene increased the NO levels by inducing cell apoptosis by two mechanisms: through the production of H(2)O(2) and ERK pathway activation at low concentrations, and through the inhibition of farnesylation of proteins and O(2)- production at high concentrations. Both mechanisms were related to the increase in total nitrites, and the production of oxidative stress intermediates was involved.
Collapse
|
28
|
Saha B, Jyothi Prasanna S, Chandrasekar B, Nandi D. Gene modulation and immunoregulatory roles of interferon gamma. Cytokine 2009; 50:1-14. [PMID: 20036577 DOI: 10.1016/j.cyto.2009.11.021] [Citation(s) in RCA: 231] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2009] [Revised: 11/02/2009] [Accepted: 11/24/2009] [Indexed: 01/19/2023]
Abstract
Interferon-gamma (IFNgamma) is a central regulator of the immune response and signals via the Janus Activated Kinase (JAK)-Signal Transducer and Activator of Transcription (STAT) pathway. Phosphorylated STAT1 homodimers translocate to the nucleus, bind to Gamma Activating Sequence (GAS) and recruit additional factors to modulate gene expression. A bioinformatics analysis revealed that greater number of putative promoters of immune related genes and also those not directly involved in immunity contain GAS compared to response elements (RE) for Interferon Regulatory Factor (IRF)1, Nuclear factor kappa B (NFkappaB) and Activator Protein (AP)1. GAS is present in putative promoters of well known IFNgamma-induced genes, IRF1, GBP1, CXCL10, and other genes identified were TLR3, VCAM1, CASP4, etc. Analysis of three microarray studies revealed that the expression of a subset of only GAS containing immune genes were modulated by IFNgamma. As a significant correlation exists between GAS containing immune genes and IFNgamma-regulated gene expression, this strategy may identify novel IFNgamma-responsive immune genes. This analysis is integrated with the literature on the roles of IFNgamma in mediating a plethora of functions: anti-microbial responses, antigen processing, inflammation, growth suppression, cell death, tumor immunity and autoimmunity. Overall, this review summarizes our present knowledge on IFNgamma mediated signaling and functions.
Collapse
Affiliation(s)
- Banishree Saha
- Department of Biochemistry, Indian Institute of Science, Bangalore, India
| | | | | | | |
Collapse
|
29
|
Davicino R, Manuele MG, Turner S, Ferraro G, Anesini C. Antiproliferative Activity ofLarrea Divaricata Cav.on Lymphoma Cell Line: Participation of Hydrogen Peroxide in Its Action. Cancer Invest 2009; 28:13-22. [DOI: 10.3109/07357900902849665] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
30
|
Liu HL, Xu JJ, Dai XM, Shi JB, Xu S, Gao J, Yao QZ, Liu F. Both DNA damage and mitochondrial dysfunction are involved in novel oxadiazolo[3,4-d]pyrimidine nucleoside derivatives-induced cancer cell death. J Appl Toxicol 2009; 29:489-95. [DOI: 10.1002/jat.1433] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
31
|
Davicino R, Manuele MG, Ferraro G, Micalizzi B, Anesini C. Modulatory effect of hydrogen peroxide on tumoral lymphocytes proliferation. Immunopharmacol Immunotoxicol 2009; 31:130-9. [DOI: 10.1080/08923970802520701] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
32
|
Lee ST, Wu TT, Yu PY, Chen RM. Apoptotic insults to human HepG2 cells induced by S-(+)-ketamine occurs through activation of a Bax-mitochondria-caspase protease pathway. Br J Anaesth 2008; 102:80-9. [PMID: 19001360 DOI: 10.1093/bja/aen322] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Ketamine is widely used as an i.v. anaesthetic agent and as a drug of abuse. Hepatocytes contribute to the metabolism of endogenous and exogenous substances. This study evaluated the toxic effects of S-(+)-ketamine and possible mechanisms using human hepatoma HepG2 cells as the experimental model. METHODS HepG2 cells were exposed to S-(+)-ketamine. Cell viability and the release of lactate dehydrogenase (LDH) and gamma-glutamyl transpeptidase (GPT) were measured to determine the toxicity of S-(+)-ketamine to HepG2 cells. Cell morphology, DNA fragmentation, and apoptotic cells were analysed to evaluate the mechanism of S-(+)-ketamine-induced cell death. Amounts of Bax, an apoptotic protein, and cytochrome c in the cytoplasm or mitochondria were quantified by immunoblotting. Cellular adenosine triphosphate levels were analysed using a bioluminescence assay. Caspases-3, -9, and -6 were measured fluorometrically. RESULTS Exposure of HepG2 cells to S-(+)-ketamine increased the release of LDH and GPT, but decreased cell viability (all P<0.01). S-(+)-Ketamine time-dependently caused shrinkage of HepG2 cells. Exposure to S-(+)-ketamine led to significant DNA fragmentation and cell apoptosis (P=0.003 and 0.002). S-(+)-Ketamine increased translocation of Bax from the cytoplasm to mitochondria, but decreased the mitochondrial membrane potential and cellular adenosine triphosphate levels (all P<0.01). Sequentially, cytosolic cytochrome c levels and activities of caspases-9, -3, and -6 were augmented after S-(+)-ketamine administration (all P<0.001). Z-VEID-FMK, an inhibitor of caspase-6, alleviated the S-(+)-ketamine-induced augmentation of caspase-6 activity, DNA fragmentation, and cell apoptosis (all P<0.001). CONCLUSIONS This study shows that S-(+)-ketamine can induce apoptotic insults to human HepG2 cells via a Bax-mitochondria-caspase protease pathway. Thus, we suggest that S-(+)-ketamine at a clinically relevant or an abused concentration may induce liver dysfunction possibly due to its toxicity to hepatocytes.
Collapse
Affiliation(s)
- S-T Lee
- Department of Pediatrics, Cathay General Hospital, Taipei, Taiwan, Republic of China
| | | | | | | |
Collapse
|
33
|
Cherng YG, Chang HC, Lin YL, Kuo ML, Chiu WT, Chen RM. Apoptotic insults to human chondrocytes induced by sodium nitroprusside are involved in sequential events, including cytoskeletal remodeling, phosphorylation of mitogen-activated protein kinase kinase kinase-1/c-Jun N-terminal kinase, and Bax-mitochondria-mediated caspase activation. J Orthop Res 2008; 26:1018-26. [PMID: 18306405 DOI: 10.1002/jor.20578] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Nitric oxide (NO) can regulate chondrocyte activities. This study was aimed to evaluate the molecular mechanisms of NO donor sodium nitroprusside (SNP)-induced insults to human chondrocytes. Exposure of human chondrocytes to SNP increased cellular NO levels but decreased cell viability in concentration- and time-dependent manners. SNP time dependently induced DNA fragmentation and cell apoptosis. Treatment with 2-phenyl-4,4,5,5-tetramethyl-imidazoline-1-oxyl 3-oxide, an NO scavenger, significantly lowered SNP-induced cell injuries. Administration of SNP interrupted F-actin and microtubule cytoskeletons and stimulated phosphorylation of mitogen-activated protein kinase kinase kinase-1 (MEKK1) and c-Jun N-terminal kinase (JNK). Similar to SNP, cytochalasin D, an inhibitor of F-actin formation, disturbed F-actin polymerization and increased MEKK1 and JNK activations. Overexpression of a dominant negative mutant of MEKK1 (dnMEK1) in human chondrocytes significantly ameliorated SNP-induced cell apoptosis. Exposure to SNP promoted Bax translocation from the cytoplasm to mitochondria, but application of dnMEKK1 lowered the translocation. SNP time dependently decreased the mitochondrial membrane potential, complex I NADH dehydrogenase activity, and cellular ATP levels, but increased the release of cytochrome c from mitochondria to the cytoplasm. Activities of caspase-9, -3, and -6 were sequentially increased by SNP administration. This study shows that SNP can induce apoptosis of human chondrocytes through sequential events, including cytoskeletal remodeling, activation of MEKK1/JNK, Bax translocation, mitochondrial dysfunction, cytochrome c release, caspase activation, and DNA fragmentation.
Collapse
Affiliation(s)
- Yih-Giun Cherng
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, No. 250 Wu-Hsing St., Taipei 110, Taiwan, Republic of China
| | | | | | | | | | | |
Collapse
|
34
|
Leon L, Jeannin JF, Bettaieb A. Post-translational modifications induced by nitric oxide (NO): implication in cancer cells apoptosis. Nitric Oxide 2008; 19:77-83. [PMID: 18474258 DOI: 10.1016/j.niox.2008.04.014] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2008] [Revised: 04/16/2008] [Accepted: 04/16/2008] [Indexed: 11/28/2022]
Abstract
Post-translational modifications of proteins can regulate the balance between survival and cell death signals. It is increasingly recognized that nitric oxide (NO) and reactive oxygen species (ROS)-induced post-translational modifications could play a role in cell death. This review provides an introduction of current knowledge of NO proteins modifications promoting or inhibiting cell death with special attention in cancer cells.
Collapse
Affiliation(s)
- Lissbeth Leon
- EPHE, Laboratoire d'immunologie et immunothérapie des cancers, Inserm U866, Dijon, F-21000, France.
| | | | | |
Collapse
|
35
|
Wu GJ, Chen TG, Chang HC, Chiu WT, Chang CC, Chen RM. Nitric oxide from both exogenous and endogenous sources activates mitochondria-dependent events and induces insults to human chondrocytes. J Cell Biochem 2008; 101:1520-31. [PMID: 17492650 DOI: 10.1002/jcb.21268] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
During inflammation, overproduction of nitric oxide (NO) can damage chondrocytes. In this study, we separately evaluated the toxic effects of exogenous and endogenous NO on human chondrocytes and their possible mechanisms. Human chondrocytes were exposed to sodium nitroprusside (SNP), an NO donor, or a combination of lipopolysaccharide (LPS) and interferon-gamma (IFN-gamma) as the exogenous and endogenous sources of NO, respectively. Administration of SNP or a combination of LPS and IFN-gamma in human chondrocytes increased cellular NO levels but decreased cell viability. Exposure to exogenous or endogenous NO significantly induced apoptosis of human chondrocytes. When treated with exogenous or endogenous NO, the mitochondrial membrane potential time-dependently decreased. Exposure to exogenous or endogenous NO significantly enhanced cellular reactive oxygen species (ROS) and cytochrome c (Cyt c) levels. Administration of exogenous or endogenous NO increased caspase-3 activity and consequently induced DNA fragmentation. Suppression of caspase-3 activation by Z-DEVD-FMK decreased NO-induced DNA fragmentation and cell apoptosis. Similar to SNP, exposure of human chondrocytes to S-nitrosoglutathione (GSNO), another NO donor, caused significant increases in Cyt c levels, caspase-3 activity, and DNA fragmentation, and induced cell apoptosis. Pretreatment with N-monomethyl arginine (NMMA), an inhibitor of NO synthase, significantly decreased cellular NO levels, and lowered endogenous NO-induced alterations in cellular Cyt c amounts, caspase-3 activity, DNA fragmentation, and cell apoptosis. Results of this study show that NO from exogenous and endogenous sources can induce apoptotic insults to human chondrocytes via a mitochondria-dependent mechanism.
Collapse
Affiliation(s)
- Gong-Jhe Wu
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | | | | | | | | | | |
Collapse
|
36
|
Parihar MS, Parihar A, Chen Z, Nazarewicz R, Ghafourifar P. mAtNOS1 regulates mitochondrial functions and apoptosis of human neuroblastoma cells. Biochim Biophys Acta Gen Subj 2008; 1780:921-6. [PMID: 18359297 DOI: 10.1016/j.bbagen.2008.02.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2008] [Revised: 02/19/2008] [Accepted: 02/19/2008] [Indexed: 01/11/2023]
Abstract
mAtNOS1 is a novel gene recently reported in mammalian cells with functions that are not fully understood. The present study generated human neuroblastoma SHSY cells over- and underexpressing mAtNOS1 and shows that mAtNOS1 is involved in regulating mitochondrial nitric oxide, mitochondrial transmembrane potential, protein tyrosine nitration, cytochrome c release, and apoptosis of those cells.
Collapse
Affiliation(s)
- Mordhwaj S Parihar
- Department of Surgery, Davis Heart & Lung Research Institute, The Ohio State University, Columbus, Ohio, USA
| | | | | | | | | |
Collapse
|
37
|
Zeini M, López-Fontal R, Través PG, Benito G, Hortelano S. Differential sensitivity to apoptosis among the cells that contribute to the atherosclerotic disease. Biochem Biophys Res Commun 2007; 363:444-50. [PMID: 17884013 DOI: 10.1016/j.bbrc.2007.09.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2007] [Accepted: 09/05/2007] [Indexed: 12/22/2022]
Abstract
Apoptosis plays an important role in a great number of pathological processes, including atherosclerotic disease. Although apoptosis occurs in the major cell types found in atherosclerotic lesions (e.g. macrophages, endothelial cells, and smooth muscle cells), the mechanism involved in this process differs depending on the stage, the localization and the cell composition of the plaque. In this study, we have compared the effects of different apoptotic inducers on the cells that form the atherosclerotic plaque. We have demonstrated that monocytes and macrophages are more susceptible to apoptosis than smooth muscle cells and endothelial cells. These findings provide insights about the potential role of apoptosis in the atherosclerotic disease and suggest strategies to treat vascular diseases by exploiting the differential sensitivity of cells to cell death.
Collapse
Affiliation(s)
- Miriam Zeini
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University, School of Medicine, Stanford, CA 94305, USA
| | | | | | | | | |
Collapse
|
38
|
Voloboueva LA, Suh SW, Swanson RA, Giffard RG. Inhibition of mitochondrial function in astrocytes: implications for neuroprotection. J Neurochem 2007; 102:1383-94. [PMID: 17488276 PMCID: PMC3175820 DOI: 10.1111/j.1471-4159.2007.04634.x] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Much evidence suggests that astrocytes protect neurons against ischemic injury. Although astrocytes are more resistant to some insults than neurons, few studies offer insight into the real time changes of astrocytic protective functions with stress. Mitochondria are one of the primary targets of ischemic injury in astrocytes. We investigated the time course of changes in astrocytic ATP levels, plasma membrane potential, and glutamate uptake, a key protective function, induced by mitochondrial inhibition. Our results show that significant functional change precedes reduction in astrocytic viability with mitochondrial inhibition. Using the mitochondrial inhibitor fluorocitrate (FC, 0.25 mmol/L) that is preferentially taken by astrocytes we found that inhibition of astrocyte mitochondria increased vulnerability of co-cultured neurons to glutamate toxicity. In our studies, the rates of FC-induced astrocytic mitochondrial depolarization were accelerated in mixed astrocyte/neuron cultures. We hypothesized that the more rapid mitochondrial depolarization was promoted by an additional energetic demand imposed be the co-cultured neurons. To test this hypothesis, we exposed pure astrocytic cultures to 0.01-1 mmol/L aspartate as a metabolic load. Aspartate application accelerated the rates of FC-induced mitochondrial depolarization, and, at 1 mmol/L, induced astrocytic death, suggesting that strong energetic demands during ischemia can compromise astrocytic function and viability.
Collapse
Affiliation(s)
- Ludmila A Voloboueva
- Department of Anesthesia, Stanford University School of Medicine, Stanford, California 94305, USA
| | | | | | | |
Collapse
|
39
|
de las Heras B, Hortelano S, Girón N, Bermejo P, Rodríguez B, Boscá L. Kaurane diterpenes protect against apoptosis and inhibition of phagocytosis in activated macrophages. Br J Pharmacol 2007; 152:249-55. [PMID: 17618303 PMCID: PMC1978262 DOI: 10.1038/sj.bjp.0707382] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND AND PURPOSE The kaurane diterpenes foliol and linearol are inhibitors of the activation of nuclear factor kappaB, a transcription factor involved in the inflammatory response. Effects of these diterpenes on apoptosis and phagocytosis have been analysed in cultured peritoneal macrophages and in the mouse macrophage cell line, RAW 264.7. EXPERIMENTAL APPROACH Macrophages were maintained in culture and activated with pro-inflammatory stimuli in the absence or presence of diterpenes. Apoptosis and the phagocytosis in these cells under these conditions were determined. KEY RESULTS Incubation of macrophages with a mixture of bacterial lipopolysaccharide (LPS)/interferon-gamma (IFN-gamma) induced apoptosis through a NO-dependent pathway, an effect significantly inhibited by foliol and linearol in the low muM range, without cytotoxic effects. Apoptosis in macrophages induced by NO donors was also inhibited. The diterpenes prevented apoptosis through a mechanism compatible with the inhibition of caspase-3 activation, release of cytochrome c to the cytosol and p53 overexpression, as well as an alteration in the levels of proteins of the Bcl-2 family, in particular, the levels of Bax. Cleavage of poly(ADP-ribose) polymerase, a well-established caspase substrate, was reduced by these diterpenes. Treatment of cells with foliol and linearol decreased phagocytosis of zymosan bioparticles by RAW 264.7 cells and to a greater extent by peritoneal macrophages. CONCLUSIONS AND IMPLICATIONS Both diterpenes protected macrophages from apoptosis and inhibited phagocytosis, resulting in a paradoxical control of macrophage function, as viability was prolonged but inflammatory and phagocytic functions were impaired.
Collapse
Affiliation(s)
- B de las Heras
- Departamento de Farmacología Facultad de Farmacia, Universidad Complutense Madrid, Spain
| | - S Hortelano
- CNIC, Melchor Fernández Almagro 3 Madrid, Spain
| | - N Girón
- Departamento de Farmacología Facultad de Farmacia, Universidad Complutense Madrid, Spain
| | - P Bermejo
- Departamento de Farmacología Facultad de Farmacia, Universidad Complutense Madrid, Spain
| | - B Rodríguez
- Instituto de Química Orgánica (CSIC), Juan de la Cierva 3 Madrid, Spain
| | - L Boscá
- CNIC, Melchor Fernández Almagro 3 Madrid, Spain
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Arturo Duperier 4 Madrid, Spain
- Author for correspondence:
| |
Collapse
|
40
|
Voloboueva LA, Suh SW, Swanson RA, Giffard RG. Inhibition of mitochondrial function in astrocytes: implications for neuroprotection. J Neurochem 2007. [PMID: 17488276 DOI: 10.1111/j.1471-4159.2007.4634.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Much evidence suggests that astrocytes protect neurons against ischemic injury. Although astrocytes are more resistant to some insults than neurons, few studies offer insight into the real time changes of astrocytic protective functions with stress. Mitochondria are one of the primary targets of ischemic injury in astrocytes. We investigated the time course of changes in astrocytic ATP levels, plasma membrane potential, and glutamate uptake, a key protective function, induced by mitochondrial inhibition. Our results show that significant functional change precedes reduction in astrocytic viability with mitochondrial inhibition. Using the mitochondrial inhibitor fluorocitrate (FC, 0.25 mmol/L) that is preferentially taken by astrocytes we found that inhibition of astrocyte mitochondria increased vulnerability of co-cultured neurons to glutamate toxicity. In our studies, the rates of FC-induced astrocytic mitochondrial depolarization were accelerated in mixed astrocyte/neuron cultures. We hypothesized that the more rapid mitochondrial depolarization was promoted by an additional energetic demand imposed be the co-cultured neurons. To test this hypothesis, we exposed pure astrocytic cultures to 0.01-1 mmol/L aspartate as a metabolic load. Aspartate application accelerated the rates of FC-induced mitochondrial depolarization, and, at 1 mmol/L, induced astrocytic death, suggesting that strong energetic demands during ischemia can compromise astrocytic function and viability.
Collapse
Affiliation(s)
- Ludmila A Voloboueva
- Department of Anesthesia, Stanford University School of Medicine, Stanford, California 94305, USA
| | | | | | | |
Collapse
|
41
|
Chen JY, Mak NK, Yow CMN, Fung MC, Chiu LC, Leung WN, Cheung NH. The Binding Characteristics and Intracellular Localization of Temoporfin (mTHPC) in Myeloid Leukemia Cells: Phototoxicity and Mitochondrial Damage¶. Photochem Photobiol 2007. [DOI: 10.1562/0031-8655(2000)0720541tbcail2.0.co2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
42
|
Carreras MC, Poderoso JJ. Mitochondrial nitric oxide in the signaling of cell integrated responses. Am J Physiol Cell Physiol 2007; 292:C1569-80. [PMID: 17496232 DOI: 10.1152/ajpcell.00248.2006] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Mitochondria are the specialized organelles for energy metabolism, but, as a typical example of system biology, they also activate a multiplicity of pathways that modulate cell proliferation and mitochondrial biogenesis or oppositely promote cell arrest and programmed cell death by a limited number of oxidative or nitrosative reactions. These reactions are influenced by matrix nitric oxide (NO) steady-state concentration, either from local production or by gas diffusion to mitochondria from the canonical sources. Likewise, in a range of ∼30–200 nM, NO turns mitochondrial O2utilization down by binding to cytochrome oxidase and elicits a burst of superoxide anion and hydrogen peroxide that diffuses outside mitochondria. Depending on NO levels and antioxidant defenses, more or less H2O2accumulates in cytosol and nucleus, and the resulting redox grading contributes to dual activation of proliferating and proapoptotic cascades, like ERK1/2 or p38 MAPK. Moreover, these sequential activating pathways participate in rat liver and brain development and in thyroid modulation of mitochondrial metabolism and contribute to hypothyroid phenotype through complex I nitration. On the contrary, lack of NO disrupts pathways like S-nitrosylation or H2O2production and likewise is a gateway to disease in amyotrophic lateral sclerosis with superoxide dismutase 1 mutations or to cancer proliferation.
Collapse
Affiliation(s)
- Maria Cecilia Carreras
- Laboratory of Oxygen Metabolism, University Hospital of Buenos Aires, Cordoba 2351, 1120 Buenos Aires, Argentina.
| | | |
Collapse
|
43
|
Holzmuller P, Bras-Gonçalves R, Lemesre JL. Phenotypical characteristics, biochemical pathways, molecular targets and putative role of nitric oxide-mediated programmed cell death in Leishmania. Parasitology 2007; 132 Suppl:S19-32. [PMID: 17018162 DOI: 10.1017/s0031182006000837] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Nitric oxide (NO) has been demonstrated to be the principal effector molecule mediating intracellular killing of Leishmania, both in vitro and in vivo. We investigated the type of cell death process induced by NO for the intracellular amastigote stage of the protozoa Leishmania. Specific detection methods revealed a rapid and extensive cell death with morphological features of apoptosis in axenic amastigotes exposed to NO donors, in intracellular amastigotes inside in vitro - activated mouse macrophages and also in activated macrophages of regressive lesions in a leishmaniasis-resistant mouse model. We extended our investigations to the dog, a natural host-reservoir of Leishmania parasites, by demonstrating that co-incubation of infected macrophages with autologous lymphocytes derived from dogs immunised with purified excreted-secreted antigens of Leishmania resulted in a significant NO-mediated apoptotic cell death of intracellular amastigotes. From the biochemical point of view, NO-mediated Leishmania amastigotes apoptosis did not seem to be controlled by caspase activity as indicated by the lack of effect of cell permeable inhibitors of caspases and cysteine proteases, in contrast to specific proteasome inhibitors, such as lactacystin or calpain inhibitor I. Moreover, addition of the products of two NO molecular targets, cis-aconitase and glyceraldehyde-3-phosphate dehydrogenase, also had an inhibitory effect on the cell death induced by NO. Interestingly, activities of these two enzymes plus 6-phosphogluconate dehydrogenase, parasitic enzymes involved in both glycolysis and respiration processes, are overexpressed in amastigotes selected for their NO resistance. This review focuses on cell death of the intracellular stage of the pathogen Leishmania induced by nitrogen oxides and gives particular attention to the biochemical pathways and the molecular targets potentially involved. Questions about the role of Leishmania amastigotes NO-mediated apoptosis in the overall infection process are raised and discussed.
Collapse
Affiliation(s)
- P Holzmuller
- Equipe 1 Rôle biologique des facteurs d'excrétion-sécrétion des leishmanies: intérêt diagnostique et immunoprophylactique, UR 008 Pathogénie des Trypanosomatidae, IRD, B.P. 64501, 911 avenue Agropolis, 34394 Montpellier cedex 5, France
| | | | | |
Collapse
|
44
|
An HJ, Maeng O, Kang KH, Lee JO, Kim YS, Paik SG, Lee H. Activation of Ras Up-regulates Pro-apoptotic BNIP3 in Nitric Oxide-induced Cell Death. J Biol Chem 2006; 281:33939-48. [PMID: 16954213 DOI: 10.1074/jbc.m605819200] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Nitric oxide (NO) produced by NO synthases causes nitration and nitrosylation of cellular factors. We have shown previously that endogenously produced or exogenously added NO induces expression of BNIP3 (Bcl-2/adenovirus E1B 19 kDa-interacting protein 3), leading to death of macrophages (Yook, Y.-H., Kang, K.-H., Maeng, O., Kim, T.-R., Lee, J.-O., Kang, K.-i., Kim, Y.-S., Paik, S.-G., and Lee, H. (2004) Biochem. Biophys. Res. Commun. 321, 298-305). We now provide evidence that Ras mediates NO-induced BNIP3 expression via the MEK/ERK/hypoxia-inducible factor (HIF)-1 pathway. (a) ras-Q61L, a constitutively active form of Ras, up-regulated BNIP3 protein expression by enhancing Bnip3 promoter activity, and ras-S17N, a dominant-negative form, and ras-C118S, an S-nitrosylation mutant, blocked NO-induced BNIP3 expression, suggesting that Ras acts downstream of NO and that NO activates Ras by nitrosylation. (b) U0126, a specific MEK inhibitor, completely abolished BNIP3 expression and the stimulation of promoter activity by NO and Ras, whereas 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one, SB203580, and wortmannin, specific inhibitors of soluble guanylyl cyclase, p38 MAPK, and phosphatidylinositol 3-kinase, respectively, had no effect. Ras, MEK1/2, and ERK1/2 were sequentially activated by NO treatment of macrophages. (c) Mutation of the HIF-1-binding site (hypoxia-response element) in the Bnip3 promoter abolished BNIP3 induction, and HIF-1alpha was strongly induced by NO. (d) Transient expression of activated Ras promoted macrophage death, as did NO, and this Ras-mediated cell death was inhibited by silencing BNIP3 expression. These results suggest that NO-induced death of macrophages is mediated, at least in part, by BNIP3 induction.
Collapse
Affiliation(s)
- Hyun-Jung An
- Department of Biology, School of Biosciences and Biotechnology, Chungnam National University, Daejeon, Korea
| | | | | | | | | | | | | |
Collapse
|
45
|
Chang CC, Liao YS, Lin YL, Chen RM. Nitric oxide protects osteoblasts from oxidative stress-induced apoptotic insults via a mitochondria-dependent mechanism. J Orthop Res 2006; 24:1917-25. [PMID: 16917919 DOI: 10.1002/jor.20244] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Nitric oxide (NO) contributes to the regulation of osteoblast activities. In this study, we evaluated the protective effects of NO pretreatment on oxidative stress-induced osteoblast apoptosis and its possible mechanism using neonatal rat calvarial osteoblasts as the experimental model. Exposure of osteoblasts to sodium nitroprusside (SNP) at a low concentration of 0.3 mM significantly increased cellular NO levels without affecting cell viability. However, when the concentration reached a high concentration of 2 mM, SNP increased the levels of intracellular reactive oxygen species and induced osteoblast injuries. Thus, administration of 0.3 and 2 mM SNP in osteoblasts were respectively used as sources of NO and oxidative stress. Pretreatment with NO for 24 h significantly ameliorated the oxidative stress-caused morphological alterations and decreases in alkaline phosphatase activity, and reduced cell death. Oxidative stress induced osteoblast death via an apoptotic mechanism, but NO pretreatment protected osteoblasts against the toxic effects. The mitochondrial membrane potential was significantly reduced following exposure to the oxidative stress. However, pretreatment with NO significantly lowered the suppressive effects. Oxidative stress increased cellular Bax protein production and cytochrome c release from mitochondria. Pretreatment with NO significantly decreased oxidative stress-caused augmentation of Bax and cytochrome c protein levels. In parallel with cytochrome c release, oxidative stress induced caspase-3 activation and DNA fragmentation. Pretreatment with NO significantly reduced the oxidative stress-enhanced caspase-3 activation and DNA damage. Results of this study show that NO pretreatment can protect osteoblasts from oxidative stress-induced apoptotic insults. The protective action involves a mitochondria-dependent mechanism.
Collapse
Affiliation(s)
- Chia-Chen Chang
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, No. 250 Wu-Hsing Street, Taipei 110, Taiwan, Republic of China
| | | | | | | |
Collapse
|
46
|
Zeini M, Través PG, López-Fontal R, Pantoja C, Matheu A, Serrano M, Boscá L, Hortelano S. Specific contribution of p19(ARF) to nitric oxide-dependent apoptosis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2006; 177:3327-36. [PMID: 16920973 DOI: 10.4049/jimmunol.177.5.3327] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
NO is an important bioactive molecule involved in a variety of physio- and pathological processes, including apoptosis induction. The proapoptotic activity of NO involves the rise in the tumor suppressor p53 and the accumulation and targeting of proapoptotic members of the Bcl-2 family, in particular Bax and the release of cytochrome c from the mitochondria. However, the exact mechanism by which NO induces p53 activation has not been fully elucidated. In this study, we describe that NO induces p19(ARF) through a transcriptional mechanism. This up-regulation of p19(ARF) activates p53, leading to apoptosis. The importance of p19(ARF) on NO-dependent apoptosis was revealed by the finding that various cell types from alternate reading frame-knockout mice exhibit a diminished response to NO-mediated apoptosis when compared with normal mice. Moreover, the biological relevance of alternative reading frame to p53 apoptosis was confirmed in in vivo models of apoptosis. Together, these results demonstrate that NO-dependent apoptosis requires, in part, the activation of p19(ARF).
Collapse
Affiliation(s)
- Miriam Zeini
- Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Kleschyov AL, Strand S, Schmitt S, Gottfried D, Skatchkov M, Sjakste N, Daiber A, Umansky V, Munzel T. Dinitrosyl-iron triggers apoptosis in Jurkat cells despite overexpression of Bcl-2. Free Radic Biol Med 2006; 40:1340-8. [PMID: 16631524 DOI: 10.1016/j.freeradbiomed.2005.12.001] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2004] [Revised: 11/17/2005] [Accepted: 12/01/2005] [Indexed: 11/26/2022]
Abstract
Cells expressing the cytokine-inducible NO synthase are known to trigger apoptosis in neighboring cells. Paramagnetic dinitrosyl nonheme iron complexes (DNIC) were found in tumor tissue about 40 years ago; however, the role of these NO(+)-bearing species is not completely understood. In the human Jurkat leukemia cell line, the application of the model complex DNIC-thiosulfate (50-200 microM) induced apoptosis (defined by phosphatidylserine externalization) in a concentration- and time-dependent manner. In Jurkat cells, the pan-caspase inhibitor, zVADfmk (50 microM), and/or stable transfection of antiapoptotic protein, Bcl-2, was unable to afford protection against DNIC-induced apoptosis. The membrane-impermeable metal chelator, N-methyl-D-glucamine dithiocarbamate (MGD; 200 microM), in the presence of DNIC significantly increased apoptosis, but had no effect on its own. Electron paramagnetic resonance studies showed that MGD led to rapid transformation of the extracellular DNIC into the stable impermeable NO-Fe-MGD complex and to a burst-type release of nitrosonium (NO(+)) equivalents in the extracellular space. These results suggest that in Jurkat cells, DNIC-thiosulfate induces Bcl-2- and caspase-independent apoptosis, which is probably secondary to local nitrosative stress at the cell surface. We hypothesize that the local release of nonheme Fe-NO species by activated macrophages may play a role in the killing of malignant cells that have high Bcl-2 levels.
Collapse
Affiliation(s)
- Andrei L Kleschyov
- II-Department of Medicine, Johannes Gutenberg University, Mainz, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Orsi NM. Embryotoxicity of the nitric oxide donor sodium nitroprusside in preimplantation bovine embryos in vitro. Anim Reprod Sci 2006; 91:225-36. [PMID: 15964159 DOI: 10.1016/j.anireprosci.2005.04.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2005] [Revised: 04/21/2005] [Accepted: 04/29/2005] [Indexed: 02/02/2023]
Abstract
Many early pregnancy complications are associated with an imbalance in pro- and anti-inflammatory cytokines, resulting in alterations in nitric oxide (NO) profile. Since very little is known about the modus operandi of this free radical in early embryos, this study characterised NO embryotoxicity in terms of bovine embryo development and metabolism. Embryos were generated by in vitro maturation and fertilisation of oocytes aspirated from abattoir-derived ovaries. Zygote to blastocyst rates were measured in SOFaaBSA in the presence and absence of the NO donor sodium nitroprusside (SNP) over the 0-50 microM range (n=10 per group). Since concentrations <10 microM SNP depressed blastocyst rate, blastocyst cell numbers (determined by bisbenzimide staining; n=22 and 20), glucose, pyruvate, lactate (measured ultramicrofluorometrically) and amino acid profiles (quantified by HPLC; n=28 and 23) were assessed at 0 and 10 microM SNP. SNP depressed cell numbers, reduced pyruvate and glucose uptake, perturbed quantitative tyrosine, threonine, phenylalanine, lysine, glycine, tryptophan, methionine and valine profiles, and decreased retention into the negative range (P<0.05). Qualitative asparagine and lysine profiles were affected by SNP, while proportional amino acid production and consumption were increased and decreased, respectively (P<0.05). These findings indicate that SNP (presumably through increases in NO profile): (i) fails to improve bovine embryo development in vitro, (ii) exerts toxic effects, likely through ATP starvation induced by cytochrome c oxidase (oxidative phosphorylation) and glyceraldehyde-3-phosphate dehydrogenase (glycolysis) inhibition, and (iii) may affect albumin endocytosis/hydrolysis or protein biosynthesis, rather than causing a loss of intracellular amino acids or simply depressing their metabolism.
Collapse
Affiliation(s)
- Nicolas M Orsi
- Perinatal Research Group, Academic Unit of Paediatrics, Obstetrics and Gynaecology, D Floor, Clarendon Wing, Leeds General Infirmary, Belmont Grove, Leeds, LS2 9NS, UK.
| |
Collapse
|
49
|
Ho WP, Chen TL, Chiu WT, Tai YT, Chen RM. Nitric Oxide Induces Osteoblast Apoptosis through a Mitochondria-Dependent Pathway. Ann N Y Acad Sci 2006; 1042:460-70. [PMID: 15965092 DOI: 10.1196/annals.1338.039] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Osteoblasts contribute to bone remodeling. Nitric oxide can regulate osteoblast activities. In this study, we attempted to evaluate the pathophysiological effects of nitric oxide on osteoblasts and its possible mechanism using neonatal rat calvarial osteoblasts as the experimental model. Exposure of osteoblasts to sodium nitroprusside, a nitric oxide donor, decreased alkaline phosphatase activities and cell viability in a concentration- and time-dependent manner. Apoptotic analysis revealed that sodium nitroprusside time-dependently increased the percentages of osteoblasts undergoing apoptosis. Administration of sodium nitroprusside reduced the mitochondrial membrane potential of osteoblasts. In parallel with the mitochondrial dysfunction, levels of intracellular reactive oxygen species and cytochrome c were significantly elevated following sodium nitroprusside administration. Exposure of osteoblasts to sodium nitroprusside significantly increased caspase-3 activity. Results of this study show that nitric oxide, decomposed from sodium nitroprusside, can induce osteoblast apoptosis through a mitochondrion-dependent cascade that causes mitochondrial dysfunction, release of intracellular reactive oxygen species and cytochrome c from mitochondria to cytoplasm, and activation of caspase-3.
Collapse
Affiliation(s)
- Wei-Pin Ho
- Department of Orthopedics, Wan-Fang Hospital, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | | | | | | | | |
Collapse
|
50
|
Kong CW, Huang CH, Hsu TG, Tsai KKC, Hsu CF, Huang MC, Chen LC. Leukocyte mitochondrial alterations after cardiac surgery involving cardiopulmonary bypass: clinical correlations. Shock 2005; 21:315-9. [PMID: 15179131 DOI: 10.1097/00024382-200404000-00005] [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] [Indexed: 11/26/2022]
Abstract
Cardiac surgery with the use of cardiopulmonary bypass (CPB) is known to initiate systemic inflammatory responses that are associated with immune dysregulations, but the pathomechanisms underlying these changes remain elusive. Mitochondrial transmembrane potential (MTP) is an important determinant of leukocytic functions and viability, and may be altered as a part of the cellular responses to systemic inflammatory insults. Therefore, we examined MTP in three subsets of peripheral leukocytes in 18 patients receiving uncomplicated cardiac surgery involving CPB. The MTP of neutrophils and lymphocytes significantly increased, whereas that of monocytes significantly declined, after the surgery. The alterations in leukocytic MTP were transient, normalizing 3 days to 1 week after the surgery, and were accompanied by transient overproduction of intracellular oxidants, including nitric oxide and superoxide. Despite these perturbations, the viability status of leukocytes remained unaltered. Positive correlations were found between the changes of leukocyte MTP and various clinical parameters, implying that leukocyte mitochondrial alterations are parts of the systemic immune perturbations induced by the bypass surgery.
Collapse
Affiliation(s)
- Chi-Woon Kong
- Division of Critical Care, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.
| | | | | | | | | | | | | |
Collapse
|