1
|
Lin H, Liang Y, Zhao W, Cao J, Wang T, Wang C. Reassessing the role of nitric oxide in the pathogenesis of sphincter of Oddi dysfunction. Gastroenterol Rep (Oxf) 2025; 13:goaf001. [PMID: 40046950 PMCID: PMC11882319 DOI: 10.1093/gastro/goaf001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 07/25/2024] [Accepted: 09/13/2024] [Indexed: 04/11/2025] Open
Abstract
The pathogenic mechanisms underlying sphincter of Oddi dysfunction (SOD) remain incompletely understood, and it often leads to severe symptoms encompassing nausea, vomiting, and abdominal pain. New evidence now suggests correlations between nitric oxide (NO) and SOD. In this review, we summarized the factors influencing SOD pathogenesis via NO and its derivative, the peroxynitrite anion. NO appears to enhance SOD progression by modulating sphincter of Oddi (SO) contractions via NO-sGC-cGMP signaling or inducing the apoptosis of enteric neurons, interstitial cells of Cajal, smooth muscle cells, and other cellular components via peroxynitrite anion-mediated organelle damage. Thus, a comprehensive understanding of SOD will provide a foundation for the identification of potential drugs and treatment approaches.
Collapse
Affiliation(s)
- Haonan Lin
- Department of General Surgery, Affiliated Hospital of Shaoxing University, Shaoxing, Zhejiang, P. R. China
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, P. R. China
| | - Yixuan Liang
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, P. R. China
- School of Integrative Medicine, Dalian Medical University, Dalian, Liaoning, P. R. China
| | - Wangqiang Zhao
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, P. R. China
- School of Integrative Medicine, Dalian Medical University, Dalian, Liaoning, P. R. China
| | - Junwei Cao
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, P. R. China
- School of Integrative Medicine, Dalian Medical University, Dalian, Liaoning, P. R. China
| | - Tianqi Wang
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, P. R. China
| | - Changmiao Wang
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, P. R. China
| |
Collapse
|
2
|
Anderle S, Bonnar O, Henderson J, Shaw K, Chagas AM, McMullan L, Webber A, McGowan K, King SL, Hall CN. APOE4 and sedentary lifestyle synergistically impair neurovascular function in the visual cortex of awake mice. Commun Biol 2025; 8:144. [PMID: 39880935 PMCID: PMC11779976 DOI: 10.1038/s42003-025-07585-z] [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: 06/07/2024] [Accepted: 01/21/2025] [Indexed: 01/31/2025] Open
Abstract
Reduced cerebral blood flow occurs early in the development of Alzheimer's disease (AD), but the factors producing this reduction are unknown. Here, we ask whether genetic and lifestyle risk factors for AD-the ε4 allele of the Apolipoprotein (APOE) gene, and physical activity-can together produce this reduction in cerebral blood flow which leads eventually to AD. Using in vivo two-photon microscopy and haemodynamic measures, we record neurovascular function from the visual cortex of physically active or sedentary mice expressing APOE3 and APOE4 in place of murine APOE. Energy supply and demand are mismatched in APOE4 mice, with smaller increases in cerebral blood flow, blood volume and blood oxygenation occurring during neuronal activation as blood vessels frequently fail to dilate. Exercise dose-dependently overall improves neurovascular function, with an increased impact of exercise apparent after longer exposure times. Several haemodynamic measures show a larger beneficial effect of exercise in APOE4 vs. APOE3 mice. Thus, APOE4 genotype in conjunction with sedentary behaviour produces the worst neurovascular function. Promotion of physical activity may therefore be particularly important to improve cerebrovascular function and reduce dementia risk in APOE4 carriers.
Collapse
Affiliation(s)
- Silvia Anderle
- School of Psychology and Sussex Neuroscience, University of Sussex, Brighton, UK
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, UK
| | - Orla Bonnar
- School of Psychology and Sussex Neuroscience, University of Sussex, Brighton, UK
- MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital and Harvard Medical School, Boston, USA
| | - Joseph Henderson
- School of Psychology and Sussex Neuroscience, University of Sussex, Brighton, UK
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Kira Shaw
- School of Psychology and Sussex Neuroscience, University of Sussex, Brighton, UK
| | - Andre M Chagas
- School of Psychology and Sussex Neuroscience, University of Sussex, Brighton, UK
| | - Letitia McMullan
- School of Psychology and Sussex Neuroscience, University of Sussex, Brighton, UK
| | - Alexandra Webber
- School of Psychology and Sussex Neuroscience, University of Sussex, Brighton, UK
| | - Kirsty McGowan
- School of Psychology and Sussex Neuroscience, University of Sussex, Brighton, UK
| | - Sarah L King
- School of Psychology and Sussex Neuroscience, University of Sussex, Brighton, UK
| | - Catherine N Hall
- School of Psychology and Sussex Neuroscience, University of Sussex, Brighton, UK.
| |
Collapse
|
3
|
Park MK, Yang HW, Woo SY, Kim DY, Son DS, Choi BY, Suh SW. Modulation of Second Messenger Signaling in the Brain Through PDE4 and PDE5 Inhibition: Therapeutic Implications for Neurological Disorders. Cells 2025; 14:86. [PMID: 39851514 PMCID: PMC11763391 DOI: 10.3390/cells14020086] [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: 12/11/2024] [Revised: 01/07/2025] [Accepted: 01/07/2025] [Indexed: 01/26/2025] Open
Abstract
Phosphodiesterase (PDE) enzymes regulate intracellular signaling pathways crucial for brain development and the pathophysiology of neurological disorders. Among the 11 PDE subtypes, PDE4 and PDE5 are particularly significant due to their regulation of cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP) signaling, respectively, which are vital for learning, memory, and neuroprotection. This review synthesizes current evidence on the roles of PDE4 and PDE5 in neurological health and disease, focusing on their regulation of second messenger pathways and their implications for brain function. Elevated PDE4 activity impairs synaptic plasticity by reducing cAMP levels and protein kinase A (PKA) activity, contributing to cognitive decline, acute brain injuries, and neuropsychiatric conditions such as bipolar disorder and schizophrenia. Similarly, PDE5 dysregulation disrupts nitric oxide (NO) signaling and protein kinase G (PKG) pathways, which are involved in cerebrovascular homeostasis, recovery after ischemic events, and neurodegenerative processes in Alzheimer's, Parkinson's, and Huntington's diseases. PDE4 and PDE5 are promising therapeutic targets for neurological disorders. Pharmacological modulation of these enzymes offers potential to enhance cognitive function and mitigate pathological mechanisms underlying brain injuries, neurodegenerative diseases, and psychiatric disorders. Further research into the regulation of PDE4 and PDE5 will advance therapeutic strategies for these conditions.
Collapse
Affiliation(s)
- Min Kyu Park
- Department of Physiology, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea; (M.K.P.); (H.W.Y.); (S.Y.W.); (D.Y.K.)
| | - Hyun Wook Yang
- Department of Physiology, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea; (M.K.P.); (H.W.Y.); (S.Y.W.); (D.Y.K.)
| | - Seo Young Woo
- Department of Physiology, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea; (M.K.P.); (H.W.Y.); (S.Y.W.); (D.Y.K.)
| | - Dong Yeon Kim
- Department of Physiology, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea; (M.K.P.); (H.W.Y.); (S.Y.W.); (D.Y.K.)
| | - Dae-Soon Son
- Division of Data Science, Data Science Convergence Research Center, Hallym University, Chuncheon 24252, Republic of Korea;
| | - Bo Young Choi
- Institute of Sport Science, Hallym University, Chuncheon 24252, Republic of Korea;
- Department of Physical Education, Hallym University, Chuncheon 24252, Republic of Korea
| | - Sang Won Suh
- Department of Physiology, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea; (M.K.P.); (H.W.Y.); (S.Y.W.); (D.Y.K.)
| |
Collapse
|
4
|
Yang P, Nie T, Sun X, Xu L, Ma C, Wang F, Long L, Chen J. Wheel-Running Exercise Alleviates Anxiety-Like Behavior via Down-Regulating S-Nitrosylation of Gephyrin in the Basolateral Amygdala of Male Rats. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400205. [PMID: 38965798 PMCID: PMC11425869 DOI: 10.1002/advs.202400205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 06/06/2024] [Indexed: 07/06/2024]
Abstract
Physical exercise has beneficial effect on anxiety disorders, but the underlying molecular mechanism remains largely unknown. Here, it is demonstrated that physical exercise can downregulate the S-nitrosylation of gephyrin (SNO-gephyrin) in the basolateral amygdala (BLA) to exert anxiolytic effects. It is found that the level of SNO-gephyrin is significantly increased in the BLA of high-anxiety rats and a downregulation of SNO-gephyrin at cysteines 212 and 284 produced anxiolytic effect. Mechanistically, inhibition of SNO-gephyrin by either Cys212 or Cys284 mutations increased the surface expression of GABAAR γ2 and the subsequent GABAergic neurotransmission, exerting anxiolytic effect in male rats. On the other side, overexpression of neuronal nitric oxide synthase in the BLA abolished the anxiolytic-like effects of physical exercise. This study reveals a key role of downregulating SNO-gephyrin in the anxiolytic effects of physical exercise, providing a new explanation for protein post-translational modifications in the brain after exercise.
Collapse
Affiliation(s)
- Ping‐Fen Yang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious DiseasesDepartment of PharmacologySchool of Basic MedicineTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
- The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei ProvinceWuhan430030China
| | - Tai‐Lei Nie
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious DiseasesDepartment of PharmacologySchool of Basic MedicineTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
- The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei ProvinceWuhan430030China
| | - Xia‐Nan Sun
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious DiseasesDepartment of PharmacologySchool of Basic MedicineTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
- The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei ProvinceWuhan430030China
| | - Lan‐Xin Xu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious DiseasesDepartment of PharmacologySchool of Basic MedicineTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
- The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei ProvinceWuhan430030China
| | - Cong Ma
- Key Laboratory of Molecular Biophysics of the Ministry of EducationCollege of Life Science and TechnologyHuazhong University of Science and TechnologyWuhan430030China
| | - Fang Wang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious DiseasesDepartment of PharmacologySchool of Basic MedicineTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
- The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei ProvinceWuhan430030China
- Hubei Shizhen LaboratoryWuhan430030China
| | - Li‐Hong Long
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious DiseasesDepartment of PharmacologySchool of Basic MedicineTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
- The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei ProvinceWuhan430030China
- Hubei Shizhen LaboratoryWuhan430030China
| | - Jian‐Guo Chen
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious DiseasesDepartment of PharmacologySchool of Basic MedicineTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
- The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei ProvinceWuhan430030China
- Hubei Shizhen LaboratoryWuhan430030China
| |
Collapse
|
5
|
Cabello MC, Chen G, Melville MJ, Osman R, Kumar GD, Domaille DW, Lippert AR. Ex Tenebris Lux: Illuminating Reactive Oxygen and Nitrogen Species with Small Molecule Probes. Chem Rev 2024; 124:9225-9375. [PMID: 39137397 DOI: 10.1021/acs.chemrev.3c00892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/15/2024]
Abstract
Reactive oxygen and nitrogen species are small reactive molecules derived from elements in the air─oxygen and nitrogen. They are produced in biological systems to mediate fundamental aspects of cellular signaling but must be very tightly balanced to prevent indiscriminate damage to biological molecules. Small molecule probes can transmute the specific nature of each reactive oxygen and nitrogen species into an observable luminescent signal (or even an acoustic wave) to offer sensitive and selective imaging in living cells and whole animals. This review focuses specifically on small molecule probes for superoxide, hydrogen peroxide, hypochlorite, nitric oxide, and peroxynitrite that provide a luminescent or photoacoustic signal. Important background information on general photophysical phenomena, common probe designs, mechanisms, and imaging modalities will be provided, and then, probes for each analyte will be thoroughly evaluated. A discussion of the successes of the field will be presented, followed by recommendations for improvement and a future outlook of emerging trends. Our objectives are to provide an informative, useful, and thorough field guide to small molecule probes for reactive oxygen and nitrogen species as well as important context to compare the ecosystem of chemistries and molecular scaffolds that has manifested within the field.
Collapse
Affiliation(s)
- Maidileyvis C Cabello
- Department of Chemistry, Southern Methodist University, Dallas, Texas 75275-0314, United States
| | - Gen Chen
- Department of Chemistry, Southern Methodist University, Dallas, Texas 75275-0314, United States
| | - Michael J Melville
- Department of Chemistry, Colorado School of Mines, Golden, Colorado 80401, United States
| | - Rokia Osman
- Department of Chemistry, Southern Methodist University, Dallas, Texas 75275-0314, United States
| | - G Dinesh Kumar
- Department of Chemistry, Colorado School of Mines, Golden, Colorado 80401, United States
| | - Dylan W Domaille
- Department of Chemistry, Colorado School of Mines, Golden, Colorado 80401, United States
| | - Alexander R Lippert
- Department of Chemistry, Southern Methodist University, Dallas, Texas 75275-0314, United States
| |
Collapse
|
6
|
Scarpellino G, Brunetti V, Berra-Romani R, De Sarro G, Guerra G, Soda T, Moccia F. The Unexpected Role of the Endothelial Nitric Oxide Synthase at the Neurovascular Unit: Beyond the Regulation of Cerebral Blood Flow. Int J Mol Sci 2024; 25:9071. [PMID: 39201757 PMCID: PMC11354477 DOI: 10.3390/ijms25169071] [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: 07/22/2024] [Revised: 08/19/2024] [Accepted: 08/21/2024] [Indexed: 09/03/2024] Open
Abstract
Nitric oxide (NO) is a highly versatile gasotransmitter that has first been shown to regulate cardiovascular function and then to exert tight control over a much broader range of processes, including neurotransmitter release, neuronal excitability, and synaptic plasticity. Endothelial NO synthase (eNOS) is usually far from the mind of synaptic neurophysiologists, who have focused most of their attention on neuronal NO synthase (nNOS) as the primary source of NO at the neurovascular unit (NVU). Nevertheless, the available evidence suggests that eNOS could also contribute to generating the burst of NO that, serving as volume intercellular messenger, is produced in response to neuronal activity in the brain parenchyma. Herein, we review the role of eNOS in both the regulation of cerebral blood flow and of synaptic plasticity and discuss the mechanisms by which cerebrovascular endothelial cells may transduce synaptic inputs into a NO signal. We further suggest that eNOS could play a critical role in vascular-to-neuronal communication by integrating signals converging onto cerebrovascular endothelial cells from both the streaming blood and active neurons.
Collapse
Affiliation(s)
- Giorgia Scarpellino
- Laboratory of General Physiology, Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, 27100 Pavia, Italy; (G.S.); (V.B.)
| | - Valentina Brunetti
- Laboratory of General Physiology, Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, 27100 Pavia, Italy; (G.S.); (V.B.)
| | - Roberto Berra-Romani
- Department of Biomedicine, School of Medicine, Benemérita Universidad Autónoma de Puebla, Puebla 72410, Mexico;
| | - Giovambattista De Sarro
- Department of Health Sciences, University of Magna Graecia, 88100 Catanzaro, Italy; (G.D.S.); (T.S.)
| | - Germano Guerra
- Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, 86100 Campobasso, Italy;
| | - Teresa Soda
- Department of Health Sciences, University of Magna Graecia, 88100 Catanzaro, Italy; (G.D.S.); (T.S.)
| | - Francesco Moccia
- Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, 86100 Campobasso, Italy;
| |
Collapse
|
7
|
Stykel MG, Ryan SD. Network analysis of S-nitrosylated synaptic proteins demonstrates unique roles in health and disease. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119720. [PMID: 38582237 DOI: 10.1016/j.bbamcr.2024.119720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 03/24/2024] [Accepted: 03/27/2024] [Indexed: 04/08/2024]
Abstract
Nitric oxide can covalently modify cysteine thiols on target proteins to alter that protein's function in a process called S-nitrosylation (SNO). S-nitrosylation of synaptic proteins plays an integral part in neurotransmission. Here we review the function of the SNO-proteome at the synapse and whether clusters of SNO-modification may predict synaptic dysfunction associated with disease. We used a systematic search strategy to concatenate SNO-proteomic datasets from normal human or murine brain samples. Identified SNO-modified proteins were then filtered against proteins reported in the Synaptome Database, which provides a detailed and experimentally verified annotation of all known synaptic proteins. Subsequently, we performed an unbiased network analysis of all known SNO-synaptic proteins to identify clusters of SNO proteins commonly involved in biological processes or with known disease associations. The resulting SNO networks were significantly enriched in biological processes related to metabolism, whereas significant gene-disease associations were related to Schizophrenia, Alzheimer's, Parkinson's and Huntington's disease. Guided by an unbiased network analysis, the current review presents a thorough discussion of how clustered changes to the SNO-proteome influence health and disease.
Collapse
Affiliation(s)
- Morgan G Stykel
- Department of Molecular and Cellular Biology, The University of Guelph, Guelph, ON, Canada
| | - Scott D Ryan
- Department of Molecular and Cellular Biology, The University of Guelph, Guelph, ON, Canada; Hotchkiss Brain Institute, Department of Clinical Neuroscience, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
8
|
Longden TA, Lederer WJ. Electro-metabolic signaling. J Gen Physiol 2024; 156:e202313451. [PMID: 38197953 PMCID: PMC10783436 DOI: 10.1085/jgp.202313451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 10/27/2023] [Accepted: 12/14/2023] [Indexed: 01/11/2024] Open
Abstract
Precise matching of energy substrate delivery to local metabolic needs is essential for the health and function of all tissues. Here, we outline a mechanistic framework for understanding this critical process, which we refer to as electro-metabolic signaling (EMS). All tissues exhibit changes in metabolism over varying spatiotemporal scales and have widely varying energetic needs and reserves. We propose that across tissues, common signatures of elevated metabolism or increases in energy substrate usage that exceed key local thresholds rapidly engage mechanisms that generate hyperpolarizing electrical signals in capillaries that then relax contractile elements throughout the vasculature to quickly adjust blood flow to meet changing needs. The attendant increase in energy substrate delivery serves to meet local metabolic requirements and thus avoids a mismatch in supply and demand and prevents metabolic stress. We discuss in detail key examples of EMS that our laboratories have discovered in the brain and the heart, and we outline potential further EMS mechanisms operating in tissues such as skeletal muscle, pancreas, and kidney. We suggest that the energy imbalance evoked by EMS uncoupling may be central to cellular dysfunction from which the hallmarks of aging and metabolic diseases emerge and may lead to generalized organ failure states-such as diverse flavors of heart failure and dementia. Understanding and manipulating EMS may be key to preventing or reversing these dysfunctions.
Collapse
Affiliation(s)
- Thomas A. Longden
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
- Laboratory of Neurovascular Interactions, Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - W. Jonathan Lederer
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
- Laboratory of Molecular Cardiology, Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
9
|
Vaccaro MG, Innocenti B, Cione E, Gallelli L, De Sarro G, Bonilla DA, Cannataro R. Acute effects of a chewable beetroot-based supplement on cognitive performance: a double-blind randomized placebo-controlled crossover clinical trial. Eur J Nutr 2024; 63:303-321. [PMID: 37875637 PMCID: PMC10799154 DOI: 10.1007/s00394-023-03265-y] [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: 09/04/2022] [Accepted: 10/03/2023] [Indexed: 10/26/2023]
Abstract
BACKGROUND Dietary nitrate (NO3-) has been shown to be useful as an ergogenic aid with potential applications in health and disease (e.g., blood pressure control). However, there is no consensus about the effects of dietary NO3- or beetroot (BR) juice supplementation on cognitive function. OBJECTIVE The aim of this study was to evaluate the effects of a single dose of a chewable BR-based supplement on cognitive performance. METHODS A double-blind randomized placebo-controlled two-period crossover clinical trial was carried out based on the extension of the CONSORT guidelines for randomized crossover trials. A total of 44 participants (24 F; 20 M; 32.7 [12.5] years; 66.3 [9.0] kg; 170 [9.2] cm; 22.8 [1.4] kg/m2) were randomly allocated to receive first either four BR-based chewable tablets (BR-CT) containing 3 g of a Beta vulgaris extract (RedNite®) or four tablets of a placebo (maltodextrin). A 4-day washout period was used before crossover. Ninety minutes after ingestion of the treatments, a neuropsychological testing battery was administered in each period. The trial was registered at clinicaltrials.gov NCT05509075. RESULTS Significant improvements with moderate effect size were found on memory consolidation at the short and long term only after BR-CT supplementation via the Rey Auditory Verbal Learning Test immediate (+ 20.69%) and delayed (+ 12.34%) recalls. Likewise, enhancement on both frontal lobe functions (+ 2.57%) and cognitive flexibility (+ 11.16%) were detected after BR-CT. There was no significant change (p < 0.05) on verbal memory of short-term digits, working memory and information processing speed. Mixed results were found on mood and anxiety through the Beck Depression Inventory-II (BDI-II) and the State-Trait Anxiety Inventory (STAI-Y1 and STAI-Y2); however, sequence and period effects were seen on STAI-Y2. CONCLUSIONS The acute administration of a chewable BR-based supplement improves certain aspects of cognitive function in healthy females and males, particularly memory capacity and frontal skills.
Collapse
Affiliation(s)
- Maria Grazia Vaccaro
- Department of Medical and Surgical Sciences, University of Magna Graecia, Catanzaro, Italy
| | | | - Erika Cione
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
- Galascreen Laboratories, University of Calabria, Rende, Italy
| | - Luca Gallelli
- Clinical Pharmacology and Pharmacovigilance Operative Unit, Department of Health Science, University of Magna Graecia, Mater Domini Hospital, Catanzaro, Italy
| | - Giovambattista De Sarro
- Clinical Pharmacology and Pharmacovigilance Operative Unit, Department of Health Science, University of Magna Graecia, Mater Domini Hospital, Catanzaro, Italy
| | - Diego A Bonilla
- Research Division, Dynamical Business and Science Society-DBSS International SAS, 110861, Bogotá, Colombia
- Research Group in Physical Activity, Sports and Health Sciences (GICAFS), Universidad de Córdoba, 230002, Montería, Colombia
- Research Group in Biochemistry and Molecular Biology, Universidad Distrital Francisco José de Caldas, 110311, Bogotá, Colombia
| | - Roberto Cannataro
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy.
- Galascreen Laboratories, University of Calabria, Rende, Italy.
- Research Division, Dynamical Business and Science Society-DBSS International SAS, 110861, Bogotá, Colombia.
| |
Collapse
|
10
|
Kimura H. Hydrogen Sulfide (H 2S)/Polysulfides (H 2S n) Signalling and TRPA1 Channels Modification on Sulfur Metabolism. Biomolecules 2024; 14:129. [PMID: 38275758 PMCID: PMC10813152 DOI: 10.3390/biom14010129] [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: 12/19/2023] [Revised: 01/11/2024] [Accepted: 01/17/2024] [Indexed: 01/27/2024] Open
Abstract
Hydrogen sulfide (H2S) and polysulfides (H2Sn, n ≥ 2) produced by enzymes play a role as signalling molecules regulating neurotransmission, vascular tone, cytoprotection, inflammation, oxygen sensing, and energy formation. H2Sn, which have additional sulfur atoms to H2S, and other S-sulfurated molecules such as cysteine persulfide and S-sulfurated cysteine residues of proteins, are produced by enzymes including 3-mercaptopyruvate sulfurtransferase (3MST). H2Sn are also generated by the chemical interaction of H2S with NO, or to a lesser extent with H2O2. S-sulfuration (S-sulfhydration) has been proposed as a mode of action of H2S and H2Sn to regulate the activity of target molecules. Recently, we found that H2S/H2S2 regulate the release of neurotransmitters, such as GABA, glutamate, and D-serine, a co-agonist of N-methyl-D-aspartate (NMDA) receptors. H2S facilitates the induction of hippocampal long-term potentiation, a synaptic model of memory formation, by enhancing the activity of NMDA receptors, while H2S2 achieves this by activating transient receptor potential ankyrin 1 (TRPA1) channels in astrocytes, potentially leading to the activation of nearby neurons. The recent findings show the other aspects of TRPA1 channels-that is, the regulation of the levels of sulfur-containing molecules and their metabolizing enzymes. Disturbance of the signalling by H2S/H2Sn has been demonstrated to be involved in various diseases, including cognitive and psychiatric diseases. The physiological and pathophysiological roles of these molecules will be discussed.
Collapse
Affiliation(s)
- Hideo Kimura
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Sanyo-Onoda City University, 1-1-1 Daigaku-Dori, Sanyo-Onoda 756-0884, Yamaguchi, Japan
| |
Collapse
|
11
|
Dutta B, Shelar SB, Nirmalraj A, Gupta S, Barick KC, Gupta J, Hassan PA. Smart Magnetic Nanocarriers for Codelivery of Nitric Oxide and Doxorubicin for Enhanced Apoptosis in Cancer Cells. ACS OMEGA 2023; 8:44545-44557. [PMID: 38046289 PMCID: PMC10688159 DOI: 10.1021/acsomega.3c03734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 10/30/2023] [Accepted: 10/31/2023] [Indexed: 12/05/2023]
Abstract
Extremely short half-life therapeutic molecule nitric oxide (NO) plays significant roles in the functioning of various physiological and pathological processes in the human body, whereas doxorubicin hydrochloride (DOX) is a clinically important anticancer drug widely used in cancer chemotherapy. Thus, the intracellular delivery of these therapeutic molecules is tremendously important to achieve their full potential. Herein, we report a novel approach for the development of highly water-dispersible magnetic nanocarriers for codelivery of NO and DOX. Primarily, bifunctional magnetic nanoparticles enriched with carboxyl and thiol groups were prepared by introducing cysteine onto the surface of citrate-functionalized Fe3O4 nanoparticles. DOX was electrostatically conjugated onto the surface of bifunctional nanoparticles via carboxyl moieties, whereas the thiol group was further nitrosated to provide NO-releasing molecules. The developed magnetic nanocarrier exhibited good aqueous colloidal stability, protein resistance behavior, and high encapsulation efficacy for NO (65.5%) and DOX (85%), as well as sustained release characteristics. Moreover, they showed superior cytotoxicity toward cancer (A549 and MCF-7) cells via apoptosis induction over normal (WI26VA4) cells. Specifically, we have developed magnetic nanocarriers having the capability of dual delivery of NO and DOX, which holds great potential for combinatorial cancer treatment.
Collapse
Affiliation(s)
- Bijaideep Dutta
- Chemistry
Division, Bhabha Atomic Research Centre,
Trombay, Mumbai 400085, India
- Homi
Bhabha National Institute, Anushaktinagar, Mumbai 400094, India
| | - Sandeep B. Shelar
- Chemistry
Division, Bhabha Atomic Research Centre,
Trombay, Mumbai 400085, India
| | - Ananya Nirmalraj
- Chemistry
Division, Bhabha Atomic Research Centre,
Trombay, Mumbai 400085, India
- Department
of Chemistry, Sunandan Divatia School of Science, SVKM’s NMIMS (Deemed-to-be University), Vile Parle (W), Mumbai 400056, India
| | - Sonali Gupta
- Chemistry
Division, Bhabha Atomic Research Centre,
Trombay, Mumbai 400085, India
- Homi
Bhabha National Institute, Anushaktinagar, Mumbai 400094, India
| | - Kanhu C. Barick
- Chemistry
Division, Bhabha Atomic Research Centre,
Trombay, Mumbai 400085, India
- Homi
Bhabha National Institute, Anushaktinagar, Mumbai 400094, India
| | - Jagriti Gupta
- Chemistry
Division, Bhabha Atomic Research Centre,
Trombay, Mumbai 400085, India
| | - Puthusserickal A. Hassan
- Chemistry
Division, Bhabha Atomic Research Centre,
Trombay, Mumbai 400085, India
- Homi
Bhabha National Institute, Anushaktinagar, Mumbai 400094, India
| |
Collapse
|
12
|
Carretero VJ, Ramos E, Segura-Chama P, Hernández A, Baraibar AM, Álvarez-Merz I, Muñoz FL, Egea J, Solís JM, Romero A, Hernández-Guijo JM. Non-Excitatory Amino Acids, Melatonin, and Free Radicals: Examining the Role in Stroke and Aging. Antioxidants (Basel) 2023; 12:1844. [PMID: 37891922 PMCID: PMC10603966 DOI: 10.3390/antiox12101844] [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: 09/05/2023] [Revised: 09/23/2023] [Accepted: 09/26/2023] [Indexed: 10/29/2023] Open
Abstract
The aim of this review is to explore the relationship between melatonin, free radicals, and non-excitatory amino acids, and their role in stroke and aging. Melatonin has garnered significant attention in recent years due to its diverse physiological functions and potential therapeutic benefits by reducing oxidative stress, inflammation, and apoptosis. Melatonin has been found to mitigate ischemic brain damage caused by stroke. By scavenging free radicals and reducing oxidative damage, melatonin may help slow down the aging process and protect against age-related cognitive decline. Additionally, non-excitatory amino acids have been shown to possess neuroprotective properties, including antioxidant and anti-inflammatory in stroke and aging-related conditions. They can attenuate oxidative stress, modulate calcium homeostasis, and inhibit apoptosis, thereby safeguarding neurons against damage induced by stroke and aging processes. The intracellular accumulation of certain non-excitatory amino acids could promote harmful effects during hypoxia-ischemia episodes and thus, the blockade of the amino acid transporters involved in the process could be an alternative therapeutic strategy to reduce ischemic damage. On the other hand, the accumulation of free radicals, specifically mitochondrial reactive oxygen and nitrogen species, accelerates cellular senescence and contributes to age-related decline. Recent research suggests a complex interplay between melatonin, free radicals, and non-excitatory amino acids in stroke and aging. The neuroprotective actions of melatonin and non-excitatory amino acids converge on multiple pathways, including the regulation of calcium homeostasis, modulation of apoptosis, and reduction of inflammation. These mechanisms collectively contribute to the preservation of neuronal integrity and functions, making them promising targets for therapeutic interventions in stroke and age-related disorders.
Collapse
Affiliation(s)
- Victoria Jiménez Carretero
- Department of Pharmacology and Therapeutic, Teófilo Hernando Institute, Faculty of Medicine, Universidad Autónoma de Madrid, Av. Arzobispo Morcillo 4, 28029 Madrid, Spain
| | - Eva Ramos
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Complutense University of Madrid, 28040 Madrid, Spain
| | - Pedro Segura-Chama
- Investigador por México-CONAHCYT, Instituto Nacional de Psiquiatría "Ramón de la Fuente Muñiz", Calzada México-Xochimilco 101, Huipulco, Tlalpan, Mexico City 14370, Mexico
| | - Adan Hernández
- Institute of Neurobiology, Universidad Nacional Autónoma of México, Juriquilla, Santiago de Querétaro 76230, Querétaro, Mexico
| | - Andrés M Baraibar
- Department of Neurosciences, Universidad del País Vasco UPV/EHU, Achucarro Basque Center for Neuroscience, Barrio Sarriena, s/n, 48940 Leioa, Spain
| | - Iris Álvarez-Merz
- Department of Pharmacology and Therapeutic, Teófilo Hernando Institute, Faculty of Medicine, Universidad Autónoma de Madrid, Av. Arzobispo Morcillo 4, 28029 Madrid, Spain
| | - Francisco López Muñoz
- Faculty of Health Sciences, University Camilo José Cela, C/Castillo de Alarcón 49, Villanueva de la Cañada, 28692 Madrid, Spain
- Neuropsychopharmacology Unit, Hospital 12 de Octubre Research Institute (i + 12), Avda. Córdoba, s/n, 28041 Madrid, Spain
| | - Javier Egea
- Molecular Neuroinflammation and Neuronal Plasticity Research Laboratory, Hospital Universitario Santa Cristina, Health Research Institute, Hospital Universitario de la Princesa, 28006 Madrid, Spain
| | - José M Solís
- Neurobiology-Research Service, Hospital Ramón y Cajal, Carretera de Colmenar Viejo, Km. 9, 28029 Madrid, Spain
| | - Alejandro Romero
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Complutense University of Madrid, 28040 Madrid, Spain
| | - Jesús M Hernández-Guijo
- Department of Pharmacology and Therapeutic, Teófilo Hernando Institute, Faculty of Medicine, Universidad Autónoma de Madrid, Av. Arzobispo Morcillo 4, 28029 Madrid, Spain
- Ramón y Cajal Institute for Health Research (IRYCIS), Hospital Ramón y Cajal, Carretera de Colmenar Viejo, Km. 9, 28029 Madrid, Spain
| |
Collapse
|
13
|
Eskandari S, Rezayof A, Asghari SM, Hashemizadeh S. Neurobiochemical characteristics of arginine-rich peptides explain their potential therapeutic efficacy in neurodegenerative diseases. Neuropeptides 2023; 101:102356. [PMID: 37390744 DOI: 10.1016/j.npep.2023.102356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 06/19/2023] [Accepted: 06/21/2023] [Indexed: 07/02/2023]
Abstract
Neurodegenerative diseases, including Alzheimer̕ s disease (AD), Parkinson̕ s disease (PD), Huntington̕ s disease (HD), and Amyotrophic Lateral Sclerosis (ALS) require special attention to find new potential treatment methods. This review aims to summarize the current knowledge of the relationship between the biochemical properties of arginine-rich peptides (ARPs) and their neuroprotective effects to deal with the harmful effects of risk factors. It seems that ARPs have portrayed a promising and fantastic landscape for treating neurodegeneration-associated disorders. With multimodal mechanisms of action, ARPs play various unprecedented roles, including as the novel delivery platforms for entering the central nervous system (CNS), the potent antagonists for calcium influx, the invader molecules for targeting mitochondria, and the protein stabilizers. Interestingly, these peptides inhibit the proteolytic enzymes and block protein aggregation to induce pro-survival signaling pathways. ARPs also serve as the scavengers of toxic molecules and the reducers of oxidative stress agents. They also have anti-inflammatory, antimicrobial, and anti-cancer properties. Moreover, by providing an efficient nucleic acid delivery system, ARPs can play an essential role in developing various fields, including gene vaccines, gene therapy, gene editing, and imaging. ARP agents and ARP/cargo therapeutics can be raised as an emergent class of neurotherapeutics for neurodegeneration. Part of the aim of this review is to present recent advances in treating neurodegenerative diseases using ARPs as an emerging and powerful therapeutic tool. The applications and progress of ARPs-based nucleic acid delivery systems have also been discussed to highlight their usefulness as a broad-acting class of drugs.
Collapse
Affiliation(s)
- Sedigheh Eskandari
- Department of Animal Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran; Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran
| | - Ameneh Rezayof
- Department of Animal Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran.
| | - S Mohsen Asghari
- Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran.
| | - Shiva Hashemizadeh
- School of Cognitive Sciences, Institute for Research in Fundamental Sciences, IPM, Tehran, Iran
| |
Collapse
|
14
|
Friebe A, Kraehling JR, Russwurm M, Sandner P, Schmidtko A. The 10th International Conference on cGMP 2022: recent trends in cGMP research and development-meeting report. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023; 396:1669-1686. [PMID: 37079081 PMCID: PMC10338386 DOI: 10.1007/s00210-023-02484-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 03/31/2023] [Indexed: 04/21/2023]
Abstract
Increasing cGMP is a unique therapeutic principle, and drugs inhibiting cGMP-degrading enzymes or stimulating cGMP production are approved for the treatment of various diseases such as erectile dysfunction, coronary artery disease, pulmonary hypertension, chronic heart failure, irritable bowel syndrome, or achondroplasia. In addition, cGMP-increasing therapies are preclinically profiled or in clinical development for quite a broad set of additional indications, e.g., neurodegenerative diseases or different forms of dementias, bone formation disorders, underlining the pivotal role of cGMP signaling pathways. The fundamental understanding of the signaling mediated by nitric oxide-sensitive (soluble) guanylyl cyclase and membrane-associated receptor (particulate) guanylyl cyclase at the molecular and cellular levels, as well as in vivo, especially in disease models, is a key prerequisite to fully exploit treatment opportunities and potential risks that could be associated with an excessive increase in cGMP. Furthermore, human genetic data and the clinical effects of cGMP-increasing drugs allow back-translation into basic research to further learn about signaling and treatment opportunities. The biannual international cGMP conference, launched nearly 20 years ago, brings all these aspects together as an established and important forum for all topics from basic science to clinical research and pivotal clinical trials. This review summarizes the contributions to the "10th cGMP Conference on cGMP Generators, Effectors and Therapeutic Implications," which was held in Augsburg in 2022 but will also provide an overview of recent key achievements and activities in the field of cGMP research.
Collapse
Affiliation(s)
- Andreas Friebe
- Institute of Physiology, University of Würzburg, Röntgenring 9, D-97070 Würzburg, Germany
| | - Jan R. Kraehling
- Pharmaceuticals, Research and Early Development, Pharma Research Center, Bayer AG, Aprather Weg 18a, D-42096 Wuppertal, Germany
| | - Michael Russwurm
- Institute of Pharmacology, Ruhr-University Bochum, Universitätsstr. 150, D-44801 Bochum, Germany
| | - Peter Sandner
- Pharmaceuticals, Research and Early Development, Pharma Research Center, Bayer AG, Aprather Weg 18a, D-42096 Wuppertal, Germany
- Institute of Pharmacology, Hannover Medical School, Carl-Neuberg-Str. 1, D-30625 Hannover, Germany
| | - Achim Schmidtko
- Institute of Pharmacology and Clinical Pharmacy, Goethe University, Max-Von-Laue-Str. 9, D-60438 Frankfurt Am Main, Germany
| |
Collapse
|
15
|
Miyazawa D, Suzuki K, Sato H, Katsurayama N, Tahira T, Mizutani H, Ohara N. Docosahexaenoic acid contributes to increased CaMKII protein expression and a tendency to increase nNOS protein expression in differentiated NG108-15 cells. Drug Discov Ther 2023:2023.01003. [PMID: 37245984 DOI: 10.5582/ddt.2023.01003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
Docosahexaenoic acid (DHA; 22:6n-3), an n-3 polyunsaturated fatty acid, has various important roles in brain functions. Nitric oxide (NO) produced by neuronal NO synthase (nNOS) and Ca2+/calmodulindependent protein kinase II (CaMKII) is also involved in brain functions. We investigated the influence of DHA on nNOS and CaMKII protein expression in differentiated NG108-15 cells. NG108-15 cells were seeded in 12-well plates, and after 24 h, the medium was replaced with Dulbecco's modified Eagle's medium containing 1% fetal bovine serum, 0.2 mM dibutyryl cyclic adenosine monophosphate and 100 nM dexamethasone as differentiation-inducing medium. When cells were cultured in differentiation-inducing medium, neurite-like outgrowths were observed on days 5 and 6. However, no significant difference in morphology was observed in cells with or without DHA treatment. With or without DHA addition, nNOS protein expression was increased on days 5 and 6 compared with day 0. This increase tended to be enhanced by DHA. CaMKII protein expression did not change after differentiation without DHA, but was significantly increased on day 6 compared with day 0 with DHA addition. These data indicate that DHA is involved in brain functions by regulating CaMKII and nNOS protein expression.
Collapse
Affiliation(s)
| | - Kinari Suzuki
- College of Pharmacy, Kinjo Gakuin University, Nagoya, Japan
| | - Hikari Sato
- College of Pharmacy, Kinjo Gakuin University, Nagoya, Japan
| | | | - Tomoko Tahira
- College of Pharmacy, Kinjo Gakuin University, Nagoya, Japan
| | | | - Naoki Ohara
- College of Pharmacy, Kinjo Gakuin University, Nagoya, Japan
| |
Collapse
|
16
|
Abstract
eNOS (endothelial nitric oxide synthase) is critically important enzyme responsible for regulation of cardiovascular homeostasis. Under physiological conditions, constitutive eNOS activity and production of endothelial nitric oxide (NO) exert essential neurovascular protective functions. In this review, we first discuss the roles of endothelial NO in prevention of neuronal amyloid accumulation and formation of neurofibrillary tangles, hallmarks of Alzheimer disease pathology. Next, we review existing evidence suggesting that NO released from endothelium prevents activation of microglia, stimulates glycolysis in astrocytes, and increases biogenesis of mitochondria. We also address major risk factors for cognitive impairment including aging and ApoE4 (apolipoprotein 4) genotype with focus on their detrimental effects on eNOS/NO signaling. Relevant to this review, recent studies suggested that aged eNOS heterozygous mice are unique model of spontaneous cerebral small vessel disease. In this regard, we review contribution of dysfunctional eNOS to deposition of Aβ (amyloid-β) into blood vessel wall leading to development of cerebral amyloid angiopathy. We conclude that endothelial dysfunction manifested by the loss of neurovascular protective functions of NO may significantly contribute to development of cognitive impairment.
Collapse
Affiliation(s)
- Zvonimir S. Katusic
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota 55902, USA
- Department of Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota 55902, USA
| | - Livius V. d’Uscio
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota 55902, USA
- Department of Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota 55902, USA
| | - Tongrong He
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota 55902, USA
- Department of Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota 55902, USA
| |
Collapse
|
17
|
Kim YL, Plank JT, Li B, Lippert AR. Kinetics-Based Quantification of Peroxynitrite Using the Oxidative Decarbonylation of Isatin. Anal Chem 2022; 94:17803-17809. [PMID: 36520991 DOI: 10.1021/acs.analchem.2c03474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Peroxynitrite and its radical decomposition products are highly reactive nitrogen and oxygen species that can influence the balance between health and disease in multiple organ systems. Despite vigorous research activity, real-time quantitative monitoring of peroxynitrite generated by donor compounds remains challenging. Here, we report a kinetics-based fluorescence method for quantitative tracking of peroxynitrite generation using the oxidative decarbonylation of isatin to form anthranilic acid as a fluorescent probe. This method relies on knowledge of the rate of the reaction of peroxynitrite with the probe, which we measure using stopped-flow fluorescence techniques. To the best of our knowledge, this is the first optical method capable of providing real-time quantitative measures of peroxynitrite concentrations generated from donor compounds, as demonstrated herein for SIN-1 and Angeli's salt.
Collapse
Affiliation(s)
- Yujin L Kim
- Department of Chemistry, Southern Methodist University, Dallas, Texas75275-0314, United States
| | - Joshua T Plank
- Department of Chemistry, Southern Methodist University, Dallas, Texas75275-0314, United States
| | - Bo Li
- Department of Chemistry, Southern Methodist University, Dallas, Texas75275-0314, United States
| | - Alexander R Lippert
- Department of Chemistry, Southern Methodist University, Dallas, Texas75275-0314, United States
| |
Collapse
|
18
|
Barandov A, Ghosh S, Jasanoff A. Probing nitric oxide signaling using molecular MRI. Free Radic Biol Med 2022; 191:241-248. [PMID: 36084790 PMCID: PMC10204116 DOI: 10.1016/j.freeradbiomed.2022.08.042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 08/27/2022] [Accepted: 08/31/2022] [Indexed: 11/18/2022]
Abstract
Wide field measurements of nitric oxide (NO) signaling could help understand and diagnose the many physiological processes in which NO plays a key role. Magnetic resonance imaging (MRI) can support particularly powerful approaches for this purpose if equipped with molecular probes sensitized to NO and NO-associated targets. In this review, we discuss the development of MRI-detectable probes that could enable studies of nitrergic signaling in animals and potentially human subjects. Major families of probes include contrast agents designed to capture and report integrated NO levels directly, as well as molecules that respond to or emulate the activity of nitric oxide synthase enzymes. For each group, we outline the relevant molecular mechanisms and discuss results that have been obtained in vitro and in animals. The most promising in vivo data described to date have been acquired using NO capture-based relaxation agents and using engineered nitric oxide synthases that provide hemodynamic readouts of NO signaling pathway activation. These advances establish a beachhead for ongoing efforts to improve the sensitivity, specificity, and clinical applicability of NO-related molecular MRI technology.
Collapse
Affiliation(s)
- Ali Barandov
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, MA, 02139, USA
| | - Souparno Ghosh
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, MA, 02139, USA
| | - Alan Jasanoff
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, MA, 02139, USA; Department of Brain & Cognitive Sciences, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, MA, 02139, USA; Department of Nuclear Science & Engineering, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, MA, 02139, USA.
| |
Collapse
|
19
|
Clinical Trials of New Drugs for Vascular Cognitive Impairment and Vascular Dementia. Int J Mol Sci 2022; 23:ijms231911067. [PMID: 36232368 PMCID: PMC9569827 DOI: 10.3390/ijms231911067] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 09/09/2022] [Accepted: 09/14/2022] [Indexed: 12/03/2022] Open
Abstract
Population aging has challenged the treatment of cognitive impairment or dementia. Vascular dementia is the second leading cause of dementia after Alzheimer’s disease. Cognitive consequences after ischemic brain injury have been recognized as a preferred target for therapeutic strategies, prompting the search for potential agents. The keyword “vascular dementia” was used to search ClinicalTrials.gov to determine agents represented in phases I, II, III, and IV. The agents were classified on the basis of their mechanisms. Of the 17 randomized controlled trials meeting our inclusion criteria, 9 were completed in the past 10 years, and 8 are ongoing or in the planning stages. We also identified one trial in phase I, nine in phase II, six in phase III, and one in phase IV. Fewer trials of new drugs for improving cognition or ameliorating the behavioral and psychological symptoms of dementia target vascular dementia than Alzheimer’s dementia. Drug trials on vascular dementia overlap with drug trials targeting functional outcomes in cerebrovascular disease. International pharmaceutical companies’ investment in new drugs targeting VCI and vascular dementia remains insufficient.
Collapse
|
20
|
Lundberg JO, Weitzberg E. Nitric oxide signaling in health and disease. Cell 2022; 185:2853-2878. [DOI: 10.1016/j.cell.2022.06.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 06/01/2022] [Accepted: 06/06/2022] [Indexed: 10/16/2022]
|
21
|
Rodkin S, Dzreyan V, Bibov M, Ermakov A, Derezina T, Kirichenko E. NO-Dependent Mechanisms of p53 Expression and Cell Death in Rat’s Dorsal Root Ganglia after Sciatic-Nerve Transection. Biomedicines 2022; 10:biomedicines10071664. [PMID: 35884967 PMCID: PMC9313305 DOI: 10.3390/biomedicines10071664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/03/2022] [Accepted: 07/09/2022] [Indexed: 11/16/2022] Open
Abstract
Peripheral-nerve injury is a frequent cause of disability. Presently, no clinically effective neuroprotectors have been found. We have studied the NO-dependent expression of p53 in the neurons and glial cells of the dorsal root ganglia (DRG) of a rat’s spinal cord, as well as the role of NO in the death of these cells under the conditions of axonal stress, using sciatic-nerve axotomy as a model. It was found out that axotomy led to the nuclear–cytoplasmic redistribution of p53 in neurons, 24 h after trauma. The NO donor led to a considerable increase in the level of p53 in nuclei and, to a smaller degree, in the cytoplasm of neurons and karyoplasm of glial cells 4 and 24 h after axotomy. Application of a selective inhibitor of inducible NO-synthase (iNOS) provided the opposite effect. Introduction of the NO donor resulted in a significant increase in cell death in the injured ipsilateral DRG, 24 h and 7 days after trauma. The selective inhibitor of iNOS demonstrated a neuroprotective effect. Axotomy was shown to upregulate the iNOS in nuclei and cytoplasm of DRG cells. The NO-dependent expression of p53, which is particularly achieved through iNOS activation, is believed to be a putative signaling mechanism of neural and glial-cell death after axotomy.
Collapse
Affiliation(s)
- Stanislav Rodkin
- Faculty of Bioengineering and Veterinary Medicine, Don State Technical University, Gagarin Square 1, 344000 Rostov-on-Don, Russia; (A.E.); (T.D.); (E.K.)
- Correspondence: ; Tel.: +7-(918)-576-2390
| | - Valentina Dzreyan
- Laboratory of Molecular Neurobiology, Academy of Biology and Biotechnology, Southern Federal University, Stachki Ave., 194/1, 344090 Rostov-on-Don, Russia;
| | - Mikhail Bibov
- Department of General and Clinical Biochemistry No. 2, Rostov State Medical University, Nakhichevansky, 29, 344022 Rostov-on-Don, Russia;
| | - Alexey Ermakov
- Faculty of Bioengineering and Veterinary Medicine, Don State Technical University, Gagarin Square 1, 344000 Rostov-on-Don, Russia; (A.E.); (T.D.); (E.K.)
| | - Tatyana Derezina
- Faculty of Bioengineering and Veterinary Medicine, Don State Technical University, Gagarin Square 1, 344000 Rostov-on-Don, Russia; (A.E.); (T.D.); (E.K.)
| | - Evgeniya Kirichenko
- Faculty of Bioengineering and Veterinary Medicine, Don State Technical University, Gagarin Square 1, 344000 Rostov-on-Don, Russia; (A.E.); (T.D.); (E.K.)
| |
Collapse
|
22
|
Cao LX, Bing YH, Xu YH, Zhang GJ, Chu CP, Hong L, Qiu DL. Nicotine Facilitates Facial Stimulation-Evoked Mossy Fiber-Granule Cell Long-Term Potentiation in vivo in Mice. Front Cell Neurosci 2022; 16:905724. [PMID: 35860314 PMCID: PMC9289189 DOI: 10.3389/fncel.2022.905724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Accepted: 06/14/2022] [Indexed: 11/13/2022] Open
Abstract
Nicotine is a psychoactive component of tobacco that plays critical roles in the regulation of neuronal circuit function and neuroplasticity and contributes to the improvement of working memory performance and motor learning function via nicotinic acetylcholine receptors (nAChRs). Under in vivo conditions, nicotine enhances facial stimulation-evoked mossy fiber-granule cell (MF-GrC) synaptic transmission, which suggests that nicotine regulates MF-GrC synaptic plasticity in the mouse cerebellar cortex. In this study, we investigated the effects of nicotine on facial stimulation-induced long-term potentiation (LTP) of MF-GrC synaptic transmission in urethane-anesthetized mice. Our results showed that facial stimulation at 20 Hz induced an MF-GrC LTP in the mouse cerebellar granular layer that was significantly enhanced by the application of nicotine (1 μM). Blockade of α4β2 nAChRs, but not α7 nAChRs, during delivery of 20 Hz facial stimulation prevented the nicotine-induced facilitation of MF-GrC LTP. Notably, the facial stimulation-induced MF-GrC LTP was abolished by an N-methyl-D-aspartate (NMDA) receptor antagonist, but it was restored by additional application of nicotine during delivery of 20 Hz facial stimulation. Furthermore, antagonism of α4β2 nAChRs, but not α7 nAChRs, during delivery of 20 Hz facial stimulation prevented nicotine-induced MF-GrC LTP. Moreover, inhibition of nitric oxide synthase (NOS) abolished the facial stimulation-induced MF-GrC LTP, as well as the effect of nicotine on it. Our results indicated that 20 Hz facial stimulation induced MF-GrC LTP via an NMDA receptor/nitric oxide (NO) cascade, but MF-GrC LTP was enhanced by nicotine through the α4β2 AChR/NO signaling pathway. These results suggest that nicotine-induced facilitation of MF-GrC LTP may play a critical role in the improvement of working memory performance and motor learning function.
Collapse
Affiliation(s)
- Li-Xin Cao
- Department of Physiology and Pathophysiology, College of Medicine, Yanbian University, Yanji, China
| | - Yan-Hua Bing
- Department of Physiology and Pathophysiology, College of Medicine, Yanbian University, Yanji, China
| | - Yin-Hua Xu
- Department of Neurology, Affiliated Hospital of Yanbian University, Yanji, China
| | - Guang-Jian Zhang
- Department of Pain, Affiliated Hospital of Yanbian University, Yanji, China
| | - Chun-Ping Chu
- Department of Physiology, College of Basic Medicine, Jilin Medical University, Jilin City, China
| | - Lan Hong
- Department of Physiology and Pathophysiology, College of Medicine, Yanbian University, Yanji, China
- *Correspondence: Lan Hong,
| | - De-Lai Qiu
- Department of Physiology, College of Basic Medicine, Jilin Medical University, Jilin City, China
- De-Lai Qiu, ,
| |
Collapse
|
23
|
Saiyasit N, Butlig EAR, Chaney SD, Traylor MK, Hawley NA, Randall RB, Bobinger HV, Frizell CA, Trimm F, Crook ED, Lin M, Hill BD, Keller JL, Nelson AR. Neurovascular Dysfunction in Diverse Communities With Health Disparities-Contributions to Dementia and Alzheimer's Disease. Front Neurosci 2022; 16:915405. [PMID: 35844216 PMCID: PMC9279126 DOI: 10.3389/fnins.2022.915405] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 05/31/2022] [Indexed: 12/13/2022] Open
Abstract
Alzheimer's disease and related dementias (ADRD) are an expanding worldwide crisis. In the absence of scientific breakthroughs, the global prevalence of ADRD will continue to increase as more people are living longer. Racial or ethnic minority groups have an increased risk and incidence of ADRD and have often been neglected by the scientific research community. There is mounting evidence that vascular insults in the brain can initiate a series of biological events leading to neurodegeneration, cognitive impairment, and ADRD. We are a group of researchers interested in developing and expanding ADRD research, with an emphasis on vascular contributions to dementia, to serve our local diverse community. Toward this goal, the primary objective of this review was to investigate and better understand health disparities in Alabama and the contributions of the social determinants of health to those disparities, particularly in the context of vascular dysfunction in ADRD. Here, we explain the neurovascular dysfunction associated with Alzheimer's disease (AD) as well as the intrinsic and extrinsic risk factors contributing to dysfunction of the neurovascular unit (NVU). Next, we ascertain ethnoregional health disparities of individuals living in Alabama, as well as relevant vascular risk factors linked to AD. We also discuss current pharmaceutical and non-pharmaceutical treatment options for neurovascular dysfunction, mild cognitive impairment (MCI) and AD, including relevant studies and ongoing clinical trials. Overall, individuals in Alabama are adversely affected by social and structural determinants of health leading to health disparities, driven by rurality, ethnic minority status, and lower socioeconomic status (SES). In general, these communities have limited access to healthcare and healthy food and other amenities resulting in decreased opportunities for early diagnosis of and pharmaceutical treatments for ADRD. Although this review is focused on the current state of health disparities of ADRD patients in Alabama, future studies must include diversity of race, ethnicity, and region to best be able to treat all individuals affected by ADRD.
Collapse
Affiliation(s)
- Napatsorn Saiyasit
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL, United States
| | - Evan-Angelo R. Butlig
- Department of Neurology, Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, Intellectual and Developmental Disabilities Research Center, University of California, Los Angeles, Los Angeles, CA, United States
| | - Samantha D. Chaney
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL, United States
| | - Miranda K. Traylor
- Department of Health, Kinesiology, and Sport, University of South Alabama, Mobile, AL, United States
| | - Nanako A. Hawley
- Department of Psychology, University of South Alabama, Mobile, AL, United States
| | - Ryleigh B. Randall
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL, United States
| | - Hanna V. Bobinger
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL, United States
| | - Carl A. Frizell
- Department of Physician Assistant Studies, University of South Alabama, Mobile, AL, United States
| | - Franklin Trimm
- College of Medicine, University of South Alabama, Mobile, AL, United States
| | - Errol D. Crook
- Department of Internal Medicine, College of Medicine, University of South Alabama, Mobile, AL, United States
| | - Mike Lin
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL, United States
| | - Benjamin D. Hill
- Department of Psychology, University of South Alabama, Mobile, AL, United States
| | - Joshua L. Keller
- Department of Health, Kinesiology, and Sport, University of South Alabama, Mobile, AL, United States
| | - Amy R. Nelson
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL, United States
| |
Collapse
|
24
|
Saulskaya NB, Burmakina MA, Trofimova NA. Effect of Activation and Blockade of Nitrergic Neurotransmission on Serotonin System Activity of the Rat Medial Prefrontal Cortex. J EVOL BIOCHEM PHYS+ 2022. [DOI: 10.1134/s0022093022020181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
25
|
Lu D, Wan P, Liu Y, Jin XH, Chu CP, Bing YH, Qiu DL. Facial Stimulation Induces Long-Term Potentiation of Mossy Fiber-Granule Cell Synaptic Transmission via GluN2A-Containing N-Methyl-D-Aspartate Receptor/Nitric Oxide Cascade in the Mouse Cerebellum. Front Cell Neurosci 2022; 16:863342. [PMID: 35431815 PMCID: PMC9005984 DOI: 10.3389/fncel.2022.863342] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 03/01/2022] [Indexed: 12/21/2022] Open
Abstract
Long-term synaptic plasticity in the cerebellar cortex is a possible mechanism for motor learning. Previous studies have demonstrated the induction of mossy fiber-granule cell (MF-GrC) synaptic plasticity under in vitro and in vivo conditions, but the mechanisms underlying sensory stimulation-evoked long-term synaptic plasticity of MF-GrC in living animals are unclear. In this study, we investigated the mechanism of long-term potentiation (LTP) of MF-GrC synaptic transmission in the cerebellum induced by train of facial stimulation at 20 Hz in urethane-anesthetized mice using electrophysiological recording, immunohistochemistry techniques, and pharmacological methods. Blockade of GABAA receptor activity and repetitive facial stimulation at 20 Hz (240 pulses) induced an LTP of MF-GrC synapses in the mouse cerebellar cortical folium Crus II, accompanied with a decrease in paired-pulse ratio (N2/N1). The facial stimulation-induced MF-GrC LTP was abolished by either an N-methyl-D-aspartate (NMDA) receptor blocker, i.e., D-APV, or a specific GluNR2A subunit-containing NMDA receptor antagonist, PEAQX, but was not prevented by selective GluNR2B or GluNR2C/D subunit-containing NMDA receptor blockers. Application of GNE-0723, a selective and brain-penetrant-positive allosteric modulator of GluN2A subunit-containing NMDA receptors, produced an LTP of N1, accompanied with a decrease in N2/N1 ratio, and occluded the 20-Hz facial stimulation-induced MF-GrC LTP. Inhibition of nitric oxide synthesis (NOS) prevented the facial stimulation-induced MF-GrC LTP, while activation of NOS produced an LTP of N1, with a decrease in N2/N1 ratio, and occluded the 20-Hz facial stimulation-induced MF-GrC LTP. In addition, GluN2A-containing NMDA receptor immunoreactivity was observed in the mouse cerebellar granular layer. These results indicate that facial stimulation at 20 Hz induced LTP of MF-GrC synaptic transmission via the GluN2A-containing NMDA receptor/nitric oxide cascade in mice. The results suggest that the sensory stimulation-evoked LTP of MF-GrC synaptic transmission in the granular layer may play a critical role in cerebellar adaptation to native mossy fiber excitatory inputs and motor learning behavior in living animals.
Collapse
Affiliation(s)
- Di Lu
- Department of Physiology and Pathophysiology, College of Medicine, Yanbian University, Yanji, China
- Department of Ophthalmology, Affiliated Hospital of Yanbian University, Yanji, China
| | - Peng Wan
- Department of Neurology, Affiliated Hospital of Yanbian University, Yanji, China
| | - Yang Liu
- Department of Physiology and Pathophysiology, College of Medicine, Yanbian University, Yanji, China
- Department of Ophthalmology, Affiliated Hospital of Yanbian University, Yanji, China
| | - Xian-Hua Jin
- Department of Neurology, Affiliated Hospital of Yanbian University, Yanji, China
| | - Chun-Ping Chu
- Department of Physiology, College of Basic Medicine, Jilin Medical University, Jilin, China
| | - Yan-Hua Bing
- Department of Physiology and Pathophysiology, College of Medicine, Yanbian University, Yanji, China
- *Correspondence: Yan-Hua Bing,
| | - De-Lai Qiu
- Department of Physiology, College of Basic Medicine, Jilin Medical University, Jilin, China
- *Correspondence: Yan-Hua Bing,
| |
Collapse
|
26
|
Abstract
Significance: Reactive sulfur and nitrogen species such as hydrogen sulfide (H2S) and nitric oxide (NO•) are ubiquitous cellular signaling molecules that play central roles in physiology and pathophysiology. A deeper understanding of these signaling pathways will offer new opportunities for therapeutic treatments and disease management. Recent Advances: Chemiluminescence methods have been fundamental in detecting and measuring biological reactive sulfur and nitrogen species, and new approaches are emerging for imaging these analytes in living intact specimens. Ozone-based and luminol-based chemiluminescence methods have been optimized for quantitative analysis of hydrogen sulfide and nitric oxide in biological samples and tissue homogenates, and caged luciferin and 1,2-dioxetanes are emerging as a versatile approach for monitoring and imaging reactive sulfur and nitrogen species in living cells and animal models. Critical Issues: This review article will cover the major chemiluminescence approaches for detecting, measuring, and imaging reactive sulfur and nitrogen species in biological systems, including a brief history of the development of the most established approaches and highlights of the opportunities provided by emerging approaches. Future Directions: Emerging chemiluminescence approaches offer new opportunities for monitoring and imaging reactive sulfur and nitrogen species in living cells, animals, and human clinical samples. Widespread adoption and translation of these approaches, however, requires an emphasis on rigorous quantitative methods, reproducibility, and effective technology transfer. Antioxid. Redox Signal. 36, 337-353.
Collapse
Affiliation(s)
- Bo Li
- Department of Chemistry, Southern Methodist University, Dallas, Texas USA
| | - Yujin Lisa Kim
- Department of Chemistry, Southern Methodist University, Dallas, Texas USA
| | - Alexander Ryan Lippert
- Department of Chemistry, Southern Methodist University, Dallas, Texas USA.,Center for Drug Discovery, Design, and Delivery (CD), Southern Methodist University, Dallas, Texas USA
| |
Collapse
|
27
|
Kasamatsu S, Tsutsuki H, Ida T, Sawa T, Watanabe Y, Akaike T, Ihara H. Regulation of nitric oxide/reactive oxygen species redox signaling by nNOS splicing variants. Nitric Oxide 2022; 120:44-52. [PMID: 35033681 DOI: 10.1016/j.niox.2022.01.004] [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: 11/22/2021] [Revised: 01/05/2022] [Accepted: 01/12/2022] [Indexed: 10/19/2022]
Abstract
We previously demonstrated different expression patterns of the neuronal nitric oxide synthase (nNOS) splicing variants, nNOS-μ and nNOS-α, in the rat brain; however, their exact functions have not been fully elucidated. In this study, we compared the enzymatic activities of nNOS-μ and nNOS-α and investigated intracellular redox signaling in nNOS-expressing PC12 cells, stimulated with a neurotoxicant, 1-methyl-4-phenylpyridinium ion (MPP+), to enhance the nNOS uncoupling reaction. Using in vitro studies, we show that nNOS-μ produced nitric oxide (NO), as did nNOS-α, in the presence of tetrahydrobiopterin (BH4), an important cofactor for the enzymatic activity. However, nNOS-μ generated more NO and less superoxide than nNOS-α in the absence of BH4. MPP + treatment induced more reactive oxygen species (ROS) production in nNOS-α-expressing PC12 cells than in those expressing nNOS-μ, which correlated with the intracellular production of 8-nitroguanosine 3',5'-cyclic monophosphate (8-nitro-cGMP), a downstream messenger of nNOS redox signaling, and apoptosis in these cells. Furthermore, post-treatment with 8-nitro-cGMP aggravated MPP+-induced cytotoxicity via activation of the H-Ras/extracellular signal-regulated kinase signaling pathway. In conclusion, our results provide strong evidence that nNOS-μ exhibits distinctive enzymatic properties of NO/ROS production, contributing to the regulation of intracellular redox signaling, including the downstream production of 8-nitro-cGMP.
Collapse
Affiliation(s)
- Shingo Kasamatsu
- Department of Biological Science, Graduate School of Science, Osaka Prefecture University, Osaka, 599-8531, Japan
| | - Hiroyasu Tsutsuki
- Department of Microbiology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, 860-8556, Japan
| | - Tomoaki Ida
- Department of Environmental Medicine and Molecular Toxicology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, 980-8575, Japan
| | - Tomohiro Sawa
- Department of Microbiology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, 860-8556, Japan
| | - Yasuo Watanabe
- Department of Pharmacology, Showa Pharmaceutical University, Tokyo, 194-8543, Japan
| | - Takaaki Akaike
- Department of Environmental Medicine and Molecular Toxicology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, 980-8575, Japan
| | - Hideshi Ihara
- Department of Biological Science, Graduate School of Science, Osaka Prefecture University, Osaka, 599-8531, Japan.
| |
Collapse
|
28
|
The effect of self-administered methamphetamine on GABAergic interneuron populations and functional connectivity of the nucleus accumbens and prefrontal cortex. Psychopharmacology (Berl) 2022; 239:2903-2919. [PMID: 35920922 PMCID: PMC9385811 DOI: 10.1007/s00213-022-06175-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 06/08/2022] [Indexed: 11/27/2022]
Abstract
INTRODUCTION Methamphetamine (METH, "ice") is a potent and addictive psychostimulant. Abuse of METH perturbs neurotransmitter systems and induces neurotoxicity; however, the neurobiological mechanisms which underlie addiction to METH are not fully understood, limiting the efficacy of available treatments. Here we investigate METH-induced changes to neuronal nitric oxide synthase (nNOS), parvalbumin and calretinin-expressing GABAergic interneuron populations within the nucleus accumbens (NAc), prefrontal cortex (PFC) and orbitofrontal cortex (OFC). We hypothesise that dysfunction or loss of these GABAergic interneuron populations may disrupt the excitatory/inhibitory balance within the brain. METHODS Male Long Evans rats (N = 32) were trained to lever press for intravenous METH or received yoked saline infusions. Following 14 days of behavioural extinction, animals were given a non-contingent injection of saline or METH (1 mg/kg, IP) to examine drug-primed reinstatement to METH-seeking behaviours. Ninety minutes post-IP injection, animals were culled and brain sections were analysed for Fos, nNOS, parvalbumin and calretinin immunoreactivity in eight distinct subregions of the NAc, PFC and OFC. RESULTS METH exposure differentially affected GABAergic populations, with METH self-administration increasing nNOS immunoreactivity at distinct locations in the prelimbic cortex and decreasing parvalbumin immunoreactivity in the NAc. METH self-administration triggered reduced calretinin immunoreactivity, whilst acute METH administration produced a significant increase in calretinin immunoreactivity. As expected, non-contingent METH-priming treatment increased Fos immunoreactivity in subregions of the NAc and PFC. CONCLUSION Here we report that METH exposure in this model may alter the function of GABAergic interneurons in more subtle ways, such as alterations in neuronal firing or synaptic connectivity.
Collapse
|
29
|
Coelho AA, Vila-Verde C, Sartim AG, Uliana DL, Braga LA, Guimarães FS, Lisboa SF. Inducible Nitric Oxide Synthase Inhibition in the Medial Prefrontal Cortex Attenuates the Anxiogenic-Like Effect of Acute Restraint Stress via CB 1 Receptors. Front Psychiatry 2022; 13:923177. [PMID: 35911236 PMCID: PMC9330908 DOI: 10.3389/fpsyt.2022.923177] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 06/06/2022] [Indexed: 11/13/2022] Open
Abstract
Stress exposure can result in several proinflammatory alterations in the brain, including overexpression of the inducible isoform of nitric oxide synthase (iNOS) in the medial prefrontal cortex (mPFC). These changes may be involved in the development of many psychiatric conditions. However, it is unknown if iNOS in mPFC plays a significant role in stress-induced behavioral changes. The endocannabinoid (ECB) system is also influenced by stress. Its activation seems to be a counter regulatory mechanism to prevent or decrease the stress-mediated neuroinflammatory consequences. However, it is unclear if the ECB system and iNOS interact to influence stress consequences. This study aimed to test the hypothesis that the anti-stress effect of iNOS inhibition in mPFC involves the local ECB system, particularly the CB1 cannabinoid receptors. Male Wistar rats with guide cannula aimed at the mPFC were submitted to acute restraint stress (RS) for 2 h. In the following morning, rats received bilateral microinjections of vehicle, AM251 (CB1 antagonist; 100 pmol), and/or 1400W (iNOS selective inhibitor; 10-4, 10-3, or 10-2 nmol) into the prelimbic area of mPFC (PL-mPFC) before being tested in the elevated plus-maze (EPM). iNOS inhibition by 1400W prevented the anxiogenic-like effect observed in animals submitted to RS. The drug did not promote behavior changes in naive animals, demonstrating a stress-dependent effect. The 1400W-anti-stress effect was prevented by local pretreatment with AM251. Our data suggest that iNOS inhibition may facilitate the local endocannabinoid signaling, attenuating stress effects.
Collapse
Affiliation(s)
- Arthur A Coelho
- Pharmacology Department, Ribeirão Preto Medical School-University of São Paulo, São Paulo, Brazil.,Biomolecular Sciences Department, School of Pharmaceutical Sciences of Ribeirão Preto-University of São Paulo, São Paulo, Brazil
| | - Carla Vila-Verde
- Pharmacology Department, Ribeirão Preto Medical School-University of São Paulo, São Paulo, Brazil
| | - Ariandra G Sartim
- Biomolecular Sciences Department, School of Pharmaceutical Sciences of Ribeirão Preto-University of São Paulo, São Paulo, Brazil
| | - Daniela L Uliana
- Pharmacology Department, Ribeirão Preto Medical School-University of São Paulo, São Paulo, Brazil.,Departments of Neuroscience, Psychiatry and Psychology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Laura A Braga
- Pharmacology Department, Ribeirão Preto Medical School-University of São Paulo, São Paulo, Brazil
| | - Francisco S Guimarães
- Pharmacology Department, Ribeirão Preto Medical School-University of São Paulo, São Paulo, Brazil
| | - Sabrina F Lisboa
- Pharmacology Department, Ribeirão Preto Medical School-University of São Paulo, São Paulo, Brazil.,Biomolecular Sciences Department, School of Pharmaceutical Sciences of Ribeirão Preto-University of São Paulo, São Paulo, Brazil
| |
Collapse
|
30
|
Melis MR, Argiolas A. Erectile Function and Sexual Behavior: A Review of the Role of Nitric Oxide in the Central Nervous System. Biomolecules 2021; 11:biom11121866. [PMID: 34944510 PMCID: PMC8699072 DOI: 10.3390/biom11121866] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 12/06/2021] [Accepted: 12/08/2021] [Indexed: 12/21/2022] Open
Abstract
Nitric oxide (NO), the neuromodulator/neurotransmitter formed from l-arginine by neuronal, endothelial and inducible NO synthases, is involved in numerous functions across the body, from the control of arterial blood pressure to penile erection, and at central level from energy homeostasis regulation to memory, learning and sexual behavior. The aim of this work is to review earlier studies showing that NO plays a role in erectile function and sexual behavior in the hypothalamus and its paraventricular nucleus and the medial preoptic area, and integrate these findings with those of recent studies on this matter. This revisitation shows that NO influences erectile function and sexual behavior in males and females by acting not only in the paraventricular nucleus and medial preoptic area but also in extrahypothalamic brain areas, often with different mechanisms. Most importantly, since these areas are strictly interconnected with the paraventricular nucleus and medial preoptic area, send to and receive neural projections from the spinal cord, in which sexual communication between brain and genital apparatus takes place, this review reveals that central NO participates in concert with neurotransmitters/neuropeptides to a neural circuit controlling both the consummatory (penile erection, copulation, lordosis) and appetitive components (sexual motivation, arousal, reward) of sexual behavior.
Collapse
|
31
|
Spiers JG, Steinert JR. Nitrergic modulation of ion channel function in regulating neuronal excitability. Channels (Austin) 2021; 15:666-679. [PMID: 34802368 PMCID: PMC8632290 DOI: 10.1080/19336950.2021.2002594] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Nitric oxide (NO) signaling in the brain provides a wide range of functional properties in response to neuronal activity. NO exerts its effects through different signaling pathways, namely, through the canonical soluble guanylyl cyclase-mediated cGMP production route and via post-translational protein modifications. The latter pathways comprise cysteine S-nitrosylation and 3-nitrotyrosination of distinct tyrosine residues. Many ion channels are targeted by one or more of these signaling routes, which leads to their functional regulation under physiological conditions or facilities their dysfunction leading to channelopathies in many pathologies. The resulting alterations in ion channel function changes neuronal excitability, synaptic transmission, and action potential propagation. Transient and activity-dependent NO production mediates reversible ion channel modifications via cGMP and S-nitrosylation signaling, whereas more pronounced and longer-term NO production during conditions of elevated oxidative stress leads to increasingly cumulative and irreversible protein 3-nitrotyrosination. The complexity of this regulation and vast variety of target ion channels and their associated functional alterations presents a challenging task in assessing and understanding the role of NO signaling in physiology and disease.
Collapse
Affiliation(s)
- Jereme G Spiers
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Australia
| | - Joern R Steinert
- School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham, UK
| |
Collapse
|
32
|
Bashkatova V. Metabotropic glutamate receptors and nitric oxide in dopaminergic neurotoxicity. World J Psychiatry 2021; 11:830-840. [PMID: 34733645 PMCID: PMC8546773 DOI: 10.5498/wjp.v11.i10.830] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 04/11/2021] [Accepted: 08/03/2021] [Indexed: 02/06/2023] Open
Abstract
Dopaminergic neurotoxicity is characterized by damage and death of dopaminergic neurons. Parkinson's disease (PD) is a neurodegenerative disorder that primarily involves the loss of dopaminergic neurons in the substantia nigra. Therefore, the study of the mechanisms, as well as the search for new targets for the prevention and treatment of neurodegenerative diseases, is an important focus of modern neuroscience. PD is primarily caused by dysfunction of dopaminergic neurons; however, other neurotransmitter systems are also involved. Research reports have indicated that the glutamatergic system is involved in different pathological conditions, including dopaminergic neurotoxicity. Over the last two decades, the important functional interplay between dopaminergic and glutamatergic systems has stimulated interest in the possible role of metabotropic glutamate receptors (mGluRs) in the development of extrapyramidal disorders. However, the specific mechanisms driving these processes are presently unclear. The participation of the universal neuronal messenger nitric oxide (NO) in the mechanisms of dopaminergic neurotoxicity has attracted increased attention. The current paper aims to review the involvement of mGluRs and the contribution of NO to dopaminergic neurotoxicity. More precisely, we focused on studies conducted on the rotenone-induced PD model. This review is also an outline of our own results obtained using the method of electron paramagnetic resonance, which allows quantitation of NO radicals in brain structures.
Collapse
Affiliation(s)
- Valentina Bashkatova
- Laboratory of Physiology Reinforcements, Anokhin Institute of Normal Physiology, Moscow 125315, Russia
| |
Collapse
|
33
|
da Silva GM, da Silva MC, Nascimento DVG, Lima Silva EM, Gouvêa FFF, de França Lopes LG, Araújo AV, Ferraz Pereira KN, de Queiroz TM. Nitric Oxide as a Central Molecule in Hypertension: Focus on the Vasorelaxant Activity of New Nitric Oxide Donors. BIOLOGY 2021; 10:1041. [PMID: 34681140 PMCID: PMC8533285 DOI: 10.3390/biology10101041] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 10/08/2021] [Accepted: 10/09/2021] [Indexed: 12/15/2022]
Abstract
Cardiovascular diseases include all types of disorders related to the heart or blood vessels. High blood pressure is an important risk factor for cardiac complications and pathological disorders. An increase in circulating angiotensin-II is a potent stimulus for the expression of reactive oxygen species and pro-inflammatory cytokines that activate oxidative stress, perpetuating a deleterious effect in hypertension. Studies demonstrate the capacity of NO to prevent platelet or leukocyte activation and adhesion and inhibition of proliferation, as well as to modulate inflammatory or anti-inflammatory reactions and migration of vascular smooth muscle cells. However, in conditions of low availability of NO, such as during hypertension, these processes are impaired. Currently, there is great interest in the development of compounds capable of releasing NO in a modulated and stable way. Accordingly, compounds containing metal ions coupled to NO are being investigated and are widely recognized as having great relevance in the treatment of different diseases. Therefore, the exogenous administration of NO is an attractive and pharmacological alternative in the study and treatment of hypertension. The present review summarizes the role of nitric oxide in hypertension, focusing on the role of new NO donors, particularly the metal-based drugs and their protagonist activity in vascular function.
Collapse
Affiliation(s)
- Gabriela Maria da Silva
- Laboratory of Nutrition, Physical Activity and Phenotypic Plasticity, Federal University of Pernambuco, Vitória de Santo Antão 55.608-680, PE, Brazil; (G.M.d.S.); (M.C.d.S.); (D.V.G.N.); (E.M.L.S.); (A.V.A.); (K.N.F.P.)
| | - Mirelly Cunha da Silva
- Laboratory of Nutrition, Physical Activity and Phenotypic Plasticity, Federal University of Pernambuco, Vitória de Santo Antão 55.608-680, PE, Brazil; (G.M.d.S.); (M.C.d.S.); (D.V.G.N.); (E.M.L.S.); (A.V.A.); (K.N.F.P.)
| | - Déborah Victória Gomes Nascimento
- Laboratory of Nutrition, Physical Activity and Phenotypic Plasticity, Federal University of Pernambuco, Vitória de Santo Antão 55.608-680, PE, Brazil; (G.M.d.S.); (M.C.d.S.); (D.V.G.N.); (E.M.L.S.); (A.V.A.); (K.N.F.P.)
| | - Ellen Mayara Lima Silva
- Laboratory of Nutrition, Physical Activity and Phenotypic Plasticity, Federal University of Pernambuco, Vitória de Santo Antão 55.608-680, PE, Brazil; (G.M.d.S.); (M.C.d.S.); (D.V.G.N.); (E.M.L.S.); (A.V.A.); (K.N.F.P.)
| | - Fabíola Furtado Fialho Gouvêa
- School of Technical Health, Health Sciences Center, Federal University of Paraíba, João Pessoa 58.051-900, PB, Brazil;
| | - Luiz Gonzaga de França Lopes
- Laboratory of Bioinorganic Chemistry, Department of Organic and Inorganic Chemistry, Federal University of Ceará, Fortaleza 60.020-181, CE, Brazil;
| | - Alice Valença Araújo
- Laboratory of Nutrition, Physical Activity and Phenotypic Plasticity, Federal University of Pernambuco, Vitória de Santo Antão 55.608-680, PE, Brazil; (G.M.d.S.); (M.C.d.S.); (D.V.G.N.); (E.M.L.S.); (A.V.A.); (K.N.F.P.)
| | - Kelli Nogueira Ferraz Pereira
- Laboratory of Nutrition, Physical Activity and Phenotypic Plasticity, Federal University of Pernambuco, Vitória de Santo Antão 55.608-680, PE, Brazil; (G.M.d.S.); (M.C.d.S.); (D.V.G.N.); (E.M.L.S.); (A.V.A.); (K.N.F.P.)
| | - Thyago Moreira de Queiroz
- Laboratory of Nutrition, Physical Activity and Phenotypic Plasticity, Federal University of Pernambuco, Vitória de Santo Antão 55.608-680, PE, Brazil; (G.M.d.S.); (M.C.d.S.); (D.V.G.N.); (E.M.L.S.); (A.V.A.); (K.N.F.P.)
| |
Collapse
|
34
|
N-Methyl-D-aspartate Glutamate Receptor Modulates Cardiovascular and Neuroendocrine Responses Evoked by Hemorrhagic Shock in Rats. BIOMED RESEARCH INTERNATIONAL 2021; 2021:1156031. [PMID: 34423030 PMCID: PMC8378978 DOI: 10.1155/2021/1156031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 07/12/2021] [Accepted: 08/04/2021] [Indexed: 12/12/2022]
Abstract
Here, we report the participation of N-methyl-D-aspartate (NMDA) glutamate receptor in the mediation of cardiovascular and circulating vasopressin responses evoked by a hemorrhagic stimulus. In addition, once NMDA receptor activation is a prominent mechanism involved in nitric oxide (NO) synthesis in the brain, we investigated whether control of hemorrhagic shock by NMDA glutamate receptor was followed by changes in NO synthesis in brain supramedullary structures involved in cardiovascular and neuroendocrine control. Thus, we observed that intraperitoneal administration of the selective NMDA glutamate receptor antagonist dizocilpine maleate (MK801, 0.3 mg/kg) delayed and reduced the magnitude of hemorrhage-induced hypotension. Besides, hemorrhage induced a tachycardia response in the posthemorrhage period (i.e., recovery period) in control animals, and systemic treatment with MK801 caused a bradycardia response during hemorrhagic shock. Hemorrhagic stimulus increased plasma vasopressin levels during the recovery period and NMDA receptor antagonism increased concentration of this hormone during both the hemorrhage and postbleeding periods in relation to control animals. Moreover, hemorrhagic shock caused a decrease in NOx levels in the paraventricular nucleus of the hypothalamus (PVN), amygdala, bed nucleus of the stria terminalis (BNST), and ventral periaqueductal gray matter (vPAG). Nevertheless, treatment with MK801 did not affect these effects. Taken together, these results indicate that the NMDA glutamate receptor is involved in the hemorrhagic shock by inhibiting circulating vasopressin release. Our data also suggest a role of the NMDA receptor in tachycardia, but not in the decreased NO synthesis in the brain evoked by hemorrhage.
Collapse
|
35
|
Cellular context shapes cyclic nucleotide signaling in neurons through multiple levels of integration. J Neurosci Methods 2021; 362:109305. [PMID: 34343574 DOI: 10.1016/j.jneumeth.2021.109305] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 07/22/2021] [Accepted: 07/29/2021] [Indexed: 02/06/2023]
Abstract
Intracellular signaling with cyclic nucleotides are ubiquitous signaling pathways, yet the dynamics of these signals profoundly differ in different cell types. Biosensor imaging experiments, by providing direct measurements in intact cellular environment, reveal which receptors are activated by neuromodulators and how the coincidence of different neuromodulators is integrated at various levels in the signaling cascade. Phosphodiesterases appear as one important determinant of cross-talk between different signaling pathways. Finally, analysis of signal dynamics reveal that striatal medium-sized spiny neuron obey a different logic than other brain regions such as cortex, probably in relation with the function of this brain region which efficiently detects transient dopamine.
Collapse
|
36
|
Delli V, Silva MSB, Prévot V, Chachlaki K. The KiNG of reproduction: Kisspeptin/ nNOS interactions shaping hypothalamic GnRH release. Mol Cell Endocrinol 2021; 532:111302. [PMID: 33964320 DOI: 10.1016/j.mce.2021.111302] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 04/20/2021] [Accepted: 04/23/2021] [Indexed: 12/21/2022]
Abstract
Gonadotropin-releasing hormone (GnRH) is the master regulator of the hypothalamic-pituitary-gonadal (HPG) axis, and therefore of fertility and reproduction. The release pattern of GnRH by the hypothalamus includes both pulses and surges. However, despite a considerable body of evidence in support of a determinant role for kisspeptin, the mechanisms regulating a GnRH pulse and surge remain a topic of debate. In this review we challenge the view of kisspeptin as an absolute "monarch", and instead present the idea of a Kisspeptin-nNOS-GnRH or "KiNG" network that is responsible for generating the "GnRH pulse" and "GnRH surge". In particular, the neuromodulator nitric oxide (NO) has opposite effects to kisspeptin on GnRH secretion in many respects, acting as the Yin to kisspeptin's Yang and creating a dynamic system in which kisspeptin provides the "ON" signal, promoting GnRH release, while NO mediates the "OFF" signal, acting as a tonic brake on GnRH secretion. This interplay between an activator and an inhibitor, which is in turn fine-tuned by the gonadal steroid environment, thus leads to the generation of GnRH pulses and surges and is crucial for the proper development and function of the reproductive axis.
Collapse
Affiliation(s)
- Virginia Delli
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S 1172, F-59000, Lille, France; FHU, 1000 Days for Health, F-59000, Lille, France
| | - Mauro S B Silva
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S 1172, F-59000, Lille, France; FHU, 1000 Days for Health, F-59000, Lille, France
| | - Vincent Prévot
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S 1172, F-59000, Lille, France; FHU, 1000 Days for Health, F-59000, Lille, France
| | - Konstantina Chachlaki
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S 1172, F-59000, Lille, France; FHU, 1000 Days for Health, F-59000, Lille, France; University Research Institute of Child Health and Precision Medicine, National and Kapodistrian University of Athens, "Aghia Sophia" Children's Hospital, Athens, Greece.
| |
Collapse
|
37
|
Scheiblich H, Steinert JR. Nitrergic modulation of neuronal excitability in the mouse hippocampus is mediated via regulation of Kv2 and voltage-gated sodium channels. Hippocampus 2021; 31:1020-1038. [PMID: 34047430 DOI: 10.1002/hipo.23366] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 04/10/2021] [Accepted: 05/19/2021] [Indexed: 12/21/2022]
Abstract
Regulation of neuronal activity is a necessity for communication and information transmission. Many regulatory processes which have been studied provide a complex picture of how neurons can respond to permanently changing functional requirements. One such activity-dependent mechanism involves signaling mediated by nitric oxide (NO). Within the brain, NO is generated in response to neuronal NO synthase (nNOS) activation but NO-dependent pathways regulating neuronal excitability in the hippocampus remain to be fully elucidated. This study was set out to systematically assess the effects of NO on ion channel activities and intrinsic excitabilities of pyramidal neurons within the CA1 region of the mouse hippocampus. We characterized whole-cell potassium and sodium currents, both involved in action potential (AP) shaping and propagation and determined NO-mediated changes in excitabilities and AP waveforms. Our data describe a novel signaling by which NO, in a cGMP-independent manner, suppresses voltage-gated Kv2 potassium and voltage-gated sodium channel activities, thereby widening AP waveforms and reducing depolarization-induced AP firing rates. Our data show that glutathione, which possesses denitrosylating activity, is sufficient to prevent the observed nitrergic effects on potassium and sodium channels, whereas inhibition of cGMP signaling is also sufficient to abolish NO modulation of sodium currents. We propose that NO suppresses both ion channel activities via redox signaling and that an additional cGMP-mediated component is required to exert effects on sodium currents. Both mechanisms result in a dampened excitability and firing ability providing new data on nitrergic activities in the context of activity-dependent regulation of neuronal function following nNOS activation.
Collapse
Affiliation(s)
- Hannah Scheiblich
- Department of Neurodegenerative Disease and Geriatric Psychiatry/Neurology, University of Bonn Medical Center, Bonn, Germany
| | - Joern R Steinert
- Faculty of Medicine and Health Sciences, University of Nottingham, School of Life Sciences, Queen's Medical Centre, Nottingham, UK
| |
Collapse
|
38
|
Correia SS, Iyengar RR, Germano P, Tang K, Bernier SG, Schwartzkopf CD, Tobin J, Lee TWH, Liu G, Jacobson S, Carvalho A, Rennie GR, Jung J, Renhowe PA, Lonie E, Winrow CJ, Hadcock JR, Jones JE, Currie MG. The CNS-Penetrant Soluble Guanylate Cyclase Stimulator CY6463 Reveals its Therapeutic Potential in Neurodegenerative Diseases. Front Pharmacol 2021; 12:656561. [PMID: 34108877 PMCID: PMC8181742 DOI: 10.3389/fphar.2021.656561] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 04/16/2021] [Indexed: 12/21/2022] Open
Abstract
Effective treatments for neurodegenerative diseases remain elusive and are critically needed since the burden of these diseases increases across an aging global population. Nitric oxide (NO) is a gasotransmitter that binds to soluble guanylate cyclase (sGC) to produce cyclic guanosine monophosphate (cGMP). Impairment of this pathway has been demonstrated in neurodegenerative diseases. Normalizing deficient NO-cGMP signaling could address multiple pathophysiological features of neurodegenerative diseases. sGC stimulators are small molecules that synergize with NO, activate sGC, and increase cGMP production. Many systemic sGC stimulators have been characterized and advanced into clinical development for a variety of non-central nervous system (CNS) pathologies. Here, we disclose the discovery of CY6463, the first brain-penetrant sGC stimulator in clinical development for the treatment of neurodegenerative diseases, and demonstrate its ability to improve neuronal activity, mediate neuroprotection, and increase cognitive performance in preclinical models. In several cellular assays, CY6463 was demonstrated to be a potent stimulator of sGC. In agreement with the known effects of sGC stimulation in the vasculature, CY6463 elicits decreases in blood pressure in both rats and mice. Relative to a non-CNS penetrant sGC stimulator, rodents treated with CY6463 had higher cGMP levels in cerebrospinal fluid (CSF), functional-magnetic-resonance-imaging-blood-oxygen-level-dependent (fMRI-BOLD) signals, and cortical electroencephalographic (EEG) gamma-band oscillatory power. Additionally, CY6463 improved cognitive performance in a model of cognitive disruption induced by the administration of a noncompetitive N-methyl-D-aspartate (NMDA) receptor antagonist. In models of neurodegeneration, CY6463 treatment increased long-term potentiation (LTP) in hippocampal slices from a Huntington’s disease mouse model and decreased the loss of dendritic spines in aged and Alzheimer’s disease mouse models. In a model of diet-induced obesity, CY6463 reduced markers of inflammation in the plasma. Furthermore, CY6463 elicited an additive increase in cortical gamma-band oscillatory power when co-administered with donepezil: the standard of care in Alzheimer’s disease. Together, these data support the clinical development of CY6463 as a novel treatment for neurodegenerative disorders.
Collapse
Affiliation(s)
| | | | - Peter Germano
- Cyclerion Therapeutics, Cambridge, MA, United States
| | - Kim Tang
- Ironwood Pharmaceuticals, Cambridge, MA, United States
| | | | | | - Jenny Tobin
- Cyclerion Therapeutics, Cambridge, MA, United States
| | | | - Guang Liu
- Cyclerion Therapeutics, Cambridge, MA, United States
| | | | | | - Glen R Rennie
- Cyclerion Therapeutics, Cambridge, MA, United States
| | - Joon Jung
- Cyclerion Therapeutics, Cambridge, MA, United States
| | | | | | | | | | - Juli E Jones
- Cyclerion Therapeutics, Cambridge, MA, United States
| | - Mark G Currie
- Cyclerion Therapeutics, Cambridge, MA, United States
| |
Collapse
|
39
|
Nitric oxide and the brain. Part 2: Effects following neonatal brain injury-friend or foe? Pediatr Res 2021; 89:746-752. [PMID: 32563184 DOI: 10.1038/s41390-020-1021-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 04/30/2020] [Accepted: 06/02/2020] [Indexed: 12/30/2022]
Abstract
Nitric oxide (NO) has critical roles in a wide variety of key biologic functions and has intricate transport mechanisms for delivery to key distal tissues under normal conditions. However, NO also plays important roles during disease processes, such as hypoxia-ischemia, asphyxia, neuro-inflammation, and retinopathy of prematurity. The effects of exogenous NO on the developing neonatal brain remain controversial. Inhaled NO (iNO) can be neuroprotective or toxic depending on a variety of factors, including cellular redox state, underlying disease processes, duration of treatment, and dose. This review identifies key gaps in knowledge that should prompt further investigation into the possible role of iNO as a therapeutic agent after injury to the brain. IMPACT: NO is a key signal mediator in the neonatal brain with neuroprotective and neurotoxic properties. iNO, a commonly used medication, has significant effects on the neonatal brain. Dosing, duration, and timing of administration of iNO can affect the developing brain. This review article summarizes the roles of NO in association with various disease processes that impact neonates, such as brain hypoxia-ischemia, asphyxia, retinopathy of prematurity, and neuroinflammation. The impact of this review is that it clearly describes gaps in knowledge, and makes the case for further, targeted studies in each of the identified areas.
Collapse
|
40
|
Petrova ON, Lamarre I, Fasani F, Grillon C, Negrerie M. Soluble Guanylate Cyclase Inhibitors Discovered among Natural Compounds. JOURNAL OF NATURAL PRODUCTS 2020; 83:3642-3651. [PMID: 33290062 DOI: 10.1021/acs.jnatprod.0c00854] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Soluble guanylate cyclase (sGC) is the human receptor of nitric oxide (NO) in numerous kinds of cells and produces the second messenger 3',5'-cyclic guanosine monophosphate (cGMP) upon NO binding to its heme. sGC is involved in many cell signaling pathways both under healthy conditions and under pathological conditions, such as angiogenesis associated with tumor growth. Addressing the selective inhibition of the NO/cGMP pathway is a strategy worthwhile to be investigated for slowing down tumoral angiogenesis or for curing vasoplegia. However, sGC inhibitors are lacking investigation. We have explored a chemical library of various natural compounds and have discovered inhibitors of sGC. The selected compounds were evaluated for their inhibition of purified sGC in vitro and sGC in endothelial cells. Six natural compounds, from various organisms, have IC50 in the range 0.2-1.5 μM for inhibiting the NO-activated synthesis of cGMP by sGC, and selected compounds exhibit a quantified antiangiogenic activity using an endothelial cell line. These sGC inhibitors can be used directly as tools to investigate angiogenesis and cell signaling or as templates for drug design.
Collapse
Affiliation(s)
- Olga N Petrova
- Laboratoire d'Optique et Biosciences, INSERM U1182, Ecole Polytechnique, Palaiseau, France
| | - Isabelle Lamarre
- Laboratoire d'Optique et Biosciences, INSERM U1182, Ecole Polytechnique, Palaiseau, France
| | - Fabienne Fasani
- Centre de Biophysique Moléculaire, UPR4301 CNRS, Orléans, France
| | | | - Michel Negrerie
- Laboratoire d'Optique et Biosciences, INSERM U1182, Ecole Polytechnique, Palaiseau, France
| |
Collapse
|
41
|
Ferreira-Junior NC, Crestani CC, Lagatta DC, Resstel LBM, Correa FMA, Alves FHF. Nitric oxide in the insular cortex modulates baroreflex responses in a cGMP-independent pathway. Brain Res 2020; 1747:147037. [PMID: 32738232 DOI: 10.1016/j.brainres.2020.147037] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 07/01/2020] [Accepted: 07/27/2020] [Indexed: 10/23/2022]
Abstract
Insular cortex is a brain structure involved in the modulation of autonomic activity and cardiovascular function. The nitric oxide/cyclic guanosine-3',5'-monophosphate pathway is a prominent signaling mechanism in the central nervous system, controlling behavioral and physiological responses. Nevertheless, despite evidence regarding the presence of nitric oxide-synthesizing neurons in the insular cortex, its role in the control of autonomic and cardiovascular function has never been reported. Thus, the present study aimed to investigate the involvement of nitric oxide/cyclic guanosine-3',5'-monophosphate pathway mediated by neuronal nitric oxide synthase (nNOS) activation within the insular cortex in the modulation of baroreflex responses in unanesthetized rats. For this, we evaluated the effect of bilateral microinjection of either the nitric oxide scavenger carboxy-PTIO, the selective neuronal nitric oxide synthase inhibitor Nω-Propyl-l-arginine or the soluble guanylate cyclase inhibitor ODQ into the insular cortex on the bradycardia evoked by blood pressure increases in response to intravenous infusion of phenylephrine, and the tachycardia caused by blood pressure decreases evoked by intravenous infusion of sodium nitroprusside. Bilateral microinjection of either NPLA or carboxy-PTIO into the insular cortex increased the reflex bradycardic response, whereas the reflex tachycardia was decreased by these treatments. Bilateral microinjection of the soluble guanylate cyclase inhibitor into the insular cortex did not affect any parameter of baroreflex function evaluated. Overall, our findings provide evidence that insular cortex nitrergic signaling, acting via neuronal nitric oxide synthase, plays a prominent role in control of baroreflex function. However, control of reflex responses seems to be independent of soluble guanylate cyclase activation.
Collapse
Affiliation(s)
- Nilson C Ferreira-Junior
- Department of Pharmacology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Carlos C Crestani
- Laboratory of Pharmacology, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, SP, Brazil
| | - Davi C Lagatta
- Department of Pharmacology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Leonardo B M Resstel
- Department of Pharmacology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Fernando M A Correa
- Department of Pharmacology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Fernando H F Alves
- Department of Health Sciences, Faculty of Medicine - Federal University of Lavras, Lavras, MG, Brazil.
| |
Collapse
|
42
|
Betolngar DB, Mota É, Fabritius A, Nielsen J, Hougaard C, Christoffersen CT, Yang J, Kehler J, Griesbeck O, Castro LRV, Vincent P. Phosphodiesterase 1 Bridges Glutamate Inputs with NO- and Dopamine-Induced Cyclic Nucleotide Signals in the Striatum. Cereb Cortex 2020; 29:5022-5036. [PMID: 30877787 DOI: 10.1093/cercor/bhz041] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 02/14/2019] [Indexed: 12/15/2022] Open
Abstract
The calcium-regulated phosphodiesterase 1 (PDE1) family is highly expressed in the brain, but its functional role in neurones is poorly understood. Using the selective PDE1 inhibitor Lu AF64196 and biosensors for cyclic nucleotides including a novel biosensor for cGMP, we analyzed the effect of PDE1 on cAMP and cGMP in individual neurones in brain slices from male newborn mice. Release of caged NMDA triggered a transient increase of intracellular calcium, which was associated with a decrease in cAMP and cGMP in medium spiny neurones in the striatum. Lu AF64196 alone did not increase neuronal cyclic nucleotide levels, but blocked the NMDA-induced reduction in cyclic nucleotides indicating that this was mediated by calcium-activated PDE1. Similar effects were observed in the prefrontal cortex and the hippocampus. Upon corelease of dopamine and NMDA, PDE1 was shown to down-regulate the D1-receptor mediated increase in cAMP. PDE1 inhibition increased long-term potentiation in rat ventral striatum, showing that PDE1 is implicated in the regulation of synaptic plasticity. Overall, our results show that PDE1 reduces cyclic nucleotide signaling in the context of glutamate and dopamine coincidence. This effect could have a therapeutic value for treating brain disorders related to dysfunctions in dopamine neuromodulation.
Collapse
Affiliation(s)
| | - Élia Mota
- Sorbonne Université, CNRS, Biological Adaptation and Ageing, Paris, France
| | - Arne Fabritius
- Max Planck Institute for Neurobiology, Tools for Bio-Imaging, Am Klopferspitz 18, Martinsried, Germany
| | | | | | | | - Jun Yang
- Shanghai Chempartner Co. Ltd., Shanghai, China
| | - Jan Kehler
- H. Lundbeck A/S, Ottiliavej 9, Valby, Denmark
| | - Oliver Griesbeck
- Max Planck Institute for Neurobiology, Tools for Bio-Imaging, Am Klopferspitz 18, Martinsried, Germany
| | - Liliana R V Castro
- Sorbonne Université, CNRS, Biological Adaptation and Ageing, Paris, France
| | - Pierre Vincent
- Sorbonne Université, CNRS, Biological Adaptation and Ageing, Paris, France
| |
Collapse
|
43
|
Araki S, Osuka K, Takata T, Tsuchiya Y, Watanabe Y. Coordination between Calcium/Calmodulin-Dependent Protein Kinase II and Neuronal Nitric Oxide Synthase in Neurons. Int J Mol Sci 2020; 21:ijms21217997. [PMID: 33121174 PMCID: PMC7662388 DOI: 10.3390/ijms21217997] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 10/21/2020] [Accepted: 10/22/2020] [Indexed: 12/15/2022] Open
Abstract
Ca2+/calmodulin (CaM)-dependent protein kinase II (CaMKII) is highly abundant in the brain and exhibits broad substrate specificity, thereby it is thought to participate in the regulation of neuronal death and survival. Nitric oxide (NO), produced by neuronal NO synthase (nNOS), is an important neurotransmitter and plays a role in neuronal activity including learning and memory processes. However, high levels of NO can contribute to excitotoxicity following a stroke and neurodegenerative disease. Aside from NO, nNOS also generates superoxide which is involved in both cell injury and signaling. CaMKII is known to activate and translocate from the cytoplasm to the post-synaptic density in response to neuronal activation where nNOS is predominantly located. Phosphorylation of nNOS at Ser847 by CaMKII decreases NO generation and increases superoxide generation. Conversely, NO-induced S-nitrosylation of CaMKII at Cys6 is a prominent determinant of the CaMKII inhibition in ATP competitive fashion. Thus, the "cross-talk" between CaMKII and NO/superoxide may represent important signal transduction pathways in brain. In this review, we introduce the molecular mechanism of and pathophysiological role of mutual regulation between CaMKII and nNOS in neurons.
Collapse
Affiliation(s)
- Shoma Araki
- Department of Pharmacology, Showa Pharmaceutical University, Machida, Tokyo 194-8543, Japan; (S.A.); (T.T.); (Y.T.)
| | - Koji Osuka
- Department of Neurological Surgery, Aichi Medical University, Aichi 480-1195, Japan;
| | - Tsuyoshi Takata
- Department of Pharmacology, Showa Pharmaceutical University, Machida, Tokyo 194-8543, Japan; (S.A.); (T.T.); (Y.T.)
- Department of Environmental Health Sciences and Molecular Toxicology, Graduate School of Medicine, Tohoku University, Miyagi 980-8575, Japan
| | - Yukihiro Tsuchiya
- Department of Pharmacology, Showa Pharmaceutical University, Machida, Tokyo 194-8543, Japan; (S.A.); (T.T.); (Y.T.)
| | - Yasuo Watanabe
- Department of Pharmacology, Showa Pharmaceutical University, Machida, Tokyo 194-8543, Japan; (S.A.); (T.T.); (Y.T.)
- Correspondence:
| |
Collapse
|
44
|
Hirooka Y. Sympathetic Activation in Hypertension: Importance of the Central Nervous System. Am J Hypertens 2020; 33:914-926. [PMID: 32374869 DOI: 10.1093/ajh/hpaa074] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 04/18/2020] [Accepted: 05/01/2020] [Indexed: 12/20/2022] Open
Abstract
The sympathetic nervous system plays a critical role in the pathogenesis of hypertension. The central nervous system (CNS) organizes the sympathetic outflow and various inputs from the periphery. The brain renin-angiotensin system has been studied in various regions involved in controlling sympathetic outflow. Recent progress in cardiovascular research, particularly in vascular biology and neuroscience, as well as in traditional physiological approaches, has advanced the field of the neural control of hypertension in which the CNS plays a vital role. Cardiovascular research relating to hypertension has focused on the roles of nitric oxide, oxidative stress, inflammation, and immunity, and the network among various organs, including the heart, kidney, spleen, gut, and vasculature. The CNS mechanisms are similarly networked with these factors and are widely studied in neuroscience. In this review, I describe the development of the conceptual flow of this network in the field of hypertension on the basis of several important original research articles and discuss potential future breakthroughs leading to clinical precision medicine.
Collapse
Affiliation(s)
- Yoshitaka Hirooka
- Department of Medical Technology and Sciences, School of Health Sciences at Fukuoka, International University of Health and Welfare, Okawa City, Fukuoka, Japan
- Department of Cardiovascular Medicine, Hypertension and Heart Failure Center, Takagi Hospital, Okawa City, Fukuoka, Japan
| |
Collapse
|
45
|
Vidanapathirana AK, Psaltis PJ, Bursill CA, Abell AD, Nicholls SJ. Cardiovascular bioimaging of nitric oxide: Achievements, challenges, and the future. Med Res Rev 2020; 41:435-463. [PMID: 33075148 DOI: 10.1002/med.21736] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 08/03/2020] [Accepted: 08/24/2020] [Indexed: 12/17/2022]
Abstract
Nitric oxide (NO) is a ubiquitous, volatile, cellular signaling molecule that operates across a wide physiological concentration range (pM-µM) in different tissues. It is a highly diffusible messenger and intermediate in various metabolic pathways. NO plays a pivotal role in maintaining optimum cardiovascular function, particularly by regulating vascular tone and blood flow. This review highlights the need for accurate, real-time bioimaging of NO in clinical diagnostic, therapeutic, monitoring, and theranostic applications within the cardiovascular system. We summarize electrochemical, optical, and nanoscale sensors that allow measurement and imaging of NO, both directly and indirectly via surrogate measurements. The physical properties of NO render it difficult to accurately measure in tissues using direct methods. There are also significant limitations associated with the NO metabolites used as surrogates to indirectly estimate NO levels. All these factors added to significant variability in the measurement of NO using available methodology have led to a lack of sensors and imaging techniques of clinical applicability in relevant vascular pathologies such as atherosclerosis and ischemic heart disease. Challenges in applying current methods to biomedical and clinical translational research, including the wide physiological range of NO and limitations due to the characteristics and toxicity of the sensors are discussed, as are potential targets and modifications for future studies. The development of biocompatible nanoscale sensors for use in combination with existing clinical imaging modalities provides a feasible opportunity for bioimaging NO within the cardiovascular system.
Collapse
Affiliation(s)
- Achini K Vidanapathirana
- Vascular Research Centre, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia.,Australian Research Council (ARC), Centre of Excellence for Nanoscale BioPhotonics (CNBP), Adelaide, Australia.,Institute for Photonics and Advanced Sensing (IPAS), University of Adelaide, Adelaide, South Australia, Australia.,Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| | - Peter J Psaltis
- Vascular Research Centre, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia.,Australian Research Council (ARC), Centre of Excellence for Nanoscale BioPhotonics (CNBP), Adelaide, Australia.,Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| | - Christina A Bursill
- Vascular Research Centre, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia.,Australian Research Council (ARC), Centre of Excellence for Nanoscale BioPhotonics (CNBP), Adelaide, Australia.,Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| | - Andrew D Abell
- Australian Research Council (ARC), Centre of Excellence for Nanoscale BioPhotonics (CNBP), Adelaide, Australia.,Institute for Photonics and Advanced Sensing (IPAS), University of Adelaide, Adelaide, South Australia, Australia.,Department of Chemistry, University of Adelaide, Adelaide, South Australia, Australia
| | - Stephen J Nicholls
- Australian Research Council (ARC), Centre of Excellence for Nanoscale BioPhotonics (CNBP), Adelaide, Australia.,Monash Cardiovascular Research Centre, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
46
|
Arrais AC, Melo LHMF, Norrara B, Almeida MAB, Freire KF, Melo AMMF, Oliveira LCD, Lima FOV, Engelberth RCGJ, Cavalcante JDS, Araújo DPD, Guzen FP, Freire MAM, Cavalcanti JRLP. S100B protein: general characteristics and pathophysiological implications in the Central Nervous System. Int J Neurosci 2020; 132:313-321. [DOI: 10.1080/00207454.2020.1807979] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Ana Cristina Arrais
- Laboratory of Experimental Neurology, Department of Biomedical Sciences, Faculty of Health Sciences, University of the State of Rio Grande do Norte, Mossoró, RN, Brazil
| | - Lívia Helena M. F. Melo
- Laboratory of Experimental Neurology, Department of Biomedical Sciences, Faculty of Health Sciences, University of the State of Rio Grande do Norte, Mossoró, RN, Brazil
| | - Bianca Norrara
- Laboratory of Experimental Neurology, Department of Biomedical Sciences, Faculty of Health Sciences, University of the State of Rio Grande do Norte, Mossoró, RN, Brazil
| | - Marina Abuquerque B. Almeida
- Laboratory of Experimental Neurology, Department of Biomedical Sciences, Faculty of Health Sciences, University of the State of Rio Grande do Norte, Mossoró, RN, Brazil
| | - Kalina Fernandes Freire
- Laboratory of Experimental Neurology, Department of Biomedical Sciences, Faculty of Health Sciences, University of the State of Rio Grande do Norte, Mossoró, RN, Brazil
| | - Acydalia Madruga M. F. Melo
- Laboratory of Experimental Neurology, Department of Biomedical Sciences, Faculty of Health Sciences, University of the State of Rio Grande do Norte, Mossoró, RN, Brazil
| | - Lucidio Clebeson de Oliveira
- Laboratory of Experimental Neurology, Department of Biomedical Sciences, Faculty of Health Sciences, University of the State of Rio Grande do Norte, Mossoró, RN, Brazil
| | - Francisca Overlânia Vieira Lima
- Laboratory of Experimental Neurology, Department of Biomedical Sciences, Faculty of Health Sciences, University of the State of Rio Grande do Norte, Mossoró, RN, Brazil
| | - Rovena Clara G. J. Engelberth
- Laboratory of Neurochemical Studies, Department of Physiology, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - Jeferson de Souza Cavalcante
- Laboratory of Neurochemical Studies, Department of Physiology, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - Dayane Pessoa de Araújo
- Laboratory of Experimental Neurology, Department of Biomedical Sciences, Faculty of Health Sciences, University of the State of Rio Grande do Norte, Mossoró, RN, Brazil
| | - Fausto Pierdoná Guzen
- Laboratory of Experimental Neurology, Department of Biomedical Sciences, Faculty of Health Sciences, University of the State of Rio Grande do Norte, Mossoró, RN, Brazil
| | - Marco Aurelio M. Freire
- Laboratory of Experimental Neurology, Department of Biomedical Sciences, Faculty of Health Sciences, University of the State of Rio Grande do Norte, Mossoró, RN, Brazil
| | - José Rodolfo L. P. Cavalcanti
- Laboratory of Experimental Neurology, Department of Biomedical Sciences, Faculty of Health Sciences, University of the State of Rio Grande do Norte, Mossoró, RN, Brazil
| |
Collapse
|
47
|
Tau induces PSD95-neuronal NOS uncoupling and neurovascular dysfunction independent of neurodegeneration. Nat Neurosci 2020; 23:1079-1089. [PMID: 32778793 PMCID: PMC7896353 DOI: 10.1038/s41593-020-0686-7] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 07/02/2020] [Indexed: 02/06/2023]
Abstract
Cerebrovascular abnormalities have emerged as a preclinical manifestation of Alzheimer’s disease and frontotemporal dementia, diseases characterized by accumulation of hyperphosphorylated forms of the microtubule associated protein tau. However, it is unclear if tau contributes to these neurovascular alterations independent of neurodegeneration. We report that mice expressing mutated tau exhibit a selective suppression of neural activity-induced cerebral blood flow increases that precedes tau pathology and cognitive impairment. The dysfunction is attributable to reduced vasodilatation of intracerebral arterioles and is reversible by turning down tau production. Mechanistically, the failure of neurovascular coupling involves tau-induced dissociation of neuronal nitric oxide synthase from post synaptic-density-95 and reduced production of the potent vasodilator nitric oxide during glutamatergic synaptic activity. The data identify glutamatergic signaling dysfunction and nitric oxide deficiency as yet-undescribed early manifestations of tau pathobiology independent of neurodegeneration, and provide a mechanism for the neurovascular alterations observed in the preclinical stages of tauopathies.
Collapse
|
48
|
Piotrowicz Z, Chalimoniuk M, Płoszczyca K, Czuba M, Langfort J. Exercise-Induced Elevated BDNF Level Does Not Prevent Cognitive Impairment Due to Acute Exposure to Moderate Hypoxia in Well-Trained Athletes. Int J Mol Sci 2020; 21:ijms21155569. [PMID: 32759658 PMCID: PMC7432544 DOI: 10.3390/ijms21155569] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 07/30/2020] [Accepted: 07/31/2020] [Indexed: 12/12/2022] Open
Abstract
Exposure to acute hypoxia causes a detrimental effect on the brain which is also manifested by a decrease in the ability to perform psychomotor tasks. Conversely, brain-derived neurotrophic factor (BDNF), whose levels are elevated in response to exercise, is a well-known factor in improving cognitive function. Therefore, the aim of our study was to investigate whether the exercise under hypoxic conditions affects psychomotor performance. For this purpose, 11 healthy young athletes performed a graded cycloergometer exercise test to volitional exhaustion under normoxia and acute mild hypoxia (FiO2 = 14.7%). Before, immediately after exercise and after a period of recovery, choice reaction time (CRT) and number of correct reactions (NCR) in relation to changes in serum BDNF were examined. Additionally, other selected factors which may modify BDNF production, i.e., cortisol (C), nitrite, catecholamines (adrenalin-A, noradrenaline-NA, dopamine-DA, serotonin-5-HT) and endothelin-1 (ET-1), were also measured. Exercise in hypoxic conditions extended CRT by 13.8% (p < 0.01) and decreased NCR (by 11.5%) compared to rest (p < 0.05). During maximal workload, NCR was lower by 9% in hypoxia compared to normoxia (p < 0.05). BDNF increased immediately after exercise in normoxia (by 29.3%; p < 0.01), as well as in hypoxia (by 50.0%; p < 0.001). There were no differences in BDNF between normoxia and hypoxia. Considering the fact that similar levels of BDNF were seen in both conditions but cognitive performance was suppressed in hypoxia, acute elevation of BDNF did not compensate for hypoxia-induced cognition impairment. Moreover, neither potentially negative effects of C nor positive effects of A, DA and NO on the brain were observed in our study.
Collapse
Affiliation(s)
- Zofia Piotrowicz
- Institute of Sport Sciences, The Jerzy Kukuczka Academy of Physical Education, 40-065 Katowice, Poland;
- Correspondence:
| | - Małgorzata Chalimoniuk
- Department of Tourism and Health in Biała Podlaska, The Józef Piłsudski University of Physical Education, 00-968 Warsaw, Poland;
| | - Kamila Płoszczyca
- Department of Kinesiology, Institute of Sport, 01-982 Warsaw, Poland; (K.P.); (M.C.)
| | - Miłosz Czuba
- Department of Kinesiology, Institute of Sport, 01-982 Warsaw, Poland; (K.P.); (M.C.)
- Faculty of Health Sciences, Jan Dlugosz University, 42-200 Czestochowa, Poland
| | - Józef Langfort
- Institute of Sport Sciences, The Jerzy Kukuczka Academy of Physical Education, 40-065 Katowice, Poland;
| |
Collapse
|
49
|
Hockley A, Berger JI, Palmer AR, Wallace MN. Nitric oxide increases gain in the ventral cochlear nucleus of guinea pigs with tinnitus. Eur J Neurosci 2020; 52:4057-4080. [PMID: 32686192 DOI: 10.1111/ejn.14913] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 07/10/2020] [Accepted: 07/11/2020] [Indexed: 12/21/2022]
Abstract
Previous work has led to the hypothesis that, during the production of noise-induced tinnitus, higher levels of nitric oxide (NO), in the ventral cochlear nucleus (VCN), increase the gain applied to a reduced input from the cochlea. To test this hypothesis, we noise-exposed 26 guinea pigs, identified evidence of tinnitus in 12 of them and then compared the effects of an iontophoretically applied NO donor or production inhibitor on VCN single unit activity. We confirmed that the mean driven firing rate for the tinnitus and control groups was the same while it had fallen in the non-tinnitus group. By contrast, the mean spontaneous rate had increased for the tinnitus group relative to the control group, while it remained the same for the non-tinnitus group. A greater proportion of units responded to exogenously applied NO in the tinnitus (56%) and non-tinnitus groups (71%) than a control population (24%). In the tinnitus group, endogenous NO facilitated the driven firing rate in 37% (7/19) of neurons and appeared to bring the mean driven rate back up to control levels by a mechanism involving N-methyl-D-aspartic acid (NMDA) receptors. By contrast, in the non-tinnitus group, endogenous NO only facilitated the driven firing rate in 5% (1/22) of neurons and there was no facilitation of driven rate in the control group. The effects of endogenous NO on spontaneous activity were unclear. These results suggest that NO is involved in increasing the gain applied to driven activity, but other factors are also involved in the increase in spontaneous activity.
Collapse
Affiliation(s)
- Adam Hockley
- Medical Research Council Institute of Hearing Research, School of Medicine, University of Nottingham, Nottingham, UK.,School of Life Sciences, University of Nottingham, Nottingham, UK.,Department of Otolaryngology, Kresge Hearing Research Institute, University of Michigan, Ann Arbor, MI, USA
| | - Joel I Berger
- Medical Research Council Institute of Hearing Research, School of Medicine, University of Nottingham, Nottingham, UK.,Department of Neurosurgery, University of Iowa, Iowa City, IA, USA
| | - Alan R Palmer
- Medical Research Council Institute of Hearing Research, School of Medicine, University of Nottingham, Nottingham, UK.,Hearing Sciences, School of Medicine, University of Nottingham, Nottingham, UK
| | - Mark N Wallace
- Medical Research Council Institute of Hearing Research, School of Medicine, University of Nottingham, Nottingham, UK.,Hearing Sciences, School of Medicine, University of Nottingham, Nottingham, UK
| |
Collapse
|
50
|
Zhu D, Sun M, Liu Q, Yue Y, Lu J, Lin X, Shi J. Angiotensin (1-7) through modulation of the NMDAR-nNOS-NO pathway and serotonergic metabolism exerts an anxiolytic-like effect in rats. Behav Brain Res 2020; 390:112671. [PMID: 32437889 DOI: 10.1016/j.bbr.2020.112671] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 04/05/2020] [Accepted: 04/21/2020] [Indexed: 01/02/2023]
Abstract
Although recent studies have shown that angiotensin (1-7) (Ang [1-7]) exerts anti-stress and anxiolytic-like effects, the underlying mechanisms remain elusive. The ventral hippocampus (VH) is proposed to be a critical brain region for mood and stress management through the N-methyl-d-aspartate receptor (NMDAR) signaling pathway. However, the role of VH NMDAR signaling in the effects of Ang (1-7) remains unclear. In the present study, Ang (1-7) was injected into the bilateral VH of stressed rats, or in combination with a Fyn kinase inhibitor, NMDAR antagonist, neuronal nitric oxide synthase (nNOS) inhibitor, or nitric oxide (NO) scavenger. Anxiety-like behaviors were assessed using the open field test and elevated plus maze test, while alterations in NMDAR-nNOS-NO signaling and serotonergic metabolism were examined in the VH. After 21 days of chronic restraint stress, anxiety-like behaviors were evident. Levels of phosphorylated NR2B (a key NMDAR subunit), its upstream kinase Fyn, as well as activity of nNOS and monoamine oxidase (MAO) were markedly reduced. In contrast, levels of serotonin were increased. Bilateral VH infusion of Ang (1-7) recovered NMDAR-nNOS-NO signaling and MAO-mediated serotonin metabolism, as well as reducing anxiety-like behaviors in stressed rats. These effects were diminished by blockade of MasR (Ang [1-7]-specific receptor), Fyn kinase, NMDAR, nNOS, or NO production. Altogether, these findings indicate that Ang (1-7) exerts anxiolytic effects through modulation of the NMDAR-nNOS-NO pathway and serotonergic metabolism. Future translational research should focus on the relationship between Ang (1-7), glutamatergic neurotransmission, and serotonergic neurotransmission in the VH.
Collapse
Affiliation(s)
- Donglin Zhu
- Department of Neurology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, PR China
| | - Ming Sun
- Emergency Department, The Affiliated Suqian Hospital of Xuzhou Medical University, Suqian, PR China
| | - Qinqin Liu
- Department of Neurology, The Affiliated Suqian Hospital of Xuzhou Medical University, Suqian, PR China
| | - Yu Yue
- Department of Neurology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, PR China
| | - Jie Lu
- Department of Neurology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, PR China
| | - Xingjian Lin
- Department of Neurology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, PR China
| | - Jingping Shi
- Department of Neurology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, PR China.
| |
Collapse
|