1
|
Hamed MA, Wasinger V, Wang Q, Biazik J, Graham P, Malouf D, Bucci J, Li Y. Optimising Extracellular Vesicle Metabolomic Methodology for Prostate Cancer Biomarker Discovery. Metabolites 2024; 14:367. [PMID: 39057690 PMCID: PMC11279087 DOI: 10.3390/metabo14070367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 06/21/2024] [Accepted: 06/26/2024] [Indexed: 07/28/2024] Open
Abstract
Conventional diagnostic tools for prostate cancer (PCa), such as prostate-specific antigen (PSA), transrectal ultrasound (TRUS), digital rectal examination (DRE), and tissue biopsy face, limitations in individual risk stratification due to invasiveness or reliability issues. Liquid biopsy is a less invasive and more accurate alternative. Metabolomic analysis of extracellular vesicles (EVs) holds a promise for detecting non-genetic alterations and biomarkers in PCa diagnosis and risk assessment. The current research gap in PCa lies in the lack of accurate biomarkers for early diagnosis and real-time monitoring of cancer progression or metastasis. Establishing a suitable approach for observing dynamic EV metabolic alterations that often occur earlier than being detectable by other omics technologies makes metabolomics valuable for early diagnosis and monitoring of PCa. Using four distinct metabolite extraction approaches, the metabolite cargo of PC3-derived large extracellular vesicles (lEVs) was evaluated using a combination of methanol, cell shearing using microbeads, and size exclusion filtration, as well as two fractionation chemistries (pHILIC and C18 chromatography) that are also examined. The unfiltered methanol-microbeads approach (MB-UF), followed by pHILIC LC-MS/MS for EV metabolite extraction and analysis, is effective. Identified metabolites such as L-glutamic acid, pyruvic acid, lactic acid, and methylmalonic acid have important links to PCa and are discussed. Our study, for the first time, has comprehensively evaluated the extraction and separation methods with a view to downstream sample integrity across omics platforms, and it presents an optimised protocol for EV metabolomics in PCa biomarker discovery.
Collapse
Affiliation(s)
- Mahmoud Assem Hamed
- St. George and Sutherland Clinical Campuses, School of Clinical Medicine, University of New South Wales (UNSW) Sydney, Kensington, NSW 2052, Australia; (M.A.H.); (Q.W.); (P.G.); (J.B.)
- Cancer Care Centre, St. George Hospital, Kogarah, NSW 2217, Australia
| | - Valerie Wasinger
- Bioanalytical Mass Spectrometry Facility, Mark Wainwright Analytical Centre, University of New South Wales (UNSW) Sydney, Kensington, NSW 2052, Australia;
| | - Qi Wang
- St. George and Sutherland Clinical Campuses, School of Clinical Medicine, University of New South Wales (UNSW) Sydney, Kensington, NSW 2052, Australia; (M.A.H.); (Q.W.); (P.G.); (J.B.)
- Cancer Care Centre, St. George Hospital, Kogarah, NSW 2217, Australia
| | - Joanna Biazik
- Electron Microscope Unit, Mark Wainwright Analytical Centre, University of New South Wales (UNSW) Sydney, Kensington, NSW 2052, Australia;
| | - Peter Graham
- St. George and Sutherland Clinical Campuses, School of Clinical Medicine, University of New South Wales (UNSW) Sydney, Kensington, NSW 2052, Australia; (M.A.H.); (Q.W.); (P.G.); (J.B.)
- Cancer Care Centre, St. George Hospital, Kogarah, NSW 2217, Australia
| | - David Malouf
- Department of Urology, St. George Hospital, Kogarah, NSW 2217, Australia;
| | - Joseph Bucci
- St. George and Sutherland Clinical Campuses, School of Clinical Medicine, University of New South Wales (UNSW) Sydney, Kensington, NSW 2052, Australia; (M.A.H.); (Q.W.); (P.G.); (J.B.)
- Cancer Care Centre, St. George Hospital, Kogarah, NSW 2217, Australia
| | - Yong Li
- St. George and Sutherland Clinical Campuses, School of Clinical Medicine, University of New South Wales (UNSW) Sydney, Kensington, NSW 2052, Australia; (M.A.H.); (Q.W.); (P.G.); (J.B.)
- Cancer Care Centre, St. George Hospital, Kogarah, NSW 2217, Australia
| |
Collapse
|
2
|
Ruperti F, Becher I, Stokkermans A, Wang L, Marschlich N, Potel C, Maus E, Stein F, Drotleff B, Schippers KJ, Nickel M, Prevedel R, Musser JM, Savitski MM, Arendt D. Molecular profiling of sponge deflation reveals an ancient relaxant-inflammatory response. Curr Biol 2024; 34:361-375.e9. [PMID: 38181793 DOI: 10.1016/j.cub.2023.12.021] [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: 08/17/2023] [Revised: 11/03/2023] [Accepted: 12/07/2023] [Indexed: 01/07/2024]
Abstract
A hallmark of animals is the coordination of whole-body movement. Neurons and muscles are central to this, yet coordinated movements also exist in sponges that lack these cell types. Sponges are sessile animals with a complex canal system for filter-feeding. They undergo whole-body movements resembling "contractions" that lead to canal closure and water expulsion. Here, we combine live 3D optical coherence microscopy, pharmacology, and functional proteomics to elucidate the sequence and detail of shape changes, the tissues and molecular physiology involved, and the control of these movements. Morphometric analysis and targeted perturbation suggest that the movement is driven by the relaxation of actomyosin stress fibers in epithelial canal cells, which leads to whole-body deflation via collapse of the incurrent and expansion of the excurrent canal system. Thermal proteome profiling and quantitative phosphoproteomics confirm the control of cellular relaxation by an Akt/NO/PKG/PKA pathway. Agitation-induced deflation leads to differential phosphorylation of proteins forming epithelial cell junctions, implying their mechanosensitive role. Unexpectedly, untargeted metabolomics detect a concomitant decrease in antioxidant molecules during deflation, reflecting an increase in reactive oxygen species. Together with the secretion of proteinases, cytokines, and granulin, this indicates an inflammation-like state of the deflating sponge reminiscent of vascular endothelial cells experiencing oscillatory shear stress. These results suggest the conservation of an ancient relaxant-inflammatory response of perturbed fluid-carrying systems in animals and offer a possible mechanism for whole-body coordination through diffusible paracrine signals and mechanotransduction.
Collapse
Affiliation(s)
- Fabian Ruperti
- Developmental Biology Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany; Collaboration for joint Ph.D. degree between EMBL and Heidelberg University, Faculty of Biosciences 69117 Heidelberg, Germany
| | - Isabelle Becher
- Genome Biology Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | | | - Ling Wang
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany.
| | - Nick Marschlich
- Developmental Biology Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany; Centre for Organismal Studies (COS), University of Heidelberg, 69120 Heidelberg, Germany
| | - Clement Potel
- Genome Biology Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Emanuel Maus
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Frank Stein
- Proteomics Core Facility, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Bernhard Drotleff
- Metabolomics Core Facility, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Klaske J Schippers
- Developmental Biology Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Michael Nickel
- Bionic consulting Dr. Michael Nickel, 71686 Remseck am Neckar, Germany
| | - Robert Prevedel
- Developmental Biology Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany; Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Jacob M Musser
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06520, USA.
| | - Mikhail M Savitski
- Genome Biology Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany; Proteomics Core Facility, European Molecular Biology Laboratory, 69117 Heidelberg, Germany.
| | - Detlev Arendt
- Developmental Biology Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany; Centre for Organismal Studies (COS), University of Heidelberg, 69120 Heidelberg, Germany.
| |
Collapse
|
3
|
Samaja M, Malavalli A, Vandegriff KD. How Nitric Oxide Hindered the Search for Hemoglobin-Based Oxygen Carriers as Human Blood Substitutes. Int J Mol Sci 2023; 24:14902. [PMID: 37834350 PMCID: PMC10573492 DOI: 10.3390/ijms241914902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 09/30/2023] [Accepted: 10/02/2023] [Indexed: 10/15/2023] Open
Abstract
The search for a clinically affordable substitute of human blood for transfusion is still an unmet need of modern society. More than 50 years of research on acellular hemoglobin (Hb)-based oxygen carriers (HBOC) have not yet produced a single formulation able to carry oxygen to hemorrhage-challenged tissues without compromising the body's functions. Of the several bottlenecks encountered, the high reactivity of acellular Hb with circulating nitric oxide (NO) is particularly arduous to overcome because of the NO-scavenging effect, which causes life-threatening side effects as vasoconstriction, inflammation, coagulopathies, and redox imbalance. The purpose of this manuscript is not to add a review of candidate HBOC formulations but to focus on the biochemical and physiological events that underly NO scavenging by acellular Hb. To this purpose, we examine the differential chemistry of the reaction of NO with erythrocyte and acellular Hb, the NO signaling paths in physiological and HBOC-challenged situations, and the protein engineering tools that are predicted to modulate the NO-scavenging effect. A better understanding of two mechanisms linked to the NO reactivity of acellular Hb, the nitrosylated Hb and the nitrite reductase hypotheses, may become essential to focus HBOC research toward clinical targets.
Collapse
Affiliation(s)
- Michele Samaja
- Department of Health Science, University of Milan, 20143 Milan, Italy
| | | | | |
Collapse
|
4
|
Chuang WH, Chou YT, Chen YH, Kuo TH, Liaw WF, Lu TT, Kao CF, Wang YM. Neuroprotective Effect of NO-Delivery Dinitrosyl Iron Complexes (DNICs) on Amyloid Pathology in the Alzheimer's Disease Cell Model. ACS Chem Neurosci 2023; 14:2922-2934. [PMID: 37533298 DOI: 10.1021/acschemneuro.3c00348] [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] [Indexed: 08/04/2023] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by cognitive impairment, memory loss, and behavioral deficits. β-amyloid1-42 (Aβ1-42) aggregation is a significant cause of the pathogenesis in AD. Despite the numerous types of research, the current treatment efficacy remains insufficient. Hence, a novel therapeutic strategy is required. Nitric oxide (NO) is a multifunctional gaseous molecule. NO displays a neuroprotective role in the central nervous system by inhibiting the Aβ aggregation and rescuing memory and learning deficit through the NO signaling pathway. Targeting the NO pathway might be a therapeutic option; however, NO has a limited half-life under the biological system. To address this issue, a biomimetic dinitrosyl iron complex [(NO)2Fe(μ-SCH2CH2COOH)2Fe(NO)2] (DNIC-COOH) that could stably deliver NO was explored in the current study. To determine whether DNIC-COOH exerts anti-AD efficacy, DNIC-COOH was added to neuron-like cells and primary cortical neurons along with Aβ1-42. This study found that DNIC-COOH protected neuronal cells from Aβ-induced cytotoxicity, potentiated neuronal functions, and facilitated Aβ1-42 degradation through the NO-sGC-cGMP-AKT-GSK3β-CREB/MMP-9 pathway.
Collapse
Affiliation(s)
- Wen-Han Chuang
- Department of Biological Science and Technology, Institute of Molecular Medicine and Bioengineering, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
- Center for Intelligent Drug Systems and Smart Bio-Devices (IDS2B), National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
| | - Yu-Ting Chou
- Department of Biological Science and Technology, Institute of Molecular Medicine and Bioengineering, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
- Center for Intelligent Drug Systems and Smart Bio-Devices (IDS2B), National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
| | - Yi-Hong Chen
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Ting-Han Kuo
- Department of Biological Science and Technology, Institute of Molecular Medicine and Bioengineering, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
| | - Wen-Feng Liaw
- Department of Chemistry, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Tsai-Te Lu
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu 300, Taiwan
- Department of Chemistry, National Tsing Hua University, Hsinchu 300, Taiwan
- Department of Chemistry, Chung Yuan Christian University, Taoyuan 32023, Taiwan
| | - Chih-Fei Kao
- Department of Biological Science and Technology, Institute of Molecular Medicine and Bioengineering, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
- Center for Intelligent Drug Systems and Smart Bio-Devices (IDS2B), National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
| | - Yun-Ming Wang
- Department of Biological Science and Technology, Institute of Molecular Medicine and Bioengineering, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
- Center for Intelligent Drug Systems and Smart Bio-Devices (IDS2B), National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
- Department of Biomedical Science and Environmental Biology, Department of Dentistry, Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| |
Collapse
|
5
|
Ruperti F, Becher I, Stokkermans A, Wang L, Marschlich N, Potel C, Maus E, Stein F, Drotleff B, Schippers K, Nickel M, Prevedel R, Musser JM, Savitski MM, Arendt D. Molecular profiling of sponge deflation reveals an ancient relaxant-inflammatory response. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.02.551666. [PMID: 37577507 PMCID: PMC10418225 DOI: 10.1101/2023.08.02.551666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
A hallmark of animals is the coordination of whole-body movement. Neurons and muscles are central to this, yet coordinated movements also exist in sponges that lack these cell types. Sponges are sessile animals with a complex canal system for filter-feeding. They undergo whole-body movements resembling "contractions" that lead to canal closure and water expulsion. Here, we combine 3D optical coherence microscopy, pharmacology, and functional proteomics to elucidate anatomy, molecular physiology, and control of these movements. We find them driven by the relaxation of actomyosin stress fibers in epithelial canal cells, which leads to whole-body deflation via collapse of the incurrent and expansion of the excurrent system, controlled by an Akt/NO/PKG/A pathway. A concomitant increase in reactive oxygen species and secretion of proteinases and cytokines indicate an inflammation-like state reminiscent of vascular endothelial cells experiencing oscillatory shear stress. This suggests an ancient relaxant-inflammatory response of perturbed fluid-carrying systems in animals.
Collapse
Affiliation(s)
- Fabian Ruperti
- Developmental Biology Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
- Collaboration for joint Ph.D. degree between EMBL and Heidelberg University, Faculty of Biosciences 69117 Heidelberg, Germany
| | - Isabelle Becher
- Genome Biology Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | | | - Ling Wang
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Nick Marschlich
- Developmental Biology Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
- Centre for Organismal Studies (COS), University of Heidelberg, 69120 Heidelberg, Germany
| | - Clement Potel
- Genome Biology Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Emanuel Maus
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Frank Stein
- Proteomics Core Facility, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Bernhard Drotleff
- Metabolomics Core Facility, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Klaske Schippers
- Developmental Biology Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Michael Nickel
- Bionic Consulting Dr. Michael Nickel, 71686 Remseck am Neckar, Germany
| | - Robert Prevedel
- Developmental Biology Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Jacob M Musser
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06520, USA
| | - Mikhail M Savitski
- Genome Biology Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
- Proteomics Core Facility, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Detlev Arendt
- Developmental Biology Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
- Centre for Organismal Studies (COS), University of Heidelberg, 69120 Heidelberg, Germany
| |
Collapse
|
6
|
Erythropoietin in Glaucoma: From Mechanism to Therapy. Int J Mol Sci 2023; 24:ijms24032985. [PMID: 36769310 PMCID: PMC9917746 DOI: 10.3390/ijms24032985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 01/26/2023] [Accepted: 01/27/2023] [Indexed: 02/05/2023] Open
Abstract
Glaucoma can cause irreversible vision loss and is the second leading cause of blindness worldwide. The disease mechanism is complex and various factors have been implicated in its pathogenesis, including ischemia, excessive oxidative stress, neurotropic factor deprivation, and neuron excitotoxicity. Erythropoietin (EPO) is a hormone that induces erythropoiesis in response to hypoxia. However, studies have shown that EPO also has neuroprotective effects and may be useful for rescuing apoptotic retinal ganglion cells in glaucoma. This article explores the relationship between EPO and glaucoma and summarizes preclinical experiments that have used EPO to treat glaucoma, with an aim to provide a different perspective from the current view that glaucoma is incurable.
Collapse
|
7
|
Duarte-Silva AT, Ximenes LGR, Guimarães-Souza M, Domith I, Paes-de-Carvalho R. Chemical signaling in the developing avian retina: Focus on cyclic AMP and AKT-dependent pathways. Front Cell Dev Biol 2022; 10:1058925. [PMID: 36568967 PMCID: PMC9780464 DOI: 10.3389/fcell.2022.1058925] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 11/28/2022] [Indexed: 12/13/2022] Open
Abstract
Communication between developing progenitor cells as well as differentiated neurons and glial cells in the nervous system is made through direct cell contacts and chemical signaling mediated by different molecules. Several of these substances are synthesized and released by developing cells and play roles since early stages of Central Nervous System development. The chicken retina is a very suitable model for neurochemical studies, including the study of regulation of signaling pathways during development. Among advantages of the model are its very well-known histogenesis, the presence of most neurotransmitter systems found in the brain and the possibility to make cultures of neurons and/or glial cells where many neurochemical functions develop in a similar way than in the intact embryonic tissue. In the chicken retina, some neurotransmitters or neuromodulators as dopamine, adenosine, and others are coupled to cyclic AMP production or adenylyl cyclase inhibition since early stages of development. Other substances as vitamin C and nitric oxide are linked to the major neurotransmitter glutamate and AKT metabolism. All these different systems regulate signaling pathways, including PKA, PKG, SRC, AKT and ERK, and the activation of the transcription factor CREB. Dopamine and adenosine stimulate cAMP accumulation in the chick embryo retina through activation of D1 and A2a receptors, respectively, but the onset of dopamine stimulation is much earlier than that of adenosine. However, adenosine can inhibit adenylyl cyclase and modulate dopamine-dependent cAMP increase since early developmental stages through A1 receptors. Dopamine stimulates different PKA as well as EPAC downstream pathways both in intact tissue and in culture as the CSK-SRC pathway modulating glutamate NMDA receptors as well as vitamin C release and CREB phosphorylation. By the other hand, glutamate modulates nitric oxide production and AKT activation in cultured retinal cells and this pathway controls neuronal survival in retina. Glutamate and adenosine stimulate the release of vitamin C and this vitamin regulates the transport of glutamate, activation of NMDA receptors and AKT phosphorylation in cultured retinal cells. In the present review we will focus on these reciprocal interactions between neurotransmitters or neuromodulators and different signaling pathways during retinal development.
Collapse
Affiliation(s)
- A. T. Duarte-Silva
- Program of Neurosciences, Institute of Biology, Fluminense Federal University, Niterói, Brazil
| | - L. G. R. Ximenes
- Program of Neurosciences, Institute of Biology, Fluminense Federal University, Niterói, Brazil
| | - M. Guimarães-Souza
- Program of Neurosciences, Institute of Biology, Fluminense Federal University, Niterói, Brazil
| | - I. Domith
- Program of Neurosciences, Institute of Biology, Fluminense Federal University, Niterói, Brazil
| | - R. Paes-de-Carvalho
- Program of Neurosciences, Institute of Biology, Fluminense Federal University, Niterói, Brazil,Department of Neurobiology, Institute of Biology, Fluminense Federal University, Niterói, Brazil,*Correspondence: R. Paes-de-Carvalho,
| |
Collapse
|
8
|
Haider AA, Rex TS, Wareham LK. cGMP Signaling in the Neurovascular Unit-Implications for Retinal Ganglion Cell Survival in Glaucoma. Biomolecules 2022; 12:1671. [PMID: 36421684 PMCID: PMC9687235 DOI: 10.3390/biom12111671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 11/07/2022] [Accepted: 11/09/2022] [Indexed: 11/16/2022] Open
Abstract
Glaucoma is a progressive age-related disease of the visual system and the leading cause of irreversible blindness worldwide. Currently, intraocular pressure (IOP) is the only modifiable risk factor for the disease, but even as IOP is lowered, the pathology of the disease often progresses. Hence, effective clinical targets for the treatment of glaucoma remain elusive. Glaucoma shares comorbidities with a multitude of vascular diseases, and evidence in humans and animal models demonstrates an association between vascular dysfunction of the retina and glaucoma pathology. Integral to the survival of retinal ganglion cells (RGCs) is functional neurovascular coupling (NVC), providing RGCs with metabolic support in response to neuronal activity. NVC is mediated by cells of the neurovascular unit (NVU), which include vascular cells, glial cells, and neurons. Nitric oxide-cyclic guanosine monophosphate (NO-cGMP) signaling is a prime mediator of NVC between endothelial cells and neurons, but emerging evidence suggests that cGMP signaling is also important in the physiology of other cells of the NVU. NO-cGMP signaling has been implicated in glaucomatous neurodegeneration in humans and mice. In this review, we explore the role of cGMP signaling in the different cell types of the NVU and investigate the potential links between cGMP signaling, breakdown of neurovascular function, and glaucoma pathology.
Collapse
Affiliation(s)
| | | | - Lauren K. Wareham
- Vanderbilt Eye Institute, Department of Ophthalmology and Visual Sciences, Vanderbilt University Medical Center, Nashville, TN 37212, USA
| |
Collapse
|
9
|
Ramírez-Patiño R, Avalos-Navarro G, Figuera LE, Varela-Hernández JJ, Bautista-Herrera LA, Muñoz-Valle JF, Gallegos-Arreola MP. Influence of nitric oxide signaling mechanisms in cancer. Int J Immunopathol Pharmacol 2022; 36:3946320221135454. [PMID: 36260949 PMCID: PMC9585559 DOI: 10.1177/03946320221135454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Nitric oxide (NO) is a molecule with multiple biological functions that is involved in various pathophysiological processes such as neurotransmission and blood vessel relaxation as well as the endocrine system, immune system, growth factors, and cancer. However, in the carcinogenesis process, it has a dual behavior; at low doses, NO regulates homeostatic functions, while at high concentrations, it promotes tissue damage or acts as an agent for immune defense against microorganisms. Thus, its participation in the carcinogenic process is controversial. Cancer is a multifactorial disease that presents complex behavior. A better understanding of the molecular mechanisms associated with the initiation, promotion, and progression of neoplastic processes is required. Some hypotheses have been proposed regarding the influence of NO in activating oncogenic pathways that trigger carcinogenic processes, because NO might regulate some signaling pathways thought to promote cancer development and more aggressive tumor growth. Additionally, NO inhibits apoptosis of tumor cells, together with the deregulation of proteins that are involved in tissue homeostasis, promoting spreading to other organs and initiating metastatic processes. This paper describes the signaling pathways that are associated with cancer, and how the concentration of NO can serve a beneficial or pathological function in the initiation and promotion of neoplastic events.
Collapse
Affiliation(s)
- R Ramírez-Patiño
- Departamento de Ciencias Médicas y de la Vida, Centro Universitario de la Ciénega (CUCIÉNEGA), Universidad de Guadalajara, Ocotlán Jalisco, México
| | - G Avalos-Navarro
- Departamento de Ciencias Médicas y de la Vida, Centro Universitario de la Ciénega (CUCIÉNEGA), Universidad de Guadalajara, Ocotlán Jalisco, México
| | - LE Figuera
- División de Génetica, Centro de Investigación Biomédica de Occidente (CIBO), Instituto Mexicano del Seguro Social (IMSS), Guadalajara Jalisco, México,Doctorado en Genética Humana, Centro Universitario de Ciencias de la Salud (CUCS), Universidad de Guadalajara, Guadalajara Jalisco, México
| | - JJ Varela-Hernández
- Departamento de Ciencias Médicas y de la Vida, Centro Universitario de la Ciénega (CUCIÉNEGA), Universidad de Guadalajara, Ocotlán Jalisco, México
| | - LA Bautista-Herrera
- Departamento de Farmacobiología, Centro Universitario de Ciencias Exactas e Ingeniería (CUCEI), Universidad de Guadalajara, Guadalajara Jalisco, México
| | - JF Muñoz-Valle
- Instituto de Investigación en Ciencias Biomédicas (IICB), Centro Universitario de Ciencias de la Salud (CUCS) Universidad de Guadalajara, Guadalajara Jalisco, México
| | - MP Gallegos-Arreola
- División de Génetica, Centro de Investigación Biomédica de Occidente (CIBO), Instituto Mexicano del Seguro Social (IMSS), Guadalajara Jalisco, México,Martha Patricia Gallegos-Arreola, División de Genética CIBO, IMSS, Sierra Mojada 800, Col, Independencia, Guadalajara, Jalisco 44340, México.
| |
Collapse
|
10
|
Akaraphutiporn E, Sunaga T, Bwalya EC, Yanlin W, Carol M, Okumura M. An Insight into the Role of Apoptosis and Autophagy in Nitric Oxide-Induced Articular Chondrocyte Cell Death. Cartilage 2021; 13:826S-838S. [PMID: 33307758 PMCID: PMC8804748 DOI: 10.1177/1947603520976768] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
OBJECTIVE To investigate the role and characterize the molecular mechanisms regulating apoptosis and autophagy in nitric oxide (NO)-induced chondrocyte cell death. DESIGN Cell apoptosis and autophagy were evaluated in chondrocytes treated with sodium nitroprusside (SNP) combined with the presence or absence of interleukin-1 beta (IL-1β) and nutrient-deprived conditions. The concentration of nitrite was determined by Griess reaction. Activation of apoptosis and autophagy were determined by immunocytochemistry, Western blot, and quantitative real-time polymerase chain reaction (qPCR) analysis. Flow cytometry and MTT assay were used to assess cell viability. RESULTS Cotreatment of chondrocytes with SNP and IL-1β under nutrient-deprived condition potentially enhanced the effect of NO-induced cell death. Immunocytochemistry, Western blot, and qPCR analysis indicated that treatment of chondrocytes with SNP significantly reduced autophagic activity, autophagic flux, and multiple autophagy-related (Atg) genes expression. These findings were associated with an increase in ERK, Akt, and mTOR phosphorylation, whereas autophagy induction through mTOR/p70S6K inhibition by rapamycin significantly suppressed NO-induced cell apoptosis. Furthermore, the cleavage of poly(ADP-ribose) polymerase (PARP) and caspase-3 activation in response to apoptosis was weakly detected. These results corresponded with a significant increase in apoptosis-inducing factor (AIF) expression, suggesting the involvement of the caspase-independent pathway. CONCLUSIONS These results demonstrate that in chondrocyte cultures with cells induced into an osteoarthritis state, NO inhibits autophagy and induces chondrocyte apoptosis mainly, but not completely through the caspase-independent pathway. Our data suggest that autophagy is a protective mechanism in the pathogenesis of osteoarthritis and could be proposed as a therapeutic target for degenerative joint diseases.
Collapse
Affiliation(s)
- Ekkapol Akaraphutiporn
- Laboratory of Veterinary Surgery,
Department of Veterinary Clinical Sciences, Graduate School of Veterinary Medicine,
Hokkaido University, Sapporo, Hokkaido, Japan
| | - Takafumi Sunaga
- Laboratory of Veterinary Surgery,
Department of Veterinary Clinical Sciences, Graduate School of Veterinary Medicine,
Hokkaido University, Sapporo, Hokkaido, Japan,Takafumi Sunaga, Laboratory of Veterinary
Surgery, Department of Veterinary Clinical Sciences, Graduate School of
Veterinary Medicine, Hokkaido University, Kita 18 Nishi 9, Kita-ku, Sapporo,
Hokkaido, 060-0818, Japan.
| | - Eugene C. Bwalya
- Department of Clinical Studies, Samora
Machel School of Veterinary Medicine, University of Zambia, Lusaka, Zambia
| | - Wang Yanlin
- Laboratory of Veterinary Surgery,
Department of Veterinary Clinical Sciences, Graduate School of Veterinary Medicine,
Hokkaido University, Sapporo, Hokkaido, Japan
| | - Mwale Carol
- Laboratory of Veterinary Surgery,
Department of Veterinary Clinical Sciences, Graduate School of Veterinary Medicine,
Hokkaido University, Sapporo, Hokkaido, Japan
| | - Masahiro Okumura
- Laboratory of Veterinary Surgery,
Department of Veterinary Clinical Sciences, Graduate School of Veterinary Medicine,
Hokkaido University, Sapporo, Hokkaido, Japan
| |
Collapse
|
11
|
Ikeda Y, Nagase N, Tsuji A, Kitagishi Y, Matsuda S. Neuroprotection by dipeptidyl-peptidase-4 inhibitors and glucagon-like peptide-1 analogs via the modulation of AKT-signaling pathway in Alzheimer’s disease. World J Biol Chem 2021; 12:104-113. [PMID: 34904048 PMCID: PMC8637616 DOI: 10.4331/wjbc.v12.i6.104] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 05/21/2021] [Accepted: 11/28/2021] [Indexed: 02/06/2023] Open
Abstract
Alzheimer’s disease (AD) is the most common reason for progressive dementia in the elderly. It has been shown that disorders of the mammalian/mechanistic target of rapamycin (mTOR) signaling pathways are related to the AD. On the other hand, diabetes mellitus (DM) is a risk factor for the cognitive dysfunction. The pathogenesis of the neuronal impairment caused by diabetic hyperglycemia is intricate, which contains neuro-inflammation and/or neurodegeneration and dementia. Glucagon-like peptide-1 (GLP1) is interesting as a possible link between metabolism and brain impairment. Modulation of GLP1 activity can influence amyloid-beta peptide aggregation via the phosphoinositide-3 kinase/AKT/mTOR signaling pathway in AD. The GLP1 receptor agonists have been shown to have favorable actions on the brain such as the improvement of neurological deficit. They might also exert a beneficial effect with refining learning and memory on the cognitive impairment induced by diabetes. Recent experimental and clinical evidence indicates that dipeptidyl-peptidase-4 (DPP4) inhibitors, being currently used for DM therapy, may also be effective for AD treatment. The DPP-4 inhibitors have demonstrated neuroprotection and cognitive improvements in animal models. Although further studies for mTOR, GLP1, and DPP4 signaling pathways in humans would be intensively required, they seem to be a promising approach for innovative AD-treatments. We would like to review the characteristics of AD pathogenesis, the key roles of mTOR in AD and the preventive and/ or therapeutic suggestions of directing the mTOR signaling pathway.
Collapse
Affiliation(s)
- Yuka Ikeda
- Food Science and Nutrition, Nara Women’s University, Nara 630-8506, Japan
| | - Nozomi Nagase
- Food Science and Nutrition, Nara Women’s University, Nara 630-8506, Japan
| | - Ai Tsuji
- Food Science and Nutrition, Nara Women’s University, Nara 630-8506, Japan
| | - Yasuko Kitagishi
- Food Science and Nutrition, Nara Women’s University, Nara 630-8506, Japan
| | - Satoru Matsuda
- Food Science and Nutrition, Nara Women’s University, Nara 630-8506, Japan
| |
Collapse
|
12
|
Schiel KA. A beneficial role for elevated extracellular glutamate in Amyotrophic Lateral Sclerosis and cerebral ischemia. Bioessays 2021; 43:e2100127. [PMID: 34585427 DOI: 10.1002/bies.202100127] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 09/03/2021] [Accepted: 09/08/2021] [Indexed: 11/06/2022]
Abstract
This hypothesis proposes that increased extracellular glutamate in Amyotrophic Lateral Sclerosis (ALS) and cerebral ischemia, currently viewed as a trigger for excitotoxicity, is actually beneficial as it stimulates the utilization of glutamate as metabolic fuel. Renewed appreciation of glutamate oxidation by ischemic neurons has raised questions regarding the role of extracellular glutamate in ischemia. Is it detrimental, as suggested by excitotoxicity in early in vitro studies, or beneficial, as suggested by its oxidation in later in vivo studies? The answer may depend on the activity of N-methyl-D-aspartate (NMDA) glutamate receptors. Early in vitro procedures co-activated NMDA receptors (NMDARs) containing 2A (GluN2A) and 2B (GluN2B) subunits, an event now believed to trigger excitotoxicity; however, during in vivo ischemia D-serine and zinc molecules are released and these ensure only GluN2B receptors are stimulated. This not only prevents excitotoxicity but also initiates signaling cascades that allow ischemic neurons to import and oxidize glutamate.
Collapse
|
13
|
Socodato R, Portugal CC, Canedo T, Rodrigues A, Almeida TO, Henriques JF, Vaz SH, Magalhães J, Silva CM, Baptista FI, Alves RL, Coelho-Santos V, Silva AP, Paes-de-Carvalho R, Magalhães A, Brakebusch C, Sebastião AM, Summavielle T, Ambrósio AF, Relvas JB. Microglia Dysfunction Caused by the Loss of Rhoa Disrupts Neuronal Physiology and Leads to Neurodegeneration. Cell Rep 2021; 31:107796. [PMID: 32579923 DOI: 10.1016/j.celrep.2020.107796] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 05/29/2019] [Accepted: 06/01/2020] [Indexed: 12/28/2022] Open
Abstract
Nervous tissue homeostasis requires the regulation of microglia activity. Using conditional gene targeting in mice, we demonstrate that genetic ablation of the small GTPase Rhoa in adult microglia is sufficient to trigger spontaneous microglia activation, producing a neurological phenotype (including synapse and neuron loss, impairment of long-term potentiation [LTP], formation of β-amyloid plaques, and memory deficits). Mechanistically, loss of Rhoa in microglia triggers Src activation and Src-mediated tumor necrosis factor (TNF) production, leading to excitotoxic glutamate secretion. Inhibiting Src in microglia Rhoa-deficient mice attenuates microglia dysregulation and the ensuing neurological phenotype. We also find that the Rhoa/Src signaling pathway is disrupted in microglia of the APP/PS1 mouse model of Alzheimer disease and that low doses of Aβ oligomers trigger microglia neurotoxic polarization through the disruption of Rhoa-to-Src signaling. Overall, our results indicate that disturbing Rho GTPase signaling in microglia can directly cause neurodegeneration.
Collapse
Affiliation(s)
- Renato Socodato
- Instituto de Investigação e Inovação em Saúde and Instituto de Biologia Molecular e Celular, Porto, Portugal
| | - Camila C Portugal
- Instituto de Investigação e Inovação em Saúde and Instituto de Biologia Molecular e Celular, Porto, Portugal
| | - Teresa Canedo
- Instituto de Investigação e Inovação em Saúde and Instituto de Biologia Molecular e Celular, Porto, Portugal
| | - Artur Rodrigues
- Instituto de Investigação e Inovação em Saúde and Instituto de Biologia Molecular e Celular, Porto, Portugal
| | - Tiago O Almeida
- Instituto de Investigação e Inovação em Saúde and Instituto de Biologia Molecular e Celular, Porto, Portugal
| | - Joana F Henriques
- Instituto de Investigação e Inovação em Saúde and Instituto de Biologia Molecular e Celular, Porto, Portugal
| | - Sandra H Vaz
- Instituto de Farmacologia e Neurociências, Lisboa, Portugal; Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - João Magalhães
- Instituto de Investigação e Inovação em Saúde and Instituto de Biologia Molecular e Celular, Porto, Portugal
| | - Cátia M Silva
- Instituto de Investigação e Inovação em Saúde and Instituto de Biologia Molecular e Celular, Porto, Portugal
| | - Filipa I Baptista
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Coimbra, Portugal; Center for Innovative Biomedicine and Biotechnology (CIBB), Faculty of Medicine, Coimbra, Portugal; Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
| | - Renata L Alves
- Instituto de Investigação e Inovação em Saúde and Instituto de Biologia Molecular e Celular, Porto, Portugal
| | - Vanessa Coelho-Santos
- Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Ana Paula Silva
- Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Roberto Paes-de-Carvalho
- Department of Neurobiology and Program of Neurosciences, Institute of Biology, Fluminense Federal University, Niterói, Brazil
| | - Ana Magalhães
- Instituto de Investigação e Inovação em Saúde and Instituto de Biologia Molecular e Celular, Porto, Portugal
| | - Cord Brakebusch
- Molecular Pathology Section, BRIC, Københavns Biocenter, Copenhagen, Denmark
| | - Ana M Sebastião
- Instituto de Farmacologia e Neurociências, Lisboa, Portugal; Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Teresa Summavielle
- Instituto de Investigação e Inovação em Saúde and Instituto de Biologia Molecular e Celular, Porto, Portugal; Escola Superior de Saúde, Politécnico do Porto, Porto, Portugal
| | - António F Ambrósio
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Coimbra, Portugal; Center for Innovative Biomedicine and Biotechnology (CIBB), Faculty of Medicine, Coimbra, Portugal; Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal; Association for Innovation and Biomedical Research on Light and Image (AIBILI), Coimbra, Portugal
| | - João B Relvas
- Instituto de Investigação e Inovação em Saúde and Instituto de Biologia Molecular e Celular, Porto, Portugal; Faculdade de Medicina, Universidade do Porto, Porto, Portugal; The Discoveries Centre for Regeneration and Precision Medicine, Porto Campus, Porto, Portugal.
| |
Collapse
|
14
|
Ding Z, Ogata D, Roszik J, Qin Y, Kim SH, Tetzlaff MT, Lazar AJ, Davies MA, Ekmekcioglu S, Grimm EA. iNOS Associates With Poor Survival in Melanoma: A Role for Nitric Oxide in the PI3K-AKT Pathway Stimulation and PTEN S-Nitrosylation. Front Oncol 2021; 11:631766. [PMID: 33643925 PMCID: PMC7907506 DOI: 10.3389/fonc.2021.631766] [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] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 01/11/2021] [Indexed: 01/28/2023] Open
Abstract
We previously showed that inducible nitric oxide synthase (iNOS) protein expression in melanoma tumor cells is associated with poor patient prognosis. Here, we analyzed the association between iNOS and the oncogenic PI3K-AKT pathway. TCGA data show that iNOS and phospho-Akt Ser473 expression were associated significantly only in the subset of tumors with genetically intact PTEN. Employing a stage III melanoma TMA, we showed that iNOS protein presence is significantly associated with shorter survival only in tumors with PTEN protein expression. These findings led to our hypothesis that the iNOS product, nitric oxide (NO), suppresses the function of PTEN and stimulates PI3K-Akt activation. Melanoma cells in response to NO exposure in vitro exhibited enhanced AKT kinase activity and substrate phosphorylation, as well as attenuated PTEN phosphatase activity. Biochemical analysis showed that NO exposure resulted in a post-translationally modified S-Nitrosylation (SNO) PTEN, which was also found in cells expressing iNOS. Our findings provide evidence that NO-rich cancers may exhibit AKT activation due to post-translational inactivation of PTEN. This unique activation of oncogenic pathway under nitrosative stress may contribute to the pathogenesis of iNOS in melanoma. Significance: Our study shows that iNOS expression is associated with increased PI3K-AKT signaling and worse clinical outcomes in melanoma patients with wt (intact) PTEN. Mutated PTEN is already inactivated. We also demonstrate that NO activates the PI3K-AKT pathway by suppressing PTEN suppressor function concurrent with the formation of PTEN-SNO. This discovery provides insight into the consequences of inflammatory NO produced in human melanoma and microenvironmental cells. It suggests that NO-driven modification provides a marker of PTEN inactivation, and represents a plausible mechanism of tumor suppressor inactivation in iNOS expressing subset of cancers.
Collapse
Affiliation(s)
- Zhen Ding
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Dai Ogata
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States.,Department of Dermatologic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Jason Roszik
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States.,Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Yong Qin
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States.,Department of Pharmaceutical Sciences, School of Pharmacy, The University of Texas at El Paso, El Paso, TX, United States
| | - Sun-Hee Kim
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Michael T Tetzlaff
- Department of Dermatopathology and Oral Pathology, University of California San Francisco, San Francisco, CA, United States.,Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Alexander J Lazar
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States.,Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Michael A Davies
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Suhendan Ekmekcioglu
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Elizabeth A Grimm
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
15
|
McCarty MF, Lerner A. Nutraceutical induction and mimicry of heme oxygenase activity as a strategy for controlling excitotoxicity in brain trauma and ischemic stroke: focus on oxidative stress. Expert Rev Neurother 2020; 21:157-168. [PMID: 33287596 DOI: 10.1080/14737175.2021.1861940] [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] [Indexed: 12/26/2022]
Abstract
Introduction: Ischemic stroke and traumatic brain injury are leading causes of acute mortality, and in the longer run, major causes of significant mental and physical impairment. Most of the brain neuronal cell death in the minutes and hours following an ischemic stroke or brain trauma is mediated by the process of excitotoxicity, in which sustained elevations of extracellular glutamate, reflecting a failure of ATP-dependent mechanism which sequester glutamate in neurons and astrocytes, drive excessive activation of NMDA receptors. Areas covered: A literature search was undertaken to clarify the molecular mechanisms whereby excessive NMDA activation leads to excitotoxic neuronal death, and to determine what safe nutraceutical agents might have practical potential for rescuing at-risk neurons by intervening in these mechanisms. Expert opinion: Activation of both NADPH oxidase and neuronal nitric oxide synthase in the microenvironment of activated NMDA receptors drives production of superoxide and highly toxic peroxynitrite. This leads to excessive activation of PARP and p38 MAP kinase, mitochondrial dysfunction, and subsequent neuronal death. Heme oxygenase-1 (HO-1) induction offers protection via inhibition of NADPH oxidase and promotion of cGMP generation. Phase 2-inductive nutraceuticals can induce HO-1, and other nutraceuticals can mimic the effects of its products biliverdin and carbon monoxide.
Collapse
Affiliation(s)
| | - Aaron Lerner
- Technion Israel Institute of Technology Ruth and Bruce Rappaport Faculty of Medicine- Research, Haifa, Israel (Retired)
| |
Collapse
|
16
|
Gladulich LFH, Peixoto-Rodrigues MC, Campello-Costa P, Paes-de-Carvalho R, Cossenza M. NMDA-induced nitric oxide generation and CREB activation in central nervous system is dependent on eukaryotic elongation factor 2 kinase. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1867:118783. [DOI: 10.1016/j.bbamcr.2020.118783] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 06/11/2020] [Accepted: 06/17/2020] [Indexed: 01/28/2023]
|
17
|
Cossenza M, Socodato R, Mejía-García TA, Domith I, Portugal CC, Gladulich LFH, Duarte-Silva AT, Khatri L, Antoine S, Hofmann F, Ziff EB, Paes-de-Carvalho R. Protein synthesis inhibition promotes nitric oxide generation and activation of CGKII-dependent downstream signaling pathways in the retina. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1867:118732. [PMID: 32360667 DOI: 10.1016/j.bbamcr.2020.118732] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 04/15/2020] [Accepted: 04/26/2020] [Indexed: 01/28/2023]
Abstract
Nitric oxide is an important neuromodulator in the CNS, and its production within neurons is modulated by NMDA receptors and requires a fine-tuned availability of L-arginine. We have previously shown that globally inhibiting protein synthesis mobilizes intracellular L-arginine "pools" in retinal neurons, which concomitantly enhances neuronal nitric oxide synthase-mediated nitric oxide production. Activation of NMDA receptors also induces local inhibition of protein synthesis and L-arginine intracellular accumulation through calcium influx and stimulation of eucariotic elongation factor type 2 kinase. We hypothesized that protein synthesis inhibition might also increase intracellular L-arginine availability to induce nitric oxide-dependent activation of downstream signaling pathways. Here we show that nitric oxide produced by inhibiting protein synthesis (using cycloheximide or anisomycin) is readily coupled to AKT activation in a soluble guanylyl cyclase and cGKII-dependent manner. Knockdown of cGKII prevents cycloheximide or anisomycin-induced AKT activation and its nuclear accumulation. Moreover, in retinas from cGKII knockout mice, cycloheximide was unable to enhance AKT phosphorylation. Indeed, cycloheximide also produces an increase of ERK phosphorylation which is abrogated by a nitric oxide synthase inhibitor. In summary, we show that inhibition of protein synthesis is a previously unanticipated driving force for nitric oxide generation and activation of downstream signaling pathways including AKT and ERK in cultured retinal cells. These results may be important for the regulation of synaptic signaling and neuronal development by NMDA receptors as well as for solving conflicting data observed when using protein synthesis inhibitors for studying neuronal survival during development as well in behavior and memory studies.
Collapse
Affiliation(s)
- Marcelo Cossenza
- Program of Neurosciences, Institute of Biology, Fluminense Federal University, Niterói, RJ, Brazil; Department of Physiology and Pharmacology, Biomedical Institute, Fluminense Federal University, Niterói, RJ, Brazil.
| | - Renato Socodato
- Program of Neurosciences, Institute of Biology, Fluminense Federal University, Niterói, RJ, Brazil; Instituto de Investigação e Inovação em Saúde (i3S) and Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Porto, Portugal
| | - Telmo A Mejía-García
- Program of Neurosciences, Institute of Biology, Fluminense Federal University, Niterói, RJ, Brazil
| | - Ivan Domith
- Program of Neurosciences, Institute of Biology, Fluminense Federal University, Niterói, RJ, Brazil
| | - Camila C Portugal
- Program of Neurosciences, Institute of Biology, Fluminense Federal University, Niterói, RJ, Brazil; Instituto de Investigação e Inovação em Saúde (i3S) and Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Porto, Portugal
| | - Luis F H Gladulich
- Program of Neurosciences, Institute of Biology, Fluminense Federal University, Niterói, RJ, Brazil
| | - Aline T Duarte-Silva
- Department of Neurobiology, Institute of Biology, Fluminense Federal University, Niterói, RJ, Brazil
| | - Latika Khatri
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY, United States
| | - Shannon Antoine
- Graduate Program in Neuroscience & Physiology, New York University School of Medicine, New York, NY, United States
| | - Franz Hofmann
- Institut für Pharmakologie und Toxikologie der TU-München, Munich, Germany
| | - Edward B Ziff
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY, United States
| | - Roberto Paes-de-Carvalho
- Program of Neurosciences, Institute of Biology, Fluminense Federal University, Niterói, RJ, Brazil; Department of Neurobiology, Institute of Biology, Fluminense Federal University, Niterói, RJ, Brazil.
| |
Collapse
|
18
|
Chen B, Yang B, Zhu J, Wu J, Sha J, Sun J, Bao E, Zhang X. Hsp90 Relieves Heat Stress-Induced Damage in Mouse Kidneys: Involvement of Antiapoptotic PKM2-AKT and Autophagic HIF-1α Signaling. Int J Mol Sci 2020; 21:ijms21051646. [PMID: 32121259 PMCID: PMC7084842 DOI: 10.3390/ijms21051646] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Accepted: 02/26/2020] [Indexed: 01/01/2023] Open
Abstract
Heat stress can particularly affect the kidney because of its high rate of adenosine triphosphate consumption. Competition between apoptosis and autophagy-mediated survival always exists in damaged tissue. And Hsp90 can enhance cellular protection to resist heat stress. However, the relationship between Hsp90 and the above competition and its underlying mechanism in the kidney are unclear. The present study found that heat stress induced obvious histopathological and oxidative injury, which was connected with cellular apoptosis and autophagy in the kidney and was associated with the levels of Hsp90 expression or function. The data showed that during heat stress, Hsp90 activated the PKM2-Akt signaling pathway to exert antiapoptotic effects and induce Hsp70 expression regulated by HSF-1, stimulated autophagy-mediated survival through the HIF-1α-BNIP3/BNIP3L pathway, and finally protected the kidney from heat-stress injury. Moreover, the nuclear translocation of PKM2, (p-) Akt, HSF-1, and HIF-1α was enhanced by heat stress, but only intranuclear p-Akt and HSF-1 were specifically influenced by Hsp90, contributing to regulate the cellular ability of resisting heat-stress damage. Our study provided new insights regarding the molecular mechanism of Hsp90 in the kidney in response to heat-stress injury, possibly contributing to finding new targets for the pharmacological regulation of human or animal acute kidney injury from heat stress in future research.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Xiaohui Zhang
- Correspondence: ; Tel.: +86-258-439-5316; Fax: +86-258-439-8669
| |
Collapse
|
19
|
Ivanova VO, Balaban PM, Bal NV. Modulation of AMPA Receptors by Nitric Oxide in Nerve Cells. Int J Mol Sci 2020; 21:ijms21030981. [PMID: 32024149 PMCID: PMC7038066 DOI: 10.3390/ijms21030981] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 01/30/2020] [Accepted: 01/30/2020] [Indexed: 12/16/2022] Open
Abstract
Nitric oxide (NO) is a gaseous molecule with a large number of functions in living tissue. In the brain, NO participates in numerous intracellular mechanisms, including synaptic plasticity and cell homeostasis. NO elicits synaptic changes both through various multi-chain cascades and through direct nitrosylation of targeted proteins. Along with the N-methyl-d-aspartate (NMDA) glutamate receptors, one of the key components in synaptic functioning are α-amino-3-hydroxy-5-methyl-4-isoxazole propionate (AMPA) receptors—the main target for long-term modifications of synaptic effectivity. AMPA receptors have been shown to participate in most of the functions important for neuronal activity, including memory formation. Interactions of NO and AMPA receptors were observed in important phenomena, such as glutamatergic excitotoxicity in retinal cells, synaptic plasticity, and neuropathologies. This review focuses on existing findings that concern pathways by which NO interacts with AMPA receptors, influences properties of different subunits of AMPA receptors, and regulates the receptors’ surface expression.
Collapse
|
20
|
Regulation of Mitochondria-Associated Membranes (MAMs) by NO/sGC/PKG Participates in the Control of Hepatic Insulin Response. Cells 2019; 8:cells8111319. [PMID: 31731523 PMCID: PMC6912364 DOI: 10.3390/cells8111319] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 10/07/2019] [Accepted: 10/22/2019] [Indexed: 12/13/2022] Open
Abstract
Under physiological conditions, nitric oxide (NO) produced by the endothelial NO synthase (eNOS) upregulates hepatic insulin sensitivity. Recently, contact sites between the endoplasmic reticulum and mitochondria named mitochondria-associated membranes (MAMs) emerged as a crucial hub for insulin signaling in the liver. As mitochondria are targets of NO, we explored whether NO regulates hepatic insulin sensitivity by targeting MAMs. In Huh7 cells, primary rat hepatocytes and mouse livers, enhancing NO concentration increased MAMs, whereas inhibiting eNOS decreased them. In vitro, those effects were prevented by inhibiting protein kinase G (PKG) and mimicked by activating soluble guanylate cyclase (sGC) and PKG. In agreement with the regulation of MAMs, increasing NO concentration improved insulin signaling, both in vitro and in vivo, while eNOS inhibition disrupted this response. Finally, inhibition of insulin signaling by wortmannin did not affect the impact of NO on MAMs, while experimental MAM disruption, using either targeted silencing of cyclophilin D or the overexpression of the organelle spacer fetal and adult testis-expressed 1 (FATE-1), significantly blunted the effects of NO on both MAMs and insulin response. Therefore, under physiological conditions, NO participates to the regulation of MAM integrity through the sGC/PKG pathway and concomitantly improves hepatic insulin sensitivity. Altogether, our data suggest that the induction of MAMs participate in the impact of NO on hepatocyte insulin response.
Collapse
|
21
|
Cirne-Santos CC, Barros CDS, Nogueira CCR, Azevedo RC, Yamamoto KA, Meira GLS, de Vasconcelos ZFM, Ratcliffe NA, Teixeira VL, Schmidt-Chanasit J, Ferreira DF, Paixão ICNDP. Inhibition by Marine Algae of Chikungunya Virus Isolated From Patients in a Recent Disease Outbreak in Rio de Janeiro. Front Microbiol 2019; 10:2426. [PMID: 31708898 PMCID: PMC6821653 DOI: 10.3389/fmicb.2019.02426] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 10/08/2019] [Indexed: 01/18/2023] Open
Abstract
Chikungunya virus (CHIKV) infection is one of the most challenging re-emergent diseases caused by a virus, and with no specific antiviral treatment it has now become a major public health concern. In this investigation, 25 blood samples were collected from patients with characteristic CHIKV symptoms and submitted to a virus isolation protocol, which detected 3 CHIKV isolates. These samples were evaluated by sequencing for the characterization of the strains and any homology to viruses circulating in Brazil during a recent outbreak. These viruses were used for the development of antiviral assays. Subsequently, the inhibitory effects of seaweed extracts on CHIKV replication were studied. The marine species of algae tested were Bryothamnion triquetrum, Caulerpa racemosa, Laurencia dendroidea, Osmundaria obtusiloba, Ulva fasciata, and Kappaphycus alvarezii, all of which are found in different countries including Brazil. The results revealed high levels of CHIKV inhibition, including extracts of O. obtusiloba with inhibition values of 1.25 μg/mL and a selectivity index of 420. Viral inhibition was dependent on the time of addition of extract of O. obtusiloba to the infected cells, with the optimal inhibition occurring up to 16 h after infection. Neuron evaluations with O. obtusiloba were performed and demonstrated low toxicity, and in infected neurons we observed high inhibitory activity in a dose-dependent manner. These results indicate that the algal extracts may be promising novel candidates for the development of therapeutic agents against CHIKV infections.
Collapse
Affiliation(s)
- Claudio Cesar Cirne-Santos
- Laboratório de Virologia Molecular e Biotecnologia Marinha, Programa de Pós-graduação em Ciências e Biotecnologia, Departamento de Biologia Celular e Molecular, Instituto de Biologia, Universidade Federal Fluminense, Niterói, Brazil.,Departamento de Ensino, Curso de Farmácia na Universidade Salgado de Oliveira, Niterói, Brazil
| | - Caroline de Souza Barros
- Laboratório de Virologia Molecular e Biotecnologia Marinha, Programa de Pós-graduação em Ciências e Biotecnologia, Departamento de Biologia Celular e Molecular, Instituto de Biologia, Universidade Federal Fluminense, Niterói, Brazil
| | - Caio Cesar Richter Nogueira
- Laboratório de Virologia Molecular e Biotecnologia Marinha, Programa de Pós-graduação em Ciências e Biotecnologia, Departamento de Biologia Celular e Molecular, Instituto de Biologia, Universidade Federal Fluminense, Niterói, Brazil.,Laboratório de Produtos Naturais de Algas Marinhas (ALGAMAR), Departamento de Biologia Marinha, Instituto de Biologia, Universidade Federal Fluminense, Niterói, Brazil
| | - Renata Campos Azevedo
- Instituto de Microbiologia Paulo de Góes (IMPPG), Departamento de Virologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Kristie Aimi Yamamoto
- Instituto de Microbiologia Paulo de Góes (IMPPG), Departamento de Virologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Guilherme Louzada Silva Meira
- Instituto de Microbiologia Paulo de Góes (IMPPG), Departamento de Virologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | | | - Valéria Laneuville Teixeira
- Laboratório de Produtos Naturais de Algas Marinhas (ALGAMAR), Departamento de Biologia Marinha, Instituto de Biologia, Universidade Federal Fluminense, Niterói, Brazil.,Laboratório de Biologia e Taxonomia de Algas (LABIOTAL), Programa de Pós-graduação em Biodiversidade Neotropical, Instituto de Biociencias, Universidade Federal do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Jonas Schmidt-Chanasit
- Bernhard Nocht Institute for Tropical Medicine, WHO Collaborating Centre for Arbovirus and Haemorrhagic Fever Reference and Research, Hamburg, Germany
| | - Davis Fernandes Ferreira
- Instituto de Microbiologia Paulo de Góes (IMPPG), Departamento de Virologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Department of Molecular and Structural Biochemistry, North Carolina State University, Raleigh, NC, United States
| | - Izabel Christina Nunes de Palmer Paixão
- Laboratório de Virologia Molecular e Biotecnologia Marinha, Programa de Pós-graduação em Ciências e Biotecnologia, Departamento de Biologia Celular e Molecular, Instituto de Biologia, Universidade Federal Fluminense, Niterói, Brazil
| |
Collapse
|
22
|
Signaling pathways involved in adaptive responses to cell membrane disruption. CURRENT TOPICS IN MEMBRANES 2019; 84:99-127. [PMID: 31610867 DOI: 10.1016/bs.ctm.2019.07.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Plasma membrane disruption occurs frequently in many animal tissues. Cell membrane disruption induces not only a rapid and massive influx of Ca2+ into the cytosol but also an efflux or release of various signaling molecules, such as ATP, from the cytosol; in turn, these signaling molecules stimulate a variety of pathways in both wounded and non-wounded neighboring cells. These signals first trigger cell membrane repair responses in the wounded cell but then induce an adaptive response, which results in faster membrane repair in the event of future wounds in both wounded and non-wounded neighboring cells. In addition, signaling pathways stimulated by membrane disruption induce other adaptive responses, including cell survival, regeneration, migration, and proliferation. This chapter summarizes the role of intra- and intercellular signaling pathways in adaptive responses triggered by cell membrane disruption.
Collapse
|
23
|
Cheng Z, Zhang T, Zheng J, Ding W, Wang Y, Li Y, Zhu L, Murray M, Zhou F. Betulinic acid derivatives can protect human Müller cells from glutamate-induced oxidative stress. Exp Cell Res 2019; 383:111509. [PMID: 31344390 DOI: 10.1016/j.yexcr.2019.111509] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 07/19/2019] [Accepted: 07/22/2019] [Indexed: 02/06/2023]
Abstract
Müller cells are the predominant retinal glial cells. One of the key roles of Müller cells is in the uptake of the neurotransmitter glutamate and in its conversion to glutamine. Müller cell dysfunction due to oxidative stress elicited by high glutamate concentrations can lead to toxicity, which promote the pathogenesis of retinal diseases like diabetic retinopathy and glaucoma. This study investigated the anti-oxidant activity and mechanisms of betulinic acid (BA) and its derivatives in human Müller cells. Human MIO-M1 Müller cells were pre-treated in the presence or absence of BA, BE as well as their derivatives (named H3-H20) followed by incubation with glutamate. Cell viability was evaluated with the MTT and calcein-AM assays. Reactive oxygen species (ROS) production in MIO-M1 cells was measured using CM-H2DCFDA and flow cytometry. The activation of cellular apoptosis and necrosis was analyzed with annexin V/PI staining and flow cytometry. The modulation of signaling pathways involved in glutamate-mediated cytotoxicity and ROS production was evaluated by immunoblotting. The BA derivatives H3, H5 and H7 exhibited minimal cytotoxicity and significant anti-oxidant activity. These compounds significantly suppressed ROS production and attenuated cellular necrosis elicited by glutamate-induced oxidative stress. The protective effects of H3, H5 and H7 in MIO-M1 cells were associated with the attenuation of Akt, Erk, and JNK signaling. The BA analogues H3, H5 and H7 are protective against glutamate-induced oxidative stress in human Müller cells, and elicit their actions by modulation of the Erk, Akt and JNK signaling pathways. These agents are potential candidate molecules for the prevention or treatment of human retinal diseases.
Collapse
Affiliation(s)
- Zhengqi Cheng
- The University of Sydney, School of Pharmacy, NSW, 2006, Australia
| | - Ting Zhang
- The University of Sydney, Save Sight Institute, Sydney, NSW, 2000, Australia
| | - Jian Zheng
- Northeast Forestry University, Center for Bioactive Products/Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education, Harbin, 150040, China
| | - Weimin Ding
- Harbin University of Science and Technology, School of Chemical and Environmental Engineering, Harbin, 150080, Heilongjiang, China
| | - Yang Wang
- Northeast Forestry University, Center for Bioactive Products/Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education, Harbin, 150040, China
| | - Yue Li
- The University of Sydney, School of Pharmacy, NSW, 2006, Australia
| | - Ling Zhu
- The University of Sydney, Save Sight Institute, Sydney, NSW, 2000, Australia
| | - Michael Murray
- The University of Sydney, Discipline of Pharmacology, Faculty of Medicine and Health, NSW, 2006, Australia
| | - Fanfan Zhou
- The University of Sydney, School of Pharmacy, NSW, 2006, Australia.
| |
Collapse
|
24
|
Portugal CC, da Encarnação TG, Domith I, Dos Santos Rodrigues A, de Oliveira NA, Socodato R, Paes-de-Carvalho R. Dopamine-Induced Ascorbate Release From Retinal Neurons Involves Glutamate Release, Activation of AMPA/Kainate Receptors and Downstream Signaling Pathways. Front Neurosci 2019; 13:453. [PMID: 31143097 PMCID: PMC6521073 DOI: 10.3389/fnins.2019.00453] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 04/23/2019] [Indexed: 01/28/2023] Open
Abstract
Ascorbate, the reduced form of Vitamin C, is one of the most abundant and important low-molecular weight antioxidants in living tissues. Most animals synthesize vitamin C, but some primates, including humans, have lost this capacity due to disruption in L-gulono-gamma-lactone oxidase gene. Because of this incapacity, those animals must obtain Vitamin C from the diet. Ascorbate is highly concentrated in the central nervous system (CNS), including the retina, and plays essential roles in neuronal physiology. Ascorbate transport into cells is controlled by Sodium Vitamin C Co-Transporters (SVCTs). There are four SVCT isoforms and SVCT2 is the major isoform controlling ascorbate transport in the CNS. Regarding ascorbate release from retinal neurons, Glutamate, by activating its ionotropic receptors leads to ascorbate release via the reversion of SVCT2. Moreover, dopamine, via activation of D1 receptor/cyclic AMP/EPAC2 pathway, also induces ascorbate release via SVCT2 reversion. Because the dopaminergic and glutamatergic systems are interconnected in the CNS, we hypothesized that dopamine could regulate ascorbate release indirectly, via the glutamatergic system. Here we reveal that dopamine increases the release of D-Aspartate from retinal neurons in a way independent on calcium ions and dependent on excitatory amino acid transporters. In addition, dopamine-dependent SVCT2 reversion leading to ascorbate release occurs by activation of AMPA/Kainate receptors and downstream ERK/AKT pathways. Overall, our data reveal a dopamine-to-glutamate signaling that regulates the bioavailability of ascorbate in neuronal cells.
Collapse
Affiliation(s)
- Camila Cabral Portugal
- Instituto de Investigação e Inovação em Saúde and Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Porto, Portugal
| | - Thaísa Godinho da Encarnação
- Department of Neurobiology and Program of Neurosciences, Institute of Biology, Fluminense Federal University, Niterói, Brazil
| | - Ivan Domith
- Department of Neurobiology and Program of Neurosciences, Institute of Biology, Fluminense Federal University, Niterói, Brazil
| | - Alexandre Dos Santos Rodrigues
- Department of Neurobiology and Program of Neurosciences, Institute of Biology, Fluminense Federal University, Niterói, Brazil
| | - Nádia Almeida de Oliveira
- Department of Neurobiology and Program of Neurosciences, Institute of Biology, Fluminense Federal University, Niterói, Brazil
| | - Renato Socodato
- Instituto de Investigação e Inovação em Saúde and Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Porto, Portugal
| | - Roberto Paes-de-Carvalho
- Department of Neurobiology and Program of Neurosciences, Institute of Biology, Fluminense Federal University, Niterói, Brazil
| |
Collapse
|
25
|
de Carvalho MAJ, Chaves-Filho A, de Souza AG, de Carvalho Lima CN, de Lima KA, Rios Vasconcelos ER, Feitosa ML, Souza Oliveira JV, de Souza DAA, Macedo DS, de Souza FCF, de França Fonteles MM. Proconvulsant effects of sildenafil citrate on pilocarpine-induced seizures: Involvement of cholinergic, nitrergic and pro-oxidant mechanisms. Brain Res Bull 2019; 149:60-74. [PMID: 31004733 DOI: 10.1016/j.brainresbull.2019.04.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 03/26/2019] [Accepted: 04/09/2019] [Indexed: 12/12/2022]
Abstract
Sildenafil is a phosphodiesterase 5 inhibitor used for the treatment of erectile dysfunction and pulmonary hypertension. Proconvulsant effect is a serious adverse event associated with sildenafil use. Here, we investigated the possible proconvulsant effects of sildenafil in pilocarpine (PILO)-induced seizures model, which mimics some aspects of temporal lobe epilepsy. We also evaluated sildenafil's effects on hippocampal markers related to PILO-induced seizure, for instance, acetylcholinesterase (AChE) activity, oxidative stress and nitric oxide (NO) markers, namely nitrite, inducible NO synthase (iNOS) and neuronal NOS (nNOS). The influences of muscarinic receptors blockade on sildenafil proconvulsant effects and brain nitrite levels were also evaluated. Male mice were submitted to single or repeated (7 days) sildenafil administration (2.5, 5, 10 and 20 mg/kg). Thirty minutes later, PILO was injected and mice were further evaluated for 1 h for seizure activity. Sildenafil induced a dose- and time-progressive proconvulsant effect in PILO-induced seizures. Sildenafil also potentiated the inhibitory effect of PILO in AChE activity and induced a further increase in nitrite levels and pro-oxidative markers, mainly in the hippocampus. Repeated sildenafil treatment also increased the hippocampal expression of iNOS and nNOS isoforms, while the blockade of muscarinic receptors attenuated both sildenafil-induced proconvulsant effect and brain nitrite changes. Our data firstly demonstrated the proconvulsant effect of sildenafil in PILO-model of seizures. This effect seems to be related to an increased cholinergic-nitrergic tone and pro-oxidative brain changes. Also, our findings advert to caution in using sildenafil for patients suffering from neurological conditions that reduces seizure threshold, such as epilepsy.
Collapse
Affiliation(s)
- Michele Albuquerque Jales de Carvalho
- Neuropsychopharmacology Laboratory, Drug Research and Development Center, Faculty of Medicine, Department of Physiology and Pharmacology, Universidade Federal do Ceará, Fortaleza, CE, Brazil
| | - Adriano Chaves-Filho
- Neuropsychopharmacology Laboratory, Drug Research and Development Center, Faculty of Medicine, Department of Physiology and Pharmacology, Universidade Federal do Ceará, Fortaleza, CE, Brazil
| | - Alana Gomes de Souza
- Neuropsychopharmacology Laboratory, Drug Research and Development Center, Faculty of Medicine, Department of Physiology and Pharmacology, Universidade Federal do Ceará, Fortaleza, CE, Brazil
| | - Camila Nayane de Carvalho Lima
- Neuropsychopharmacology Laboratory, Drug Research and Development Center, Faculty of Medicine, Department of Physiology and Pharmacology, Universidade Federal do Ceará, Fortaleza, CE, Brazil
| | - Klistenes Alves de Lima
- Neuropsychopharmacology Laboratory, Drug Research and Development Center, Faculty of Medicine, Department of Physiology and Pharmacology, Universidade Federal do Ceará, Fortaleza, CE, Brazil
| | - Emiliano Ricardo Rios Vasconcelos
- Neuropsychopharmacology Laboratory, Drug Research and Development Center, Faculty of Medicine, Department of Physiology and Pharmacology, Universidade Federal do Ceará, Fortaleza, CE, Brazil
| | - Mariana Lima Feitosa
- Neuropsychopharmacology Laboratory, Drug Research and Development Center, Faculty of Medicine, Department of Physiology and Pharmacology, Universidade Federal do Ceará, Fortaleza, CE, Brazil
| | - João Victor Souza Oliveira
- Neuropsychopharmacology Laboratory, Drug Research and Development Center, Faculty of Medicine, Department of Physiology and Pharmacology, Universidade Federal do Ceará, Fortaleza, CE, Brazil
| | - Denia Alves Albuquerque de Souza
- Neuropsychopharmacology Laboratory, Drug Research and Development Center, Faculty of Medicine, Department of Physiology and Pharmacology, Universidade Federal do Ceará, Fortaleza, CE, Brazil
| | - Danielle S Macedo
- Neuropsychopharmacology Laboratory, Drug Research and Development Center, Faculty of Medicine, Department of Physiology and Pharmacology, Universidade Federal do Ceará, Fortaleza, CE, Brazil; National Institute for Translational Medicine (INCT-TM, CNPq), Ribeirão Preto, Brazil
| | - Francisca Cléa Florenço de Souza
- Neuropsychopharmacology Laboratory, Drug Research and Development Center, Faculty of Medicine, Department of Physiology and Pharmacology, Universidade Federal do Ceará, Fortaleza, CE, Brazil
| | - Marta Maria de França Fonteles
- Neuropsychopharmacology Laboratory, Drug Research and Development Center, Faculty of Medicine, Department of Physiology and Pharmacology, Universidade Federal do Ceará, Fortaleza, CE, Brazil; Pharmacy Department, Faculty of Dentistry, Nursing and Pharmacy, Universidade Federal do Ceará, Fortaleza, CE, Brazil.
| |
Collapse
|
26
|
Firat PG, Demirel EE, Demirel S, Dikci S, Turkoz Y, Ozyalın F. Increased Aqueous Humor Symmetric Dimethylarginine Level in Patients with Primary Open Angle Glaucoma. Curr Eye Res 2019; 44:619-622. [PMID: 30707048 DOI: 10.1080/02713683.2019.1578378] [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] [Indexed: 01/09/2023]
Abstract
Purpose: To evaluate the asymmetric dimethylarginine (ADMA), symmetric dimethylarginine (SDMA) and L-arginine (L-Arg) levels both in plasma and aqueous humor of primary open angle glaucoma (POAG) patients and matched controls. Patients and Methods: 25 primary open angle glaucoma patients and 42 control cases with senile cataract were included in the study. Plasma and aqueous humor ADMA, SDMA and L-Arg levels of the participants were measured by using liquid chromatography-tandem mass spectrometry. Results: A significant increase in aqueous humor SDMA level was detected in POAG patients compared with controls (p = 0.0115). No significant difference was detected in plasma and aqueous humor ADMA, L-Arg levels. Conclusion: The aqueous humor levels of SDMA are found to be associated with POAG. The result of this current study supports the role of nitric oxide pathway in glaucoma.
Collapse
Affiliation(s)
- Penpe Gül Firat
- a Department of Ophthalmology , Inonu University School of Medicine , Malatya , Turkey
| | | | - Soner Demirel
- a Department of Ophthalmology , Inonu University School of Medicine , Malatya , Turkey
| | - Seyhan Dikci
- a Department of Ophthalmology , Inonu University School of Medicine , Malatya , Turkey
| | - Yusuf Turkoz
- c Department of Biochemistry , Inonu University School of Medicine , Malatya , Turkey
| | - Fatma Ozyalın
- c Department of Biochemistry , Inonu University School of Medicine , Malatya , Turkey
| |
Collapse
|
27
|
Cirne-Santos CC, Barros CDS, Nogueira CCR, Azevedo RC, Yamamoto KA, Meira GLS, de Vasconcelos ZFM, Ratcliffe NA, Teixeira VL, Schmidt-Chanasit J, Ferreira DF, Paixão ICNDP. Inhibition by Marine Algae of Chikungunya Virus Isolated From Patients in a Recent Disease Outbreak in Rio de Janeiro. Front Microbiol 2019. [PMID: 31708898 DOI: 10.3389/fmicb201902426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/06/2023] Open
Abstract
Chikungunya virus (CHIKV) infection is one of the most challenging re-emergent diseases caused by a virus, and with no specific antiviral treatment it has now become a major public health concern. In this investigation, 25 blood samples were collected from patients with characteristic CHIKV symptoms and submitted to a virus isolation protocol, which detected 3 CHIKV isolates. These samples were evaluated by sequencing for the characterization of the strains and any homology to viruses circulating in Brazil during a recent outbreak. These viruses were used for the development of antiviral assays. Subsequently, the inhibitory effects of seaweed extracts on CHIKV replication were studied. The marine species of algae tested were Bryothamnion triquetrum, Caulerpa racemosa, Laurencia dendroidea, Osmundaria obtusiloba, Ulva fasciata, and Kappaphycus alvarezii, all of which are found in different countries including Brazil. The results revealed high levels of CHIKV inhibition, including extracts of O. obtusiloba with inhibition values of 1.25 μg/mL and a selectivity index of 420. Viral inhibition was dependent on the time of addition of extract of O. obtusiloba to the infected cells, with the optimal inhibition occurring up to 16 h after infection. Neuron evaluations with O. obtusiloba were performed and demonstrated low toxicity, and in infected neurons we observed high inhibitory activity in a dose-dependent manner. These results indicate that the algal extracts may be promising novel candidates for the development of therapeutic agents against CHIKV infections.
Collapse
Affiliation(s)
- Claudio Cesar Cirne-Santos
- Laboratório de Virologia Molecular e Biotecnologia Marinha, Programa de Pós-graduação em Ciências e Biotecnologia, Departamento de Biologia Celular e Molecular, Instituto de Biologia, Universidade Federal Fluminense, Niterói, Brazil
- Departamento de Ensino, Curso de Farmácia na Universidade Salgado de Oliveira, Niterói, Brazil
| | - Caroline de Souza Barros
- Laboratório de Virologia Molecular e Biotecnologia Marinha, Programa de Pós-graduação em Ciências e Biotecnologia, Departamento de Biologia Celular e Molecular, Instituto de Biologia, Universidade Federal Fluminense, Niterói, Brazil
| | - Caio Cesar Richter Nogueira
- Laboratório de Virologia Molecular e Biotecnologia Marinha, Programa de Pós-graduação em Ciências e Biotecnologia, Departamento de Biologia Celular e Molecular, Instituto de Biologia, Universidade Federal Fluminense, Niterói, Brazil
- Laboratório de Produtos Naturais de Algas Marinhas (ALGAMAR), Departamento de Biologia Marinha, Instituto de Biologia, Universidade Federal Fluminense, Niterói, Brazil
| | - Renata Campos Azevedo
- Instituto de Microbiologia Paulo de Góes (IMPPG), Departamento de Virologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Kristie Aimi Yamamoto
- Instituto de Microbiologia Paulo de Góes (IMPPG), Departamento de Virologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Guilherme Louzada Silva Meira
- Instituto de Microbiologia Paulo de Góes (IMPPG), Departamento de Virologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | | | - Valéria Laneuville Teixeira
- Laboratório de Produtos Naturais de Algas Marinhas (ALGAMAR), Departamento de Biologia Marinha, Instituto de Biologia, Universidade Federal Fluminense, Niterói, Brazil
- Laboratório de Biologia e Taxonomia de Algas (LABIOTAL), Programa de Pós-graduação em Biodiversidade Neotropical, Instituto de Biociencias, Universidade Federal do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Jonas Schmidt-Chanasit
- Bernhard Nocht Institute for Tropical Medicine, WHO Collaborating Centre for Arbovirus and Haemorrhagic Fever Reference and Research, Hamburg, Germany
| | - Davis Fernandes Ferreira
- Instituto de Microbiologia Paulo de Góes (IMPPG), Departamento de Virologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Department of Molecular and Structural Biochemistry, North Carolina State University, Raleigh, NC, United States
| | - Izabel Christina Nunes de Palmer Paixão
- Laboratório de Virologia Molecular e Biotecnologia Marinha, Programa de Pós-graduação em Ciências e Biotecnologia, Departamento de Biologia Celular e Molecular, Instituto de Biologia, Universidade Federal Fluminense, Niterói, Brazil
| |
Collapse
|
28
|
Brusson M, De Grandis M, Cochet S, Bigot S, Marin M, Leduc M, Guillonneau F, Mayeux P, Peyrard T, Chomienne C, Le Van Kim C, Cassinat B, Kiladjian JJ, El Nemer W. Impact of hydroxycarbamide and interferon-α on red cell adhesion and membrane protein expression in polycythemia vera. Haematologica 2018; 103:972-981. [PMID: 29599206 PMCID: PMC6058771 DOI: 10.3324/haematol.2017.182303] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 03/21/2018] [Indexed: 01/13/2023] Open
Abstract
Polycythemia vera is a chronic myeloproliferative neoplasm characterized by the JAK2V617F mutation, elevated blood cell counts and a high risk of thrombosis. Although the red cell lineage is primarily affected by JAK2V617F, the impact of mutated JAK2 on circulating red blood cells is poorly documented. Recently, we showed that in polycythemia vera, erythrocytes had abnormal expression of several proteins including Lu/BCAM adhesion molecule and proteins from the endoplasmic reticulum, mainly calreticulin and calnexin. Here we investigated the effects of hydroxycarbamide and interferon-α treatments on the expression of erythroid membrane proteins in a cohort of 53 patients. Surprisingly, while both drugs tended to normalize calreticulin expression, proteomics analysis showed that hydroxycarbamide deregulated the expression of 53 proteins in red cell ghosts, with overexpression and downregulation of 37 and 16 proteins, respectively. Within over-expressed proteins, hydroxycarbamide was found to enhance the expression of adhesion molecules such as Lu/BCAM and CD147, while interferon-α did not. In addition, we found that hydroxycarbamide increased Lu/BCAM phosphorylation and exacerbated red cell adhesion to its ligand laminin. Our study reveals unexpected adverse effects of hydroxycarbamide on red cell physiology in polycythemia vera and provides new insights into the effects of this molecule on gene regulation and protein recycling or maturation during erythroid differentiation. Furthermore, our study shows deregulation of Lu/BCAM and CD147 that are two ubiquitously expressed proteins linked to progression of solid tumors, paving the way for future studies to address the role of hydroxycarbamide in tissues other than blood cells in myeloproliferative neoplasms.
Collapse
Affiliation(s)
- Mégane Brusson
- Biologie Intégrée du Globule Rouge UMR_S1134, Inserm, Université Paris Diderot, Sorbonne Paris Cité, Université de la Réunion, Université des Antilles.,Institut National de la Transfusion Sanguine, F-75015 Paris.,Laboratoire d'Excellence GR-Ex, Paris
| | - Maria De Grandis
- Biologie Intégrée du Globule Rouge UMR_S1134, Inserm, Université Paris Diderot, Sorbonne Paris Cité, Université de la Réunion, Université des Antilles.,Institut National de la Transfusion Sanguine, F-75015 Paris.,Laboratoire d'Excellence GR-Ex, Paris
| | - Sylvie Cochet
- Biologie Intégrée du Globule Rouge UMR_S1134, Inserm, Université Paris Diderot, Sorbonne Paris Cité, Université de la Réunion, Université des Antilles.,Institut National de la Transfusion Sanguine, F-75015 Paris.,Laboratoire d'Excellence GR-Ex, Paris
| | - Sylvain Bigot
- Biologie Intégrée du Globule Rouge UMR_S1134, Inserm, Université Paris Diderot, Sorbonne Paris Cité, Université de la Réunion, Université des Antilles.,Institut National de la Transfusion Sanguine, F-75015 Paris.,Laboratoire d'Excellence GR-Ex, Paris
| | - Mickaël Marin
- Biologie Intégrée du Globule Rouge UMR_S1134, Inserm, Université Paris Diderot, Sorbonne Paris Cité, Université de la Réunion, Université des Antilles.,Institut National de la Transfusion Sanguine, F-75015 Paris.,Laboratoire d'Excellence GR-Ex, Paris
| | - Marjorie Leduc
- Plateforme de Protéomique de l'Université Paris Descartes (3P5), Institut Cochin, INSERM U1016, CNRS UMR 8104, Université Sorbonne Paris Cité, Laboratoire d'Excellence GR-Ex, Paris
| | - François Guillonneau
- Plateforme de Protéomique de l'Université Paris Descartes (3P5), Institut Cochin, INSERM U1016, CNRS UMR 8104, Université Sorbonne Paris Cité, Laboratoire d'Excellence GR-Ex, Paris
| | - Patrick Mayeux
- Plateforme de Protéomique de l'Université Paris Descartes (3P5), Institut Cochin, INSERM U1016, CNRS UMR 8104, Université Sorbonne Paris Cité, Laboratoire d'Excellence GR-Ex, Paris
| | - Thierry Peyrard
- Biologie Intégrée du Globule Rouge UMR_S1134, Inserm, Université Paris Diderot, Sorbonne Paris Cité, Université de la Réunion, Université des Antilles.,Institut National de la Transfusion Sanguine, F-75015 Paris.,Laboratoire d'Excellence GR-Ex, Paris
| | - Christine Chomienne
- Université Sorbonne Paris Cité, Université Paris Diderot, Inserm UMR-S1131, Hôpital Saint Louis, Institut Universitaire d'Hématologie, Laboratoire de Biologie Cellulaire, Paris.,AP-HP, Hôpital Saint-Louis, Laboratoire de Biologie Cellulaire, Paris
| | - Caroline Le Van Kim
- Biologie Intégrée du Globule Rouge UMR_S1134, Inserm, Université Paris Diderot, Sorbonne Paris Cité, Université de la Réunion, Université des Antilles.,Institut National de la Transfusion Sanguine, F-75015 Paris.,Laboratoire d'Excellence GR-Ex, Paris
| | - Bruno Cassinat
- AP-HP, Hôpital Saint-Louis, Laboratoire de Biologie Cellulaire, Paris
| | - Jean-Jacques Kiladjian
- Centre d'Investigations Cliniques, Hôpital Saint-Louis, Université Paris Diderot, Paris, France
| | - Wassim El Nemer
- Biologie Intégrée du Globule Rouge UMR_S1134, Inserm, Université Paris Diderot, Sorbonne Paris Cité, Université de la Réunion, Université des Antilles .,Institut National de la Transfusion Sanguine, F-75015 Paris.,Laboratoire d'Excellence GR-Ex, Paris
| |
Collapse
|
29
|
Dopamine Promotes Ascorbate Release from Retinal Neurons: Role of D1 Receptors and the Exchange Protein Directly Activated by cAMP type 2 (EPAC2). Mol Neurobiol 2018; 55:7858-7871. [DOI: 10.1007/s12035-018-0962-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 02/12/2018] [Indexed: 12/14/2022]
|
30
|
Neuroprotective potential of high-dose biotin. Med Hypotheses 2017; 109:145-149. [PMID: 29150274 DOI: 10.1016/j.mehy.2017.10.012] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 09/28/2017] [Accepted: 10/13/2017] [Indexed: 01/28/2023]
Abstract
A recent controlled trial has established that high-dose biotin supplementation - 100 mg, three times daily - has a stabilizing effect on progression of multiple sclerosis (MS). Although this effect has been attributed to an optimization of biotin's essential cofactor role in the brain, a case can be made that direct stimulation of soluble guanylate cyclase (sGC) by pharmacological concentrations of biotin plays a key role in this regard. The utility of high-dose biotin in MS might reflect an anti-inflammatory effect of cGMP on the cerebral microvasculature, as well on oligodendrocyte differentiation and on Schwann cell production of neurotrophic factors thought to have potential for managing MS. But biotin's ability to boost cGMP synthesis in the brain may have broader neuroprotective potential. In many types of neurons and neural cells, cGMP exerts neurotrophic-mimetic effects - entailing activation of the PI3K-Akt and Ras-ERK pathways - that promote neuron survival and plasticity. Hippocampal long term potentiation requires nitric oxide synthesis, which in turn promotes an activating phosphorylation of CREB via a pathway involving cGMP and protein kinase G (PKG). In Alzheimer's disease (AD), amyloid beta suppresses this mechanism by inhibiting sGC activity; agents which exert a countervailing effect by boosting cGMP levels tend to restore effective long-term potentiation in rodent models of AD. Moreover, NO/cGMP suppresses amyloid beta production within the brain by inhibiting expression of amyloid precursor protein and BACE1. In conjunction with cGMP's ability to oppose neuron apoptosis, these effects suggest that high-dose biotin might have potential for the prevention and management of AD. cGMP also promotes neurogenesis, and may lessen stroke risk by impeding atherogenesis and hypertrophic remodeling in the cerebral vasculature. The neuroprotective potential of high-dose biotin likely could be boosted by concurrent administration of brain-permeable phosphodiesterase-5 inhibitors.
Collapse
|
31
|
Díaz-Lucena D, Gutierrez-Mecinas M, Moreno B, Martínez-Sánchez JL, Pifarré P, García A. Mechanisms Involved in the Remyelinating Effect of Sildenafil. J Neuroimmune Pharmacol 2017; 13:6-23. [PMID: 28776122 DOI: 10.1007/s11481-017-9756-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 07/20/2017] [Indexed: 12/19/2022]
Abstract
Remyelination occurs in demyelinated lesions in multiple sclerosis (MS) and pharmacological treatments that enhance this process will critically impact the long term functional outcome in the disease. Sildenafil, a cyclic GMP (cGMP)-specific phosphodiesterase 5 inhibitor (PDE5-I), is an oral vasodilator drug extensively used in humans for treatment of erectile dysfunction and pulmonary arterial hypertension. PDE5 is expressed in central nervous system (CNS) neuronal and glial populations and in endothelial cells and numerous studies in rodent models of neurological disease have evidenced the neuroprotective potential of PDE5-Is. Using myelin oligodendrocyte glycoprotein (MOG)-induced experimental autoimmune encephalomyelitis (EAE) as a MS model, we previously showed that daily administration of sildenafil starting at peak disease rapidly ameliorates clinical symptoms while administration at symptoms onset prevents disease progression. These beneficial effects of the drug involved down-regulation of adaptive and innate immune responses, protection of axons and oligodendrocytes (OLs) and promotion of remyelination. In this work we have investigated mechanisms involved in the remyelinating effect of sildenafil. Using demyelinated organotypic cerebellar slice cultures we demonstrate that sildenafil stimulates remyelination by direct effects on CNS cells in a nitric oxide (NO)-cGMP-protein kinase G (PKG)-dependent manner. We also show that sildenafil treatment enhances OL maturation and induces expression of the promyelinating factor ciliary neurotrophic factor (CNTF) in spinal cord of EAE mice and in cerebellar slice cultures. Furthermore, we demonstrate that sildenafil promotes a M2 phenotype in bone marrow derived macrophages (BMDM) and increases myelin phagocytosis in these cells and in M2 microglia/macrophages in the spinal cord of EAE mice. Taken together these data indicate that promotion of OL maturation directly or through induction of growth factor expression, regulation of microglia/macrophage inflammatory phenotype and clearance of myelin debris may be relevant mechanisms involved in sildenafil enhancement of remyelination in demyelinated tissue and further support the contention that this well tolerated drug could be useful for ameliorating MS pathology.
Collapse
Affiliation(s)
- Daniela Díaz-Lucena
- Institute of Biotechnology and Biomedicine and Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, Bellaterra, 08193, Barcelona, Spain.,Institute of Neuropathology, IDIBELL-Hospital Universitari de Bellvitge, Hospitalet de Llobregat, 08097, Barcelona, Spain
| | - María Gutierrez-Mecinas
- Institute of Biotechnology and Biomedicine and Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, Bellaterra, 08193, Barcelona, Spain.,Institute of Neuroscience and Psychology, University of Glasgow, G12 8QQ, Glasgow, UK
| | - Beatriz Moreno
- Institute of Biotechnology and Biomedicine and Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, Bellaterra, 08193, Barcelona, Spain.,Basic Sciences Department, Universitat Internacional de Catalunya, Sant Cugat del Vallès, 08195, Barcelona, Spain
| | - José Lupicinio Martínez-Sánchez
- Institute of Biotechnology and Biomedicine and Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, Bellaterra, 08193, Barcelona, Spain.,Barts Cancer Institute, London, EC1M 6BQ, UK
| | - Paula Pifarré
- Institute of Biotechnology and Biomedicine and Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, Bellaterra, 08193, Barcelona, Spain. .,Centre for Genomic Regulation CRG, PRBB Building, 08003, Barcelona, Spain.
| | - Agustina García
- Institute of Biotechnology and Biomedicine and Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, Bellaterra, 08193, Barcelona, Spain.
| |
Collapse
|
32
|
Portugal CC, Socodato R, Canedo T, Silva CM, Martins T, Coreixas VSM, Loiola EC, Gess B, Röhr D, Santiago AR, Young P, Minshall RD, Paes-de-Carvalho R, Ambrósio AF, Relvas JB. Caveolin-1-mediated internalization of the vitamin C transporter SVCT2 in microglia triggers an inflammatory phenotype. Sci Signal 2017; 10:10/472/eaal2005. [PMID: 28351945 DOI: 10.1126/scisignal.aal2005] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Vitamin C is essential for the development and function of the central nervous system (CNS). The plasma membrane sodium-vitamin C cotransporter 2 (SVCT2) is the primary mediator of vitamin C uptake in neurons. SVCT2 specifically transports ascorbate, the reduced form of vitamin C, which acts as a reducing agent. We demonstrated that ascorbate uptake through SVCT2 was critical for the homeostasis of microglia, the resident myeloid cells of the CNS that are essential for proper functioning of the nervous tissue. We found that depletion of SVCT2 from the plasma membrane triggered a proinflammatory phenotype in microglia and resulted in microglia activation. Src-mediated phosphorylation of caveolin-1 on Tyr14 in microglia induced the internalization of SVCT2. Ascorbate treatment, SVCT2 overexpression, or blocking SVCT2 internalization prevented the activation of microglia. Overall, our work demonstrates the importance of the ascorbate transport system for microglial homeostasis and hints that dysregulation of ascorbate transport might play a role in neurological disorders.
Collapse
Affiliation(s)
- Camila C Portugal
- Instituto de Investigação e Inovação em Saúde and Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Rua Alfredo Allen, 208 4200-135 Porto, Portugal.
| | - Renato Socodato
- Instituto de Investigação e Inovação em Saúde and Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Rua Alfredo Allen, 208 4200-135 Porto, Portugal
| | - Teresa Canedo
- Instituto de Investigação e Inovação em Saúde and Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Rua Alfredo Allen, 208 4200-135 Porto, Portugal
| | - Cátia M Silva
- Instituto de Investigação e Inovação em Saúde and Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Rua Alfredo Allen, 208 4200-135 Porto, Portugal
| | - Tânia Martins
- Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, Health Sciences Campus, Azinhaga Santa Comba, 3000-548 Coimbra, Portugal.,Center for Neuroscience and Cell Biology, Institute for Biomedical Imaging and Life Sciences (CNC.IBILI) Consortium, University of Coimbra, Coimbra, Portugal
| | - Vivian S M Coreixas
- Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, Health Sciences Campus, Azinhaga Santa Comba, 3000-548 Coimbra, Portugal.,Department of Neurobiology and Program of Neurosciences, Institute of Biology, Fluminense Federal University, Rua Outeiro São João Batista, 24020-971 Niterói, Rio de Janeiro, Brazil
| | - Erick C Loiola
- Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, Health Sciences Campus, Azinhaga Santa Comba, 3000-548 Coimbra, Portugal.,Department of Neurobiology and Program of Neurosciences, Institute of Biology, Fluminense Federal University, Rua Outeiro São João Batista, 24020-971 Niterói, Rio de Janeiro, Brazil
| | - Burkhard Gess
- Department of Neurology, University of Muenster, 48149 Muenster, Germany.,Department of Neurology, RWTH Aachen University, Pauwelstrasse 30, 52074 Aachen, Germany
| | - Dominik Röhr
- Department of Neurology, University of Muenster, 48149 Muenster, Germany
| | - Ana R Santiago
- Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, Health Sciences Campus, Azinhaga Santa Comba, 3000-548 Coimbra, Portugal.,Center for Neuroscience and Cell Biology, Institute for Biomedical Imaging and Life Sciences (CNC.IBILI) Consortium, University of Coimbra, Coimbra, Portugal
| | - Peter Young
- Department of Neurology, University of Muenster, 48149 Muenster, Germany
| | - Richard D Minshall
- Department of Pharmacology and Center for Lung and Vascular Biology and Department of Anesthesiology, University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Roberto Paes-de-Carvalho
- Department of Neurobiology and Program of Neurosciences, Institute of Biology, Fluminense Federal University, Rua Outeiro São João Batista, 24020-971 Niterói, Rio de Janeiro, Brazil
| | - António F Ambrósio
- Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, Health Sciences Campus, Azinhaga Santa Comba, 3000-548 Coimbra, Portugal.,Center for Neuroscience and Cell Biology, Institute for Biomedical Imaging and Life Sciences (CNC.IBILI) Consortium, University of Coimbra, Coimbra, Portugal
| | - João B Relvas
- Instituto de Investigação e Inovação em Saúde and Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Rua Alfredo Allen, 208 4200-135 Porto, Portugal.
| |
Collapse
|
33
|
Olivares-González L, Martínez-Fernández de la Cámara C, Hervás D, Marín MP, Lahoz A, Millán JM, Rodrigo R. cGMP-Phosphodiesterase Inhibition Prevents Hypoxia-Induced Cell Death Activation in Porcine Retinal Explants. PLoS One 2016; 11:e0166717. [PMID: 27861632 PMCID: PMC5115799 DOI: 10.1371/journal.pone.0166717] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 11/02/2016] [Indexed: 12/11/2022] Open
Abstract
Retinal hypoxia and oxidative stress are involved in several retinal degenerations including diabetic retinopathy, glaucoma, central retinal artery occlusion, or retinopathy of prematurity. The second messenger cyclic guanosine monophosphate (cGMP) has been reported to be protective for neuronal cells under several pathological conditions including ischemia/hypoxia. The purpose of this study was to evaluate whether the accumulation of cGMP through the pharmacological inhibition of phosphodiesterase (PDE) with Zaprinast prevented retinal degeneration induced by mild hypoxia in cultures of porcine retina. Exposure to mild hypoxia (5% O2) for 24h reduced cGMP content and induced retinal degeneration by caspase dependent and independent (PARP activation) mechanisms. Hypoxia also produced a redox imbalance reducing antioxidant response (superoxide dismutase and catalase activities) and increasing superoxide free radical release. Zaprinast reduced mild hypoxia-induced cell death through inhibition of caspase-3 or PARP activation depending on the cell layer. PDE inhibition also ameliorated the effects of mild hypoxia on antioxidant response and the release of superoxide radical in the photoreceptor layer. The use of a PKG inhibitor, KT5823, suggested that cGMP-PKG pathway is involved in cell survival and antioxidant response. The inhibition of PDE, therefore, could be useful for reducing retinal degeneration under hypoxic/ischemic conditions.
Collapse
Affiliation(s)
- Lorena Olivares-González
- Grupo de Biomedicina Molecular, Celular y Genómica, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
- CIBER de Enfermedades Raras (CIBERER), Madrid, Spain
| | | | - David Hervás
- Unidad de Bioestadística, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
| | - María Pilar Marín
- Unidad de Microscopía, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
| | - Agustin Lahoz
- Unidad de Hepatología Experimental, Unidad Analítica, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
| | - José María Millán
- Grupo de Biomedicina Molecular, Celular y Genómica, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
- CIBER de Enfermedades Raras (CIBERER), Madrid, Spain
| | - Regina Rodrigo
- Grupo de Biomedicina Molecular, Celular y Genómica, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
- CIBER de Enfermedades Raras (CIBERER), Madrid, Spain
- * E-mail:
| |
Collapse
|
34
|
Li L, Zhao D, Jin Z, Zhang J, Paul C, Wang Y. Phosphodiesterase 5a Inhibition with Adenoviral Short Hairpin RNA Benefits Infarcted Heart Partially through Activation of Akt Signaling Pathway and Reduction of Inflammatory Cytokines. PLoS One 2015; 10:e0145766. [PMID: 26709517 PMCID: PMC4692549 DOI: 10.1371/journal.pone.0145766] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 12/08/2015] [Indexed: 01/28/2023] Open
Abstract
Introduction Treatment with short hairpin RNA (shRNA) interference therapy targeting phosphodiesterase 5a after myocardial infarction (MI) has been shown to mitigate post-MI heart failure. We investigated the mechanisms that underpin the beneficial effects of PDE5a inhibition through shRNA on post-MI heart failure. Methods An adenoviral vector with an shRNA sequence inserted was adopted for the inhibition of phosphodiesterase 5a (Ad-shPDE5a) in vivo and in vitro. Myocardial infarction (MI) was induced in male C57BL/6J mice by left coronary artery ligation, and immediately after that, the Ad-shPDE5a was injected intramyocardially around the MI region and border areas. Results Four weeks post-MI, the Ad-shPDE5a-treated mice showed significant mitigation of the left ventricular (LV) dilatation and dysfunction compared to control mice. Infarction size and fibrosis were also significantly reduced in Ad-shPDE5a-treated mice. Additionally, Ad-shPDE5a treatment decreased the MI-induced inflammatory cytokines interleukin (IL)-1β, IL-6, tumor necrosis factor-α, and transforming growth factor-β1, which was confirmed in vitro in Ad-shPDE5a transfected myofibroblasts cultured under oxygen glucose deprivation. Finally, Ad-shPDE5a treatment was found to activate the myocardial Akt signaling pathway in both in vivo and in vitro experiments. Conclusion These findings indicate that PDE5a inhibition by Ad-shPDE5a via the Akt signal pathway could be of significant value in the design of future therapeutics for post-MI heart failure.
Collapse
Affiliation(s)
- Longhu Li
- Department of Cardiology, the First Hospital of Qiqihaer City, Qiqihaer, China
- Department of Cardiology, the Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Dong Zhao
- Collaborative Innovation Center of Judicial Civilization, China, Key Laboratory of Evidence Science (China University of Political Science and Law), Ministry of Education, Beijing, China
| | - Zhe Jin
- Department of Cardiology, the First Hospital of Qiqihaer City, Qiqihaer, China
| | - Jian Zhang
- Department of EICU, the Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Christian Paul
- Department of Pathology and Lab Medicine, University of Cincinnati Medical Center, 231 Albert Sabin Way, Cincinnati, Ohio, 45267, United States of America
| | - Yigang Wang
- Department of Pathology and Lab Medicine, University of Cincinnati Medical Center, 231 Albert Sabin Way, Cincinnati, Ohio, 45267, United States of America
- * E-mail:
| |
Collapse
|
35
|
Wu X, Su J, Chen L, Ma B, Gu X, Zhu L. Ginkgolide B Protects Neurons from Ischemic Injury by Inhibiting the Expression of RTP801. Cell Mol Neurobiol 2015; 35:943-52. [PMID: 25869596 DOI: 10.1007/s10571-015-0189-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 03/25/2015] [Indexed: 01/28/2023]
Abstract
RTP801 (also known as REDD1), a stress-related protein, is induced by several environmental stresses such as ischemia and cigarette smoke. Although ischemia can dramatically up-regulate RTP801 expression in brain ischemia, up to now, the exact relation between RTP801 and neuronal death in ischemia is poorly understood. In the current study, using oxygen and glucose deprivation as an in vitro ischemic model in primary cultured cortical neurons, we found that the expression of RTP801 increased progressively with prolongation of ischemic duration, in which the expression of RTP801 is positively correlated with the release of lactate dehydrogenase (LDH) in neurons, and knockdown of RTP801 promoted neuronal survival in ischemia-reperfusion. It was further found that ginkgolide B (GB) could significantly increase cell viability and decrease LDH release, and at the same time reduce the levels of RTP801 mRNA and protein in neurons after ischemia and reperfusion. Moreover, GB-induced reduction in expression of RTP801 was blocked by application of LY294002, a specific inhibitor of phosphatidylinositol 3-kinase (PI3K). These results demonstrate that RTP801 could play a detrimental role on neurons in ischemia, and GB might protect neurons against ischemic injury by inhibiting RTP801 expression via PI3K pathway.
Collapse
Affiliation(s)
- Xiaomei Wu
- Department of Neurochemistry, Institute for Nautical Medicine, Nantong University, 9 Seyuan Road, Nantong, 226019, China
| | - Jianyou Su
- Medical Laboratory Center, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong, 226001, China
| | - Lei Chen
- Department of Neurochemistry, Institute for Nautical Medicine, Nantong University, 9 Seyuan Road, Nantong, 226019, China
| | - Baodong Ma
- Department of Neurochemistry, Institute for Nautical Medicine, Nantong University, 9 Seyuan Road, Nantong, 226019, China
| | - Xiaosu Gu
- Department of Neurology, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong, 226001, China
| | - Li Zhu
- Department of Neurochemistry, Institute for Nautical Medicine, Nantong University, 9 Seyuan Road, Nantong, 226019, China.
| |
Collapse
|
36
|
Socodato R, Portugal CC, Canedo T, Domith I, Oliveira NA, Paes-de-Carvalho R, Relvas JB, Cossenza M. c-Src deactivation by the polyphenol 3-O-caffeoylquinic acid abrogates reactive oxygen species-mediated glutamate release from microglia and neuronal excitotoxicity. Free Radic Biol Med 2015; 79:45-55. [PMID: 25486178 DOI: 10.1016/j.freeradbiomed.2014.11.019] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2014] [Revised: 11/10/2014] [Accepted: 11/26/2014] [Indexed: 12/13/2022]
Abstract
3-O-caffeoylquinic acid (3-CQA) is an isomer of chlorogenic acid, which has been shown to regulate lipopolysaccharide-induced tumor necrosis factor production in microglia. Whereas overactivation of microglia is associated with neuronal loss in brain diseases via reactive oxygen species (ROS) production and glutamate excitotoxicity, naïve (nonactivated) microglia are believed to generate little ROS under basal conditions, contributing to the modulation of synaptic activity and nerve tissue repair. However, the signaling pathways controlling basal ROS homeostasis in microglial cells are still poorly understood. Here we used time-lapse microscopy coupled with highly sensitive FRET biosensors (for detecting c-Src activation, ROS generation, and glutamate release) and lentivirus-mediated shRNA delivery to study the pathways involved in antioxidant-regulated ROS generation and how this associates with microglia-induced neuronal cell death. We report that 3-CQA abrogates the acquisition of an amoeboid morphology in microglia triggered by Aβ oligomers or the HIV Tat peptide. Moreover, 3-CQA deactivates c-Src tyrosine kinase and abrogates c-Src activation during proinflammatory microglia stimulation, which shuts off ROS production in these cells. Moreover, forced increment of c-Src catalytic activity by overexpressing an inducible c-Src heteromerization construct in microglia increases ROS production, abrogating the 3-CQA effects. Whereas oxidant (hydrogen peroxide) stimulation dramatically enhances glutamate release from microglia, such release is diminished by the 3-CQA inhibition of c-Src/ROS generation, significantly alleviating cell death in cultures from embryonic neurons. Overall, we provide further mechanistic insight into the modulation of ROS production in cortical microglia, indicating antioxidant-regulated c-Src function as a pathway for controlling microglia-triggered oxidative damage.
Collapse
Affiliation(s)
- Renato Socodato
- Glial Cell Biology Laboratory Instituto de Biologia Molecular e Celular, Universidade do Porto, 4150-180 Porto, Portugal.
| | - Camila C Portugal
- Glial Cell Biology Laboratory Instituto de Biologia Molecular e Celular, Universidade do Porto, 4150-180 Porto, Portugal
| | - Teresa Canedo
- Addiction Biology Laboratory, Instituto de Biologia Molecular e Celular, Universidade do Porto, 4150-180 Porto, Portugal
| | - Ivan Domith
- Neurosciences Program, Fluminense Federal University, Niterói 24210-130, Brazil
| | - Nadia A Oliveira
- Neurosciences Program, Fluminense Federal University, Niterói 24210-130, Brazil
| | - Roberto Paes-de-Carvalho
- Neurosciences Program, Fluminense Federal University, Niterói 24210-130, Brazil; Department of Neurobiology, Institute of Biology, Fluminense Federal University, Niterói 24210-130, Brazil
| | - João B Relvas
- Glial Cell Biology Laboratory Instituto de Biologia Molecular e Celular, Universidade do Porto, 4150-180 Porto, Portugal
| | - Marcelo Cossenza
- Neurosciences Program, Fluminense Federal University, Niterói 24210-130, Brazil; Department of Physiology and Pharmacology, Biomedical Institute, Fluminense Federal University, Niterói 24210-130, Brazil.
| |
Collapse
|
37
|
Socodato R, Portugal CC, Domith I, Oliveira NA, Coreixas VSM, Loiola EC, Martins T, Santiago AR, Paes-de-Carvalho R, Ambrósio AF, Relvas JB. c-Src function is necessary and sufficient for triggering microglial cell activation. Glia 2014; 63:497-511. [PMID: 25421817 DOI: 10.1002/glia.22767] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Accepted: 11/05/2014] [Indexed: 12/22/2022]
Abstract
Microglial cells are the resident macrophages of the central nervous system. Their function is essential for neuronal tissue homeostasis. After inflammatory stimuli, microglial cells become activated changing from a resting and highly ramified cell shape to an amoeboid-like morphology. These morphological changes are associated with the release of proinflammatory cytokines and glutamate, as well as with high phagocytic activity. The acquisition of such phenotype has been associated with activation of cytoplasmic tyrosine kinases, including those of the Src family (SFKs). In this study, using both in vivo and in vitro inflammation models coupled to FRET-based time-lapse microscopy, lentiviruses-mediated shRNA delivery and genetic gain-of-function experiments, we demonstrate that among SFKs c-Src function is necessary and sufficient for triggering microglia proinflammatory signature, glutamate release, microglia-induced neuronal loss, and phagocytosis. c-Src inhibition in retinal neuroinflammation experimental paradigms consisting of intravitreal injection of LPS or ischemia-reperfusion injury significantly reduced microglia activation changing their morphology to a more resting phenotype and prevented neuronal apoptosis. Our data demonstrate an essential role for c-Src in microglial cell activation.
Collapse
Affiliation(s)
- Renato Socodato
- Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Porto, Portugal; Program of Neurosciences, Fluminense Federal University, Niterói, Rio de Janeiro, Brazil; Department of Neurobiology, Institute of Biology, Fluminense Federal University, Niterói, Rio de Janeiro, Brazil; Faculty of Medicine, Centre of Ophthalmology and Vision Sciences, Institute for Biomedical Imaging and Life Sciences (IBILI), University of Coimbra, Coimbra, Portugal
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Insulin resistance in chronic kidney disease is ameliorated by spironolactone in rats and humans. Kidney Int 2014; 87:749-60. [PMID: 25337775 DOI: 10.1038/ki.2014.348] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Revised: 08/24/2014] [Accepted: 08/28/2014] [Indexed: 12/27/2022]
Abstract
In this study, we examined the association between chronic kidney disease (CKD) and insulin resistance. In a patient cohort with nondiabetic stages 2-5 CKD, estimated glomerular filtration rate (eGFR) was negatively correlated and the plasma aldosterone concentration was independently associated with the homeostasis model assessment of insulin resistance. Treatment with the mineralocorticoid receptor blocker spironolactone ameliorated insulin resistance in patients, and impaired glucose tolerance was partially reversed in fifth/sixth nephrectomized rats. In these rats, insulin-induced signal transduction was attenuated, especially in the adipose tissue. In the adipose tissue of nephrectomized rats, nuclear mineralocorticoid receptor expression, expression of the mineralocorticoid receptor target molecule SGK-1, tissue aldosterone content, and expression of the aldosterone-producing enzyme CYP11B2 increased. Mineralocorticoid receptor activation in the adipose tissue was reversed by spironolactone. In the adipose tissue of nephrectomized rats, asymmetric dimethylarginine (ADMA; an uremic substance linking uremia and insulin resistance) increased, the expression of the ADMA-degrading enzymes DDAH1 and DDAH2 decreased, and the oxidative stress increased. All of these changes were reversed by spironolactone. In mature adipocytes, aldosterone downregulated both DDAH1 and DDAH2 expression, and ADMA inhibited the insulin-induced cellular signaling. Thus, activation of mineralocorticoid receptor and resultant ADMA accumulation in adipose tissue has, in part, a relevant role in the development of insulin resistance in CKD.
Collapse
|
39
|
The nitric oxide-cGKII system relays death and survival signals during embryonic retinal development via AKT-induced CREB1 activation. Cell Death Differ 2014; 21:915-28. [PMID: 24531539 DOI: 10.1038/cdd.2014.11] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Revised: 12/21/2013] [Accepted: 01/08/2014] [Indexed: 01/28/2023] Open
Abstract
During early neurogenesis, retinal neuronal cells display a conserved differentiation program in vertebrates. Previous studies established that nitric oxide (NO) and cGMP accumulation regulate essential events in retinal physiology. Here we used pharmacological and genetic loss-of-function to investigate the effects of NO and its downstream signaling pathway in the survival of developing avian retinal neurons in vitro and in vivo. Six-day-old (E6) chick retinal cells displayed increased calcium influx and produced higher amounts of NO when compared with E8 cells. L-arginine (substrate for NO biosynthesis) and S-nitroso-N-acetyl-D,L-penicillamine (SNAP; a nitrosothiol NO donor) promoted extensive cell death in E6 retinas, whereas in E8 both substances decreased apoptosis. The effect of NO at both periods was mediated by soluble guanylyl cyclase (sGC) and cGMP-dependent kinase (cGK) activation. In addition, shRNA-mediated cGKII knockdown prevented NO-induced cell death (E6) and cell survival (E8). This, NO-induced cell death or cell survival was not correlated with an early inhibition of retinal cell proliferation. E6 cells also responded differentially from E8 neurons regarding cyclic AMP-responsive element-binding protein (CREB) activation in the retina in vivo. NO strongly decreased nuclear phospho-CREB staining in E6 but it robustly enhanced CREB phosphorylation in the nuclei of E8 neurons, an effect that was completely abrogated by cGKII shRNAs at both embryonic stages. The ability of NO in regulating CREB differentially during retinal development relied on the capacity of cGKII in decreasing (E6) or increasing (E8) nuclear AKT (V-Akt murine thymoma viral oncogene) activation. Accordingly, inhibiting AKT prevented both cGKII shRNA-mediated CREB upregulation in E6 and SNAP-induced CREB activation in E8. Furthermore, shRNA-mediated in vivo cGKII or in vitro CREB1 knockdown confirmed that NO/cGKII dualistically regulated the downstream CREB1 pathway and caspase activation in the chick retina to modulate neuronal viability. These data demonstrate that NO-mediated cGKII signaling may function to control the viability of neuronal cells during early retinal development via AKT/CREB1 activity.
Collapse
|
40
|
Cossenza M, Socodato R, Portugal CC, Domith ICL, Gladulich LFH, Encarnação TG, Calaza KC, Mendonça HR, Campello-Costa P, Paes-de-Carvalho R. Nitric oxide in the nervous system: biochemical, developmental, and neurobiological aspects. VITAMINS AND HORMONES 2014; 96:79-125. [PMID: 25189385 DOI: 10.1016/b978-0-12-800254-4.00005-2] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Nitric oxide (NO) is a very reactive molecule, and its short half-life would make it virtually invisible until its discovery. NO activates soluble guanylyl cyclase (sGC), increasing 3',5'-cyclic guanosine monophosphate levels to activate PKGs. Although NO triggers several phosphorylation cascades due to its ability to react with Fe II in heme-containing proteins such as sGC, it also promotes a selective posttranslational modification in cysteine residues by S-nitrosylation, impacting on protein function, stability, and allocation. In the central nervous system (CNS), NO synthesis usually requires a functional coupling of nitric oxide synthase I (NOS I) and proteins such as NMDA receptors or carboxyl-terminal PDZ ligand of NOS (CAPON), which is critical for specificity and triggering of selected pathways. NO also modulates CREB (cAMP-responsive element-binding protein), ERK, AKT, and Src, with important implications for nerve cell survival and differentiation. Differences in the regulation of neuronal death or survival by NO may be explained by several mechanisms involving localization of NOS isoforms, amount of NO being produced or protein sets being modulated. A number of studies show that NO regulates neurotransmitter release and different aspects of synaptic dynamics, such as differentiation of synaptic specializations, microtubule dynamics, architecture of synaptic protein organization, and modulation of synaptic efficacy. NO has also been associated with synaptogenesis or synapse elimination, and it is required for long-term synaptic modifications taking place in axons or dendrites. In spite of tremendous advances in the knowledge of NO biological effects, a full description of its role in the CNS is far from being completely elucidated.
Collapse
Affiliation(s)
- Marcelo Cossenza
- Programa de Neurociências, Instituto de Biologia, Universidade Federal Fluminense, Niterói, RJ, Brazil; Departamento de Fisiologia e Farmacologia, Instituto Biomédico, Universidade Federal Fluminense, Rio de Janeiro, Brazil
| | - Renato Socodato
- Programa de Neurociências, Instituto de Biologia, Universidade Federal Fluminense, Niterói, RJ, Brazil
| | - Camila C Portugal
- Programa de Neurociências, Instituto de Biologia, Universidade Federal Fluminense, Niterói, RJ, Brazil
| | - Ivan C L Domith
- Programa de Neurociências, Instituto de Biologia, Universidade Federal Fluminense, Niterói, RJ, Brazil
| | - Luis F H Gladulich
- Programa de Neurociências, Instituto de Biologia, Universidade Federal Fluminense, Niterói, RJ, Brazil
| | - Thaísa G Encarnação
- Programa de Neurociências, Instituto de Biologia, Universidade Federal Fluminense, Niterói, RJ, Brazil
| | - Karin C Calaza
- Programa de Neurociências, Instituto de Biologia, Universidade Federal Fluminense, Niterói, RJ, Brazil; Departamento de Neurobiologia, Instituto de Biologia, Universidade Federal Fluminense, Niterói, RJ, Brazil
| | - Henrique R Mendonça
- Programa de Neurociências, Instituto de Biologia, Universidade Federal Fluminense, Niterói, RJ, Brazil
| | - Paula Campello-Costa
- Programa de Neurociências, Instituto de Biologia, Universidade Federal Fluminense, Niterói, RJ, Brazil; Departamento de Neurobiologia, Instituto de Biologia, Universidade Federal Fluminense, Niterói, RJ, Brazil
| | - Roberto Paes-de-Carvalho
- Programa de Neurociências, Instituto de Biologia, Universidade Federal Fluminense, Niterói, RJ, Brazil; Departamento de Neurobiologia, Instituto de Biologia, Universidade Federal Fluminense, Niterói, RJ, Brazil.
| |
Collapse
|