1
|
Titov VY, Dolgorukova AM, Osipov AN, Kochish II. DNIC is a Structure Providing Specificity of Physiological Effects of NO. Bull Exp Biol Med 2023; 176:160-164. [PMID: 38194076 DOI: 10.1007/s10517-024-05987-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Indexed: 01/10/2024]
Abstract
Metabolism of nitric oxide (NO) donors: dinitrosyl iron complexes (DNIC), nitrosothiols (RSNO), and nitroprusside was studied on a chick embryo model. The obtained results give reason to assume that DNIC constituting the main pool of nitroso compounds in the vast majority of tissues are NO donors immediately interacting with the physiological target of NO, and other NO donors can perform this function after their transformation into DNIC. NO is released from DNIC not spontaneously, but under a joint influence of a factor destroying the complex and a target having chemical affinity for NO. A similar mechanism is apparently implicated in NO passage through the cell membrane.
Collapse
Affiliation(s)
- V Yu Titov
- Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, Moscow, Russia.
- Federal Scientific Center "All-Russian Research and Technological Poultry Institute, Russian Academy of Sciences, Sergiev Posad, Moscow region, Russia.
- K. I. Skryabin Moscow State Academy of Veterinary Medicine and Biotechnology, Moscow, Russia.
| | - A M Dolgorukova
- Federal Scientific Center "All-Russian Research and Technological Poultry Institute, Russian Academy of Sciences, Sergiev Posad, Moscow region, Russia
| | - A N Osipov
- Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, Moscow, Russia
| | - I I Kochish
- K. I. Skryabin Moscow State Academy of Veterinary Medicine and Biotechnology, Moscow, Russia
| |
Collapse
|
2
|
Yu Y, Wang P, Ren Z, Xue Y, Jia Y, Wang W, Liu M, Pan K, Xiao L, Ji D, Wang X. A low-salt diet with candesartan administration is associated with acute kidney injury in nephritis by increasing nitric oxide. Biomed Pharmacother 2023; 161:114484. [PMID: 36921530 DOI: 10.1016/j.biopha.2023.114484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 02/28/2023] [Accepted: 03/07/2023] [Indexed: 03/18/2023] Open
Abstract
A low-salt diet may activate the renin-angiotensin-aldosterone system (RAAS) and is often applied simultaneously with RAAS inhibitors, especially for treatment of proteinuric nephritis. To explore the effect of a low-salt diet combined with angiotensin receptor blockers (ARB) on kidney function, the proteinuric nephritis model was induced by single intravenous injection of doxorubicin, and then the SD rats were administrated with candesartan intraperitoneal injection and fed with different salt diets. Rats with low-salt plus candesartan, not either alone, experienced acute kidney injury (AKI) at day 7 and could not self-restore when extending the experiment time from 7 days to 21 days, unless switching low-salt to normal-salt. Among three nitric oxide synthetases (NOS), endothelial NOS (eNOS) was obviously elevated and PI3K-Akt-eNOS signal pathway was activated. NG-Nitro-L-Arginine Methyl Ester (L-NAME), an eNOS inhibitor, reversed the decreased blood pressure and recovered the kidney dysfunction induced by low-salt with candesartan. The increased TUNEL-positive cells, Bax/Bcl-2 and cleaved-caspase3 protein abundance was ameliorated by L-NAME in vivo. In vitro, sodium nitroprusside, a nitric oxide donor, can also increase Bax/Bcl-2 and cleaved-caspase3 protein level in HK-2 cell. Thus, low-salt diet combined with candesartan in nephritis rats led to AKI, and the mechanism involved the increase of eNOS/NO, which linked to the decrease of blood pressure and the increase of apoptosis. This study provides practical guidance for salt intake in cases of RAS inhibitor usage clinically.
Collapse
Affiliation(s)
- Yanting Yu
- Department of Nephrology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, China; Department of Nephrology, Nanjing BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing, China
| | - Ping Wang
- The Core Laboratory, Nanjing BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing, China
| | - Zhiyun Ren
- The Core Laboratory, Nanjing BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing, China
| | - Ying Xue
- The Core Laboratory, Nanjing BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing, China
| | - Yutao Jia
- Department of Nephrology, Nanjing BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing, China
| | - Weiwan Wang
- The Core Laboratory, Nanjing BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing, China
| | - Mingda Liu
- The Core Laboratory, Nanjing BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing, China
| | - Kueiching Pan
- Department of Nursing, Nanjing BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing, China
| | - Leijuan Xiao
- Department of Nephrology, Nanjing BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing, China.
| | - Daxi Ji
- Department of Nephrology, Nanjing BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing, China.
| | - Xiaoyan Wang
- Department of Nephrology, Nanjing BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing, China; The Core Laboratory, Nanjing BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
3
|
Kochish II, Titov VY, Nikonov IN, Brazhnik EA, Vorobyov NI, Korenyuga MV, Myasnikova OV, Dolgorukova AM, Griffin DK, Romanov MN. Unraveling signatures of chicken genetic diversity and divergent selection in breed-specific patterns of early myogenesis, nitric oxide metabolism and post-hatch growth. Front Genet 2023; 13:1092242. [PMID: 36712856 PMCID: PMC9874007 DOI: 10.3389/fgene.2022.1092242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 12/27/2022] [Indexed: 01/13/2023] Open
Abstract
Introduction: Due to long-term domestication, breeding and divergent selection, a vast genetic diversity in poultry currently exists, with various breeds being characterized by unique phenotypic and genetic features. Assuming that differences between chicken breeds divergently selected for economically and culturally important traits manifest as early as possible in development and growth stages, we aimed to explore breed-specific patterns and interrelations of embryo myogenesis, nitric oxide (NO) metabolism and post-hatch growth rate (GR). Methods: These characteristics were explored in eight breeds of different utility types (meat-type, dual purpose, egg-type, game, and fancy) by incubating 70 fertile eggs per breed. To screen the differential expression of seven key myogenesis associated genes (MSTN, GHR, MEF2C, MYOD1, MYOG, MYH1, and MYF5), quantitative real-time PCR was used. Results: We found that myogenesis associated genes expressed in the breast and thigh muscles in a coordinated manner showing breed specificity as a genetic diversity signature among the breeds studied. Notably, coordinated ("accord") expression patterns of MSTN, GHR, and MEFC2 were observed both in the breast and thigh muscles. Also, associated expression vectors were identified for MYOG and MYOD1 in the breast muscles and for MYOG and MYF5 genes in the thigh muscles. Indices of NO oxidation and post-hatch growth were generally concordant with utility types of breeds, with meat-types breeds demonstrating higher NO oxidation levels and greater GR values as compared to egg-type, dual purpose, game and fancy breeds. Discussion: The results of this study suggest that differences in early myogenesis, NO metabolism and post-hatch growth are breed-specific; they appropriately reflect genetic diversity and accurately capture the evolutionary history of divergently selected chicken breeds.
Collapse
Affiliation(s)
- Ivan I. Kochish
- K. I. Skryabin Moscow State Academy of Veterinary Medicine and Biotechnology, Moscow, Russia
| | - Vladimir Yu. Titov
- K. I. Skryabin Moscow State Academy of Veterinary Medicine and Biotechnology, Moscow, Russia
- Federal Scientific Center “All-Russian Poultry Research and Technological Institute” of the Russian Academy of Sciences, Sergiev Posad, Moscow Oblast, Russia
| | - Ilya N. Nikonov
- K. I. Skryabin Moscow State Academy of Veterinary Medicine and Biotechnology, Moscow, Russia
| | | | - Nikolai I. Vorobyov
- All-Russia Institute for Agricultural Microbiology, Pushkin, St. Petersburg, Russia
| | - Maxim V. Korenyuga
- K. I. Skryabin Moscow State Academy of Veterinary Medicine and Biotechnology, Moscow, Russia
| | - Olga V. Myasnikova
- K. I. Skryabin Moscow State Academy of Veterinary Medicine and Biotechnology, Moscow, Russia
| | - Anna M. Dolgorukova
- Federal Scientific Center “All-Russian Poultry Research and Technological Institute” of the Russian Academy of Sciences, Sergiev Posad, Moscow Oblast, Russia
| | - Darren K. Griffin
- School of Biosciences, University of Kent, Canterbury, United Kingdom
| | - Michael N. Romanov
- K. I. Skryabin Moscow State Academy of Veterinary Medicine and Biotechnology, Moscow, Russia
- School of Biosciences, University of Kent, Canterbury, United Kingdom
| |
Collapse
|
4
|
Lehnert N, Kim E, Dong HT, Harland JB, Hunt AP, Manickas EC, Oakley KM, Pham J, Reed GC, Alfaro VS. The Biologically Relevant Coordination Chemistry of Iron and Nitric Oxide: Electronic Structure and Reactivity. Chem Rev 2021; 121:14682-14905. [PMID: 34902255 DOI: 10.1021/acs.chemrev.1c00253] [Citation(s) in RCA: 101] [Impact Index Per Article: 33.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Nitric oxide (NO) is an important signaling molecule that is involved in a wide range of physiological and pathological events in biology. Metal coordination chemistry, especially with iron, is at the heart of many biological transformations involving NO. A series of heme proteins, nitric oxide synthases (NOS), soluble guanylate cyclase (sGC), and nitrophorins, are responsible for the biosynthesis, sensing, and transport of NO. Alternatively, NO can be generated from nitrite by heme- and copper-containing nitrite reductases (NIRs). The NO-bearing small molecules such as nitrosothiols and dinitrosyl iron complexes (DNICs) can serve as an alternative vehicle for NO storage and transport. Once NO is formed, the rich reaction chemistry of NO leads to a wide variety of biological activities including reduction of NO by heme or non-heme iron-containing NO reductases and protein post-translational modifications by DNICs. Much of our understanding of the reactivity of metal sites in biology with NO and the mechanisms of these transformations has come from the elucidation of the geometric and electronic structures and chemical reactivity of synthetic model systems, in synergy with biochemical and biophysical studies on the relevant proteins themselves. This review focuses on recent advancements from studies on proteins and model complexes that not only have improved our understanding of the biological roles of NO but also have provided foundations for biomedical research and for bio-inspired catalyst design in energy science.
Collapse
Affiliation(s)
- Nicolai Lehnert
- Department of Chemistry and Department of Biophysics, University of Michigan, Ann Arbor, Michigan 48109-1055, United States
| | - Eunsuk Kim
- Department of Chemistry, Brown University, Providence, Rhode Island 02912, United States
| | - Hai T Dong
- Department of Chemistry and Department of Biophysics, University of Michigan, Ann Arbor, Michigan 48109-1055, United States
| | - Jill B Harland
- Department of Chemistry and Department of Biophysics, University of Michigan, Ann Arbor, Michigan 48109-1055, United States
| | - Andrew P Hunt
- Department of Chemistry and Department of Biophysics, University of Michigan, Ann Arbor, Michigan 48109-1055, United States
| | - Elizabeth C Manickas
- Department of Chemistry and Department of Biophysics, University of Michigan, Ann Arbor, Michigan 48109-1055, United States
| | - Kady M Oakley
- Department of Chemistry, Brown University, Providence, Rhode Island 02912, United States
| | - John Pham
- Department of Chemistry, Brown University, Providence, Rhode Island 02912, United States
| | - Garrett C Reed
- Department of Chemistry and Department of Biophysics, University of Michigan, Ann Arbor, Michigan 48109-1055, United States
| | - Victor Sosa Alfaro
- Department of Chemistry and Department of Biophysics, University of Michigan, Ann Arbor, Michigan 48109-1055, United States
| |
Collapse
|
5
|
Titov VY, Dolgorukova AM, Osipov AN, Kochish II. Putative Role of Ligands of DNIC in the Physiological Action of the Complex. Bull Exp Biol Med 2021; 171:606-610. [PMID: 34617179 DOI: 10.1007/s10517-021-05278-1] [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: 02/26/2021] [Indexed: 10/20/2022]
Abstract
In a relatively isolated system of avian embryo, the metabolism of NO, a component of the dinitrosyl iron complexes (DNIC), the main NO donor in most tissues, depends on the ligands that make up the complex. This fact corroborates the earlier hypothesis that these ligands perform a regulatory function in NO metabolism. It is also shown that nitrite injected into the embryo is not oxidized to nitrate like NO in DNIC, but is accumulated outside the amniotic sac. Normally, nitrite is present in an embryo in trace amounts. These facts suggest that NO in the embryo is transferred from the donor molecule to a target in the embryo tissues further transformed with minimum oxidation to nitrite.
Collapse
Affiliation(s)
- V Yu Titov
- All-Russian Research and Technological Institute of Poultry, Russian Academy of Sciences, Sergiev Posad, Moscow region, Russia. .,N. I. Pirogov Russian National Research Medical University, Ministry of the Health of the Russian Federation, Moscow, Russia. .,K. I. Skryabin Moscow State Academy of Veterinary Medicine and Biotechnology, Moscow, Russia.
| | - A M Dolgorukova
- All-Russian Research and Technological Institute of Poultry, Russian Academy of Sciences, Sergiev Posad, Moscow region, Russia
| | - A N Osipov
- N. I. Pirogov Russian National Research Medical University, Ministry of the Health of the Russian Federation, Moscow, Russia
| | - I I Kochish
- K. I. Skryabin Moscow State Academy of Veterinary Medicine and Biotechnology, Moscow, Russia
| |
Collapse
|
6
|
Vanin AF, Tronov VA, Borodulin RR. Nitrosonium Cation as a Cytotoxic Component of Dinitrosyl Iron Complexes with Thiol-containing Ligands (based on the Experimental Work on MCF7 Human Breast Cancer Cell Culture). Cell Biochem Biophys 2021; 79:93-102. [PMID: 33492647 PMCID: PMC7829092 DOI: 10.1007/s12013-020-00962-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/07/2020] [Indexed: 12/02/2022]
Abstract
Here we demonstrate that binuclear dinitrosyl iron complexes with thiol-containing ligands (glutathione and mercaptosuccinate, B-DNIC-GSH and B-DNIC-MS, respectively) exert cytotoxic effects on MCF7 human breast cancer cells. We showed that they are mediated by nitrosonium cations released from these complexes (NO+). This finding is supported by the cytotoxic effect of both B-DNICs on MCF7 cells evidenced to retain or was even promoted in the presence of N-Methyl-D-glucamine dithiocarbamate (MGD). MGD recruits an iron nitrosyl group [Fe(NO)] from the iron-dinitrosyl fragment [Fe(NO)2] of B-DNIC-MS forming stable mononitrosyl complexes of iron with MGD and releasing NO+ cations from a [Fe(NO)2] fragment.
Collapse
Affiliation(s)
- Anatoly F Vanin
- Semenov Federal Research Centre of Chemical Physics, Russian Academy of Sciences, Moscow, Russia.
- Institute of Regenerative Medicine, Sechenov Medical University, Moscow, Russia.
| | - Viktor A Tronov
- Semenov Federal Research Centre of Chemical Physics, Russian Academy of Sciences, Moscow, Russia
| | - Rostislav R Borodulin
- Semenov Federal Research Centre of Chemical Physics, Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
7
|
Weitzner O, Seraya-Bareket C, Biron-Shental T, Fishamn A, Yagur Y, Tzadikevitch-Geffen K, Farladansky-Gershnabel S, Kidron D, Ellis M, Ashur-Fabian O. Enhanced expression of αVβ3 integrin in villus and extravillous trophoblasts of placenta accreta. Arch Gynecol Obstet 2020; 303:1175-1183. [PMID: 33112993 DOI: 10.1007/s00404-020-05844-4] [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: 09/05/2020] [Accepted: 10/13/2020] [Indexed: 11/29/2022]
Abstract
BACKGROUND Placenta accreta is one of the most serious complications in obstetrics and gynecology. Villous trophoblasts (VT) and extravillous trophoblasts (EVT) play a central role in normal placentation. Placenta accreta is characterized by abnormal invasion of EVT cells through the uterine layers, due to changes in several parameters, including adhesion proteins. Although αvβ3 integrin is a central adhesion molecule, participating in multiple invasive pathological conditions including cancer, data on placenta accreta are lacking. OBJECTIVE To study the expression pattern of αvβ3 integrin in placenta accreta in comparison with normal placentas. STUDY DESIGN We collected tissue samples from placentas defined as percreta, the most severe presentation of placenta accreta and from normal control placentas (n = 10 each). The samples underwent protein extractions for analyses of αvβ3 expression by Western blots (WB) and a parallel tissue assessment by immunohistochemistry (IHC). RESULTS WB results indicated significantly elevated αvβ3 integrin expression in the percreta samples compared to normal placentas. These elevated levels were mainly contributed by EVT cells, as demonstrated by IHC. αvβ3 integrin demonstrated a classical membranal expression in the VT cells, whereas a uniformly distributed expression was documented in the EVT cells. These patterns of the αvβ3 integrin localization were similar in both accreta and normal placental samples. CONCLUSIONS Enhanced αvβ3 integrin expression, mainly in extra villous trophoblasts of placenta percreta, implies for a role of this adhesion molecule in pathological placentation.
Collapse
Affiliation(s)
- Omer Weitzner
- Translational Hemato-Oncology Laboratory, Hematology Institute and Blood Bank, Meir Medical Center, Kfar-Saba, Israel. .,Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel. .,Department of Obstetrics and Gynecology, Meir Medical Center, Kfar Saba, Israel.
| | - Chen Seraya-Bareket
- Translational Hemato-Oncology Laboratory, Hematology Institute and Blood Bank, Meir Medical Center, Kfar-Saba, Israel.,Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.,Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Tal Biron-Shental
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.,Department of Obstetrics and Gynecology, Meir Medical Center, Kfar Saba, Israel
| | - Ami Fishamn
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.,Gynecological Oncology Unit, Meir Medical Center, Kfar Saba, Israel
| | - Yael Yagur
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.,Department of Obstetrics and Gynecology, Meir Medical Center, Kfar Saba, Israel
| | - Keren Tzadikevitch-Geffen
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.,Department of Obstetrics and Gynecology, Meir Medical Center, Kfar Saba, Israel
| | - Sivan Farladansky-Gershnabel
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.,Department of Obstetrics and Gynecology, Meir Medical Center, Kfar Saba, Israel
| | - Debora Kidron
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.,Department of Pathology, Sackler Faculty of Medicine, Meir Hospital, Kfar Saba, Israel
| | - Martin Ellis
- Translational Hemato-Oncology Laboratory, Hematology Institute and Blood Bank, Meir Medical Center, Kfar-Saba, Israel.,Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Osnat Ashur-Fabian
- Translational Hemato-Oncology Laboratory, Hematology Institute and Blood Bank, Meir Medical Center, Kfar-Saba, Israel.,Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.,Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
8
|
Hypothetical mechanism of light action on nitric oxide physiological effects. Lasers Med Sci 2020; 36:1389-1395. [PMID: 33104897 DOI: 10.1007/s10103-020-03169-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 10/21/2020] [Indexed: 10/23/2022]
Abstract
Nitric oxide is a universal cellular mediator. It is involved in many physiological processes, including those induced by light. The disability for complete analysis of the nitric oxide metabolites in tissues prevents the exact understanding of the role of NO in a particular process. The sensitivity and selectivity of an enzymatic sensor developed in our lab is based on the detection of all NO groups that carry a positive charge or acquire it in the chemical processes. Using this sensor, we have shown that dinitrosyl iron complexes (DNIC), being principal nitric oxide donors in most of living tissues, undergo transformations under light irradiation in the wavelength range of 400-700 nm. These changes are not associated with nitric oxide release to the environment. But a nitrosyl iron complex without thiol ligands (Fe(NO)n) is produced. Moreover, in the moment of the complex reorganization, the chemical bond between the NO group and other components apparently weakens and, in the presence of a substance possessing chemical affinity to the NO group, the latter acquires the ability of transition from the complex to this substance. Therefore, the efficiency of NO donors first of all depends on the existence of the NO target and its status including that under the action of light. The activation of a donor compound by light can facilitate the transfer of NO to the target. Transfer of NO from the donor to the target occurs without releasing NO, or with a minimum time of its stay in the unbound state.
Collapse
|
9
|
Žatko D, Vašková J, Perjési P, Haus M, Vaško L. Pro-oxidative and antioxidant effects of salicylates. CHEMICAL PAPERS 2020. [DOI: 10.1007/s11696-020-01152-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
10
|
Spotlight on ROS and β3-Adrenoreceptors Fighting in Cancer Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:6346529. [PMID: 31934266 PMCID: PMC6942895 DOI: 10.1155/2019/6346529] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 11/26/2019] [Indexed: 02/07/2023]
Abstract
The role of ROS and RNS is a long-standing debate in cancer. Increasing the concentration of ROS reaching the toxic threshold can be an effective strategy for the reduction of tumor cell viability. On the other hand, cancer cells, by maintaining intracellular ROS concentration at an intermediate level called “mild oxidative stress,” promote the activation of signaling that favors tumor progression by increasing cell viability and dangerous tumor phenotype. Many chemotherapeutic treatments induce cell death by rising intracellular ROS concentration. The persistent drug stimulation leads tumor cells to simulate a process called hormesis by which cancer cells exhibit a biphasic response to exposure to drugs used. After a first strong response to a low dose of chemotherapeutic agent, cancer cells start to decrease the response even if high doses of drugs were used. In this framework, β3-adrenoreceptors (β3-ARs) fit with an emerging antioxidant role in cancer. β3-ARs are involved in tumor proliferation, angiogenesis, metastasis, and immune tolerance. Its inhibition, by the selective β3-ARs antagonist (SR59230A), leads cancer cells to increase ROS concentration thus inducing cell death and to decrease NO levels thus inhibiting angiogenesis. In this review, we report an overview on reactive oxygen biology in cancer cells focusing on β3-ARs as new players in the antioxidant pathway.
Collapse
|
11
|
Chen S, Wang X, Nisar MF, Lin M, Zhong JL. Heme Oxygenases: Cellular Multifunctional and Protective Molecules against UV-Induced Oxidative Stress. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:5416728. [PMID: 31885801 PMCID: PMC6907065 DOI: 10.1155/2019/5416728] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 10/29/2019] [Indexed: 12/20/2022]
Abstract
Ultraviolet (UV) irradiation can be considered as a double-edged sword: not only is it a crucial environmental factor that can cause skin-related disorders but it can also be used for phototherapy of skin diseases. Inducible heme oxygenase-1 (HO-1) in response to a variety of stimuli, including UV exposure, is vital to maintain cell homeostasis. Heme oxygenase-2 (HO-2), another member of the heme oxygenase family, is constitutively expressed. In this review, we discuss how heme oxygenase (HO), a vital rate-limiting enzyme, participates in heme catabolism and cytoprotection. Phylogenetic analysis showed that there may exist a functional differentiation between HO-1 and HO-2 during evolution. Furthermore, depending on functions in immunomodulation and antioxidation, HO-1 participates in disease progression, especially in pathogenesis of skin diseases, such as vitiligo and psoriasis. To further investigate the particular role of HO-1 in diseases, we summarized the profile of the HO enzyme system and its related signaling pathways, such as Nrf2 and endoplasmic reticulum crucial signaling, both known to regulate HO-1 expression. Furthermore, we report on a C-terminal truncation of HO-1, which is generally considered as a signal molecule. Also, a newly identified alternative splice isoform of HO-1 not only provides us a novel perspective on comprehensive HO-1 alternative splicing but also offers us a basis to clarify the relationship between HO-1 transcripts and oxidative diseases. To conclude, the HO system is not only involved in heme catabolism but also involved in biological processes related to the pathogenesis of certain diseases, even though the mechanism of disease progression still remains sketchy. Further understanding the role of the HO system and its relationship to UV is helpful for revealing the HO-related signaling networks and the pathogenesis of many diseases.
Collapse
Affiliation(s)
- ShiDa Chen
- The Base of “111 Project” for Biomechanics & Tissue Repair Engineering; Key Laboratory of Biorheological Science and Technology, Ministry of Education, Bioengineering College, Chongqing University, Chongqing 400044, China
| | - XiaoYu Wang
- The Base of “111 Project” for Biomechanics & Tissue Repair Engineering; Key Laboratory of Biorheological Science and Technology, Ministry of Education, Bioengineering College, Chongqing University, Chongqing 400044, China
| | - Muhammad Farrukh Nisar
- The Base of “111 Project” for Biomechanics & Tissue Repair Engineering; Key Laboratory of Biorheological Science and Technology, Ministry of Education, Bioengineering College, Chongqing University, Chongqing 400044, China
| | - Mao Lin
- Chongqing Traditional Chinese Medicine Hospital, Chongqing 400011, China
| | - Julia Li Zhong
- The Base of “111 Project” for Biomechanics & Tissue Repair Engineering; Key Laboratory of Biorheological Science and Technology, Ministry of Education, Bioengineering College, Chongqing University, Chongqing 400044, China
- Chongqing Traditional Chinese Medicine Hospital, Chongqing 400011, China
| |
Collapse
|
12
|
Vanin AF. What is the Mechanism of Nitric Oxide Conversion into Nitrosonium Ions Ensuring S-Nitrosating Processes in Living Organisms. Cell Biochem Biophys 2019; 77:279-292. [PMID: 31586291 DOI: 10.1007/s12013-019-00886-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 09/12/2019] [Indexed: 12/29/2022]
Abstract
Here, I present the data testifying that the conversion of free radical NO molecules to nitrosonium ions (NO+), which are necessary for the realization of one of NO biological effects (S-nitrosation), may occur in living organisms after binding NO molecules to loosely bound iron (Fe2+ ions) with the subsequent mutual one-electron oxidation-reduction of NO molecules (their disproportionation). Inclusion of thiol-containing substances as iron ligands into this process prevents hydrolysis of NO+ ions bound to iron thus providing the formation of stable dinitrosyl iron complexes (DNIC) with thiol ligands. Such complexes act in living organisms as donors of NO and NO+, providing stabilization and transfer of these agents via the autocrine and paracrine pathways. Without loosely bound iron (labile iron pool) and thiols participating in the DNIC formation, NO functioning as one of universal regulators of diverse metabolic processes would be impossible.
Collapse
Affiliation(s)
- Anatoly F Vanin
- N.N. Semenov Federal Research Center of Chemical Physics, Russian Academy of Sciences; Institute for Regenerative Medicine, I.M. Sechenov First Moscow State Medical University, Kosygin Str.4, Moscow, 119991, Russia.
| |
Collapse
|
13
|
Nitric oxide (NO) in bird embryogenesis: physiological role and ability of practical use. WORLD POULTRY SCI J 2019. [DOI: 10.1017/s0043933912000098] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
14
|
|
15
|
Stoyanovsky DA, Tyurina YY, Shrivastava I, Bahar I, Tyurin VA, Protchenko O, Jadhav S, Bolevich SB, Kozlov AV, Vladimirov YA, Shvedova AA, Philpott CC, Bayir H, Kagan VE. Iron catalysis of lipid peroxidation in ferroptosis: Regulated enzymatic or random free radical reaction? Free Radic Biol Med 2019; 133:153-161. [PMID: 30217775 PMCID: PMC6555767 DOI: 10.1016/j.freeradbiomed.2018.09.008] [Citation(s) in RCA: 205] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 09/06/2018] [Accepted: 09/09/2018] [Indexed: 12/14/2022]
Abstract
Duality of iron as an essential cofactor of many enzymatic metabolic processes and as a catalyst of poorly controlled redox-cycling reactions defines its possible biological beneficial and hazardous role in the body. In this review, we discuss these two "faces" of iron in a newly conceptualized program of regulated cell death, ferroptosis. Ferroptosis is a genetically programmed iron-dependent form of regulated cell death driven by enhanced lipid peroxidation and insufficient capacity of thiol-dependent mechanisms (glutathione peroxidase 4, GPX4) to eliminate hydroperoxy-lipids. We present arguments favoring the enzymatic mechanisms of ferroptotically engaged non-heme iron of 15-lipoxygenases (15-LOX) in complexes with phosphatidylethanolamine binding protein 1 (PEBP1) as a catalyst of highly selective and specific oxidation reactions of arachidonoyl- (AA) and adrenoyl-phosphatidylethanolamines (PE). We discuss possible role of iron chaperons as control mechanisms for guided iron delivery directly to their "protein clients" thus limiting non-enzymatic redox-cycling reactions. We also consider opportunities of loosely-bound iron to contribute to the production of pro-ferroptotic lipid oxidation products. Finally, we propose a two-stage iron-dependent mechanism for iron in ferroptosis by combining its catalytic role in the 15-LOX-driven production of 15-hydroperoxy-AA-PE (HOO-AA-PE) as well as possible involvement of loosely-bound iron in oxidative cleavage of HOO-AA-PE to oxidatively truncated electrophiles capable of attacking nucleophilic targets in yet to be identified proteins leading to cell demise.
Collapse
Affiliation(s)
- D A Stoyanovsky
- Center for Free Radical and Antioxidant Heath, Department of Environmental Health, University of Pittsburgh, USA
| | - Y Y Tyurina
- Center for Free Radical and Antioxidant Heath, Department of Environmental Health, University of Pittsburgh, USA
| | - I Shrivastava
- Center for Free Radical and Antioxidant Heath, Department of Environmental Health, University of Pittsburgh, USA; Department of Computational and Systems Biology, University of Pittsburgh, USA
| | - I Bahar
- Department of Computational and Systems Biology, University of Pittsburgh, USA
| | - V A Tyurin
- Center for Free Radical and Antioxidant Heath, Department of Environmental Health, University of Pittsburgh, USA
| | - O Protchenko
- Genetics and Metabolism Section, Liver Diseases Branch, NIDDK, NIH, Bethesda, USA
| | - S Jadhav
- Genetics and Metabolism Section, Liver Diseases Branch, NIDDK, NIH, Bethesda, USA
| | - S B Bolevich
- Laboratory of Navigational Redox Lipidomics and Department of Human Pathology, IM Sechenov Moscow State Medical University, Russian Federation
| | - A V Kozlov
- L Boltzmann Institute for Experimental and Clinical Traumatology in AUVA Research Center, Vienna, Austria; Laboratory of Navigational Redox Lipidomics and Department of Human Pathology, IM Sechenov Moscow State Medical University, Russian Federation
| | - Y A Vladimirov
- Laboratory of Navigational Redox Lipidomics and Department of Human Pathology, IM Sechenov Moscow State Medical University, Russian Federation
| | - A A Shvedova
- Exposure Assessment Branch, NIOSH/CDC, Morgantown, WV, USA
| | - C C Philpott
- Genetics and Metabolism Section, Liver Diseases Branch, NIDDK, NIH, Bethesda, USA
| | - H Bayir
- Center for Free Radical and Antioxidant Heath, Department of Environmental Health, University of Pittsburgh, USA; Departments of Critical Care Medicine, University of Pittsburgh, USA
| | - V E Kagan
- Center for Free Radical and Antioxidant Heath, Department of Environmental Health, University of Pittsburgh, USA; Laboratory of Navigational Redox Lipidomics and Department of Human Pathology, IM Sechenov Moscow State Medical University, Russian Federation; Departments of Chemistry, University of Pittsburgh, USA; Departments of Pharmacology and Chemical Biology, University of Pittsburgh, USA; Departments of Radiation Oncology, University of Pittsburgh, USA.
| |
Collapse
|
16
|
Pisarenko O, Studneva I, Timoshin A, Veselova O. Protective efficacy of dinitrosyl iron complexes with reduced glutathione in cardioplegia and reperfusion. Pflugers Arch 2019; 471:583-593. [PMID: 30613864 DOI: 10.1007/s00424-018-02251-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 12/14/2018] [Accepted: 12/27/2018] [Indexed: 12/21/2022]
Abstract
Disturbed homeostasis of nitric oxide (NO) is one of the causes of myocardial ischemia/reperfusion (I/R) injury during open-heart surgery. This study was designed to explore mechanisms of action of dinitrosyl iron complexes with reduced glutathione ({(GS-)2Fe+(NO+)2}+, DNIC-GS) added to crystalloid cardioplegia or reperfusion solution in isolated working rat hearts. Hearts of male Wistar rats were subjected to cardioplegic arrest by St. Thomas' Hospital cardioplegic solution (STH) and normothermic global ischemia followed by reperfusion. DNIC-GS were used with STH or during early reperfusion. Lactate dehydrogenase (LDH) activity in the coronary effluent and myocardial contents of adenine nucleotides, phosphocreatine, and lactate were determined spectrophotometrically. Reactive oxygen species (ROS) formation in the coronary effluent and myocardial DNIC content was assessed by EPR technique. Cardioplegia or reperfusion with DNIC-GS significantly improved recovery of coronary flow and cardiac function compared with control. Carboxy-[2-(4-carboxyphenyl)-4,4,5,5-tetramethyl-imidozoline-1-oxy-3-oxide] (C-PTIO), a selective NO scavenger, reduced/abolished protective action of DNIC-GS. Enhanced recovery of cardiac function with DNIC-GS reduced LDH release in the coronary effluent, augmented recovery of myocardial energy state, and decreased formation of ROS-generating systems at reperfusion. Beneficial effects of DNIC-GS were related to the transfer of [Fe(NO)2] cores to thiol groups of myocardial proteins to form intracellular DNIC pools. The study concluded that DNIC-GS is a promising adjunct agent for metabolic and antioxidant protection of the heart during cardioplegic arrest and reperfusion.
Collapse
Affiliation(s)
- Oleg Pisarenko
- Laboratory for Myocardial Metabolism, National Medical Research Center for Cardiology, 3rd Cherepkovskaya Str., 15A, Moscow, Russian Federation, 121552.
| | - Irina Studneva
- Laboratory for Myocardial Metabolism, National Medical Research Center for Cardiology, 3rd Cherepkovskaya Str., 15A, Moscow, Russian Federation, 121552
| | - Alexander Timoshin
- Laboratory for Myocardial Metabolism, National Medical Research Center for Cardiology, 3rd Cherepkovskaya Str., 15A, Moscow, Russian Federation, 121552
| | - Oksana Veselova
- Laboratory for Myocardial Metabolism, National Medical Research Center for Cardiology, 3rd Cherepkovskaya Str., 15A, Moscow, Russian Federation, 121552
| |
Collapse
|
17
|
Dolgorukova AM, Titov VY, Petrov VA, Osipov AN, Slesarenko NA, Kochish II. Mechanisms of Specific Embryonic Effects of Nitrogen Oxide. Bull Exp Biol Med 2018; 165:635-639. [PMID: 30225706 DOI: 10.1007/s10517-018-4230-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Indexed: 11/28/2022]
Abstract
The study of NO metabolism in chicken embryos showed that the intensity of oxidation of both endogenous and exogenous for the embryo NO donors to nitrate is determined by the presence or state of NO targets, rather than donor concentration. The mechanism of this oxidation and its physiological role are discussed. It was also shown that oxidation product nitrate is actively eliminated from the amnionic sac.
Collapse
Affiliation(s)
- A M Dolgorukova
- Federal Research Centre All-Russian Research and Technology Institute of Poultry Industry, Russian Academy of Sciences, Moscow, Russia
| | - V Yu Titov
- Federal Research Centre All-Russian Research and Technology Institute of Poultry Industry, Russian Academy of Sciences, Moscow, Russia. .,N. I. Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, Moscow, Russia.
| | - V A Petrov
- N. I. Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, Moscow, Russia
| | - A N Osipov
- N. I. Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, Moscow, Russia
| | - N A Slesarenko
- K. I. Skryabin Moscow State Academy of Veterinary and Biotechnology, Moscow, Russia
| | - I I Kochish
- K. I. Skryabin Moscow State Academy of Veterinary and Biotechnology, Moscow, Russia
| |
Collapse
|
18
|
Mukosera GT, Liu T, Ishtiaq Ahmed AS, Li Q, Sheng MHC, Tipple TE, Baylink DJ, Power GG, Blood AB. Detection of dinitrosyl iron complexes by ozone-based chemiluminescence. Nitric Oxide 2018; 79:57-67. [PMID: 30059767 PMCID: PMC6277231 DOI: 10.1016/j.niox.2018.07.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 07/23/2018] [Accepted: 07/23/2018] [Indexed: 12/19/2022]
Abstract
Dinitrosyl iron complexes (DNICs) are important intermediates in the metabolism of nitric oxide (NO). They have been considered to be NO storage adducts able to release NO, scavengers of excess NO during inflammatory hypotensive shock, and mediators of apoptosis in cancer cells, among many other functions. Currently, all studies of DNICs in biological matrices use electron paramagnetic resonance (EPR) for both detection and quantification. EPR is limited, however, by its ability to detect only paramagnetic mononuclear DNICs even though EPR-silent binuclear are likely to be prevalent. Furthermore, physiological concentrations of mononuclear DNICs are usually lower than the EPR detection limit (1 μM). We have thus developed a chemiluminescence-based method for the selective detection of both DNIC forms at physiological, pathophysiological, and pharmacologic conditions. We have also demonstrated the use of the new method in detecting DNIC formation in the presence of nitrite and nitrosothiols within biological fluids and tissue. This new method, which can be used alone or in tandem with EPR, has the potential to offer insight about the involvement of DNICs in many NO-dependent pathways.
Collapse
Affiliation(s)
- George T Mukosera
- Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, CA, 92354, USA
| | - Taiming Liu
- Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, CA, 92354, USA
| | - Abu Shufian Ishtiaq Ahmed
- Regenerative Medicine Division, Department of Medicine, Loma Linda University School of Medicine, Loma Linda, CA, 92354, USA; Center for Dental Research, Loma Linda University School of Dentistry, Loma Linda, CA, 92350, USA
| | - Qian Li
- Neonatal Redox Biology Laboratory, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Matilda H-C Sheng
- Regenerative Medicine Division, Department of Medicine, Loma Linda University School of Medicine, Loma Linda, CA, 92354, USA
| | - Trent E Tipple
- Neonatal Redox Biology Laboratory, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - David J Baylink
- Regenerative Medicine Division, Department of Medicine, Loma Linda University School of Medicine, Loma Linda, CA, 92354, USA
| | - Gordon G Power
- Lawrence D. Longo Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, CA, 92354, USA
| | - Arlin B Blood
- Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, CA, 92354, USA; Lawrence D. Longo Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, CA, 92354, USA.
| |
Collapse
|
19
|
Abstract
Urinary tract infection (UTI) is one of the most common bacterial infections in humans, and the majority are caused by uropathogenic Escherichia coli (UPEC). The rising antibiotic resistance among UPEC and the frequent failure of antibiotics to effectively treat recurrent UTI and catheter-associated UTI motivate research on alternative ways of managing UTI. Abundant evidence indicates that the toxic radical nitric oxide (NO), formed by activation of the inducible nitric oxide synthase, plays an important role in host defence to bacterial infections, including UTI. The major source of NO production during UTI is from inflammatory cells, especially neutrophils, and from the uroepithelial cells that are known to orchestrate the innate immune response during UTI. NO and reactive nitrogen species have a wide range of antibacterial targets, including DNA, heme proteins, iron-sulfur clusters, and protein thiol groups. However, UPEC have acquired a variety of defence mechanisms for protection against NO, such as the NO-detoxifying enzyme flavohemoglobin and the NO-tolerant cytochrome bd-I respiratory oxidase. The cytotoxicity of NO-derived intermediates is nonspecific and may be detrimental to host cells, and a balanced NO production is crucial to maintain the tissue integrity of the urinary tract. In this review, we will give an overview of how NO production from host cells in the urinary tract is activated and regulated, the effect of NO on UPEC growth and colonization, and the ability of UPEC to protect themselves against NO. We also discuss the attempts that have been made to develop NO-based therapeutics for UTI treatment.
Collapse
|
20
|
Ferraz LS, Watashi CM, Colturato-Kido C, Pelegrino MT, Paredes-Gamero EJ, Weller RB, Seabra AB, Rodrigues T. Antitumor Potential of S-Nitrosothiol-Containing Polymeric Nanoparticles against Melanoma. Mol Pharm 2018; 15:1160-1168. [DOI: 10.1021/acs.molpharmaceut.7b01001] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Letícia S. Ferraz
- Center for Natural and Human Sciences (CCNH), Federal University of ABC (UFABC), 09210-580 Santo André, São Paulo, Brazil
| | - Carolina M. Watashi
- Center for Natural and Human Sciences (CCNH), Federal University of ABC (UFABC), 09210-580 Santo André, São Paulo, Brazil
| | - Carina Colturato-Kido
- Center for Natural and Human Sciences (CCNH), Federal University of ABC (UFABC), 09210-580 Santo André, São Paulo, Brazil
| | - Milena T. Pelegrino
- Center for Natural and Human Sciences (CCNH), Federal University of ABC (UFABC), 09210-580 Santo André, São Paulo, Brazil
| | - Edgar J. Paredes-Gamero
- Interdisciplinary Center for Biochemistry Investigation (CIIB), University of Mogi das Cruzes (UMC), 08780-911 Mogi das Cruzes, São Paulo, Brazil
| | - Richard B. Weller
- Medical Research Council Centre for Inflammation Research, Queen’s Medical Research Institute, University of Edinburgh, EH16 4TJ Edinburgh, U.K
| | - Amedea B. Seabra
- Center for Natural and Human Sciences (CCNH), Federal University of ABC (UFABC), 09210-580 Santo André, São Paulo, Brazil
- Nanomedicine Research Unit (NANOMED), Federal University of ABC (UFABC), Santo André, São Paulo, Brazil
| | - Tiago Rodrigues
- Center for Natural and Human Sciences (CCNH), Federal University of ABC (UFABC), 09210-580 Santo André, São Paulo, Brazil
- Nanomedicine Research Unit (NANOMED), Federal University of ABC (UFABC), Santo André, São Paulo, Brazil
| |
Collapse
|
21
|
Abstract
SIGNIFICANCE Glutathione (GSH) is the most abundant cellular low-molecular-weight thiol in the majority of organisms in all kingdoms of life. Therefore, functions of GSH and disturbed regulation of its concentration are associated with numerous physiological and pathological situations. Recent Advances: The function of GSH as redox buffer or antioxidant is increasingly being questioned. New functions, especially functions connected to the cellular iron homeostasis, were elucidated. Via the formation of iron complexes, GSH is an important player in all aspects of iron metabolism: sensing and regulation of iron levels, iron trafficking, and biosynthesis of iron cofactors. The variety of GSH coordinated iron complexes and their functions with a special focus on FeS-glutaredoxins are summarized in this review. Interestingly, GSH analogues that function as major low-molecular-weight thiols in organisms lacking GSH resemble the functions in iron homeostasis. CRITICAL ISSUES Since these iron-related functions are most likely also connected to thiol redox chemistry, it is difficult to distinguish between mechanisms related to either redox or iron metabolisms. FUTURE DIRECTIONS The ability of GSH to coordinate iron in different complexes with or without proteins needs further investigation. The discovery of new Fe-GSH complexes and their physiological functions will significantly advance our understanding of cellular iron homeostasis. Antioxid. Redox Signal. 27, 1235-1251.
Collapse
Affiliation(s)
- Carsten Berndt
- 1 Department of Neurology, Medical Faculty, Life Science Center , Heinrich-Heine-Universität, Düsseldorf, Germany
| | - Christopher Horst Lillig
- 2 Institute for Medical Biochemistry and Molecular Biology, University Medicine Greifswald , Greifswald, Germany
| |
Collapse
|
22
|
The Role of Nitric Oxide from Neurological Disease to Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1007:71-88. [DOI: 10.1007/978-3-319-60733-7_5] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
23
|
Lepka K, Volbracht K, Bill E, Schneider R, Rios N, Hildebrandt T, Ingwersen J, Prozorovski T, Lillig CH, van Horssen J, Steinman L, Hartung HP, Radi R, Holmgren A, Aktas O, Berndt C. Iron-sulfur glutaredoxin 2 protects oligodendrocytes against damage induced by nitric oxide release from activated microglia. Glia 2017; 65:1521-1534. [PMID: 28618115 DOI: 10.1002/glia.23178] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 05/15/2017] [Accepted: 05/24/2017] [Indexed: 02/06/2023]
Abstract
Demyelinated brain lesions, a hallmark of autoimmune neuroinflammatory diseases like multiple sclerosis, result from oligodendroglial cell damage. Activated microglia are considered a major source of nitric oxide and subsequent peroxynitrite-mediated damage of myelin. Here, we provide biochemical and biophysical evidence that the oxidoreductase glutaredoxin 2 inhibits peroxynitrite formation by transforming nitric oxide into dinitrosyl-diglutathionyl-iron-complexes. Glutaredoxin 2 levels influence both survival rates of primary oligodendrocyte progenitor cells and preservation of myelin structure in cerebellar organotypic slice cultures challenged with activated microglia or nitric oxide donors. Of note, glutaredoxin 2-mediated protection is not linked to its enzymatic activity as oxidoreductase, but to the disassembly of its uniquely coordinated iron-sulfur cluster using glutathione as non-protein ligand. The protective effect of glutaredoxin 2 is connected to decreased protein carbonylation and nitration. In line, brain lesions of mice suffering from experimental autoimmune encephalomyelitis, an animal model of multiple sclerosis, show decreased glutaredoxin 2 expression and increased nitrotyrosine formation indicating that this type of protection is missing in the inflamed central nervous system. Our findings link inorganic biochemistry to neuroinflammation and identify glutaredoxin 2 as a protective factor against neuroinflammation-mediated myelin damage. Thus, improved availability of glutathione-coordinated iron-sulfur clusters emerges as a potential therapeutic approach in inflammatory demyelination.
Collapse
Affiliation(s)
- Klaudia Lepka
- Department of Neurology, Medical Faculty, Heinrich-Heine Universität, Düsseldorf, 40225, Germany
| | - Katrin Volbracht
- Department of Neurology, Medical Faculty, Heinrich-Heine Universität, Düsseldorf, 40225, Germany
| | - Eckhard Bill
- Max-Planck-Institut für Chemische Energiekonversion, Mülheim/Ruhr, 45470, Germany
| | - Reiner Schneider
- Department of Neurology, Medical Faculty, Heinrich-Heine Universität, Düsseldorf, 40225, Germany
| | - Natalia Rios
- Departmento de Bioquímica and Center for Free Radical and Biomedical Research, Facultad de Medicina, Universidad de la República, Montevideo, CP 11800, Uruguay
| | - Thomas Hildebrandt
- Department of Neurology, Medical Faculty, Heinrich-Heine Universität, Düsseldorf, 40225, Germany
| | - Jens Ingwersen
- Department of Neurology, Medical Faculty, Heinrich-Heine Universität, Düsseldorf, 40225, Germany
| | - Timur Prozorovski
- Department of Neurology, Medical Faculty, Heinrich-Heine Universität, Düsseldorf, 40225, Germany
| | - Christopher Horst Lillig
- Universitätsmedizin Greifswald, Institute for Medical Biochemistry and Molecular Biology, Greifswald, 17475, Germany
| | - Jack van Horssen
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam, MB, 1007, The Netherlands
| | - Lawrence Steinman
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, 94305-5316, USA
| | - Hans-Peter Hartung
- Department of Neurology, Medical Faculty, Heinrich-Heine Universität, Düsseldorf, 40225, Germany
| | - Rafael Radi
- Departmento de Bioquímica and Center for Free Radical and Biomedical Research, Facultad de Medicina, Universidad de la República, Montevideo, CP 11800, Uruguay
| | - Arne Holmgren
- Department for Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, 17177, Sweden
| | - Orhan Aktas
- Department of Neurology, Medical Faculty, Heinrich-Heine Universität, Düsseldorf, 40225, Germany
| | - Carsten Berndt
- Department of Neurology, Medical Faculty, Heinrich-Heine Universität, Düsseldorf, 40225, Germany
| |
Collapse
|
24
|
Vanin AF, Ostrovskaya LA, Korman DB, Rykova VA, Blyuchterova NV, Fomina MM. The antitumor effect of dinitrosyl iron complexes with glutathione in a murine solid-tumor model. Biophysics (Nagoya-shi) 2017. [DOI: 10.1134/s000635091703023x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
25
|
Lewandowska H, Sadło J, Męczyńska S, Stępkowski TM, Wójciuk G, Kruszewski M. Formation of glutathionyl dinitrosyl iron complexes protects against iron genotoxicity. Dalton Trans 2015; 44:12640-52. [PMID: 26079708 DOI: 10.1039/c5dt00927h] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Dinitrosyl iron(i) complexes (DNICs), intracellular NO donors, are important factors in nitric oxide-dependent regulation of cellular metabolism and signal transduction. It has been shown that NO diminishes the toxicity of iron ions and vice versa. To gain insight into the possible role of DNIC in this phenomenon, we examined the effect of GS-DNIC formation on the ability of iron ions to mediate DNA damage, by treatment of the pUC19 plasmid with physiologically relevant concentrations of GS-DNIC. It was shown that GS-DNIC formation protects against the genotoxic effect of iron ions alone and iron ions in the presence of a naturally abundant antioxidant, GSH. This sheds new light on the iron-related protective effect of NO under the circumstances of oxidative stress.
Collapse
Affiliation(s)
- Hanna Lewandowska
- The Institute of Nuclear Chemistry and Technology, 16 Dorodna Str., 03-195 Warsaw, Poland.
| | | | | | | | | | | |
Collapse
|
26
|
Kus K, Walczak M, Maslak E, Zakrzewska A, Gonciarz-Dytman A, Zabielski P, Sitek B, Wandzel K, Kij A, Chabowski A, Holland RJ, Saavedra JE, Keefer LK, Chlopicki S. Hepatoselective Nitric Oxide (NO) Donors, V-PYRRO/NO and V-PROLI/NO, in Nonalcoholic Fatty Liver Disease: A Comparison of Antisteatotic Effects with the Biotransformation and Pharmacokinetics. Drug Metab Dispos 2015; 43:1028-36. [PMID: 25870102 PMCID: PMC11024901 DOI: 10.1124/dmd.115.063388] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2015] [Accepted: 04/09/2015] [Indexed: 04/20/2024] Open
Abstract
V-PYRRO/NO [O(2)-vinyl-1-(pyrrolidin-1-yl)diazen-1-ium-1,2-diolate] and V-PROLI/NO (O2-vinyl-[2-(carboxylato)pyrrolidin-1-yl]diazen-1-ium-1,2-diolate), two structurally similar diazeniumdiolate derivatives, were designed as liver-selective prodrugs that are metabolized by cytochrome P450 isoenzymes, with subsequent release of nitric oxide (NO). Yet, their efficacy in the treatment of nonalcoholic fatty liver disease (NAFLD) and their comparative pharmacokinetic and metabolic profiles have not been characterized. The aim of the present work was to compare the effects of V-PYRRO/NO and V-PROLI/NO on liver steatosis, glucose tolerance, and liver fatty acid composition in C57BL/6J mice fed a high-fat diet, as well as to comprehensively characterize the ADME (absorption, distribution, metabolism and excretion) profiles of both NO donors. Despite their similar structure, V-PYRRO/NO and V-PROLI/NO showed differences in pharmacological efficacy in the murine model of NAFLD. V-PYRRO/NO, but not V-PROLI/NO, attenuated liver steatosis, improved glucose tolerance, and favorably modified fatty acid composition in the liver. Both compounds were characterized by rapid absorption following i.p. administration, rapid elimination from the body, and incomplete bioavailability. However, V-PYRRO/NO was eliminated mainly by the liver, whereas V-PROLI/NO was excreted mostly in unchanged form by the kidney. V-PYRRO/NO was metabolized by CYP2E1, CYP2C9, CYP1A2, and CYP3A4, whereas V-PROLI/NO was metabolized mainly by CYP1A2. Importantly, V-PYRRO/NO was a better NO releaser in vivo and in the isolated, perfused liver than V-PROLI/NO, an effect compatible with the superior antisteatotic activity of V-PYRRO/NO. In conclusion, V-PYRRO/NO displayed a pronounced antisteatotic effect associated with liver-targeted NO release, whereas V-PROLI/NO showed low effectiveness, was not taken up by the liver, and was eliminated mostly in unchanged form by the kidney.
Collapse
Affiliation(s)
- Kamil Kus
- Jagiellonian Centre for Experimental Therapeutics (K.K., M.W., E.M., A.Z., A.G.-D., B.S., K.W., A.K., S.Ch.), Department of Pharmacokinetics and Physical Pharmacy, Medical College (K.K., M.W., A.G.-D., A.K.), and Department of Experimental Pharmacology, Chair of Pharmacology, Medical College (S.Ch.), Jagiellonian University, Krakow, Poland; Department of Physiology, Medical University of Bialystok, Bialystok, Poland (P.Z., A.Ch.); Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland (J.E.S.); and Chemical Biology Laboratory, National Cancer Institute, Frederick, Maryland (R.J.H., L.K.K.)
| | - Maria Walczak
- Jagiellonian Centre for Experimental Therapeutics (K.K., M.W., E.M., A.Z., A.G.-D., B.S., K.W., A.K., S.Ch.), Department of Pharmacokinetics and Physical Pharmacy, Medical College (K.K., M.W., A.G.-D., A.K.), and Department of Experimental Pharmacology, Chair of Pharmacology, Medical College (S.Ch.), Jagiellonian University, Krakow, Poland; Department of Physiology, Medical University of Bialystok, Bialystok, Poland (P.Z., A.Ch.); Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland (J.E.S.); and Chemical Biology Laboratory, National Cancer Institute, Frederick, Maryland (R.J.H., L.K.K.)
| | - Edyta Maslak
- Jagiellonian Centre for Experimental Therapeutics (K.K., M.W., E.M., A.Z., A.G.-D., B.S., K.W., A.K., S.Ch.), Department of Pharmacokinetics and Physical Pharmacy, Medical College (K.K., M.W., A.G.-D., A.K.), and Department of Experimental Pharmacology, Chair of Pharmacology, Medical College (S.Ch.), Jagiellonian University, Krakow, Poland; Department of Physiology, Medical University of Bialystok, Bialystok, Poland (P.Z., A.Ch.); Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland (J.E.S.); and Chemical Biology Laboratory, National Cancer Institute, Frederick, Maryland (R.J.H., L.K.K.)
| | - Agnieszka Zakrzewska
- Jagiellonian Centre for Experimental Therapeutics (K.K., M.W., E.M., A.Z., A.G.-D., B.S., K.W., A.K., S.Ch.), Department of Pharmacokinetics and Physical Pharmacy, Medical College (K.K., M.W., A.G.-D., A.K.), and Department of Experimental Pharmacology, Chair of Pharmacology, Medical College (S.Ch.), Jagiellonian University, Krakow, Poland; Department of Physiology, Medical University of Bialystok, Bialystok, Poland (P.Z., A.Ch.); Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland (J.E.S.); and Chemical Biology Laboratory, National Cancer Institute, Frederick, Maryland (R.J.H., L.K.K.)
| | - Anna Gonciarz-Dytman
- Jagiellonian Centre for Experimental Therapeutics (K.K., M.W., E.M., A.Z., A.G.-D., B.S., K.W., A.K., S.Ch.), Department of Pharmacokinetics and Physical Pharmacy, Medical College (K.K., M.W., A.G.-D., A.K.), and Department of Experimental Pharmacology, Chair of Pharmacology, Medical College (S.Ch.), Jagiellonian University, Krakow, Poland; Department of Physiology, Medical University of Bialystok, Bialystok, Poland (P.Z., A.Ch.); Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland (J.E.S.); and Chemical Biology Laboratory, National Cancer Institute, Frederick, Maryland (R.J.H., L.K.K.)
| | - Piotr Zabielski
- Jagiellonian Centre for Experimental Therapeutics (K.K., M.W., E.M., A.Z., A.G.-D., B.S., K.W., A.K., S.Ch.), Department of Pharmacokinetics and Physical Pharmacy, Medical College (K.K., M.W., A.G.-D., A.K.), and Department of Experimental Pharmacology, Chair of Pharmacology, Medical College (S.Ch.), Jagiellonian University, Krakow, Poland; Department of Physiology, Medical University of Bialystok, Bialystok, Poland (P.Z., A.Ch.); Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland (J.E.S.); and Chemical Biology Laboratory, National Cancer Institute, Frederick, Maryland (R.J.H., L.K.K.)
| | - Barbara Sitek
- Jagiellonian Centre for Experimental Therapeutics (K.K., M.W., E.M., A.Z., A.G.-D., B.S., K.W., A.K., S.Ch.), Department of Pharmacokinetics and Physical Pharmacy, Medical College (K.K., M.W., A.G.-D., A.K.), and Department of Experimental Pharmacology, Chair of Pharmacology, Medical College (S.Ch.), Jagiellonian University, Krakow, Poland; Department of Physiology, Medical University of Bialystok, Bialystok, Poland (P.Z., A.Ch.); Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland (J.E.S.); and Chemical Biology Laboratory, National Cancer Institute, Frederick, Maryland (R.J.H., L.K.K.)
| | - Krystyna Wandzel
- Jagiellonian Centre for Experimental Therapeutics (K.K., M.W., E.M., A.Z., A.G.-D., B.S., K.W., A.K., S.Ch.), Department of Pharmacokinetics and Physical Pharmacy, Medical College (K.K., M.W., A.G.-D., A.K.), and Department of Experimental Pharmacology, Chair of Pharmacology, Medical College (S.Ch.), Jagiellonian University, Krakow, Poland; Department of Physiology, Medical University of Bialystok, Bialystok, Poland (P.Z., A.Ch.); Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland (J.E.S.); and Chemical Biology Laboratory, National Cancer Institute, Frederick, Maryland (R.J.H., L.K.K.)
| | - Agnieszka Kij
- Jagiellonian Centre for Experimental Therapeutics (K.K., M.W., E.M., A.Z., A.G.-D., B.S., K.W., A.K., S.Ch.), Department of Pharmacokinetics and Physical Pharmacy, Medical College (K.K., M.W., A.G.-D., A.K.), and Department of Experimental Pharmacology, Chair of Pharmacology, Medical College (S.Ch.), Jagiellonian University, Krakow, Poland; Department of Physiology, Medical University of Bialystok, Bialystok, Poland (P.Z., A.Ch.); Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland (J.E.S.); and Chemical Biology Laboratory, National Cancer Institute, Frederick, Maryland (R.J.H., L.K.K.)
| | - Adrian Chabowski
- Jagiellonian Centre for Experimental Therapeutics (K.K., M.W., E.M., A.Z., A.G.-D., B.S., K.W., A.K., S.Ch.), Department of Pharmacokinetics and Physical Pharmacy, Medical College (K.K., M.W., A.G.-D., A.K.), and Department of Experimental Pharmacology, Chair of Pharmacology, Medical College (S.Ch.), Jagiellonian University, Krakow, Poland; Department of Physiology, Medical University of Bialystok, Bialystok, Poland (P.Z., A.Ch.); Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland (J.E.S.); and Chemical Biology Laboratory, National Cancer Institute, Frederick, Maryland (R.J.H., L.K.K.)
| | - Ryan J Holland
- Jagiellonian Centre for Experimental Therapeutics (K.K., M.W., E.M., A.Z., A.G.-D., B.S., K.W., A.K., S.Ch.), Department of Pharmacokinetics and Physical Pharmacy, Medical College (K.K., M.W., A.G.-D., A.K.), and Department of Experimental Pharmacology, Chair of Pharmacology, Medical College (S.Ch.), Jagiellonian University, Krakow, Poland; Department of Physiology, Medical University of Bialystok, Bialystok, Poland (P.Z., A.Ch.); Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland (J.E.S.); and Chemical Biology Laboratory, National Cancer Institute, Frederick, Maryland (R.J.H., L.K.K.)
| | - Joseph E Saavedra
- Jagiellonian Centre for Experimental Therapeutics (K.K., M.W., E.M., A.Z., A.G.-D., B.S., K.W., A.K., S.Ch.), Department of Pharmacokinetics and Physical Pharmacy, Medical College (K.K., M.W., A.G.-D., A.K.), and Department of Experimental Pharmacology, Chair of Pharmacology, Medical College (S.Ch.), Jagiellonian University, Krakow, Poland; Department of Physiology, Medical University of Bialystok, Bialystok, Poland (P.Z., A.Ch.); Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland (J.E.S.); and Chemical Biology Laboratory, National Cancer Institute, Frederick, Maryland (R.J.H., L.K.K.)
| | - Larry K Keefer
- Jagiellonian Centre for Experimental Therapeutics (K.K., M.W., E.M., A.Z., A.G.-D., B.S., K.W., A.K., S.Ch.), Department of Pharmacokinetics and Physical Pharmacy, Medical College (K.K., M.W., A.G.-D., A.K.), and Department of Experimental Pharmacology, Chair of Pharmacology, Medical College (S.Ch.), Jagiellonian University, Krakow, Poland; Department of Physiology, Medical University of Bialystok, Bialystok, Poland (P.Z., A.Ch.); Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland (J.E.S.); and Chemical Biology Laboratory, National Cancer Institute, Frederick, Maryland (R.J.H., L.K.K.)
| | - Stefan Chlopicki
- Jagiellonian Centre for Experimental Therapeutics (K.K., M.W., E.M., A.Z., A.G.-D., B.S., K.W., A.K., S.Ch.), Department of Pharmacokinetics and Physical Pharmacy, Medical College (K.K., M.W., A.G.-D., A.K.), and Department of Experimental Pharmacology, Chair of Pharmacology, Medical College (S.Ch.), Jagiellonian University, Krakow, Poland; Department of Physiology, Medical University of Bialystok, Bialystok, Poland (P.Z., A.Ch.); Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland (J.E.S.); and Chemical Biology Laboratory, National Cancer Institute, Frederick, Maryland (R.J.H., L.K.K.)
| |
Collapse
|
27
|
Beyazit Y, Kocak E, Tanoglu A, Kekilli M. Oxidative stress might play a role in low serum vitamin D associated liver fibrosis among patients with autoimmune hepatitis. Dig Dis Sci 2015; 60:1106-8. [PMID: 25586086 DOI: 10.1007/s10620-015-3526-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2014] [Accepted: 01/05/2015] [Indexed: 12/09/2022]
Affiliation(s)
- Yavuz Beyazit
- Department of Gastroenterology, Canakkale State Hospital, Canakkale, Turkey
| | | | | | | |
Collapse
|
28
|
Nitrosative stress and apoptosis in non-anemic healthy rats induced by intravenous iron sucrose similars versus iron sucrose originator. Biometals 2015; 28:279-92. [DOI: 10.1007/s10534-015-9822-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Accepted: 01/07/2015] [Indexed: 01/01/2023]
|
29
|
Pereira JCM, Iretskii AV, Han RM, Ford PC. Dinitrosyl Iron Complexes with Cysteine. Kinetics Studies of the Formation and Reactions of DNICs in Aqueous Solution. J Am Chem Soc 2014; 137:328-36. [DOI: 10.1021/ja510393q] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- José Clayston Melo Pereira
- Department
of Chemistry and Biochemistry, University of California, Santa Barbara, Santa
Barbara, California 93106-9510, United States
- Departamento
de Química Geral e Inorgânica, Instituto de Química
de Araraquara, UNESP − Universidade Estadual Paulista, Araraquara, São Paulo 14801−970, Brazil
| | - Alexei V. Iretskii
- Department
of Chemistry and Biochemistry, University of California, Santa Barbara, Santa
Barbara, California 93106-9510, United States
- Department
of Chemistry and Environmental Sciences, Lake Superior State University, Sault Sainte Marie, Michigan 49783, United States
| | - Rui-Min Han
- Department
of Chemistry and Biochemistry, University of California, Santa Barbara, Santa
Barbara, California 93106-9510, United States
- Department
of Chemistry, Renmin University of China, 59 ZhongGuanCun St., Beijing, 100872, China
| | - Peter C. Ford
- Department
of Chemistry and Biochemistry, University of California, Santa Barbara, Santa
Barbara, California 93106-9510, United States
| |
Collapse
|
30
|
Lok HC, Sahni S, Richardson V, Kalinowski DS, Kovacevic Z, Lane DJR, Richardson DR. Glutathione S-transferase and MRP1 form an integrated system involved in the storage and transport of dinitrosyl-dithiolato iron complexes in cells. Free Radic Biol Med 2014; 75:14-29. [PMID: 25035074 DOI: 10.1016/j.freeradbiomed.2014.07.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Revised: 06/26/2014] [Accepted: 07/01/2014] [Indexed: 12/11/2022]
Abstract
Nitrogen monoxide (NO) is vital for many essential biological processes as a messenger and effector molecule. The physiological importance of NO is the result of its high affinity for iron in the active sites of proteins such as guanylate cyclase. Indeed, NO possesses a rich coordination chemistry with iron and the formation of dinitrosyl-dithiolato iron complexes (DNICs) is well documented. In mammals, NO generated by cytotoxic activated macrophages has been reported to play a role as a cytotoxic effector against tumor cells by binding and releasing intracellular iron. Studies from our laboratory have shown that two proteins traditionally involved in drug resistance, namely multidrug-resistance protein 1 and glutathione S-transferase, play critical roles in intracellular NO transport and storage through their interaction with DNICs (R.N. Watts et al., Proc. Natl. Acad. Sci. USA 103:7670-7675, 2006; H. Lok et al., J. Biol. Chem. 287:607-618, 2012). Notably, DNICs are present at high concentrations in cells and are biologically available. These complexes have a markedly longer half-life than free NO, making them an ideal "common currency" for this messenger molecule. Considering the many critical roles NO plays in health and disease, a better understanding of its intracellular trafficking mechanisms will be vital for the development of new therapeutics.
Collapse
Affiliation(s)
- H C Lok
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, NSW 2006, Australia
| | - S Sahni
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, NSW 2006, Australia
| | - V Richardson
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, NSW 2006, Australia
| | - D S Kalinowski
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, NSW 2006, Australia
| | - Z Kovacevic
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, NSW 2006, Australia
| | - D J R Lane
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, NSW 2006, Australia
| | - D R Richardson
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, NSW 2006, Australia.
| |
Collapse
|
31
|
Muñoz-Sánchez J, Chánez-Cárdenas ME. A review on hemeoxygenase-2: focus on cellular protection and oxygen response. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2014; 2014:604981. [PMID: 25136403 PMCID: PMC4127239 DOI: 10.1155/2014/604981] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Accepted: 04/30/2014] [Indexed: 12/13/2022]
Abstract
Hemeoxygenase (HO) system is responsible for cellular heme degradation to biliverdin, iron, and carbon monoxide. Two isoforms have been reported to date. Homologous HO-1 and HO-2 are microsomal proteins with more than 45% residue identity, share a similar fold and catalyze the same reaction. However, important differences between isoforms also exist. HO-1 isoform has been extensively studied mainly by its ability to respond to cellular stresses such as hemin, nitric oxide donors, oxidative damage, hypoxia, hyperthermia, and heavy metals, between others. On the contrary, due to its apparently constitutive nature, HO-2 has been less studied. Nevertheless, its abundance in tissues such as testis, endothelial cells, and particularly in brain, has pointed the relevance of HO-2 function. HO-2 presents particular characteristics that made it a unique protein in the HO system. Since attractive results on HO-2 have been arisen in later years, we focused this review in the second isoform. We summarize information on gene description, protein structure, and catalytic activity of HO-2 and particular facts such as its cellular impact and activity regulation. Finally, we call attention on the role of HO-2 in oxygen sensing, discussing proposed hypothesis on heme binding motifs and redox/thiol switches that participate in oxygen sensing as well as evidences of HO-2 response to hypoxia.
Collapse
Affiliation(s)
- Jorge Muñoz-Sánchez
- Laboratorio de Patología Vascular Cerebral, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, 14269 Delegación Tlalpan, DF, Mexico
| | - María Elena Chánez-Cárdenas
- Laboratorio de Patología Vascular Cerebral, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, 14269 Delegación Tlalpan, DF, Mexico
| |
Collapse
|
32
|
Borodulin RR, Dereven'kov IА, Burbaev DS, Makarov SV, Mikoyan VD, Serezhenkov VА, Kubrina LN, Ivanovic-Burmazovic I, Vanin AF. Redox activities of mono- and binuclear forms of low-molecular and protein-bound dinitrosyl iron complexes with thiol-containing ligands. Nitric Oxide 2014; 40:100-9. [PMID: 24997418 DOI: 10.1016/j.niox.2014.06.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Revised: 06/05/2014] [Accepted: 06/12/2014] [Indexed: 10/25/2022]
Abstract
EPR, optical, electrochemical and stopped-flow methods were used to demonstrate that Fe(NO)2 fragments in paramagnetic mononuclear and diamagnetic binuclear forms of dinitrosyl iron complexes with glutathione are reversibly reduced by a two-electron mechanism to be further transformed from the initial state with d(7) configuration into states with the d(8) and d(9) electronic configurations of the iron atom. Under these conditions, both forms of DNIC display identical optical and EPR characteristics in state d(9) suggesting that reduction of the binuclear form of DNIC initiates their reversible decomposition into two mononuclear dinitrosyl iron fragments, one of which is EPR-silent (d(8)) and the other one is EPR-active (d(9)). Both forms of DNIC produce EPR signals with the following values of the g-factor: g⊥=2.01, g||=1.97, gaver.=2.0. M-DNIC with glutathione manifest an ability to pass into state d(9), however, only in solutions with a low content of free glutathione. Similar transitions were established for protein-bound М- and B-DNIC with thiol-containing ligands.
Collapse
Affiliation(s)
- Rostislav R Borodulin
- N.N.Semenov Institute of Chemical Physics, Russian Academy of Sciences, Moscow, Russia
| | - Ilia А Dereven'kov
- Ivanovo State University of Chemistry and Technology, Ivanovo, Russia; Department of Chemistry and Pharmacy, University of Erlangen-Nürnberg, Germany
| | - Dosymzhan Sh Burbaev
- N.N.Semenov Institute of Chemical Physics, Russian Academy of Sciences, Moscow, Russia
| | - Sergei V Makarov
- Ivanovo State University of Chemistry and Technology, Ivanovo, Russia
| | - Vasak D Mikoyan
- N.N.Semenov Institute of Chemical Physics, Russian Academy of Sciences, Moscow, Russia
| | | | - Lyudmila N Kubrina
- N.N.Semenov Institute of Chemical Physics, Russian Academy of Sciences, Moscow, Russia
| | | | - Anatoly F Vanin
- N.N.Semenov Institute of Chemical Physics, Russian Academy of Sciences, Moscow, Russia.
| |
Collapse
|
33
|
Burgova EN, Tkachev NА, Adamyan LV, Mikoyan VD, Paklina OV, Stepanyan AA, Vanin AF. Dinitrosyl iron complexes with glutathione suppress experimental endometriosis in rats. Eur J Pharmacol 2014; 727:140-7. [DOI: 10.1016/j.ejphar.2014.01.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Revised: 12/20/2013] [Accepted: 01/07/2014] [Indexed: 11/26/2022]
|
34
|
Niedbala W, Besnard AG, Jiang HR, Alves-Filho JC, Fukada SY, Nascimento D, Mitani A, Pushparaj P, Alqahtani MH, Liew FY. Nitric oxide-induced regulatory T cells inhibit Th17 but not Th1 cell differentiation and function. THE JOURNAL OF IMMUNOLOGY 2013; 191:164-70. [PMID: 23720815 DOI: 10.4049/jimmunol.1202580] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
NO is a free radical with pleiotropic functions. We have shown earlier that NO induces a population of CD4(+)CD25(+)Foxp3(-) regulatory T cells (NO-Tregs) that suppress the functions of CD4(+)CD25(-) effector T cells in vitro and in vivo. We report in this study an unexpected finding that NO-Tregs suppressed Th17 but not Th1 cell differentiation and function. In contrast, natural Tregs (nTregs), which suppressed Th1 cells, failed to suppress Th17 cells. Consistent with this observation, NO-Tregs inhibited the expression of retinoic acid-related orphan receptor γt but not T-bet, whereas nTregs suppressed T-bet but not retinoic acid-related orphan receptor γt expression. The NO-Treg-mediated suppression of Th17 was partially cell contact-dependent and was associated with IL-10. In vivo, adoptively transferred NO-Tregs potently attenuated experimental autoimmune encephalomyelitis. The disease suppression was accompanied by a reduction of Th17, but not Th1 cells in the draining lymph nodes, and a decrease in the production of IL-17, but an increase in IL-10 synthesis. Our results therefore demonstrate the differential suppressive function between NO-Tregs and nTregs and indicate specialization of the regulatory mechanism of the immune system.
Collapse
Affiliation(s)
- Wanda Niedbala
- Centre of Immunology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Fu J, Liu L, Huang Z, Lai Y, Ji H, Peng S, Tian J, Zhang Y. Hybrid Molecule from O2-(2,4-Dinitrophenyl)diazeniumdiolate and Oleanolic Acid: A Glutathione S-Transferase π-Activated Nitric Oxide Prodrug with Selective Anti-Human Hepatocellular Carcinoma Activity and Improved Stability. J Med Chem 2013; 56:4641-55. [DOI: 10.1021/jm400393u] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Junjie Fu
- State Key
Laboratory of Natural
Medicines, China Pharmaceutical University, Nanjing 210009, PR China
- Center of Drug Discovery, China Pharmaceutical University, Nanjing 210009, PR
China
| | - Ling Liu
- State Key
Laboratory of Natural
Medicines, China Pharmaceutical University, Nanjing 210009, PR China
- Department of Pharmacology,
Medical
College, Henan University of Science and Technology, Luoyang 471003, PR China
| | - Zhangjian Huang
- State Key
Laboratory of Natural
Medicines, China Pharmaceutical University, Nanjing 210009, PR China
- Center of Drug Discovery, China Pharmaceutical University, Nanjing 210009, PR
China
| | - Yisheng Lai
- State Key
Laboratory of Natural
Medicines, China Pharmaceutical University, Nanjing 210009, PR China
- Center of Drug Discovery, China Pharmaceutical University, Nanjing 210009, PR
China
| | - Hui Ji
- State Key
Laboratory of Natural
Medicines, China Pharmaceutical University, Nanjing 210009, PR China
| | - Sixun Peng
- State Key
Laboratory of Natural
Medicines, China Pharmaceutical University, Nanjing 210009, PR China
- Center of Drug Discovery, China Pharmaceutical University, Nanjing 210009, PR
China
| | - Jide Tian
- Department of Molecular and
Medical Pharmacology, University of California, Los Angeles, California 90095, United States
| | - Yihua Zhang
- State Key
Laboratory of Natural
Medicines, China Pharmaceutical University, Nanjing 210009, PR China
- Center of Drug Discovery, China Pharmaceutical University, Nanjing 210009, PR
China
| |
Collapse
|
36
|
|
37
|
Vanin AF, Borodulin RR, Kubrina LN, Mikoyan VD, Burbaev DS. Physicochemical features of dinitrosyl iron complexes with natural thiol-containing ligands underlying the biological activities of these complexes. Biophysics (Nagoya-shi) 2013. [DOI: 10.1134/s0006350913010168] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
38
|
Neal MD, Raval JS, Triulzi DJ, Simmons RL. Innate immune activation after transfusion of stored red blood cells. Transfus Med Rev 2013; 27:113-8. [PMID: 23434246 DOI: 10.1016/j.tmrv.2013.01.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2012] [Revised: 11/26/2012] [Accepted: 01/02/2013] [Indexed: 01/28/2023]
Abstract
The transfusion of red blood cells (RBCs), although necessary for treatment of anemia and blood loss, has also been linked to increased morbidity and mortality. RBCs stored for longer durations and transfused in larger volumes are often cited as contributory to adverse outcomes. The potential mechanisms underlying deleterious effects of RBC transfusion are just beginning to be elucidated. In this narrative review, we explore the hypothesis that prolonged RBC storage results in elaboration of substances which may function as danger associated molecular pattern molecules that activate the innate immune system with consequences unfavorable to healthy homeostasis. The nature of these chemical mediators and the biological responses to them offers insight into the mechanisms of these pathological responses. Three major areas of activation of the innate immune apparatus by stored RBCs have been tentatively identified: RBC hemolysis, recipient neutrophil priming, and reactive oxygen species production. The possible mechanisms by which each might perturb the innate immune response are reviewed in a search for potential novel pathways through which transfusion can lead to an altered inflammatory response.
Collapse
Affiliation(s)
- Matthew D Neal
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | | | | | | |
Collapse
|
39
|
Sengupta R, Holmgren A. Thioredoxin and thioredoxin reductase in relation to reversible S-nitrosylation. Antioxid Redox Signal 2013; 18:259-69. [PMID: 22702224 DOI: 10.1089/ars.2012.4716] [Citation(s) in RCA: 116] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
SIGNIFICANCE Nitric oxide (NO) regulates a diverse range of cellular processes, including vasodilation, neurotransmission, and antimicrobial and anti-tumor activities. S-nitrosylation with the formation of S-nitrosothiols (RSNOs) is an important feature of NO signaling regulating protein function. In mammalian cells, glutathione (GSH), S-nitrosoglutathione reductase (GSNOR), and thioredoxin (Trx) have been identified as the major protein denitrosylases. RECENT ADVANCES Human cytosolic/nuclear Trx1 in the disulfide form can be nitrosylated at Cys73 and transnitrosylate target proteins, including caspase 3. Thus, similar to GSH, which by forming S-nitrosoglutathione (GSNO) can transnitrosylate proteins, Trx can either denitrosylate or nitrosylate proteins depending on its oxidation state. CRITICAL ISSUES In this review, we discuss the regulation of cellular processes by reversible S-nitrosylation and Trx-mediated cellular homeostasis of RSNOs and S-nitrosoproteins. FUTURE DIRECTIONS Functions of RSNOs in vivo and their pharmacological uses have not yet been fully studied. Further investigations on the role of Trx systems in relation to biologically relevant RSNOs, their functions, and the mechanisms of denitrosylation will facilitate the development of drugs and therapies. Antioxid. Redox Signal. 18, 259-269.
Collapse
Affiliation(s)
- Rajib Sengupta
- Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | | |
Collapse
|
40
|
Mechanisms of Nitric Oxide Reactions Mediated by Biologically Relevant Metal Centers. NITROSYL COMPLEXES IN INORGANIC CHEMISTRY, BIOCHEMISTRY AND MEDICINE II 2013. [DOI: 10.1007/430_2013_117] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
41
|
Gardner PR. Hemoglobin: a nitric-oxide dioxygenase. SCIENTIFICA 2012; 2012:683729. [PMID: 24278729 PMCID: PMC3820574 DOI: 10.6064/2012/683729] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2012] [Accepted: 10/04/2012] [Indexed: 05/09/2023]
Abstract
Members of the hemoglobin superfamily efficiently catalyze nitric-oxide dioxygenation, and when paired with native electron donors, function as NO dioxygenases (NODs). Indeed, the NOD function has emerged as a more common and ancient function than the well-known role in O2 transport-storage. Novel hemoglobins possessing a NOD function continue to be discovered in diverse life forms. Unique hemoglobin structures evolved, in part, for catalysis with different electron donors. The mechanism of NOD catalysis by representative single domain hemoglobins and multidomain flavohemoglobin occurs through a multistep mechanism involving O2 migration to the heme pocket, O2 binding-reduction, NO migration, radical-radical coupling, O-atom rearrangement, nitrate release, and heme iron re-reduction. Unraveling the physiological functions of multiple NODs with varying expression in organisms and the complexity of NO as both a poison and signaling molecule remain grand challenges for the NO field. NOD knockout organisms and cells expressing recombinant NODs are helping to advance our understanding of NO actions in microbial infection, plant senescence, cancer, mitochondrial function, iron metabolism, and tissue O2 homeostasis. NOD inhibitors are being pursued for therapeutic applications as antibiotics and antitumor agents. Transgenic NOD-expressing plants, fish, algae, and microbes are being developed for agriculture, aquaculture, and industry.
Collapse
Affiliation(s)
- Paul R. Gardner
- Miami Valley Biotech, 1001 E. 2nd Street, Suite 2445, Dayton, OH 45402, USA
| |
Collapse
|
42
|
Yuan L, Wang J, Xiao H, Xiao C, Wang Y, Liu X. Isoorientin induces apoptosis through mitochondrial dysfunction and inhibition of PI3K/Akt signaling pathway in HepG2 cancer cells. Toxicol Appl Pharmacol 2012; 265:83-92. [PMID: 23026832 DOI: 10.1016/j.taap.2012.09.022] [Citation(s) in RCA: 88] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2012] [Revised: 09/19/2012] [Accepted: 09/21/2012] [Indexed: 12/21/2022]
Abstract
Isoorientin (ISO) is a flavonoid compound that can be extracted from several plant species, such as Phyllostachys pubescens, Patrinia, and Drosophyllum lusitanicum; however, its biological activity remains poorly understood. The present study investigated the effects and putative mechanism of apoptosis induced by ISO in human hepatoblastoma cancer (HepG2) cells. The results showed that ISO induced cell death in a dose-dependent manner in HepG2 cells, but no toxicity in human liver cells (HL-7702) and buffalo rat liver cells (BRL-3A) treated with ISO at the indicated concentrations. ISO-induced cell death included apoptosis which characterized by the appearance of nuclear shrinkage, the cleavage of poly (ADP-ribose) polymerase (PARP) and DNA fragmentation. ISO significantly (p<0.01) increased the Bax/Bcl-2 ratio, disrupted the mitochondrial membrane potential (MMP), increased the release of cytochrome c, activated caspase-3, and enhanced intracellular levels of reactive oxygen species (ROS) and nitric oxide (NO). In addition, ISO effectively inhibited the phosphorylation of Akt and increased FoxO4 expression. The PI3K/Akt inhibitor LY294002 enhanced the apoptosis-inducing effect of ISO. However, LY294002 markedly quenched ROS and NO generation and diminished the protein expression of heme peroxidase enzyme (HO-1) and inducible nitric oxide synthase (iNOS). Furthermore, the addition of a ROS inhibitor (N-acetyl cysteine, NAC) or iNOS inhibitor (N-[3-(aminomethyl) benzyl] acetamidine, dihydrochloride, 1400W) significantly diminished the apoptosis induced by ISO and also blocked the phosphorylation of Akt. These results demonstrated for the first time that ISO induces apoptosis in HepG2 cells and indicate that this apoptosis might be mediated through mitochondrial dysfunction and PI3K/Akt signaling pathway, and has no toxicity in normal liver cells, suggesting that ISO may have good potential as a therapeutic and chemopreventive agent for liver cancer.
Collapse
Affiliation(s)
- Li Yuan
- Laboratory of Nutrition and Functional Factors, College of Food Science and Engineering, Northwest A & F University, Yangling, China
| | | | | | | | | | | |
Collapse
|
43
|
Gelam honey scavenges peroxynitrite during the immune response. Int J Mol Sci 2012; 13:12113-12129. [PMID: 23109904 PMCID: PMC3472796 DOI: 10.3390/ijms130912113] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2012] [Revised: 09/06/2012] [Accepted: 09/09/2012] [Indexed: 01/12/2023] Open
Abstract
Monocytes and macrophages are part of the first-line defense against bacterial, fungal, and viral infections during host immune responses; they express high levels of proinflammatory cytokines and cytotoxic molecules, including nitric oxide, reactive oxygen species, and their reaction product peroxynitrite. Peroxynitrite is a short-lived oxidant and a potent inducer of cell death. Honey, in addition to its well-known sweetening properties, is a natural antioxidant that has been used since ancient times in traditional medicine. We examined the ability of Gelam honey, derived from the Gelam tree (Melaleuca spp.), to scavenge peroxynitrite during immune responses mounted in the murine macrophage cell line RAW 264.7 when stimulated with lipopolysaccharide/interferon-γ (LPS/IFN-γ) and in LPS-treated rats. Gelam honey significantly improved the viability of LPS/IFN-γ-treated RAW 264.7 cells and inhibited nitric oxide production-similar to the effects observed with an inhibitor of inducible nitric oxide synthase (1400W). Furthermore, honey, but not 1400W, inhibited peroxynitrite production from the synthetic substrate 3-morpholinosydnonimine (SIN-1) and prevented the peroxynitrite-mediated conversion of dihydrorhodamine 123 to its fluorescent oxidation product rhodamine 123. Honey inhibited peroxynitrite synthesis in LPS-treated rats. Thus, honey may attenuate inflammatory responses that lead to cell damage and death, suggesting its therapeutic uses for several inflammatory disorders.
Collapse
|
44
|
Jhurry ND, Chakrabarti M, McCormick SP, Holmes-Hampton GP, Lindahl PA. Biophysical investigation of the ironome of human jurkat cells and mitochondria. Biochemistry 2012; 51:5276-84. [PMID: 22726227 DOI: 10.1021/bi300382d] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The speciation of iron in intact human Jurkat leukemic cells and their isolated mitochondria was assessed using biophysical methods. Large-scale cultures were grown in medium enriched with (57)Fe citrate. Mitochondria were isolated anaerobically to prevent oxidation of iron centers. 5 K Mössbauer spectra of cells were dominated by a sextet due to ferritin. They also exhibited an intense central quadrupole doublet due to S = 0 [Fe(4)S(4)](2+) clusters and low-spin (LS) Fe(II) heme centers. Spectra of isolated mitochondria were largely devoid of ferritin but contained the central doublet and features arising from what appear to be Fe(III) oxyhydroxide (phosphate) nanoparticles. Spectra from both cells and mitochondria contained a low-intensity doublet from non-heme high-spin (NHHS) Fe(II) species. A portion of these species may constitute the "labile iron pool" (LIP) proposed in cellular Fe trafficking. Such species might engage in Fenton chemistry to generate reactive oxygen species. Electron paramagnetic resonance spectra of cells and mitochondria exhibited signals from reduced Fe/S clusters, and HS Fe(III) heme and non-heme species. The basal heme redox state of mitochondria within cells was reduced; this redox poise was unaltered during the anaerobic isolation of the organelle. Contributions from heme a, b, and c centers were quantified using electronic absorption spectroscopy. Metal concentrations in cells and mitochondria were measured using inductively coupled plasma mass spectrometry. Results were collectively assessed to estimate the concentrations of various Fe-containing species in mitochondria and whole cells - the first "ironome" profile of a human cell.
Collapse
Affiliation(s)
- Nema D Jhurry
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX 77843-2128, USA
| | | | | | | | | |
Collapse
|
45
|
Burgova EN, Adamyan LV, Tkachev NA, Stepanyan AA, Vanin AF. Dinitrosyl iron complexes with cysteine suppress the development of experimental endometriosis in rats. Biophysics (Nagoya-shi) 2012. [DOI: 10.1134/s0006350912010071] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
46
|
Shupik MA, Vanin AF, Alessenko AV. Interaction of the nitric oxide signaling system with the sphingomyelin cycle and peroxidation on transmission of toxic signal of tumor necrosis factor-α in ischemia-reperfusion. BIOCHEMISTRY (MOSCOW) 2012; 76:1197-209. [PMID: 22117546 DOI: 10.1134/s0006297911110010] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
This review discusses the functional role of nitric oxide in ischemia-reperfusion injury and mechanisms of signal transduction of apoptosis, which accompanies ischemic damage to organs and tissues. On induction of apoptosis an interaction is observed of the nitric oxide signaling system with the sphingomyelin cycle, which is a source of a proapoptotic agent ceramide. Evidence is presented of an interaction of the sphingomyelin cycle enzymes and ceramide with nitric oxide and enzymes synthesizing nitric oxide. The role of a proinflammatory cytokine TNF-α in apoptosis and ischemia-reperfusion and mechanisms of its cytotoxic action, which involve nitric oxide, the sphingomyelin cycle, and lipid peroxidation are discussed. A comprehensive study of these signaling systems provides insight into the molecular mechanism of apoptosis during ischemia and allows us to consider new approaches for treatment of diseases associated with the activation of apoptosis.
Collapse
Affiliation(s)
- M A Shupik
- Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, Moscow, Russia
| | | | | |
Collapse
|
47
|
Konstantinova TS, Bugrova AE, Shevchenko TF, Vanin AF, Kalamkarov GR. Variation of nitric oxide content regulates the development of apoptosis in the retina. Biophysics (Nagoya-shi) 2012. [DOI: 10.1134/s0006350912020121] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
48
|
Sadli N, Ackland ML, De Mel D, Sinclair AJ, Suphioglu C. Effects of zinc and DHA on the epigenetic regulation of human neuronal cells. Cell Physiol Biochem 2012; 29:87-98. [PMID: 22415078 DOI: 10.1159/000337590] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/16/2011] [Indexed: 12/14/2022] Open
Abstract
Dietary intake of zinc and omega-3 fatty acids (DHA) have health benefits for a number of human diseases. However, the molecular basis of these health benefits remains unclear. Recently, we reported that zinc and DHA affect expression levels of histones H3 and H4 in human neuronal M17 cells. Here, using immunoblotting and densitometric analysis, we aimed to investigate the effect of zinc and DHA on post-translational modifications of histone H3 in M17 cells. In response to increase in zinc concentration, we observed increase in deacetylation, methylation and phosphorylation of H3 and decrease in acetylation. We also investigated the role of zinc in apoptosis, and found that zinc reduced the levels of the anti-apoptotic marker Bcl-2 while increasing the apoptotic marker caspase-3 levels, correlating with cell viability assays. Conversely, DHA treatment resulted in increase in acetylation of H3 and Bcl-2 levels and decrease in deacetylation, methylation, phosphorylation of H3 and caspase-3 levels, suggesting that DHA promotes gene expression and neuroprotection. Our novel findings show the opposing effects of zinc and DHA on the epigenetic regulation of human neuronal cells and highlight the potential benefit of dietary intake of DHA for management of neurodegenerative diseases.
Collapse
Affiliation(s)
- Nadia Sadli
- NeuroAllergy Research Laboratory (NARL), School of Life and Environmental Sciences, Deakin University, Geelong, Victoria, Australia
| | | | | | | | | |
Collapse
|
49
|
Saini MK, Sanyal SN, Vaiphei K. Piroxicam and C-phycocyanin mediated apoptosis in 1,2-dimethylhydrazine dihydrochloride induced colon carcinogenesis: exploring the mitochondrial pathway. Nutr Cancer 2012; 64:409-18. [PMID: 22369161 DOI: 10.1080/01635581.2012.655402] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Apoptosis is a synchronized procedure of cell death that is regulated by caspases and proapoptotic proteins. During apoptosis, translocation of cytochrome c, an electron carrier, from mitochondria into the cytosol is regulated by Bcl-2 family members. Cytochrome c in association with an apoptotic protease activating factor (Apaf), a proapoptotic protein essential for cell differentiation and procaspase-9 form the apoptosome complex, which consecutively activates effector caspase, caspase-3, and coordinate the implementation of apoptosis. In the current study, an attempt has been made to gain insight into piroxicam, a traditional nonsteroidal antiinflammatory drug and c-phycocyanin, a biliprotein from Spirulina platensis (cyanobacterium) mediated apoptosis in DMH-induced colon cancer. Male Sprague-Dawley rats were segregated into 5 groups: control, DMH, DMH + piroxicam, DMH + c-phycocyanin, and DMH + piroxicam + c-phycocyanin. Results illustrated that piroxicam and c-phycocyanin treatments stimulate cytochrome c release by downregulating the Bcl-2 (an antiapoptotic protein) expression significantly, while promoting the level of Bax (a proapoptotic protein), thereby activating caspases (caspases-9 and -3) and Apaf-1. The outcomes of the present study clearly signify that piroxicam and c-phycocyanin may mediate mitochondrial-dependent apoptosis in DMH-induced colon cancer. Moreover, apoptosis induction was more apparent in the combination regimen of piroxicam and c-phycocyanin than the individual drugs alone.
Collapse
Affiliation(s)
- Manpreet Kaur Saini
- Department of Histopathology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | | | | |
Collapse
|
50
|
Serezhenkov VA, Kuznetsov IS, Romantsova TI, Kuznetsova MI, Vanin AF. Antidiabetes drug metformin is a donor of nitric oxide: EPR measurement of efficiency. Biophysics (Nagoya-shi) 2012. [DOI: 10.1134/s0006350911060169] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|