1
|
Andrianov VV, Kulchitsky VA, Yafarova GG, Bazan LV, Bogodvid TK, Deryabina IB, Muranova LN, Silantyeva DI, Arslanov AI, Paveliev MN, Fedorova EV, Filipovich TA, Nagibov AV, Gainutdinov KL. Investigation of NO Role in Neural Tissue in Brain and Spinal Cord Injury. Molecules 2023; 28:7359. [PMID: 37959778 PMCID: PMC10650517 DOI: 10.3390/molecules28217359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 10/19/2023] [Accepted: 10/23/2023] [Indexed: 11/15/2023] Open
Abstract
Nitric oxide (NO) production in injured and intact brain regions was compared by EPR spectroscopy in a model of brain and spinal cord injury in Wistar rats. The precentral gyrus of the brain was injured, followed by the spinal cord at the level of the first lumbar vertebra. Seven days after brain injury, a reduction in NO content of 84% in injured brain regions and 66% in intact brain regions was found. The difference in NO production in injured and uninjured brain regions persisted 7 days after injury. The copper content in the brain remained unchanged one week after modeling of brain and spinal cord injury. The data obtained in the experiments help to explain the problems in the therapy of patients with combined brain injury.
Collapse
Affiliation(s)
- Viacheslav V. Andrianov
- Zavoisky Physical-Technical Institute of the Russian Academy of Sciences, 420000 Kazan, Russia; (V.V.A.); (G.G.Y.); (L.V.B.)
- Department of Human and Animals, Institute of Fundamental Medicine and Biology, Kazan Federal University, 420000 Kazan, Russia; (T.K.B.); (I.B.D.); (L.N.M.); (D.I.S.); (A.I.A.)
| | - Vladimir A. Kulchitsky
- Brain Center, Institute of Physiology, National Academy of Sciences, 220012 Minsk, Belarus; (V.A.K.); (E.V.F.); (T.A.F.); (A.V.N.)
| | - Guzel G. Yafarova
- Zavoisky Physical-Technical Institute of the Russian Academy of Sciences, 420000 Kazan, Russia; (V.V.A.); (G.G.Y.); (L.V.B.)
- Department of Human and Animals, Institute of Fundamental Medicine and Biology, Kazan Federal University, 420000 Kazan, Russia; (T.K.B.); (I.B.D.); (L.N.M.); (D.I.S.); (A.I.A.)
| | - Leah V. Bazan
- Zavoisky Physical-Technical Institute of the Russian Academy of Sciences, 420000 Kazan, Russia; (V.V.A.); (G.G.Y.); (L.V.B.)
| | - Tatiana K. Bogodvid
- Department of Human and Animals, Institute of Fundamental Medicine and Biology, Kazan Federal University, 420000 Kazan, Russia; (T.K.B.); (I.B.D.); (L.N.M.); (D.I.S.); (A.I.A.)
- Department of Biomedical Sciences, Volga Region State University of Physical Culture, Sport and Tourism, 420000 Kazan, Russia
| | - Irina B. Deryabina
- Department of Human and Animals, Institute of Fundamental Medicine and Biology, Kazan Federal University, 420000 Kazan, Russia; (T.K.B.); (I.B.D.); (L.N.M.); (D.I.S.); (A.I.A.)
| | - Lyudmila N. Muranova
- Department of Human and Animals, Institute of Fundamental Medicine and Biology, Kazan Federal University, 420000 Kazan, Russia; (T.K.B.); (I.B.D.); (L.N.M.); (D.I.S.); (A.I.A.)
| | - Dinara I. Silantyeva
- Department of Human and Animals, Institute of Fundamental Medicine and Biology, Kazan Federal University, 420000 Kazan, Russia; (T.K.B.); (I.B.D.); (L.N.M.); (D.I.S.); (A.I.A.)
| | - Almaz I. Arslanov
- Department of Human and Animals, Institute of Fundamental Medicine and Biology, Kazan Federal University, 420000 Kazan, Russia; (T.K.B.); (I.B.D.); (L.N.M.); (D.I.S.); (A.I.A.)
| | | | - Ekaterina V. Fedorova
- Brain Center, Institute of Physiology, National Academy of Sciences, 220012 Minsk, Belarus; (V.A.K.); (E.V.F.); (T.A.F.); (A.V.N.)
| | - Tatiana A. Filipovich
- Brain Center, Institute of Physiology, National Academy of Sciences, 220012 Minsk, Belarus; (V.A.K.); (E.V.F.); (T.A.F.); (A.V.N.)
| | - Aleksei V. Nagibov
- Brain Center, Institute of Physiology, National Academy of Sciences, 220012 Minsk, Belarus; (V.A.K.); (E.V.F.); (T.A.F.); (A.V.N.)
| | - Khalil L. Gainutdinov
- Zavoisky Physical-Technical Institute of the Russian Academy of Sciences, 420000 Kazan, Russia; (V.V.A.); (G.G.Y.); (L.V.B.)
- Department of Human and Animals, Institute of Fundamental Medicine and Biology, Kazan Federal University, 420000 Kazan, Russia; (T.K.B.); (I.B.D.); (L.N.M.); (D.I.S.); (A.I.A.)
| |
Collapse
|
2
|
Parshukova OI, Varlamova NG, Potolitsyna NN, Lyudinina AY, Bojko ER. Features of Metabolic Support of Physical Performance in Highly Trained Cross-Country Skiers of Different Qualifications during Physical Activity at Maximum Load. Cells 2021; 11:cells11010039. [PMID: 35011601 PMCID: PMC8750590 DOI: 10.3390/cells11010039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 12/17/2021] [Accepted: 12/21/2021] [Indexed: 11/27/2022] Open
Abstract
The purpose of our study was to identify the features of metabolic regulation in highly trained cross-country skiers of different qualifications at different stages of the maximum load test. We examined 124 highly trained cross-country skiers (male, ages 17–24). The group consisted of two subgroups based on their competition performance: 61 nonelite athletes (Group I) and 63 elite athletes (group II), who were current members of the national team of the Komi Republic and Russia. The bicycle ergometer test was performed by using the OxyconPro system (Erich Jaeger, Hoechberg, Germany). All the examined athletes performed the exercise test on a cycle ergometer “until exhaustion”. The results of our research indicate that the studied groups of athletes with high, but different levels of sports qualifications are a convenient model for studying the molecular mechanisms of adaptation to physical loads of maximum intensity. Athletes of higher qualifications reveal additional adaptive mechanisms of metabolic regulation, which is manifested in the independence of serum lactate indicators under conditions of submaximal and maximum power from maximal oxygen uptake, and they have an NO-dependent mechanism for regulating lactate levels during aerobic exercise, including work at the anaerobic threshold.
Collapse
|
3
|
Hypercapnia Modulates the Activity of Adenosine A1 Receptors and mitoK +ATP-Channels in Rat Brain When Exposed to Intermittent Hypoxia. Neuromolecular Med 2021; 24:155-168. [PMID: 34115290 DOI: 10.1007/s12017-021-08672-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Accepted: 06/05/2021] [Indexed: 10/21/2022]
Abstract
The mechanisms and signaling pathways of the neuroprotective effects of hypercapnia and its combination with hypoxia are not studied sufficiently. The study aims to test the hypothesis of the potentiating effect of hypercapnia on the systems of adaptation to hypoxia, directly associated with A1-adenosine receptors and mitochondrial ATP-dependent K+ -channels (mitoK+ATP-channels). We evaluated the relative number of A1-adenosine receptors and mitoK+ATP-channels in astrocytes obtained from male Wistar rats exposed to various respiratory conditions (15 times of hypoxia and/or hypercapnia). In addition, the relative number of these molecules in astrocytes was evaluated on an in vitro model of chemical hypoxia, as well as in the cerebral cortex after photothrombotic damage. This study indicates an increase in the relative number of A1-adenosine receptors in astrocytes and in cells next to the stroke region of the cerebral cortex in rats exposed to hypoxia and hypercapnic hypoxia, but not hypercapnia alone. Hypercapnia and hypoxia increase the relative number of mitoK+ATP-channels in astrocytes and in cells of the peri-infarct region of the cerebral cortex in rats. In an in vitro study, hypercapnia mitigates the effects of acute chemical hypoxia observed in astrocytes for A1-adenosine receptors and mitoK+ATP-channels. Hypercapnia, unlike hypoxia, does not affect the relative number of A1 receptors to adenosine. At the same time, both hypercapnia and hypoxia increase the relative number of mitoK+ATP-channels, which can potentiate their protective effects with combined exposure.
Collapse
|
4
|
Böger R, Hannemann J. Dual role of the L-arginine-ADMA-NO pathway in systemic hypoxic vasodilation and pulmonary hypoxic vasoconstriction. Pulm Circ 2020; 10:2045894020918850. [PMID: 32313645 PMCID: PMC7153195 DOI: 10.1177/2045894020918850] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 03/20/2020] [Indexed: 02/06/2023] Open
Abstract
In healthy vascular endothelium, nitric oxide acts as a vasodilator paracrine mediator on adjacent smooth muscle cells. By activating soluble guanylyl cyclase, nitric oxide stimulates cyclic guanosine monophosphate (cGMP) which causes relaxation of vascular smooth muscle (vasodilation) and inhibition of platelet aggregation. This mechanism is active in both, the systemic and pulmonary circulation. In the systemic circulation, hypoxia results in local vasodilation, which has been shown to be brought about by stabilization of hypoxia-inducible factor-1α (HIF1α) and concomitant upregulation of endothelial nitric oxide synthase. By contrast, the physiological response to hypoxia in the pulmonary circulation is vasoconstriction. Hypoxia in the lung primarily results from hypoventilation of circumscript areas of the lung, e.g. by bronchial tree obstruction or inflammatory infiltration. Therefore, hypoxic pulmonary vasoconstriction is a mechanism preventing distribution of blood to hypoventilated areas of the lungs, thereby maintaining maximal oxygenation of blood. The exact molecular mechanism of hypoxic pulmonary vasoconstriction is less well understood than hypoxic vasodilation in the systemic circulation. While alveolar epithelial cells may be key in sensing low oxygen concentration, and pulmonary vascular smooth muscle cells obviously are the effectors of vasoconstriction, the pulmonary vascular endothelium plays a crucial role as an intermediate between these cell types. Indeed, dysfunctional endothelial nitric oxide release was observed in humans exposed to acute hypoxia, and animal studies suggest that hypoxic pulmonary vasoconstriction is enhanced by nitric oxide synthase inhibition. This may be caused, in part, by elevation of asymmetric dimethylarginine, an endogenous inhibitor of nitric oxide synthesis. High asymmetric dimethylarginine levels are associated with endothelial dysfunction, vascular disease, and hypertension.
Collapse
Affiliation(s)
- Rainer Böger
- Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Institute DECIPHER, German-Chilean Institute for Research on Pulmonary Hypoxia and its Health Sequelae, Hamburg, Germany
| | - Juliane Hannemann
- Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Institute DECIPHER, German-Chilean Institute for Research on Pulmonary Hypoxia and its Health Sequelae, Hamburg, Germany
| |
Collapse
|
5
|
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: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
6
|
|
7
|
Mir JM, Malik BA, Maurya RC. Nitric oxide-releasing molecules at the interface of inorganic chemistry and biology: a concise overview. REV INORG CHEM 2019. [DOI: 10.1515/revic-2018-0017] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
AbstractThe useful aspects of nitric oxide (NO) are nowadays widely known. Due to the need for this molecule in the maintenance of homeostasis, NO-releasing compounds are tested every year to optimize its levels in a patient suffering from low NO production. This manuscript is an update of some important historical concerns about nitrosyl complexes having the ability to act as NO-releasing compounds under the influence of different chemically modified environments. At present, the search for efficient and less harmful NO-releasing molecules at desirable targets and concentrations has gained considerable momentum in nitrosyl chemistry. Iron, ruthenium, and manganese nitrosyls have been investigated elitely to disentangle their electronic transition (excitation) under visible light to act as NO donors without harming the healthy cells of a target. There is much evidence supporting the increase of NO lability if amino acids are used as complexing ligands, the design of a reduction center close to an NO grouping, and the development of porphyrin system-based nitrosyl complexes. From the overall survey, it may be concluded that the desirable properties of such scaffolds need to be evaluated further to complement the biological milieu.
Collapse
Affiliation(s)
- Jan Mohammad Mir
- Coordination, Bioinorganic and Computational Chemistry Laboratory, Department of Post Graduate Studies and Research in Chemistry and Pharmacy, Rani Durgavati University, Jabalpur 482001, Madhya Pradesh, India
- Department of Chemistry, Islamic University of Science and Technology, Awantipora 192322, Jammu and Kashmir
| | - Bashir Ahmad Malik
- Coordination, Bioinorganic and Computational Chemistry Laboratory, Department of Post Graduate Studies and Research in Chemistry and Pharmacy, Rani Durgavati University, Jabalpur 482001, Madhya Pradesh, India
- Department of Chemistry, Islamic University of Science and Technology, Awantipora 192322, Jammu and Kashmir
| | - Ram Charitra Maurya
- Coordination, Bioinorganic and Computational Chemistry Laboratory, Department of Post Graduate Studies and Research in Chemistry and Pharmacy, Rani Durgavati University, Jabalpur 482001, Madhya Pradesh, India
| |
Collapse
|
8
|
Sprick JD, Mallet RT, Przyklenk K, Rickards CA. Ischaemic and hypoxic conditioning: potential for protection of vital organs. Exp Physiol 2019; 104:278-294. [PMID: 30597638 DOI: 10.1113/ep087122] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 12/20/2018] [Indexed: 12/13/2022]
Abstract
NEW FINDINGS What is the topic of this review? Remote ischaemic preconditioning (RIPC) and hypoxic preconditioning as novel therapeutic approaches for cardiac and neuroprotection. What advances does it highlight? There is improved understanding of mechanisms and signalling pathways associated with ischaemic and hypoxic preconditioning, and potential pitfalls with application of these therapies to clinical trials have been identified. Novel adaptations of preconditioning paradigms have also been developed, including intermittent hypoxia training, RIPC training and RIPC-exercise, extending their utility to chronic settings. ABSTRACT Myocardial infarction and stroke remain leading causes of death worldwide, despite extensive resources directed towards developing effective treatments. In this Symposium Report we highlight the potential applications of intermittent ischaemic and hypoxic conditioning protocols to combat the deleterious consequences of heart and brain ischaemia. Insights into mechanisms underlying the protective effects of intermittent hypoxia training are discussed, including the activation of hypoxia-inducible factor-1 and Nrf2 transcription factors, synthesis of antioxidant and ATP-generating enzymes, and a shift in microglia from pro- to anti-inflammatory phenotypes. Although there is little argument regarding the efficacy of remote ischaemic preconditioning (RIPC) in pre-clinical models, this strategy has not consistently translated into the clinical arena. This lack of translation may be related to the patient populations targeted thus far, and the anaesthetic regimen used in two of the major RIPC clinical trials. Additionally, we do not fully understand the mechanism through which RIPC protects the vital organs, and co-morbidities (e.g. hypercholesterolemia, diabetes) may interfere with its efficacy. Finally, novel adaptations have been made to extend RIPC to more chronic settings. One adaptation is RIPC-exercise (RIPC-X), an innovative paradigm that applies cyclical RIPC to blood flow restriction exercise (BFRE). Recent findings suggest that this novel exercise modality attenuates the exaggerated haemodynamic responses that may limit the use of conventional BFRE in some clinical settings. Collectively, intermittent ischaemic and hypoxic conditioning paradigms remain an exciting frontier for the protection against ischaemic injuries.
Collapse
Affiliation(s)
- Justin D Sprick
- Division of Renal Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA, 30307, USA.,Department of Physiology & Anatomy, University of North Texas Health Science Center, Fort Worth, TX, 76107, USA
| | - Robert T Mallet
- Department of Physiology & Anatomy, University of North Texas Health Science Center, Fort Worth, TX, 76107, USA
| | - Karin Przyklenk
- Cardiovascular Research Institute, Wayne State University School of Medicine, Detroit, MI, 48201, USA.,Department of Physiology, Wayne State University School of Medicine, Detroit, MI, 48201, USA.,Department of Emergency Medicine, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Caroline A Rickards
- Department of Physiology & Anatomy, University of North Texas Health Science Center, Fort Worth, TX, 76107, USA
| |
Collapse
|
9
|
Manukhina EB, Downey HF, Mallet RT. Role of Nitric Oxide in Cardiovascular Adaptation to Intermittent Hypoxia. Exp Biol Med (Maywood) 2016; 231:343-65. [PMID: 16565431 DOI: 10.1177/153537020623100401] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Hypoxia is one of the most frequently encountered stresses in health and disease. The duration, frequency, and severity of hypoxic episodes are critical factors determining whether hypoxia is beneficial or harmful. Adaptation to intermittent hypoxia has been demonstrated to confer cardiovascular protection against more severe and sustained hypoxia, and, moreover, to protect against other stresses, including ischemia. Thus, the direct and cross protective effects of adaptation to intermittent hypoxia have been used for treatment and prevention of a variety of diseases and to increase efficiency of exercise training. Evidence is mounting that nitric oxide (NO) plays a central role in these adaptive mechanisms. NO-dependent protective mechanisms activated by intermittent hypoxia include stimulation of NO synthesis as well as restriction of NO overproduction. In addition, alternative, nonenzymic sources of NO and negative feedback of NO synthesis are important factors in optimizing NO concentrations. The adaptive enhancement of NO synthesis and/or availability activates or increases expression of other protective factors, including heat shock proteins, antioxidants and prostaglandins, making the protection more robust and sustained. Understanding the role of NO in mechanisms of adaptation to hypoxia will support development of therapies to prevent and treat hypoxic or ischemic damage to organs and cells and to increase adaptive capabilities of the organism.
Collapse
|
10
|
Wu SC, Lu CY, Chen YL, Lo FC, Wang TY, Chen YJ, Yuan SS, Liaw WF, Wang YM. Water-Soluble Dinitrosyl Iron Complex (DNIC): a Nitric Oxide Vehicle Triggering Cancer Cell Death via Apoptosis. Inorg Chem 2016; 55:9383-92. [DOI: 10.1021/acs.inorgchem.6b01562] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Affiliation(s)
- Shou-Cheng Wu
- Department of Chemistry and Frontier Research Center on Fundamental
and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Chung-Yen Lu
- Department of Chemistry and Frontier Research Center on Fundamental
and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Yi-Lin Chen
- Department of Chemistry and Frontier Research Center on Fundamental
and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Feng-Chun Lo
- Department of Chemistry and Frontier Research Center on Fundamental
and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Ting-Yin Wang
- Department of Chemistry and Frontier Research Center on Fundamental
and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Yu-Jen Chen
- Department
of Biological Science and Technology, Institute of Molecular Medicine
and Bioengineering, National Chiao Tung University, Hsinchu 30013, Taiwan
| | - Shyng-Shiou Yuan
- Translational Research
Center, Department of Medical Research, and Department of Obstetrics
and Gynecology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Wen-Feng Liaw
- Department of Chemistry and Frontier Research Center on Fundamental
and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Yun-Ming Wang
- Department
of Biological Science and Technology, Institute of Molecular Medicine
and Bioengineering, National Chiao Tung University, Hsinchu 30013, Taiwan
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| |
Collapse
|
11
|
Lu H, Wang R, Xiong J, Xie H, Kayser B, Jia ZP. In search for better pharmacological prophylaxis for acute mountain sickness: looking in other directions. Acta Physiol (Oxf) 2015; 214:51-62. [PMID: 25778288 DOI: 10.1111/apha.12490] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Revised: 02/23/2015] [Accepted: 03/11/2015] [Indexed: 12/15/2022]
Abstract
Despite decades of research, the exact pathogenic mechanisms underlying acute mountain sickness (AMS) are still poorly understood. This fact frustrates the search for novel pharmacological prophylaxis for AMS. The prevailing view is that AMS results from an insufficient physiological response to hypoxia and that prophylaxis should aim at stimulating the response. Starting off from the opposite hypothesis that AMS may be caused by an initial excessive response to hypoxia, we suggest that directly or indirectly blunting-specific parts of the response might provide promising research alternatives. This reasoning is based on the observations that (i) humans, once acclimatized, can climb Mt Everest experiencing arterial partial oxygen pressures (PaO2) as low as 25 mmHg without AMS symptoms; (ii) paradoxically, AMS usually develops at much higher PaO2 levels; and (iii) several biomarkers, suggesting initial activation of specific pathways at such PaO2, are correlated with AMS. Apart from looking for substances that stimulate certain hypoxia triggered effects, such as the ventilatory response to hypoxia, we suggest to also investigate pharmacological means aiming at blunting certain other specific hypoxia-activated pathways, or stimulating their agonists, in the quest for better pharmacological prophylaxis for AMS.
Collapse
Affiliation(s)
- H Lu
- Key Laboratory of the Plateau of Environmental Damage Control, Lanzhou General Hospital of Lanzhou Military Command, Lanzhou, China
| | | | | | | | | | | |
Collapse
|
12
|
Lu H, Wang R, Xiong J, Xie H, Kayser B, Jia ZP. In search for better pharmacological prophylaxis for acute mountain sickness: looking in other directions. Acta Physiol (Oxf) 2015; 214:51-62. [PMID: 25753758 DOI: 10.1111/apha.12486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Revised: 02/27/2015] [Accepted: 03/05/2015] [Indexed: 11/29/2022]
Abstract
Despite decades of research, the exact pathogenic mechanisms underlying acute mountain sickness (AMS) are still poorly understood. This fact frustrates the search for novel pharmacological prophylaxis for AMS. The prevailing view is that AMS results from an insufficient physiological response to hypoxia and that prophylaxis should aim at stimulating the response. Starting off from the opposite hypothesis that AMS may be caused by an initial excessive response to hypoxia we suggest that directly or indirectly blunting specific parts of the response might provide promising research alternatives. This reasoning is based on the observations that 1) humans, once acclimatized, can climb Mt Everest experiencing arterial partial oxygen pressures (PaO2 ) as low as 25 mmHg without AMS symptoms, 2) paradoxically AMS usually develops at much higher PaO2 levels, and 3) several biomarkers, suggesting initial activation of specific pathways at such PaO2 , are correlated with AMS. Apart from looking for substances that stimulate certain hypoxia triggered effects, such as the ventilatory response to hypoxia, we suggest to also investigate pharmacological means aiming at blunting certain other specific hypoxia activated pathways, or stimulating their agonists, in the quest for better pharmacological prophylaxis for AMS. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Hui Lu
- Key Laboratory of the plateau of environmental damage control, Lanzhou General Hospital of Lanzhou Military Command, Lanzhou, China
| | | | | | | | | | | |
Collapse
|
13
|
Mitochondrial function in rat cerebral cortex and hippocampus after short- and long-term hypobaric hypoxia. Brain Res 2014; 1598:66-75. [PMID: 25527397 DOI: 10.1016/j.brainres.2014.12.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Revised: 12/02/2014] [Accepted: 12/07/2014] [Indexed: 01/19/2023]
Abstract
Taking into account the importance of aerobic metabolism in brain, the aim of the present work was to evaluate mitochondrial function in cerebral cortex and hippocampus in a model of sustained hypobaric hypoxia (5000 m simulated altitude) during a short (1 mo) and a long (7 mo) term period, in order to precise the mechanisms involved in hypoxia acclimatization. Hippocampal mitochondria from rats exposed to short-term hypobaric hypoxia showed lower respiratory rates than controls in both states 4 (45%) and 3 (41%), and increased NO production (1.3 fold) as well as eNOS and nNOS expression associated to mitochondrial membranes, whereas mitochondrial membrane potential decreased (7%). No significant changes were observed in cortical mitochondria after 1 mo hypobaric hypoxia in any of the mitochondrial functionality parameters evaluated. After 7 mo hypobaric hypoxia, oxygen consumption was unchanged as compared with control animals both in hippocampal and cortical mitochondria, but mitochondrial membrane potential decreased by 16% and 8% in hippocampus and cortex respectively. Also, long-term hypobaric hypoxia induced an increase in hippocampal NO production (0.7 fold) and in eNOS expression. A clear tendency to decrease in H2O2 production was observed in both tissues. Results suggest that after exposure to hypobaric hypoxia, hippocampal mitochondria display different responses than cortical mitochondria. Also, the mechanisms responsible for acclimatization to hypoxia would be time-dependent, according to the physiological functions of the brain studied areas. Nitric oxide metabolism and membrane potential changes would be involved as self-protective mechanisms in high altitude environment.
Collapse
|
14
|
Paltsyn AA, Manukhina EB, Goryacheva AV, Downey HF, Dubrovin IP, Komissarova SV, Kubatiev AA. Intermittent hypoxia stimulates formation of binuclear neurons in brain cortex—A role of cell fusion in neuroprotection? Exp Biol Med (Maywood) 2014; 239:595-600. [DOI: 10.1177/1535370214523898] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Oligodendrocyte fusion with neurons in the brain cortex is a part of normal ontogenesis and is a possible means of neuroregeneration. Following such fusion, the oligodendrocyte nucleus undergoes neuron-specific reprogramming, resulting in the formation of binuclear neurons, which doubles the functional capability of the neuron. In this study, we tested the hypothesis that the formation of binuclear neurons is involved in long-term adaptation of the brain to intermittent hypobaric hypoxia, which is known to be neuroprotective. Rats were adapted to hypoxia in an altitude chamber at a simulated altitude of 4000 m above sea level for 14 days (30 min increasing to 4 h, daily). One micrometer sections of the left motor cortex were analyzed by light microscopy. Phases of the fusion and reprogramming process were recorded, and the number of binuclear neurons was counted for all section areas containing pyramidal neurons of layers III–V. For the control group subjected to sham hypoxia, the density of binuclear neurons was 4.49 ± 0.32 mm2. In the hypoxia-adapted group, this density increased to 5.71 ± 0.39 mm2 ( P < 0.04). In a subgroup of rats exposed to only one hypoxia session, the number of binuclear neurons did not differ from the number observed in the control group. We suggest that the increased content of binuclear neurons may serve as a structural basis for the neuroprotective effects of the adaptation to hypoxia.
Collapse
Affiliation(s)
- Alexander A Paltsyn
- Institute of General Pathology and Pathophysiology, Moscow 125315, Russia
- Russian Medical Academy of Postgraduate Education, Moscow 125315, Russia
| | - Eugenia B Manukhina
- Institute of General Pathology and Pathophysiology, Moscow 125315, Russia
- University of North Texas Health Science Center, Fort Worth, Texas 76107, USA
| | - Anna V Goryacheva
- Institute of General Pathology and Pathophysiology, Moscow 125315, Russia
| | - H Fred Downey
- University of North Texas Health Science Center, Fort Worth, Texas 76107, USA
| | - Ivan P Dubrovin
- Institute of General Pathology and Pathophysiology, Moscow 125315, Russia
| | | | - Aslan A Kubatiev
- Institute of General Pathology and Pathophysiology, Moscow 125315, Russia
- Russian Medical Academy of Postgraduate Education, Moscow 125315, Russia
| |
Collapse
|
15
|
Tannheimer M, Hornung K, Gasche M, Kuehlmuss B, Mueller M, Welsch H, Landgraf K, Guger C, Schmidt R, Steinacker JM. Decrease of asymmetric dimethylarginine predicts acute mountain sickness. J Travel Med 2012; 19:338-43. [PMID: 23379703 DOI: 10.1111/j.1708-8305.2012.00652.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
INTRODUCTION Each year, 40 million tourists worldwide are at risk of getting acute mountain sickness (AMS), because they travel to altitudes of over 2500 m. As asymmetric dimethylarginine (ADMA) is a nitric oxide synthase (NOS) inhibitor, it should increase pulmonary artery pressure (PAP) and raise the risk of acute mountain sickness and high-altitude pulmonary edema (HAPE). With this in mind, we investigated whether changes in ADMA levels (Δ-ADMA) at an altitude of 4000 m can predict an individual's susceptibility to AMS or HAPE. METHODS Twelve subjects spent two nights in a hypobaric chamber, the first night without exposure to altitude conditions and the second night at a simulated altitude of 4000 m. At identical time points during both nights (after 2, 5, and 11 hours), we determined ADMA serum levels, PAP by Doppler echocardiography and estimated hypoxia related symptoms by Lake Louise Score (LLS). RESULTS Contrary to our initial hypothesis, subjects with a marked increase in ADMA at 4000 m showed PAP levels below the critical threshold for HAPE and were not affected by AMS. By contrast, subjects with a decrease in ADMA suffered from AMS and had PAP levels above 40 mmHg. After 2 hours of hypoxia we found a significant relationship between Δ-PAP t(2) (Spearmans ρ = 0.30, p ≤ 0.05) respectively Δ-ADMA t(2) (ρ = -0.92, p ≤ 0.05) and LLS. CONCLUSION After 2 hours of hypoxia, the Δ-ADMA (positive or negative) can predict an LLS of >5 with a sensitivity of 80% and a specificity of 100% and can help assess the risk of an increase in PAP to more than 40 mmHg and thus the risk of HAPE (ϕ coefficient: 0.69; p ≤ 0.05).
Collapse
Affiliation(s)
- Markus Tannheimer
- Departments of General and Thoracic Surgery, German Armed Forces Hospital of Ulm, Ulm, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Martínez-Romero R, Cañuelo A, Siles E, Oliver FJ, Martínez-Lara E. Nitric oxide modulates hypoxia-inducible factor-1 and poly(ADP-ribose) polymerase-1 cross talk in response to hypobaric hypoxia. J Appl Physiol (1985) 2012; 112:816-23. [DOI: 10.1152/japplphysiol.00898.2011] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The physiological response to hypobaric hypoxia represents a complex network of biochemical pathways in which the nitrergic system plays an important role. Previous studies have provided evidence for an interplay between the hypoxia-inducible factor-1 (HIF-1) and poly(ADP-ribose) polymerase-1 (PARP-1) under hypoxia. Here, we evaluate the potential involvement of nitric oxide (NO) in the cross talk between these two proteins. With this aim, we studied comparatively the effect of pharmacological inhibitors of NO production or PARP activity in the response of the mouse cerebral cortex to 4 h of exposure to a simulated altitude of 31,000 ft. Particularly, we analyzed the NO and reactive oxygen species production, the expression of NO synthase (NOS) isoforms, PARP-1 activity, HIF-1α expression and HIF-1 transcriptional activity, the protein level of the factor inhibiting HIF, and, finally, beclin-1 and fractin expression, as markers of cellular damage. Our results demonstrate that the reduction of NO level did not affect reactive oxygen species production but significantly 1) dampened the posthypoxic increase in neuronal NOS and inducible NOS expression without altering endothelial NOS protein level; 2) prevented PARP activation; 3) decreased HIF-1α response to hypoxia; 4) achieved a higher long-term HIF-1 transcriptional activity by reducing factor inhibiting HIF expression; and 5) reduced hypoxic damage. The pharmacological inhibition of PARP reproduced the NOS expression pattern and the HIF-1α response observed in NOS-inhibited mice, supporting its involvement in the NO-dependent regulation of hypoxia. As a whole, these results provide new data about the molecular mechanism underlying the beneficial effects of controlling NO production under hypobaric hypoxic conditions.
Collapse
Affiliation(s)
| | - Ana Cañuelo
- Department of Experimental Biology, University of Jaén, Jaén; and
| | - Eva Siles
- Department of Experimental Biology, University of Jaén, Jaén; and
| | - F. Javier Oliver
- Institute of Parasitology and Biomedicine, Consejo Superior de Investigaciones Científicas, Granada, Spain
| | | |
Collapse
|
17
|
Tang Q, Ma J, Zhang P, Wan W, Kong L, Wu L. Persistent sodium current and Na+/H+ exchange contributes to the augmentation of the reverse Na+/Ca2+ exchange during hypoxia or acute ischemia in ventricular myocytes. Pflugers Arch 2012; 463:513-22. [PMID: 22234427 DOI: 10.1007/s00424-011-1070-y] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2011] [Accepted: 12/22/2011] [Indexed: 12/19/2022]
Abstract
The increases in persistent sodium currents (I (Na.P)) and Na(+)/H(+) exchange (NHE) causes intracellular Ca(2+) overload. The objective of this study was to determine the contribution of I (Na.P) and NHE on the hypoxia- or acute ischemia-induced increase in the reverse Na(+)/Ca(2+) exchange current (HIR- or AIR-I (NCX)). I (Na.P) and I (NCX) in rabbit ventricular myocytes were recorded during hypoxia or acute ischemia, combination of acidosis (pH values were 6.0 intracellularly and 6.8 extracellularly) and hypoxia, using whole-cell patch-clamp techniques. The results indicate that (1) under hypoxic condition, the augmentation of both HIR-I (NCX) and I (Na.P) was inhibited by TTX (2 to 8 μM) in a concentration-dependent manner. The inhibitions of I (Na,P) and HIR-I (NCX) reached maximum in the presence of either 4 μM TTX or 10 μM KR-32568 (a NHE inhibitor), respectively. The maximal inhibitions of HIR-I (NCX) by 4 μM TTX and 10 μM KR-32568 were 72.54% and 16.89%, respectively. (2) Administration of 2 μM TTX and 10 μM KR-32568 in either order in the same cells decreased HIR-I (NCX) by 64.83% and 16.94%, respectively. (3) I (Na.P) and the reverse I (NCX) were augmented during acute ischemia. TTX (4 μM) and KR-32568 (10 μM) reduced AIR-I (NCX) by 73.39% and 24.13%, respectively. (4) Under normoxic condition, veratridine (20 μM) significantly increased I (Na.P) and the reverse I (NCX), which was reversed by 4 μM TTX. In conclusion, during hypoxia or acute ischemia, both increased I (Na.P) and NHE contribute to the HIR- or AIR-I (NCX) with the former playing a major role comparing with the latter.
Collapse
Affiliation(s)
- Qiong Tang
- Cardio-Electrophysiological Research Laboratory, Medical College, Wuhan University of Science and Technology, Wuhan, Hubei, China
| | | | | | | | | | | |
Collapse
|
18
|
Goryacheva AV, Kruglov SV, Pshennikova MG, Smirin BV, Malyshev IY, Barskov IV, Viktorov IV, Downey HF, Manukhina EB. Adaptation to intermittent hypoxia restricts nitric oxide overproduction and prevents beta-amyloid toxicity in rat brain. Nitric Oxide 2010; 23:289-99. [DOI: 10.1016/j.niox.2010.08.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2010] [Revised: 08/18/2010] [Accepted: 08/24/2010] [Indexed: 11/17/2022]
|
19
|
A study of NO trafficking from dinitrosyl-iron complexes to the recombinant E. coli transcriptional factor SoxR. J Biol Inorg Chem 2008; 13:961-72. [PMID: 18449575 DOI: 10.1007/s00775-008-0383-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2008] [Accepted: 04/18/2008] [Indexed: 10/22/2022]
Abstract
SoxR is a transcriptional factor in Escherichia coli that induces the expression of SoxS to initiate the production of enzymes in response to oxidative stress. In addition to superoxide, SoxR is also sensitive to cellular NO to produce a protein-bound dinitrosyl-iron complex (DNIC) with a characteristic electron paramagnetic resonance (EPR) signal at g(av)=2.03. Toward developing a strategy for NO sensing based on this property of SoxR, we have overexpressed and purified the recombinant His-tagged SoxR protein. Upon treatment of the purified protein under anaerobic conditions with (1) NO solution, (2) S-nitrosothiol (RSNO), and (3) chemically synthesized low molecular weight DNICs (LMW-DNICs), we have observed enhancement of the EPR signal at g(av)=2.03 from the protein-bound DNICs over time, reflecting the redistribution of NO from the NO solution, RSNO and LMW-DNICs to the SoxR. We have exploited this NO exchange to investigate the kinetics and mechanisms of release and delivery of NO from various LMW-DNICs to an isopropyl-beta-D-thiogalactopyranoside-dependent SoxR expressed in E. coli cells. These experiments revealed that the NO from RSNO and LMW-DNICs could cross the biological membrane and enter the cytoplasm of the cell to form the SoxR protein-bound DNIC complex. For comparison, we have also studied the direct NO transfer from the LMW-DNICs to the SoxR protein in buffer. The NO transfer was found to be rapid. From the kinetic data derived, we showed that LMW-DNICs with bidentate thiolate ligands displayed greater stability in aqueous solution but exhibited more facile NO delivery to cytoplasmic SoxR in whole cells.
Collapse
|
20
|
Wei IH, Huang CC, Tseng CY, Chang HM, Tu HC, Tsai MH, Wen CY, Shieh JY. Mild hypoxic preconditioning attenuates injury-induced NADPH-d/nNOS expression in brainstem motor neurons of adult rats. J Chem Neuroanat 2007; 35:123-32. [PMID: 17942275 DOI: 10.1016/j.jchemneu.2007.08.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2007] [Revised: 08/21/2007] [Accepted: 08/24/2007] [Indexed: 01/30/2023]
Abstract
Excessive production of nitric oxide (NO) might have detrimental effects on the hypoxia-related neuropathology. This study aimed to test if mild hypoxic preconditioning (MHPC) would attenuate the pathological changes in the brainstem motoneurons having a different functional component after peripheral nerve crush injury (PNCI). Prior to PNCI treatment, young adult rats were caged in the mild hypoxic altitude chamber with 79Torr of the partial oxygen concentration ( pO(2)) (i.e., 0.5atm at 5500m in height) for 4 weeks to adapt the environmental changes. After that, all the animals having successfully crushed both the hypoglossal and vagus nerves (left-side) were allowed to survive for 3, 7, 14, 30 and 60 successive days in normoxic condition. Nicotinamine adenine dinucleotide phosphate-diaphorase (NADPH-d) histochemistry and neuronal nitric oxide synthase (nNOS) immunohistochemistry revealed that MHPC reduces NADPH-d/nNOS expression in the hypoglossal nucleus (HN) and the dorsal motor nucleus of the vagus (DMN) at different time points after PNCI. The morphological findings were further ascertained by Western blot analysis of nNOS and nitrite assay for NO production. Both the morphological and quantitative results peaked at 7 days in HN, whereas for those in DMN were progressively increased up to 60 days following PNCI. The staining intensity of NADPH-d/nNOS(+) neurons, expression of nNOS protein, NO production levels as well as the neuronal loss in HN and DMN of MHPC rats following PNCI were attenuated, especially for those having a longer survival period over 14 days. The MHPC treatment might induce minute amounts of NO to alter the state of milieu of the experimental animals to protect against the PNCI.
Collapse
Affiliation(s)
- I-Hua Wei
- Department of Anatomy and Cell Biology, College of Medicine, China Medical University, Taichung, Taiwan.
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Laudi S, Steudel W, Jonscher K, Schöning W, Schniedewind B, Kaisers U, Christians U, Trump S. Comparison of lung proteome profiles in two rodent models of pulmonary arterial hypertension. Proteomics 2007; 7:2469-78. [PMID: 17623304 DOI: 10.1002/pmic.200600848] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
We studied the lung proteome changes in two widely used models of pulmonary arterial hypertension (PAH): monocrotaline (MCT) injection and chronic hypoxia (CH); untreated rats were used as controls (n = 6/group). After 28 days, invasive right ventricular systolic pressure (RVSP) was measured. Lungs were immunostained for alpha-smooth muscle actin (alphaSMA). 2-DE (n = 4/group) followed by nano-LC-MS/MS was applied for protein identification. Western blotting was used additionally if possible. RVSP was significantly increased in MCT- and CH-rats (MCT 62.5 +/- 4.4 mmHg, CH 62.2 +/- 4.1 mmHg, control 25.0 +/- 1.7 mmHg, p<0.001). This was associated with an increase of alphaSMA positive vessels. In both groups, there was a significantly increased expression of proteins associated with the contractile apparatus (diphosphoHsp27 (p<0.001), Septin2 (p<0.001), F-actin capping protein (p<0.01), and tropomyosin beta (p<0.02)). In CH, proteins of the nitric oxide (Hsc70; p = 0.002), carbon monoxide (biliverdin reductase; p = 0.005), and vascular endothelial growth factor (VEGF) pathway (annexin 3; p<0.001) were significantly increased. In MCT, proteins involved in serotonin synthesis (14-3-3; p = 0.02), the enhanced unfolded protein response (ERp57; p = 0.02), and intracellular chloride channels (CLIC 1; p = 0.002) were significantly elevated. Therefore, MCT- and CH-induced vasoconstriction and remodeling seemed to be mediated via different signaling pathways. These differences should be considered in future studies using either PAH model.
Collapse
Affiliation(s)
- Sven Laudi
- University of Leipzig Medical Faculty, Department of Anesthesiology and Intensive Care Medicine, Leipzig, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Manukhina EB, Vanin AF, Markov KM, Malyshev IY. Formation and Role of Nitric Oxide Stores in Adaptation to Hypoxia. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2006; 578:35-40. [PMID: 16927667 DOI: 10.1007/0-387-29540-2_6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Affiliation(s)
- Eugenia B Manukhina
- Institute of General Pathology and Pathophysiology, Baltijskaya 8, Moscow 125315, Russia
| | | | | | | |
Collapse
|
23
|
Song MY, Zwemer CF, Whitesall SE, D'Alecy LG. Acute and conditioned hypoxic tolerance augmented by endothelial nitric oxide synthase inhibition in mice. J Appl Physiol (1985) 2006; 102:610-5. [PMID: 17068215 DOI: 10.1152/japplphysiol.00894.2006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
To identify a possible role for nitric oxide (NO) in acute hypoxic tolerance (HT) we measured hypoxic survival time (HST), effect of hypoxic conditioning (HC), and survival following hypoxic conditioning while blocking or mimicking the action of nitric oxide synthase (NOS). To inhibit NOS, CD-1 mice were given supplemental endogenous NOS inhibitor asymmetrical dimethylarginine (ADMA) or a synthetic NOS inhibitor N(omega)-nitro-L-arginine (L-NNA), both of which nonselectively inhibit three of the isoforms of NOS [inducible (iNOS), neuronal (nNOS), and endothelial NOS (eNOS)]. ADMA (10 mg/kg i.p.) or saline vehicle was given 5 min before HST testing. L-NNA was given orally at 1 g/l in drinking water with tap water as the control for 48 h before testing. Both ADMA and L-NNA significantly increased HST and augmented the HC effect on HST. Neither the nNOS selective inhibitor 7-nitroindazole (7-NI) nor the iNOS selective inhibitor N-{[3-(aminomethyl)phenyl]methyl}-enthanimidamide (1400W) had a statistically significant effect on HST or HT. The NO donor, 3-morpholinosydnoeimine, when given alone did not significantly decrease HT, but it did mitigate the increased HT effect of L-NNA. These data confirm that acute hypoxic conditioning increases HT and that NOS inhibition by endogenous (ADMA) and a synthetic NOS inhibitor (L-NNA) further increases HT, whereas iNOS and nNOS inhibition does not, suggesting that it is the inhibition of eNOS that mediates enhancement of HT.
Collapse
Affiliation(s)
- Michael Y Song
- College of Literature, Science and the Arts, University of Michigan, Ann Arbor, Michigan, USA
| | | | | | | |
Collapse
|
24
|
Serrano J, Encinas JM, Fernández AP, Rodrigo J, Martínez A. Effects of acute hypobaric hypoxia on the nitric oxide system of the rat cerebral cortex: Protective role of nitric oxide inhibitors. Neuroscience 2006; 142:799-808. [PMID: 16952423 DOI: 10.1016/j.neuroscience.2006.07.046] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2006] [Revised: 07/25/2006] [Accepted: 07/30/2006] [Indexed: 12/21/2022]
Abstract
Exposure to hypobaric hypoxia produces neuropsychological disorders. The brain nitrergic system was investigated following hypobaric hypoxia in the presence or absence of nitric oxide synthase (NOS) inhibitors. Adult rats were exposed to a simulated altitude of 8325 m (27,000 ft) for 7 h and killed after 0, 1, 3, 5, and 10 days of recovery. In addition to normobaric controls, three experimental groups were studied: i) subjected to hypobaric hypoxia without inhibitors; ii) subjected to hypobaric hypoxia and treated with 7-nitroindazole; iii) subjected to hypobaric hypoxia and treated with N(omega)-nitro-l-arginine methyl ester (l-NAME). Cerebral cortex was assayed by immunohistochemistry, Western blotting, and enzymatic assays. In animals subjected to hypobaric hypoxia without inhibitors, there was an increase in neuronal nitric oxide synthase (nNOS) immunoreactivity and Ca(2+)-dependent NOS activity from 0 to 1 days of reoxygenation. In these animals, inducible nitric oxide synthase (iNOS) expression and Ca(2+)-independent activity were undetectable, but nitrotyrosine immunoreactivity was found in some neurons. Administration of either inhibitor prevented the increase in nNOS immunoreactivity and enzymatic activity provoked by hypobaric hypoxia. Concomitantly, nitrotyrosine immunoreactivity decreased progressively. In conclusion, activation of the nitrergic system constitutes a cortical response to hypobaric hypoxia and the administration of NOS inhibitors could provide new therapeutic avenues to prevent and/or treat the symptoms produced by hypobaric hypoxia.
Collapse
Affiliation(s)
- J Serrano
- Department of Neuroanatomy and Cell Biology, Instituto Cajal, CSIC, Avd. Doctor Arce, 37, 28002 Madrid, Spain
| | | | | | | | | |
Collapse
|
25
|
Mashina SY, Aleksandrin VV, Goryacheva AV, Vlasova MA, Vanin AF, Malyshev IY, Manukhina EB. Adaptation to hypoxia prevents disturbances in cerebral blood flow during neurodegenerative process. Bull Exp Biol Med 2006; 142:169-72. [PMID: 17369930 DOI: 10.1007/s10517-006-0318-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
The rats with neurodegenerative brain disorder induced by administration of a toxic fragment of beta-amyloid demonstrate weakened endothelium-dependent dilation of cerebral vessels, which attested to impaired production of endothelial NO. At the same time, toxic beta-amyloid fragment induced the formation of NO depots in the walls of cerebral vessels, which indirectly attests to NO overproduction in the brain tissue. Preadaptation to hypoxia prevented endothelial dysfunction and improved the efficiency of NO storage. Our results suggest that adaptation to hypoxia protects the brain from various changes in NO production during neurodegenerative damage.
Collapse
Affiliation(s)
- S Yu Mashina
- Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, Moscow
| | | | | | | | | | | | | |
Collapse
|
26
|
Wei IH, Huang CC, Chang HM, Tseng CY, Tu HC, Wen CY, Shieh JY. Neuronal NADPH-d/NOS expression in the nodose ganglion of severe hypoxic rats with or without mild hypoxic preconditioning. J Chem Neuroanat 2005; 29:149-56. [PMID: 15652701 DOI: 10.1016/j.jchemneu.2004.11.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2004] [Revised: 09/20/2004] [Accepted: 11/22/2004] [Indexed: 12/25/2022]
Abstract
This study aimed to test the hypothesis that mild hypoxic preconditioning (MHPC)-induced NOS expression would attenuate the neuropathological changes in the nodose ganglion (NG) of severe hypoxic exposure (SHE) rats. Thus, the young adult rats were caged in the altitude chamber for 4 weeks prior to SHE for 4 h to gain hypoxic preconditioning. The altitude chamber was used to set the height at the level from 5500 m (0.50 atm; pO2=79 Torr) to 10,000 m (0.27 atm; pO2=43 Torr) for MHPC and SHE, respectively. The experimental animals were allowed to survive for 0, 7, 14, 30 and 60 successive days, respectively. Nicotinamide adenine dinucleotide phosphate-diaphorase (NADPH-d) histochemistry and neuronal nitric oxide synthase (nNOS) immunohistochemistry were used to detect NADPH-d/nNOS reactivity in the NG at various time points following hypoxic exposure. The present results showed that about 38% of the neurons in the NG displayed NADPH-d/nNOS positive [NADPH-d/nNOS(+)] in normoxic rats. In SHE rats, a peak in the percentage (71%) and staining intensity (230%) of NADPH-d/nNOS(+) nodose neurons at 0 day, which then gradually decreased at 7-60 days. About 25% of the nodose neurons died 60 days after SHE. However, in MHPC rats subjected to SHE, NADPH-d/nNOS(+) neurons peaked in the percentage (51%) and staining intensity (171%) at 0 day, which then decreased at 7-60 days. In addition, neuronal survival was markedly increased by MHPC. These results suggested that MHPC might have a neuroprotective effect that reduces the susceptibility of the nodose neurons to NOS mediated neuropathy subsequent to SHE.
Collapse
Affiliation(s)
- I-Hua Wei
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | | | | | | | | | | | | |
Collapse
|
27
|
Encinas JM, Fernández AP, Salas E, Castro-Blanco S, Muñoz P, Rodrigo J, Serrano J. Nitric oxide synthase and NADPH-diaphorase after acute hypobaric hypoxia in the rat caudate putamen. Exp Neurol 2004; 186:33-45. [PMID: 14980808 DOI: 10.1016/j.expneurol.2003.09.024] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2003] [Revised: 09/25/2003] [Accepted: 09/26/2003] [Indexed: 10/26/2022]
Abstract
Changes in the production system of nitric oxide (NO), a multifunctional biological messenger known to participate in blood-flow regulation, neuromodulation, and neuroprotection or neurotoxicity, were investigated in the caudate putamen of adult rats submitted to hypobaric hypoxia. Employing immunohistochemistry, Western blotting, enzymatic assay, and NADPH-diaphorase staining, we demonstrate that neuronal nitric oxide synthase (nNOS) expression and constitutive nitric oxide synthase (cNOS) activity were transiently activated by 7 h of exposure to a simulated altitude of 8325 m (27,000 ft). In addition, endothelial nitric oxide synthase (eNOS) immunoreactivity and blood vessel NADPH-diaphorase staining peaked immediately after the hypoxic stimulus, whereas inducible nitric oxide synthase (iNOS) expression and activity remained unaltered. Nitrotyrosine formation, a marker of protein nitration, was evaluated by immunohistochemistry and Western blotting, and was found to increase parallel to nitric oxide synthesis. We conclude that the nitric oxide system undergoes significant transient alterations in the caudate putamen of adult rats submitted to acute hypobaric hypoxia.
Collapse
Affiliation(s)
- Juan Manuel Encinas
- Departamento de Neuroanatomía y Biología Celular, Instituto de Neurobiología Ramón y Cajal, CSIC, E-28002 Madrid, Spain
| | | | | | | | | | | | | |
Collapse
|
28
|
De Maria F, Pedersen JZ, Caccuri AM, Antonini G, Turella P, Stella L, Lo Bello M, Federici G, Ricci G. The specific interaction of dinitrosyl-diglutathionyl-iron complex, a natural NO carrier, with the glutathione transferase superfamily: suggestion for an evolutionary pressure in the direction of the storage of nitric oxide. J Biol Chem 2003; 278:42283-93. [PMID: 12871945 DOI: 10.1074/jbc.m305568200] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The interaction of dinitrosyl-diglutathionyl-iron complex (DNDGIC), a natural carrier of nitric oxide, with representative members of the human glutathione transferase (GST) superfamily, i.e. GSTA1-1, GSTM2-2, GSTP1-1, and GSTT2-2, has been investigated by means of pre-steady and steady state kinetics, fluorometry, electron paramagnetic resonance, and radiometric experiments. This complex binds with extraordinary affinity to the active site of all these dimeric enzymes; GSTA1-1 shows the strongest interaction (KD congruent with 10-10 m), whereas GSTM2-2 and GSTP1-1 display similar and slightly lower affinities (KD congruent with 10-9 m). Binding of the complex to GSTA1-1 triggers structural intersubunit communication, which lowers the affinity for DNDGIC in the vacant subunit and also causes a drastic loss of enzyme activity. Negative cooperativity is also found in GSTM2-2 and GSTP1-1, but it does not affect the catalytic competence of the second subunit. Stopped-flow and fluorescence data fit well to a common minimal binding mechanism, which includes an initial interaction with GSH and a slower bimolecular interaction of DNDGIC with one high and one low affinity binding site. Interestingly, the Theta class GSTT2-2, close to the ancestral precursor of GSTs, shows very slow binding kinetics and hundred times lowered affinity (KD congruent with 10-7 m), whereas the bacterial GSTB1-1 is not inhibited by DNDGIC. Molecular modeling and EPR data reveal structural details that may explain the observed kinetic data. The optimized interaction with this NO carrier, developed in the more recently evolved GSTs, may be related to the acquired capacity to utilize NO as a signal messenger.
Collapse
Affiliation(s)
- Francesca De Maria
- Department of Chemical Sciences and Technologies, University of Rome Tor Vergata, Via della Ricerca Scientifica, 00133 Rome, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Encinas JM, Serrano J, Bentura ML, Castro-Blanco S, Fernández AP, Rodrigo J. Nitric Oxide System and Protein Nitration are Modified by an Acute Hypobaric Hypoxia in the Adult Rat Hippocampus. J Neuropathol Exp Neurol 2003; 62:863-77. [PMID: 14503642 DOI: 10.1093/jnen/62.8.863] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Changes in the nitric oxide system of the hippocampus from rats submitted to hypobaric hypoxia were investigated. Adult rats were exposed to a simulated altitude of 8,325 m (27,000 ft) for 7 h and killed after 0 h, 1, 3, 5, 10 and 20 days of reoxygenation. The number of neuronal nitric oxide synthase immunoreactive neurons and their dendritic plexus, as well as neuronal nitric oxide synthase immunoblotting densitometry and calcium-dependent activity increased from 0 h to 3 days of reoxygenation. In addition, endothelial nitric oxide synthase immunoreactivity peaked after 7 h of hypobaric hypoxia. Nitrotyrosine immunoreactivity showed an increase in the pyramidal cells of CA2-CA3 and in glial cells surrounding the blood vessels after 0 h, 1 and 3 days of reoxygenation. Immunoblotting densitometry of 1 of the 2 nitrotyrosine-immunoreactive bands detected also increased after 0 h and 1 day of reoxygenation. Inducible nitric oxide synthase immunoreactivity was found only in some blood vessels after 0 h, 1 and 3 days of reoxygenation, but no changes in inducible nitric oxide synthase activity or immunoblotting were detected. We conclude that transient activation of the nitric oxide system constitutes a hippocampal response to hypobaric hypoxia.
Collapse
Affiliation(s)
- Juan Manuel Encinas
- Department of Neuroanatomy and Cell Biology of the Instituto de Neurobiología Ramón y Cajal, CSIC, Madrid, Spain
| | | | | | | | | | | |
Collapse
|
30
|
Castro-Blanco S, Encinas JM, Serrano J, Alonso D, Gómez MB, Sánchez J, Ríos-Tejada F, Fernández-Vizarra P, Fernández AP, Martínez-Murillo R, Rodrigo J. Expression of nitrergic system and protein nitration in adult rat brains submitted to acute hypobaric hypoxia. Nitric Oxide 2003; 8:182-201. [PMID: 12826067 DOI: 10.1016/s1089-8603(03)00003-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Changes in the nitric oxide (NO) system of the rat cerebral cortex were investigated by immunohistochemistry, immunoblotting, and NO synthase (NOS) activity assays in adult rats submitted for 30 min to hypoxia, in a hypobaric chamber at a simulated altitude of 38,000 ft (11000 m) (154.9 mm Hg). The cerebral cortex was studied after different survival times, 0 and 24 h, 5, 8, 15, and 30 days of reoxygenation. This situation led to morphological alterations in the large type I interneurons, as well as immunoreactive changes in the appearance and number of the small neurons (type II), both containing neuronal NOS (nNOS). Some of these small neurons showed immunoreactive cytoplasm and short processes; others, the more numerous during all reoxygenation periods, contained the immunoreactive product mainly related to a perinuclear ring. Ultrastructurally, these small neurons exhibited changes in nuclear structures as in the shape of the nuclear membrane, in the distribution of heterochromatin, and in the nucleolar morphology. The reaction product for nitrotyrosine, as a marker of protein nitration, showed modifications in distribution of the immunoreactive product. No expression was found for inducible NOS (iNOS). All these modifications were accompanied by increased nNOS and nitrotyrosine production as demonstrated by Western blotting and calcium-dependent activity, returning to control conditions after 30 days of reoxygenation, suggesting a reversible NO mechanism of action.
Collapse
Affiliation(s)
- Susana Castro-Blanco
- Department of Neuroanatomy and Cell Biology, Instituto de Neurobiologia Santiago Ramón y Cajal, CSIC, E-28002 Madrid, Spain
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Kametas NA, Savvidou MD, Donald AE, McAuliffe F, Nicolaides KH. Flow-mediated dilatation of the brachial artery in pregnancy at high altitude. BJOG 2002; 109:930-7. [PMID: 12197374 DOI: 10.1111/j.1471-0528.2002.01160.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
OBJECTIVE Pregnancy at high altitude has been associated with increased prevalence of pre-eclampsia and reduced maternal oestrogen levels, factors that have been associated with endothelial dysfunction. The aim of this study was to examine the effect of high altitude (4370 m above sea level) on endothelial function during pregnancy as assessed by a non-invasive method. DESIGN Cross-sectional study. SETTING Two maternity units providing routine antenatal care: one at high altitude (District General Hospital--IPSS in Cerro de Pasco, Peru) and one at sea level (Instituto Materno-Perinatal in Lima, Peru). POPULATION Sixty pregnant women at 6-42 weeks of gestation resident at high altitude (Cerro de Pasco, Peru, 4370 m above sea level) and 54 at sea level (Lima, Peru). Comparisons were performed also in 11 and 14 non-pregnant women at each altitude, respectively. METHODS Endothelial function was assessed by flow-mediated dilatation of the brachial artery using high-resolution ultrasound. MAIN OUTCOME MEASURES Differences in flow mediated dilatation of the brachial artery in two groups of pregnant women, one at high altitude and one at sea level. RESULTS Both at high altitude and sea level flow-mediated dilatation of the brachial artery increased in the first two trimesters to levels 32% higher than non-pregnant controls. However, in the third trimester, flow-mediated dilatation of the brachial artery was lower than non-pregnant levels. Resting vessel size increased during pregnancy by 15% compared with non-pregnant controls at term, with no difference between the two populations at high and low altitude. Pregnancy at high altitude, compared with sea level, was associated with 59% lower baseline blood flow and 76% higher reactive hyperaemia. Similarly, non-pregnant controls at high altitude compared with sea level demonstrated similar flow-mediated dilatation of the brachial artery and 40% lower resting blood flow of the brachial artery. However, the difference in reactive hyperaemia did not reach statistical significance. CONCLUSION These data suggest that, during pregnancy at high altitude, endothelial function, as assessed by flow-mediated dilatation of the brachial artery, is not impaired.
Collapse
Affiliation(s)
- Nikos A Kametas
- Harris Birthright Research Centre for Fetal Medicine, King's College Hospital, Denmark Hill, London SE5 9RS, UK
| | | | | | | | | |
Collapse
|
32
|
Xi L, Tekin D, Gursoy E, Salloum F, Levasseur JE, Kukreja RC. Evidence that NOS2 acts as a trigger and mediator of late preconditioning induced by acute systemic hypoxia. Am J Physiol Heart Circ Physiol 2002; 283:H5-12. [PMID: 12063268 DOI: 10.1152/ajpheart.00920.2001] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Chronic systemic hypoxia (SH) enhances myocardial ischemic tolerance in mammals. We studied the delayed cardioprotection caused by acute SH and associated signaling mechanism. Conscious adult male mice were exposed to one or two cycles of hypoxia (H; 10% O(2)) or normoxia (21% O(2)) for various durations (30 min, 2 h, 4 h) followed by 24 h of reoxygenation. Hearts were isolated 24 h later and subjected to ischemia-reperfusion in a Langendorff model. Infarct size was reduced in mice pretreated with one (H4h) or two cycles (H4hx2) of 4 h SH compared with normoxia mice (P < 0.05), which was abolished by an inducible nitric oxide synthase (NOS2) inhibitor (S-methylisothiourea, 3 mg/kg) given before SH or ischemia. H4hx2 also failed to reduce infarct size in NOS2 knockout mice. Cyclooxygenase-2 (COX-2) inhibitor (NS-398, 10 mg/kg) did not block the protection given either before H4hx2 or ischemia. A two- to three fold increase in myocardial NOS2 expression was observed in H4h, H2hx2, and H4hx2 (P < 0.05), whereas endothelial NOS (NOS3) or COX-2 remained unchanged. We conclude that acute SH induces delayed cardioprotection, which is triggered and mediated by NOS2, but not by NOS3 or COX-2.
Collapse
Affiliation(s)
- Lei Xi
- Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia 23298-0281, USA
| | | | | | | | | | | |
Collapse
|
33
|
Muller B, Kleschyov AL, Alencar JL, Vanin A, Stoclet JC. Nitric oxide transport and storage in the cardiovascular system. Ann N Y Acad Sci 2002; 962:131-9. [PMID: 12076970 DOI: 10.1111/j.1749-6632.2002.tb04063.x] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Despite short halflife in biological fluids, nitric oxide (NO) can produce remote or long lasting effect in the cardiovascular system. Long distance transport or local storage of NO might explain these effects. In blood, recent findings suggest that in addition to being a major consumption pathway, interaction of NO with hemoglobin may permit O(2)-governed transport of NO (as S-nitrosohemoglobin) to tissues in which NO may be released together with O(2), via transnitrosation of a transport protein. In blood vessels, two different putative NO stores have been characterized. The first is the photosensitive store, formed from endothelium-derived NO. The mechanism of NO release from this store in the body (in absence of light) and its physiological relevance are unknown. The second store is generated in conditions of high tissue NO levels, as a consequence of the inducible NO synthase activity or in various stress conditions. This NO store involves formation of protein-bound dinitrosyl iron complexes or S-nitrosated proteins, or both. Low molecular weight thiols can displace NO from these stores and probably transfer it to target membrane protein(s) such as K(+) channels, via transnitrosation reactions. These stores may be involved in defence mechanisms against inflammation or stress. Thus, NO transport and storage mechanisms may be implicated in a variety of NO effects. The mechanisms of their formation and of NO release and their physiologic and pathophysiologic relevance deserve further investigations.
Collapse
Affiliation(s)
- Bernard Muller
- Université Louis Pasteur, CNRS UMR 7034, Faculté de Pharmacie, BP 24, 67401 ILLKIRCH Cedex, France.
| | | | | | | | | |
Collapse
|
34
|
Freitas I, Griffini P, Bertone V, Bertone R, Fenoglio C, Milliery R, Vairetti M. In situ detection of reactive oxygen species and nitric oxide production in normal and pathological tissues: improvement by differential interference contrast. Exp Gerontol 2002; 37:591-602. [PMID: 11830363 DOI: 10.1016/s0531-5565(01)00226-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Reactive oxygen species (ROS), among which nitric oxide (NO) is currently included, play a plethora of (patho)physiological roles. Harman's free radical theory of aging put forth over 40 years ago received full support since then. A nitric oxide hypothesis of aging recently proposed by McCann, is very likely to be the object of widespread investigation in the near future. Therefore, the possibility of localizing at the (sub)cellular level under the light microscope the sites of ROS and NO production with simple and reliable methods appears as a powerful tool for analytic cytology and pathology. Various histochemical methods were developed for visualizing ROS production; a recently improved version to localize superoxide (and possibly also singlet oxygen), based on a DAB-Mn2+ -Co2+ reaction, appears very promising. Since the direct detection of NO is still very difficult, the action sites of NO are currently localized by the identification of NO synthase (NOS). The most widespread method to reveal the catalytic activity of NOS is that of demonstrating the fixation-resistant NADPH diaphorase activity with the tetrazolium salt method. We have improved this method by using a tetrazolium salt whose formazan particles are very thin and lipid insoluble (tetranitroblue tetrazolium, TNBT) and by including a tissue protectant, polyvinyl alcohol, in the incubation medium. Here significant examples of application of the DAB-Mn2+ -Co2+ technique for ROS and the TNBT-PVA method for NOS to normal liver and brain and to solid tumors are presented. We further document the usefulness of Nomarkis's differential interference contrast (DIC) to analyze wide tissue areas where ROS production or NOS activity is low or even nil. The improved version for NOS allowed for the first time to demonstrate NOS activity in liver fat-storing cells and in astrocyte-like cells in the brain.
Collapse
Affiliation(s)
- Isabel Freitas
- Department of Animal Biology and C.N.R. Center for Histochemistry, University of Pavia, 27100 Pavia, Italy.
| | | | | | | | | | | | | |
Collapse
|
35
|
Wang PG, Xian M, Tang X, Wu X, Wen Z, Cai T, Janczuk AJ. Nitric oxide donors: chemical activities and biological applications. Chem Rev 2002; 102:1091-134. [PMID: 11942788 DOI: 10.1021/cr000040l] [Citation(s) in RCA: 1000] [Impact Index Per Article: 43.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Peng George Wang
- Department of Chemistry, Wayne State University, Detroit, Michigan 48202, USA.
| | | | | | | | | | | | | |
Collapse
|
36
|
Galleguillos M, Valenzuela MA, Riquelme R, Sanhueza E, Sánchez G, Figueroa JP, Llanos AJ. Nitric oxide synthase activity in brain tissues from llama fetuses submitted to hypoxemia. Comp Biochem Physiol A Mol Integr Physiol 2001; 129:605-14. [PMID: 11423330 DOI: 10.1016/s1095-6433(01)00299-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The fetal llama (Lama glama; a species adapted to live in chronic hypoxia in the highlands of the Andes) did not increase cerebral blood flow and reduce the brain oxygen uptake during hypoxemia. Although nitric oxide (NO) is a normal mediator in the regulation of vascular tone and synaptic transmission, NO overproduction by hypoxemia could produce neuronal damage. We hypothesized that nitric oxide synthase (NOS) activity is either maintained or reduced in the central nervous system of the llama fetuses submitted to chronic hypoxemia. Approximately 85% of the Ca(2+)-dependent NOS activity was soluble, at least 12% was associated with the mitochondrial fraction, and less than 5% remains associated with microsomes. To understand the role of NO in chronic hypoxemia, we determined the effect of 24-h hypoxemia on NOS activity in the central nervous system. No changes in activity or the subcellular distribution of NOS activity in brain tissues after hypoxemia were found. We proposed that the lack of changes in NOS activity in the llama under hypoxemia could be a cytoprotective mechanism inherent to the llama, against possible toxic effects of NO.
Collapse
Affiliation(s)
- M Galleguillos
- Departamento de Ciencias Biológicas Animales, Facultad de Ciencias Veterinarias y Pecuarias, Universidad de Chile, Santiago, Chile
| | | | | | | | | | | | | |
Collapse
|