1
|
Akintunde J, Akomolafe V, Ugbaja R, Olude A, Folayan A. Development of therapeutic supplement using roasted-cashew-nut to protect cerebral vasoconstriction injury triggered by mixture of petroleum hydrocarbons in the hypothalamus and hippocampus of rat model. Toxicol Rep 2025; 14:101943. [PMID: 39996038 PMCID: PMC11848775 DOI: 10.1016/j.toxrep.2025.101943] [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] [Received: 07/03/2024] [Revised: 01/28/2025] [Accepted: 01/31/2025] [Indexed: 02/26/2025] Open
Abstract
Petroleum-related activities have been a health global risk concern, particularly in the limbic disorders. The study aims to investigate the neuroprotection of roasted cashew nuts (RCN) on brain vasoconstriction injury induced by a mixture of petroleum hydrocarbons (MFPP). Seventy Male Wistar rats ranging 160 ± 10 g were randomized into seven groups. Group I was given distilled water. Group II was exposed to 0.2 ml MFPP. Group III, IV and V were exposed to 0.2 ml MFPP followed by treatment with 50 mg/kg atenolol, 10 % RCN and 20 % RCN, respectively. Group VI and VII were treated with 10 % RCN and 20 % RCN, respectively. The regimen period was 28 days. Cell pathological evaluation was done using hematoxylin and eosin staining and visualized under the microscope. Biochemical and molecular markers of brain vasoconstriction injury (BVI) were evaluated using spectrophotometer and RT-PCR analyzer, respectively. Student-T-test and one-way analysis of variance (ANOVA) were used to analyze the results. Sub-chronic exposure to MFPP induced BVI as evident in neuroinflammation and derangements in the histology of the hippocampus and hypothalamus coupled with momentous alterations in the neurons. Post treatment with RCN supplement remarkably modulated the effects by depleting the inflammatory mediators including HIF-1, p53 and MCP-1. Also, adenosinergic, purigenic and cholinergic of the hypothalamus and hippocampus were normalized by the supplement. It is pertinent to conclude that treatment with RCN inhibited BVI in rats via the NO-cAMP-PKA signaling pathway by reversing neuroinflammation, normalizing the purinergic and cholinergic neurotransmission in the hypothalamus and hippocampus, and stabilizing NO level coupled with brain histology improvement.
Collapse
Affiliation(s)
- J.K. Akintunde
- Molecular Toxicology and Biomedical Research Group, Department of Biochemistry, College of Biosciences, Federal University of Agriculture, Abeokuta, Nigeria
| | - V.O. Akomolafe
- Molecular Toxicology and Biomedical Research Group, Department of Biochemistry, College of Biosciences, Federal University of Agriculture, Abeokuta, Nigeria
- Department of Biochemistry, College of Natural and Applied Sciences, Chrisland University, Ajebo, Abeokuta, Nigeria
| | - R.N. Ugbaja
- Molecular Toxicology and Biomedical Research Group, Department of Biochemistry, College of Biosciences, Federal University of Agriculture, Abeokuta, Nigeria
| | - A.M. Olude
- Veterinary Anatomy (Neuroscience Unit), College of Veterinary Medicine, Federal University of Agriculture, Abeokuta, Nigeria
| | - A.D. Folayan
- Molecular Toxicology and Biomedical Research Group, Department of Biochemistry, College of Biosciences, Federal University of Agriculture, Abeokuta, Nigeria
| |
Collapse
|
2
|
Doghish AS, Elsakka EGE, Moustafa HAM, Ashraf A, Mageed SSA, Mohammed OA, Abdel-Reheim MA, Zaki MB, Elimam H, Rizk NI, Omran SA, Farag SA, Youssef DG, Abulsoud AI. Harnessing the power of miRNAs for precision diagnosis and treatment of male infertility. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:3271-3296. [PMID: 39535597 DOI: 10.1007/s00210-024-03594-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024]
Abstract
Infertility is a multifactorial reproductive system disorder, and most infertility cases occur in men. Semen testing is now thought to be the most important diagnostic test for infertile men; nonetheless, because of its limitations, the cause of infertility remains unknown for 40% of infertile men. Semen assessment's shortcomings indicate the need for improved and innovative diagnostic techniques and biomarkers worldwide. Non-coding RNAs with a length of roughly 18-22 nucleotides are called microRNAs (miRNAs). Most of our protein-coding genes are post-transcriptionally regulated by them. These molecules are unusual in bodily fluids, and aberrant variations in their expression can point to specific conditions like infertility. As a result, fresh potential biomarkers for the diagnosis and prognosis of various forms of male infertility may be represented by miRNAs. This review examined the most recent research revealing the association between different miRNAs' functions in male infertility and their expression patterns. Also, it aims to figure out the most recent strategies that could be applied for using such miRNAs as possible therapeutic targets for infertility treatment.
Collapse
Affiliation(s)
- Ahmed S Doghish
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City , 11829, Cairo, Egypt.
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, 11231, Cairo, Egypt.
| | - Elsayed G E Elsakka
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, 11231, Cairo, Egypt
| | - Hebatallah Ahmed Mohamed Moustafa
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, 11829, Cairo, Egypt
| | - Alaa Ashraf
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, 11829, Cairo, Egypt
| | - Sherif S Abdel Mageed
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, 11829, Cairo, Egypt
| | - Osama A Mohammed
- Department of Pharmacology, College of Medicine, University of Bisha, 61922, Bisha, Saudi Arabia
| | | | - Mohamed Bakr Zaki
- Department of Biochemistry, Faculty of Pharmacy, University of Sadat City, Sadat City, 32897, Menoufia, Egypt
| | - Hanan Elimam
- Department of Biochemistry, Faculty of Pharmacy, University of Sadat City, Sadat City, 32897, Menoufia, Egypt
| | - Nehal I Rizk
- Department of Biochemistry, Faculty of Pharmacy and Drug Technology, Egyptian Chinese University, Nasr City, 11786, Egypt, Cairo
| | - Sarah A Omran
- Pharmacognosy Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, 11829, Cairo, Egypt
| | - Shimaa A Farag
- School of Biotechnology, Badr University in Cairo (BUC), Badr City, 11829, Cairo, Egypt
| | - Donia G Youssef
- School of Biotechnology, Badr University in Cairo (BUC), Badr City, 11829, Cairo, Egypt
| | - Ahmed I Abulsoud
- Department of Biochemistry, Faculty of Pharmacy, Heliopolis University, El-Salam City, Cairo, 11785, Egypt
- Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, 11231, Cairo, Egypt
| |
Collapse
|
3
|
Barbagallo F, Assenza MR, Messina A. In the Brain of Phosphodiesterases: Potential Therapeutic Targets for Schizophrenia. CLINICAL PSYCHOPHARMACOLOGY AND NEUROSCIENCE : THE OFFICIAL SCIENTIFIC JOURNAL OF THE KOREAN COLLEGE OF NEUROPSYCHOPHARMACOLOGY 2025; 23:15-31. [PMID: 39820109 PMCID: PMC11747726 DOI: 10.9758/cpn.24.1229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 10/19/2024] [Accepted: 11/14/2024] [Indexed: 01/19/2025]
Abstract
Intracellular cyclic nucleotides (cyclic adenosine monophosphate and cyclic guanosine monophosphate) and downstream cellular signal transduction are regulated by phosphodiesterases (PDEs). The neuroplasticity, neurotransmitter pathways, and neuroinflammation-controlling functions of PDEs were demonstrated in numerous in vitro and animal model studies. We comprehensively reviewed the literature regarding the expression of PDEs in various brain regions. Subsequently, articles regarding schizophrenia and PDEs were examined. The pathophysiological mechanisms of schizophrenia and PDEs in preclinical and clinical investigations are briefly reviewed. Particularly for those who do not respond to conventional antipsychotics, specific PDE inhibitors may offer innovative therapeutic alternatives. Although the connection between schizophrenia and PDEs is intriguing, additional research is required. Comprehending the brain's PDE isoforms, their therapeutic potential, and any adverse effects of inhibiting them is essential for progress in this field.
Collapse
Affiliation(s)
| | - Maria Rita Assenza
- Department of Medicine and Surgery, Kore University of Enna, Enna, Italy
| | - Antonino Messina
- Department of Mental Health of Enna, Psychiatry Unity, Enna Hospital, Enna, Italy
| |
Collapse
|
4
|
Garcia-Rivas V, Soares AR, Thomas MA, Na JJ, Smith A, Picciotto MR, Mineur YS. Alcohol drinking is attenuated by PDE4 inhibition but partial microglia depletion is not sufficient to block stress-induced escalation of alcohol intake in female mice. Alcohol 2025; 122:31-42. [PMID: 39725336 DOI: 10.1016/j.alcohol.2024.12.004] [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: 10/09/2024] [Revised: 12/13/2024] [Accepted: 12/13/2024] [Indexed: 12/28/2024]
Abstract
Stress is a major contributing factor to binge drinking and development of alcohol use disorders (AUD), particularly in women. Both stress and chronic ethanol can enhance neuroinflammatory processes, which may dysregulate limbic circuits involved in ethanol reinforcement. Clinical and preclinical studies have identified sex differences in alcohol intake in response to neuroinflammatory triggers. Since both cyclic AMP (cAMP) signaling and microglial activation contribute to neuroinflammation, we explored their contribution to stress-induced ethanol drinking in mice. To this end, we first trained C57BL/6J male and female mice to volitionally drink ethanol through a modified version of the "Drinking-in-the-Dark" paradigm. We then assessed whether exposure to foot shock stress followed by repeated exposure to the previously stress-paired context might alter volitional ethanol drinking. We observed that stress exposure resulted in a delayed increase in ethanol intake, but only in female mice. The anti-inflammatory drug Apremilast, an inhibitor of phosphodiesterase type 4 (PDE4; the primary enzyme for cAMP degradation in the brain), reduced ethanol intake and decreased preference for ethanol regardless of stress exposure in females. In contrast, a partial pharmacological depletion of microglia via PLX3397 treatment did not significantly alter baseline ethanol drinking or stress-induced ethanol drinking in female mice. This study shows that female mice are more susceptible to stress-induced ethanol drinking than males, and that this occurs even after partial microglial depletion. In addition, modulation of cAMP signaling by Apremilast administration reduced ethanol drinking regardless of stress exposure, supporting the idea that it might be useful for treatment of AUD.
Collapse
Affiliation(s)
- Vernon Garcia-Rivas
- Department of Psychiatry, Yale University, 34 Park Street, 3rd Floor Research, New Haven, CT 06508, USA
| | - Alexa R Soares
- Department of Psychiatry, Yale University, 34 Park Street, 3rd Floor Research, New Haven, CT 06508, USA; Yale Interdepartmental Neuroscience Program, USA
| | - Merrilee A Thomas
- Department of Psychiatry, Yale University, 34 Park Street, 3rd Floor Research, New Haven, CT 06508, USA
| | - Jessica J Na
- Department of Psychiatry, Yale University, 34 Park Street, 3rd Floor Research, New Haven, CT 06508, USA
| | - Asia Smith
- Department of Psychiatry, Yale University, 34 Park Street, 3rd Floor Research, New Haven, CT 06508, USA; Department of Biology, Howard University, Washington DC, USA
| | - Marina R Picciotto
- Department of Psychiatry, Yale University, 34 Park Street, 3rd Floor Research, New Haven, CT 06508, USA; Yale Interdepartmental Neuroscience Program, USA.
| | - Yann S Mineur
- Department of Psychiatry, Yale University, 34 Park Street, 3rd Floor Research, New Haven, CT 06508, USA
| |
Collapse
|
5
|
Kim F, Singh P, Jo H, Xi T, Song DK, Ku SK, Choung JJ. Therapeutic effects of mirodenafil, a phosphodiesterase 5 inhibitor, on stroke models in rats. Neurotherapeutics 2025; 22:e00463. [PMID: 39393981 PMCID: PMC11742844 DOI: 10.1016/j.neurot.2024.e00463] [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: 05/10/2024] [Revised: 08/27/2024] [Accepted: 09/26/2024] [Indexed: 10/13/2024] Open
Abstract
Mirodenafil is a phosphodiesterase 5 (PDE5) inhibitor with high specificity for its target and good blood-brain barrier permeability. The drug, which is currently used for treatment of erectile dysfunction, reduces Aβ and pTau levels and improves cognitive function in mouse models of Alzheimer's disease. In the present study, we investigated the effect of mirodenafil in the transient and permanent middle cerebral artery occlusion (tMCAO and pMCAO) models of stroke in rats. Starting 24 h after cerebral artery occlusion, mirodenafil was administered subcutaneously at doses of 0.5, 1, and 2 mg/kg per day for 9 days in the tMCAO model and for 28 days in the pMCAO model. Mirodenafil significantly increased sensorimotor and cognitive recovery of tMCAO and pMCAO rats compared to saline control rats, and significantly decreased the amount of degenerative cells and cleaved caspase-3 and cleaved PARP immunoreactive cells. Effects were seen in a dose-dependent manner up to 1 mg/kg mirodenafil. The benefits of mirodenafil treatment increased with longer treatment duration, and the largest improvements over control were typically observed on the last assessment day. There was no effect of mirodenafil on infarct volume in both tMCAO and pMCAO rats. In an experiment to determine the treatment window for mirodenafil effects, a protective effect was observed when treatment was delayed 72 h after MCAO, although the most improvement was observed with shorter treatment windows. Using pMCAO and tMCAO rat models of stroke, we determined that mirodenafil improves the recovery of sensorimotor and cognitive functions after MCAO and protects cortical cells from apoptosis and degeneration. Greater benefit was observed with longer duration of treatment, and improvement was seen even when treatment was delayed.
Collapse
Affiliation(s)
- Fred Kim
- AriBio Co. Ltd., Seongnam-si 13535, Republic of Korea
| | | | - Hyunji Jo
- AriBio Co. Ltd., Seongnam-si 13535, Republic of Korea
| | - Tianyang Xi
- AriBio Co. Ltd., Seongnam-si 13535, Republic of Korea
| | | | - Sae Kwang Ku
- College of Korean Medicine, Daegu Haany University, Gyeongsan-si 38610, Republic of Korea.
| | | |
Collapse
|
6
|
Bai Z, Li P, Gao X, Zu G, Jiang A, Wu K, Mechawar N, Turecki G, Lehnert K, Snell RG, Zhou J, Hu J, Yan B, Chen L, Li W, Chen Y, Liu S, Zhu Y, You L. Exploring PDE5A upregulation in bipolar disorder: insights from single-nucleus RNA sequencing of human basal ganglia. Transl Psychiatry 2024; 14:494. [PMID: 39695100 DOI: 10.1038/s41398-024-03202-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 12/02/2024] [Accepted: 12/10/2024] [Indexed: 12/20/2024] Open
Abstract
Basal ganglia is proposed to mediate symptoms underlying bipolar disorder (BD). To understand the cell type-specific gene expression and network changes of BD basal ganglia, we performed single-nucleus RNA sequencing of 30,752 nuclei from caudate, putamen, globus pallidus, and substantia nigra of control human postmortem brain and 24,672 nuclei from BD brain. Differential expression analysis revealed major difference lying in caudate, with BD medium spiny neurons (MSNs) expressing significantly higher PDE5A, a cGMP-specific phosphodiesterase. Gene co-expression analysis (WGCNA) showed a strong correlation of caudate MSNs and gene module green, with a PDE5A-containing hub gene network. Gene regulatory network analysis (SCENIC) indicated key regulons among different cell types and basal ganglia regions, with downstream targets of key transcriptional factors showing overlapping genes such as PDEs. Upregulation of PDE5A was further validated in 7 pairs of control and BD caudate sections. Overexpression of PDE5A in primary cultured lateral ganglion eminence-derived striatal neurons led to decreased dendrite complexity, increased apoptosis, and enhanced neuronal excitability and membrane resistance. This effect could be rescued by PDE5 specific inhibitor, tadalafil. Overexpression of PDE5A in mouse striatum by stereotaxic injection caused a decreased cGMP level, an increased gene expression profile of neuroinflammation, and BD-like behaviors. Collectively, our findings provided cell type-specific gene expression profile, and indicated a causative role of PDE5A upregulation in BD basal ganglia. This study provides a single-nucleus transcriptomic profile of human control and bipolar disorder (BD) basal ganglia. Differential expression, gene co-expression, and gene regulatory network analyses collectively indicated upregulation of PDE5A in BD caudate medium spiny neurons (MSNs), which was further validated in another cohort of BD brains. The causative role of PDE5A upregulation in BD etiology is supported by the effects of PDE5A overexpression in cultured mouse MSNs in vitro and in adult mouse striatum in vivo. The former led to reduced dendrite complexity, increased apoptosis, and neuronal hyper-excitability, which could be rescued by PDE5 specific inhibitor tadalafil. The latter caused lower cGMP levels, upregulated genes associated with neuroinflammation, and BD-like behaviors.
Collapse
Affiliation(s)
- Zhixin Bai
- Department of Human Anatomy & Histoembryology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Peilong Li
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, and Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Xu Gao
- Shanghai Key Laboratory of Brain Functional Genomics (Ministry of Education), Affiliated Mental Health Center (ECNU), School of Psychology and Cognitive Science, East China Normal University, Shanghai, China
- Shanghai Changning Mental Health Center, Shanghai, China
- NYU-ECNU Institute of Brain and Cognitive Science at NYU Shanghai, Shanghai, China
| | - Gaoyu Zu
- Department of Human Anatomy & Histoembryology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Andrew Jiang
- Applied Translational Genetics Group, School of Biological Sciences, the University of Auckland, Auckland, New Zealand
| | - Keting Wu
- Department of Human Anatomy & Histoembryology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Naguib Mechawar
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Verdun, QC, Canada
- Department of Psychiatry, McGill University, Montreal, QC, Canada
| | - Gustavo Turecki
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Verdun, QC, Canada
- Department of Psychiatry, McGill University, Montreal, QC, Canada
| | - Klaus Lehnert
- Applied Translational Genetics Group, School of Biological Sciences, the University of Auckland, Auckland, New Zealand
| | - Russell G Snell
- Applied Translational Genetics Group, School of Biological Sciences, the University of Auckland, Auckland, New Zealand
| | - Jin Zhou
- Shanghai Yangpu District Mental Health Center, Mental Health Center Affiliated to Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Jia Hu
- Shanghai Yangpu District Mental Health Center, Mental Health Center Affiliated to Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Bingbing Yan
- Neo-Biotechnology Limited Company, Shanghai, China
| | - Liang Chen
- Department of Human Anatomy & Histoembryology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Wensheng Li
- Department of Human Anatomy & Histoembryology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - You Chen
- Shanghai Yangpu District Mental Health Center, Mental Health Center Affiliated to Shanghai University of Medicine & Health Sciences, Shanghai, China.
| | - Shuai Liu
- Shanghai Key Laboratory of Brain Functional Genomics (Ministry of Education), Affiliated Mental Health Center (ECNU), School of Psychology and Cognitive Science, East China Normal University, Shanghai, China.
- Shanghai Changning Mental Health Center, Shanghai, China.
- NYU-ECNU Institute of Brain and Cognitive Science at NYU Shanghai, Shanghai, China.
| | - Ying Zhu
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, and Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China.
| | - Linya You
- Department of Human Anatomy & Histoembryology, School of Basic Medical Sciences, Fudan University, Shanghai, China.
- Key Laboratory of Medical Imaging Computing and Computer Assisted Intervention of Shanghai, Fudan University, Shanghai, China.
| |
Collapse
|
7
|
Singh NK, Singh P, Varshney P, Singh A, Bhushan B. Multimodal action of phosphodiesterase 5 inhibitors against neurodegenerative disorders: An update review. J Biochem Mol Toxicol 2024; 38:e70021. [PMID: 39425458 DOI: 10.1002/jbt.70021] [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: 05/10/2024] [Revised: 09/03/2024] [Accepted: 10/02/2024] [Indexed: 10/21/2024]
Abstract
Phosphodiesterase type 5 (PDE5) is an enzyme primarily found in the smooth muscle of the corpus cavernosum and also highly expressed in the substantia nigra, cerebellum, caudate, hippocampal regions and cerebellar purkinje cells, responsible for selectively breaking down cyclic guanosine monophosphate (cGMP) into 5'-GMP and regulate intracellular cGMP levels. As a second messenger, cyclic GMP enhances signals at postsynaptic receptors and triggers downstream effector molecules, leading to changes in gene expression and neuronal responses. Additionally, cGMP signaling transduction cascade, present in the brain, is also essential for learning and memory processes. Mechanistically, PDE5 inhibitors share structural similarities with cGMP, competitively binding to PDE5 and inhibiting cGMP hydrolysis. This action enhances the effects of nitric oxide, resulting in anti-inflammatory and neuroprotective effects. Neurodegenerative disorders entail the progressive loss of neuron structure, culminating in neuronal cell death, with currently available drugs providing only limited symptomatic relief, rendering neurodegeneration considered incurable. PDE5 inhibitors have recently emerged as a potential therapeutic approach for neurodegeneration, neuroinflammation, and diseases involving cognitive impairment. This review elucidates the principal roles of 3',5'-cyclic adenosine monophosphate (cAMP) and cGMP signaling pathways in neuronal functions, believed to play pivotal roles in the pathogenesis of various neurodegenerative disorders. It provides an updated assessment of PDE5 inhibitors as disease-modifying agents for conditions such as Alzheimer's disease, Parkinson's disease, multiple sclerosis, cerebral ischemia, Huntington's disease, and neuroinflammation. The paper aims to review the current understanding of PDE5 inhibitors, which concurrently regulate both cAMP and cGMP signaling pathways, positing that they may exert complementary and synergistic effects in modifying neurodegeneration, thus presenting a novel direction in therapeutic discovery. Moreover, the review provides critical about biological functions, therapeutic potentials, limitations, challenges, and emerging applications of selective PDE5 inhibitors. This comprehensive overview aims to guide future academic and industrial endeavors in this field.
Collapse
Affiliation(s)
- Niraj Kumar Singh
- Division of Pharmacology, Institute of Pharmaceutical Research, GLA University, Chaumuhan, Mathura, India
| | - Pranjul Singh
- Division of Pharmacology, Institute of Pharmaceutical Research, GLA University, Chaumuhan, Mathura, India
| | - Prachi Varshney
- Division of Pharmacology, Institute of Pharmaceutical Research, GLA University, Chaumuhan, Mathura, India
| | - Ashini Singh
- Division of Pharmacology, Institute of Pharmaceutical Research, GLA University, Chaumuhan, Mathura, India
| | - Bharat Bhushan
- Division of Pharmacology, Institute of Pharmaceutical Research, GLA University, Chaumuhan, Mathura, India
| |
Collapse
|
8
|
Mayo P, Pascual J, Crisman E, Domínguez C, López MG, León R. Innovative pathological network-based multitarget approaches for Alzheimer's disease treatment. Med Res Rev 2024; 44:2367-2419. [PMID: 38678582 DOI: 10.1002/med.22045] [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: 11/02/2023] [Revised: 02/02/2024] [Accepted: 04/14/2024] [Indexed: 05/01/2024]
Abstract
Alzheimer's disease (AD) is the most prevalent neurodegenerative disease and is a major health threat globally. Its prevalence is forecasted to exponentially increase during the next 30 years due to the global aging population. Currently, approved drugs are merely symptomatic, being ineffective in delaying or blocking the relentless disease advance. Intensive AD research describes this disease as a highly complex multifactorial disease. Disclosure of novel pathological pathways and their interconnections has had a major impact on medicinal chemistry drug development for AD over the last two decades. The complex network of pathological events involved in the onset of the disease has prompted the development of multitarget drugs. These chemical entities combine pharmacological activities toward two or more drug targets of interest. These multitarget-directed ligands are proposed to modify different nodes in the pathological network aiming to delay or even stop disease progression. Here, we review the multitarget drug development strategy for AD during the last decade.
Collapse
Affiliation(s)
- Paloma Mayo
- Departamento de desarrollo preclínico, Fundación Teófilo Hernando, Las Rozas, Madrid, Spain
- Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
- Instituto de Química Médica, Consejo Superior de Investigaciones Científicas (IQM-CSIC), Madrid, Spain
| | - Jorge Pascual
- Departamento de desarrollo preclínico, Fundación Teófilo Hernando, Las Rozas, Madrid, Spain
- Instituto de Química Médica, Consejo Superior de Investigaciones Científicas (IQM-CSIC), Madrid, Spain
| | - Enrique Crisman
- Instituto de Química Médica, Consejo Superior de Investigaciones Científicas (IQM-CSIC), Madrid, Spain
| | - Cristina Domínguez
- Instituto de Química Médica, Consejo Superior de Investigaciones Científicas (IQM-CSIC), Madrid, Spain
| | - Manuela G López
- Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - Rafael León
- Instituto de Química Médica, Consejo Superior de Investigaciones Científicas (IQM-CSIC), Madrid, Spain
| |
Collapse
|
9
|
Crescioli C, Paronetto MP. The Emerging Role of Phosphodiesterase 5 Inhibition in Neurological Disorders: The State of the Art. Cells 2024; 13:1720. [PMID: 39451238 PMCID: PMC11506759 DOI: 10.3390/cells13201720] [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: 08/03/2024] [Revised: 10/13/2024] [Accepted: 10/15/2024] [Indexed: 10/26/2024] Open
Abstract
Growing evidence suggests that neuroinflammation is not just a consequence of neurodegeneration in pathologies such as Alzheimer's disease, Parkinson's disease, Huntington's disease or Amyotrophic lateral sclerosis, but it is rather a determinant factor, which plays a pivotal role in the onset and progression of these disorders. Neuroinflammation can affect cells and processes in the central nervous system (CNS) as well as immune cells, and might precede protein aggregation, which is a hallmark of the neurodegenerative process. Standard treatment methods are far from being able to counteract inflammation and delay neurodegeneration. Remarkably, phosphodiesterase 5 inhibitors (PDE5is), which represent potent vasoactive drugs used as a first-line treatment for erectile dysfunction (ED), display important anti-inflammatory effects through cyclic guanosine monophosphate (cGMP) level stabilization. Since PDE5 hydrolyzes cGMP, several studies positioned PDE5 as a therapeutic target, and more specifically, PDE5is as potential alternative strategies for the treatment of a variety of neurological disorders. Indeed, PDE5is can limit neuroinflammation and enhance synaptic plasticity, with beneficial effects on cognitive function and memory. The aim of this review is to provide an overview of some of the main processes underlying neuroinflammation and neurodegeneration which may be potential targets for PDE5is, focusing on sildenafil, the most extensively studied. Current strategies using PDEis for the treatment of neurodegenerative diseases will be summarized.
Collapse
Affiliation(s)
- Clara Crescioli
- Department of Movement, Human and Health Sciences, University of Rome Foro Italico, Piazza Lauro de Bosis, 6, 00135 Rome, Italy
| | - Maria Paola Paronetto
- Department of Movement, Human and Health Sciences, University of Rome Foro Italico, Piazza Lauro de Bosis, 6, 00135 Rome, Italy
- Laboratory of Molecular and Cellular Neurobiology, Fondazione Santa Lucia IRCCS, Via del Fosso di Fiorano, 64, 00143 Rome, Italy
| |
Collapse
|
10
|
Kumar A, Kim F, Song DK, Choung JJ. Polypharmacological Potential of Phosphodiesterase 5 Inhibitors for the Treatment of Neurocognitive Disorders. Aging Dis 2024; 15:2008-2014. [PMID: 38270120 PMCID: PMC11346399 DOI: 10.14336/ad.2023.1129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 11/29/2023] [Indexed: 01/26/2024] Open
Abstract
The prevalence of neurocognitive disorders (NCD) increases every year as the population continues to age, leading to significant global health concerns. Overcoming this challenge requires identifying biomarkers, risk factors, and effective therapeutic interventions that might provide meaningful clinical benefits. For Alzheimer's disease (AD), one of the most studied NCD, approved drugs include acetylcholinesterase inhibitors (rivastigmine, donepezil, and galantamine), an NMDA receptor antagonist (memantine), and anti-amyloid monoclonal antibodies (aducanumab and lecanemab). These drugs offer limited relief, targeting singular pathological processes of the AD. Given the multifactorial nature of the NCDs, a poly-pharmacological strategy may lead to improved outcomes compared to the current standard of care. In this regard, phosphodiesterase 5 (PDE5) inhibitors emerged as promising drug candidates for the treatment of neurocognitive disorders. These inhibitors increase cGMP levels and CREB signaling, thus enhancing learning, memory and neuroprotection, while reducing Aβ deposition, tau phosphorylation, oxidative stress, and neuroinflammation. In the present article, we evaluate the therapeutic potential of different PDE5 inhibitors to outline their multifaceted impact in the NCDs.
Collapse
|
11
|
Yu YH, Kim GW, Lee YR, Park DK, Song B, Kim DS. Effects of Sildenafil on Cognitive Function Recovery and Neuronal Cell Death Protection after Transient Global Cerebral Ischemia in Gerbils. Biomedicines 2024; 12:2077. [PMID: 39335590 PMCID: PMC11429064 DOI: 10.3390/biomedicines12092077] [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: 08/12/2024] [Revised: 08/30/2024] [Accepted: 09/04/2024] [Indexed: 09/30/2024] Open
Abstract
Cerebral ischemic stroke is a major cause of death worldwide due to brain cell death resulting from ischemia-reperfusion injury. However, effective treatment approaches for patients with ischemic stroke are still lacking in clinical practice. This study investigated the potential neuroprotective effects of sildenafil, a phosphodiesterase-5 inhibitor, in a gerbil model of global brain ischemia. We investigated the effects of sildenafil on the expression of glial fibrillary acidic protein and aquaporin-4, which are markers related to astrocyte activation and water homeostasis, respectively. Immunofluorescence analysis showed that the number of cells co-expressing these markers, which was elevated in the ischemia-induced group, was significantly reduced in the sildenafil-treated groups. This suggests that sildenafil may have a potential mitigating effect on astrocyte activation induced by ischemia. Additionally, we performed various behavioral tests, including the open-field test, novel object recognition, Barnes maze, Y-maze, and passive avoidance tests, to evaluate sildenafil's effect on cognitive function impaired by ischemia. Overall, the results suggest that sildenafil may serve as a neuroprotective agent, potentially alleviating delayed neuronal cell death and improving cognitive function impaired by ischemia.
Collapse
Affiliation(s)
- Yeon Hee Yu
- Department of Anatomy, College of Medicine, Soonchunhyang University, Cheonan 31151, Republic of Korea
| | - Gun Woo Kim
- Research Supporting Center for Medical Science, College of Medicine, Dong-A, Busan 49201, Republic of Korea
| | - Yu Ran Lee
- Department of Anatomy, College of Medicine, Soonchunhyang University, Cheonan 31151, Republic of Korea
| | - Dae-Kyoon Park
- Department of Anatomy, College of Medicine, Soonchunhyang University, Cheonan 31151, Republic of Korea
| | - Beomjong Song
- Department of Anatomy, College of Medicine, Soonchunhyang University, Cheonan 31151, Republic of Korea
| | - Duk-Soo Kim
- Department of Anatomy, College of Medicine, Soonchunhyang University, Cheonan 31151, Republic of Korea
| |
Collapse
|
12
|
Duarte-Silva E, Oriá AC, Mendonça IP, Paiva IHR, Leuthier Dos Santos K, Sales AJ, de Souza JRB, Maes M, Meuth SG, Peixoto CA. The Antidepressant- and Anxiolytic-Like Effects of the Phosphodiesterase Type-5 Inhibitor Tadalafil are Associated with the Modulation of the Gut-Brain Axis During CNS Autoimmunity. J Neuroimmune Pharmacol 2024; 19:45. [PMID: 39158758 DOI: 10.1007/s11481-024-10148-4] [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: 10/10/2023] [Accepted: 08/07/2024] [Indexed: 08/20/2024]
Abstract
Multiple Sclerosis (MS) is a debilitating disease that severely affects the central nervous system (CNS). Apart from neurological symptoms, it is also characterized by neuropsychiatric comorbidities, such as anxiety and depression. Phosphodiesterase-5 inhibitors (PDE5Is) such as Sildenafil and Tadalafil have been shown to possess antidepressant-like effects, but the mechanisms underpinning such effects are not fully characterized. To address this question, we used the EAE model of MS, behavioral tests, immunofluorescence, immunohistochemistry, western blot, and 16 S rRNA sequencing. Here, we showed that depressive-like behavior in Experimental Autoimmune Encephalomyelitis (EAE) mice is due to neuroinflammation, reduced synaptic plasticity, dysfunction in glutamatergic neurotransmission, glucocorticoid receptor (GR) resistance, increased blood-brain barrier (BBB) permeability, and immune cell infiltration to the CNS, as well as inflammation, increased intestinal permeability, and immune cell infiltration in the distal colon. Furthermore, 16 S rRNA sequencing revealed that behavioral dysfunction in EAE mice is associated with changes in the gut microbiota, such as an increased abundance of Firmicutes and Saccharibacteria and a reduction in Proteobacteria, Parabacteroides, and Desulfovibrio. Moreover, we detected an increased abundance of Erysipelotrichaceae and Desulfovibrionaceae and a reduced abundance of Lactobacillus johnsonii. Surprisingly, we showed that Tadalafil likely exerts antidepressant-like effects by targeting all aforementioned disease aspects. In conclusion, our work demonstrated that anxiety- and depressive-like behavior in EAE is associated with a plethora of neuroimmune and gut microbiota-mediated mechanisms and that Tadalafil exerts antidepressant-like effects probably by targeting these mechanisms. Harnessing the knowledge of these mechanisms of action of Tadalafil is important to pave the way for future clinical trials with depressed patients.
Collapse
Affiliation(s)
- Eduardo Duarte-Silva
- Laboratory of Ultrastructure, Aggeu Magalhães Institute (IAM), Recife, PE, Brazil.
- Postgraduate Program in Biosciences and Biotechnology for Health (PPGBBS), Oswaldo Cruz Foundation (FIOCRUZ-PE)/ Aggeu Magalhães Institute (IAM), Recife, PE, Brazil.
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Recife, PE, Brazil.
- Department of Neurology, University Hospital Düsseldorf, 40255, Düsseldorf, Germany.
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil.
- Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
| | | | - Ingrid Prata Mendonça
- Laboratory of Ultrastructure, Aggeu Magalhães Institute (IAM), Recife, PE, Brazil
- Postgraduate Program in Biological Sciences (PPGCB), Federal University of Pernambuco (UFPE), Recife, PE, Brazil
| | - Igor Henrique Rodrigues Paiva
- Laboratory of Ultrastructure, Aggeu Magalhães Institute (IAM), Recife, PE, Brazil
- Postgraduate Program in Biological Sciences (PPGCB), Federal University of Pernambuco (UFPE), Recife, PE, Brazil
| | | | - Amanda Juliana Sales
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | | | - Michael Maes
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
- Cognitive Impairment and Dementia Research Unit, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
- Department of Psychiatry, Medical University of Plovdiv, Plovdiv, 4002, Bulgaria
- Research Institute, Medical University of Plovdiv, Plovdiv, 4002, Bulgaria
- IMPACT, the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Barwon Health, Geelong, VIC, Australia
- Mental Health Center, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Sven Guenther Meuth
- Department of Neurology, University Hospital Düsseldorf, 40255, Düsseldorf, Germany
| | - Christina Alves Peixoto
- Laboratory of Ultrastructure, Aggeu Magalhães Institute (IAM), Recife, PE, Brazil.
- Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil.
| |
Collapse
|
13
|
AlRuwaili R, Al-Kuraishy HM, Alruwaili M, Khalifa AK, Alexiou A, Papadakis M, Saad HM, Batiha GES. The potential therapeutic effect of phosphodiesterase 5 inhibitors in the acute ischemic stroke (AIS). Mol Cell Biochem 2024; 479:1267-1278. [PMID: 37395897 PMCID: PMC11116240 DOI: 10.1007/s11010-023-04793-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 06/16/2023] [Indexed: 07/04/2023]
Abstract
Acute ischemic stroke (AIS) is a focal neurological disorder that accounts for 85% of all stroke types, due to occlusion of cerebral arteries by thrombosis and emboli. AIS is also developed due to cerebral hemodynamic abnormality. AIS is associated with the development of neuroinflammation which increases the severity of AIS. Phosphodiesterase enzyme (PDEs) inhibitors have neuro-restorative and neuroprotective effects against the development of AIS through modulation of the cerebral cyclic adenosine monophosphate (cAMP)/cyclic guanosine monophosphate (cGMP)/nitric oxide (NO) pathway. PDE5 inhibitors through mitigation of neuroinflammation may decrease the risk of long-term AIS-induced complications. PDE5 inhibitors may affect the hemodynamic properties and coagulation pathway which are associated with thrombotic complications in AIS. PDE5 inhibitors reduce activation of the pro-coagulant pathway and improve the microcirculatory level in patients with hemodynamic disturbances in AIS. PDE5 inhibitors mainly tadalafil and sildenafil improve clinical outcomes in AIS patients through the regulation of cerebral perfusion and cerebral blood flow (CBF). PDE5 inhibitors reduced thrombomodulin, P-selectin, and tissue plasminogen activator. Herein, PDE5 inhibitors may reduce activation of the pro-coagulant pathway and improve the microcirculatory level in patients with hemodynamic disturbances in AIS. In conclusion, PDE5 inhibitors may have potential roles in the management of AIS through modulation of CBF, cAMP/cGMP/NO pathway, neuroinflammation, and inflammatory signaling pathways. Preclinical and clinical studies are recommended in this regard.
Collapse
Affiliation(s)
- Raed AlRuwaili
- Department of Internal Medicine, College of Medicine, Jouf University, Sakaka, Saudi Arabia
| | - Hayder M Al-Kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, ALmustansiriyia University, Baghdad, Iraq
| | - Mubarak Alruwaili
- Department of Internal Medicine, College of Medicine, Jouf University, Sakaka, Saudi Arabia
| | - Amira Karam Khalifa
- Department of Medical Pharmacology, Kasr El-Ainy School of Medicine, Cairo University, El Manial, Cairo, 11562, Egypt
- Lecturer of Medical Pharmacology, Nahda Faculty of Medicine, Beni Suef, Egypt
| | - Athanasios Alexiou
- Department of Science and Engineering, Novel Global Community Educational Foundation, Hebersham, NSW, 2770, Australia
- AFNP Med, 1030, Vienna, Austria
| | - Marios Papadakis
- Department of Surgery II, University Hospital Witten-Herdecke, University of Witten-Herdecke, Heusnerstrasse 40, 42283, Wuppertal, Germany.
| | - Hebatallah M Saad
- Department of Pathology, Faculty of Veterinary Medicine, Matrouh University, Marsa Matrouh, 51744, Egypt
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, 22511, AlBeheira, Egypt
| |
Collapse
|
14
|
Han R, Gaurav A, Mai CW, Gautam V, Gabriel Akyirem A. Phosphodiesterase Inhibitors of Natural Origin. THE NATURAL PRODUCTS JOURNAL 2024; 14. [DOI: 10.2174/0122103155251390230927064442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 08/03/2023] [Accepted: 08/21/2023] [Indexed: 01/09/2025]
Abstract
Abstract:
Phosphodiesterases (PDEs) function to hydrolyze intracellular cyclic adenosine monophosphate
(cAMP) and cyclic guanosine monophosphate (cGMP), regulating a variety of intracellular
signal transduction and physiological activities. PDEs can be divided into 11 families
(PDE1~11) and the diversity and complex expression of PDE family genes suggest that different
subtypes may have different mechanisms. PDEs are involved in various disease pathologies such
as inflammation, asthma, depression, and erectile dysfunction and are thus targets of interest for
several drug discovery campaigns. Natural products have always been an important source of bioactive
compounds for drug discovery, over the years several natural compounds have shown potential
as inhibitors of PDEs. In this article, phosphodiesterase inhibitors of natural origin have been
reviewed with emphasis on their chemistry and biological activities.
Collapse
Affiliation(s)
- Rui Han
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, UCSI University, Taman Connaught,
Cheras Kuala Lumpur, 56000, Malaysia
| | - Anand Gaurav
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, UCSI University, Taman Connaught,
Cheras Kuala Lumpur, 56000, Malaysia
- Department of Pharmaceutical Sciences, School of Health Sciences and
Technology, UPES, Dehradun, 248007, Uttarakhand, India
- Faculty of Health Sciences, Villa College, QI Campus,
Rahdhebai Hingun, Male', 20373, Republic of Maldives
| | - Chun-Wai Mai
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, UCSI University, Taman Connaught,
Cheras Kuala Lumpur, 56000, Malaysia
| | - Vertika Gautam
- Institute of Pharmaceutical Research, GLA University,
Mathura, 281406, Uttar Pradesh, India
| | - Akowuah Gabriel Akyirem
- School of Pharmacy, Monash University Malaysia Jalan Lagoon Selatan,
47500 Bandar Sunway, Selangor Darul Ehsan, Malaysia
| |
Collapse
|
15
|
Delhaye S, Jarjat M, Boulksibat A, Sanchez C, Tempio A, Turtoi A, Giorgi M, Lacas-Gervais S, Baj G, Rovere C, Trezza V, Pellegrini M, Maurin T, Lalli E, Bardoni B. Defects in AMPAR trafficking and microglia activation underlie socio-cognitive deficits associated to decreased expression of phosphodiesterase 2 a. Neurobiol Dis 2024; 191:106393. [PMID: 38154608 DOI: 10.1016/j.nbd.2023.106393] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 12/21/2023] [Accepted: 12/21/2023] [Indexed: 12/30/2023] Open
Abstract
Phosphodiesterase 2 A (PDE2A) is an enzyme involved in the homeostasis of cAMP and cGMP and is the most highly expressed PDE in human brain regions critical for socio-cognitive behavior. In cerebral cortex and hippocampus, PDE2A expression level is upregulated in Fmr1-KO mice, a model of the Fragile X Syndrome (FXS), the most common form of inherited intellectual disability (ID) and autism spectrum disorder (ASD). Indeed, PDE2A translation is negatively modulated by FMRP, whose functional absence causes FXS. While the pharmacological inhibition of PDE2A has been associated to its pro-cognitive role in normal animals and in models of ID and ASD, homozygous PDE2A mutations have been identified in patients affected by ID, ASD and epilepsy. To clarify this apparent paradox about the role of PDE2A in brain development, we characterized here Pde2a+/- mice (homozygote animals being not viable) at the behavioral, cellular, molecular and electrophysiological levels. Pde2a+/- females display a milder form of the disorder with reduced cognitive performance in adulthood, conversely males show severe socio-cognitive deficits throughout their life. In males, these phenotypes are associated with microglia activation, elevated glutathione levels and increased externalization of Glutamate receptor (GluR1) in CA1, producing reduced mGluR-dependent Long-term Depression. Overall, our results reveal molecular targets of the PDE2A-dependent pathway underlying socio-cognitive performance. These results clarify the mechanism of action of pro-cognitive drugs based on PDE2A inactivation, which have been shown to be promising therapeutic approaches for Alzheimer's disease, schizophrenia, FXS as well as other forms of ASD.
Collapse
Affiliation(s)
- Sébastien Delhaye
- CNRS UMR7275, Inserm U1323, Université Côte d'Azur, Institut de Pharmacologie Moléculaire et Cellulaire, 06560 Valbonne, France
| | - Marielle Jarjat
- CNRS UMR7275, Inserm U1323, Université Côte d'Azur, Institut de Pharmacologie Moléculaire et Cellulaire, 06560 Valbonne, France
| | - Asma Boulksibat
- CNRS UMR7275, Inserm U1323, Université Côte d'Azur, Institut de Pharmacologie Moléculaire et Cellulaire, 06560 Valbonne, France
| | - Clara Sanchez
- CNRS UMR7275, Inserm U1323, Université Côte d'Azur, Institut de Pharmacologie Moléculaire et Cellulaire, 06560 Valbonne, France
| | - Alessandra Tempio
- CNRS UMR7275, Inserm U1323, Université Côte d'Azur, Institut de Pharmacologie Moléculaire et Cellulaire, 06560 Valbonne, France
| | - Andrei Turtoi
- Inserm U1194, Université Montpellier, Institut de Recherche en Cancérologie de Montpellier, 34298 Montpellier Cedex 5, France
| | - Mauro Giorgi
- Department of Anatomical, Histological, Forensic and Orthopaedic Sciences, DAHFMO, Sapienza University of Rome, 00161 Rome, Italy
| | - Sandra Lacas-Gervais
- Université Côte d'Azur, Centre Commun de Microscopie Appliquée, 06100 Nice, France
| | - Gabriele Baj
- Department of Life Science, University of Trieste, 34100 Trieste, Italy
| | - Carole Rovere
- CNRS UMR7275, Inserm U1323, Université Côte d'Azur, Institut de Pharmacologie Moléculaire et Cellulaire, 06560 Valbonne, France
| | | | - Manuela Pellegrini
- Department of Anatomical, Histological, Forensic and Orthopaedic Sciences, DAHFMO, Sapienza University of Rome, 00161 Rome, Italy; Institute of Biochemistry and Cell Biology, IBBC-CNR, 00015 Monterotondo Scalo, Rome, Italy
| | - Thomas Maurin
- CNRS UMR7275, Inserm U1323, Université Côte d'Azur, Institut de Pharmacologie Moléculaire et Cellulaire, 06560 Valbonne, France
| | - Enzo Lalli
- CNRS UMR7275, Inserm U1323, Université Côte d'Azur, Institut de Pharmacologie Moléculaire et Cellulaire, 06560 Valbonne, France
| | - Barbara Bardoni
- CNRS UMR7275, Inserm U1323, Université Côte d'Azur, Institut de Pharmacologie Moléculaire et Cellulaire, 06560 Valbonne, France.
| |
Collapse
|
16
|
Gusev E, Sarapultsev A. Interplay of G-proteins and Serotonin in the Neuroimmunoinflammatory Model of Chronic Stress and Depression: A Narrative Review. Curr Pharm Des 2024; 30:180-214. [PMID: 38151838 DOI: 10.2174/0113816128285578231218102020] [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: 10/04/2023] [Accepted: 11/29/2023] [Indexed: 12/29/2023]
Abstract
INTRODUCTION This narrative review addresses the clinical challenges in stress-related disorders such as depression, focusing on the interplay between neuron-specific and pro-inflammatory mechanisms at the cellular, cerebral, and systemic levels. OBJECTIVE We aim to elucidate the molecular mechanisms linking chronic psychological stress with low-grade neuroinflammation in key brain regions, particularly focusing on the roles of G proteins and serotonin (5-HT) receptors. METHODS This comprehensive review of the literature employs systematic, narrative, and scoping review methodologies, combined with systemic approaches to general pathology. It synthesizes current research on shared signaling pathways involved in stress responses and neuroinflammation, including calcium-dependent mechanisms, mitogen-activated protein kinases, and key transcription factors like NF-κB and p53. The review also focuses on the role of G protein-coupled neurotransmitter receptors (GPCRs) in immune and pro-inflammatory responses, with a detailed analysis of how 13 of 14 types of human 5-HT receptors contribute to depression and neuroinflammation. RESULTS The review reveals a complex interaction between neurotransmitter signals and immunoinflammatory responses in stress-related pathologies. It highlights the role of GPCRs and canonical inflammatory mediators in influencing both pathological and physiological processes in nervous tissue. CONCLUSION The proposed Neuroimmunoinflammatory Stress Model (NIIS Model) suggests that proinflammatory signaling pathways, mediated by metabotropic and ionotropic neurotransmitter receptors, are crucial for maintaining neuronal homeostasis. Chronic mental stress can disrupt this balance, leading to increased pro-inflammatory states in the brain and contributing to neuropsychiatric and psychosomatic disorders, including depression. This model integrates traditional theories on depression pathogenesis, offering a comprehensive understanding of the multifaceted nature of the condition.
Collapse
Affiliation(s)
- Evgenii Gusev
- Laboratory of Inflammation Immunology, Institute of Immunology and Physiology, Ural Branch of the Russian Academy of Science, Ekaterinburg 620049, Russia
- Russian-Chinese Education and Research Center of System Pathology, South Ural State University, Chelyabinsk 454080, Russia
| | - Alexey Sarapultsev
- Russian-Chinese Education and Research Center of System Pathology, South Ural State University, Chelyabinsk 454080, Russia
- Laboratory of Immunopathophysiology, Institute of Immunology and Physiology, Ural Branch of the Russian Academy of Science, Ekaterinburg 620049, Russia
| |
Collapse
|
17
|
Sadeghi MA, Hemmati S, Yousefi-Manesh H, Foroutani L, Nassireslami E, Yousefi Zoshk M, Hosseini Y, Abbasian K, Dehpour AR, Chamanara M. Cilostazol pretreatment prevents PTSD-related anxiety behavior through reduction of hippocampal neuroinflammation. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:133-144. [PMID: 37382600 DOI: 10.1007/s00210-023-02578-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 06/13/2023] [Indexed: 06/30/2023]
Abstract
Current pharmacological treatments against post-traumatic stress disorder (PTSD) lack adequate efficacy. As a result, intense research has focused on identifying other molecular pathways mediating the pathogenesis of this condition. One such pathway is neuroinflammation, which has demonstrated a role in PTSD pathogenesis by causing synaptic dysfunction, neuronal death, and functional impairment in the hippocampus. Phosphodiesterase (PDE) inhibitors (PDEIs) have emerged as promising therapeutic agents against neuroinflammation in other neurological conditions. Furthermore, PDEIs have shown some promise in animal models of PTSD. However, the current model of PTSD pathogenesis, which is based on dysregulated fear learning, implies that PDE inhibition in neurons should enhance the acquisition of fear memory from the traumatic event. As a result, we hypothesized that PDEIs may improve PTSD symptoms through inhibiting neuroinflammation rather than long-term potentiation-related mechanisms. To this end, we tested the therapeutic efficacy of cilostazol, a selective inhibitor of PDE3, on PTSD-related anxiety symptoms in the underwater trauma model of PTSD. PDE3 is expressed much more richly in microglia and astrocytes compared to neurons in the murine brain. Furthermore, we used hippocampal indolamine 2,3-dioxygenase 1 (IDO) expression and interleukin 1 beta (IL-1β) concentration as indicators of neuroinflammation. We observed that cilostazol pretreatment prevented the development of anxiety symptoms and the increase in hippocampal IDO and IL-1β following PTSD induction. As a result, PDE3 inhibition ameliorated the neuroinflammatory processes involved in the development of PTSD symptoms. Therefore, cilostazol and other PDEIs may be promising candidates for further investigation as pharmacological therapies against PTSD.
Collapse
Affiliation(s)
- Mohammad Amin Sadeghi
- Toxicology Research Center, AJA University of Medical Sciences, Tehran, Iran
- Department of Pharmacology, School of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Sara Hemmati
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hasan Yousefi-Manesh
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Laleh Foroutani
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ehsan Nassireslami
- Toxicology Research Center, AJA University of Medical Sciences, Tehran, Iran
- Department of Pharmacology, School of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Mojtaba Yousefi Zoshk
- Trauma Research Center, AJA University of Medical Sciences, Tehran, Iran
- Department of Pediatrics, AJA University of Medical Sciences, Tehran, Iran
| | - Yasaman Hosseini
- Cognitive Neuroscience Center, School of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Kourosh Abbasian
- Management and Health Economics Department, AJA University of Medical Sciences, Tehran, Iran
| | - Ahmad Reza Dehpour
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohsen Chamanara
- Toxicology Research Center, AJA University of Medical Sciences, Tehran, Iran.
- Department of Pharmacology, School of Medicine, AJA University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
18
|
Singh N, Wanjari A, Sinha AH. Effects of Nicotine on the Central Nervous System and Sleep Quality in Relation to Other Stimulants: A Narrative Review. Cureus 2023; 15:e49162. [PMID: 38130519 PMCID: PMC10733894 DOI: 10.7759/cureus.49162] [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: 10/02/2023] [Accepted: 11/20/2023] [Indexed: 12/23/2023] Open
Abstract
Nicotine is used extensively across the globe despite the common awareness of the fact that it might stimulate the neurological system in those who indulge in its consumption. Nicotine can be consumed in a wide number of various forms and can also be delivered in a wide variety of different ways. After it has been heated, it can be smoked, consumed sublingually, or brought into touch with mucosal surfaces, with the buccal mucosa being the most popular one. These three methods of consumption account for the vast majority of its use. It has been demonstrated without a reasonable doubt that people who partake in nicotine do, in fact, experience an increase in their levels of alertness, wakefulness, attention, and focus. The half-life of the substance, in addition to its effects, is highly variable depending on the forms in which it is consumed, viz. cigarettes, tobacco, gums, lozenges, and the manner in which it is administered. This is the case regardless of whether the chemical is administered orally or intravenously. It is common for a person to require multiple "hits" of the chemical throughout the course of the day, though the frequency of these needs and the intervals between them can vary greatly. The time interval between each of these "hits" can range anywhere from a few hours to a few minutes. The user has the potential to develop a tolerance to the neurostimulatory and systemic effects of nicotine, as well as a heightened sensitivity to those effects, and even hereditary predispositions to specific adverse consequences. There is also a possibility that the user will develop an addiction to nicotine. This literature review aims to explore the relationship between nicotine consumption and its effects on the central nervous system, especially on sleep.
Collapse
Affiliation(s)
- Nihaal Singh
- Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Anil Wanjari
- Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Arya Harshyt Sinha
- Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| |
Collapse
|
19
|
Rowhanirad S, Taherianfard M. The neuroprotective effects of Chalcones from Ashitaba on cuprizone-induced demyelination via modulation of brain-derived neurotrophic factor and tumor necrosis factor α. Brain Behav 2023; 13:e3144. [PMID: 37403256 PMCID: PMC10498084 DOI: 10.1002/brb3.3144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 06/04/2023] [Accepted: 06/21/2023] [Indexed: 07/06/2023] Open
Abstract
INTRODUCTION Multiple sclerosis (MS) is the most common demyelinating disease of the central nervous system. However, the limitations of available therapeutic strategies are frustrating, both in terms of their low efficacy and multiple side effects. Previous studies showed that natural compounds such as Chalcones possess neuroprotective effects on neurodegenerative disorders. However, few studies have so far been published on the potential effects of Chalcones on treating demyelinating disease. The present study was designed to investigate the effects of Chalcones from Ashitaba (ChA) on cuprizone-induced noxious changes in the C57BL6 mice model of MS. METHODS The mice received normal diets (Control group: CNT), or Cuprizone-supplemented diets either without ChA (Cuprizone group: CPZ) or with low or high (300, 600 mg/kg/day) doses of ChA (ChA-treated groups: CPZ+ChA300/600). Brain-derived neurotrophic factor (BDNF) and tumor necrosis factor alpha (TNFα) levels, demyelination scores in the corpus callosum (CC), and cognitive impairment were evaluated using the enzyme-linked immunosorbent assay, histological, and Y-maze tests, respectively. RESULTS The findings showed that ChA Co-treatment significantly reduced the extent of demyelination in the CC and the serum and brain levels of TNFα in the ChA-treated groups compared to the CPZ group. Besides, treatment with a higher dose of ChA significantly improved the behavioral responses and BDNF levels in the serum and brain of the CPZ+ChA600 group when compared with the CPZ group. CONCLUSION The present study provided evidence for the neuroprotective effects of ChA on cuprizone-induced demyelination and behavioral dysfunction in C57BL/6 mice, possibly by modulating TNFα secretion and BDNF expression.
Collapse
Affiliation(s)
- Soodeh Rowhanirad
- Division of Physiology, Department of Basic Science, School of Veterinary MedicineShiraz UniversityShirazIran
| | - Mahnaz Taherianfard
- Division of Physiology, Department of Basic Science, School of Veterinary MedicineShiraz UniversityShirazIran
| |
Collapse
|
20
|
Friebe A, Kraehling JR, Russwurm M, Sandner P, Schmidtko A. The 10th International Conference on cGMP 2022: recent trends in cGMP research and development-meeting report. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023; 396:1669-1686. [PMID: 37079081 PMCID: PMC10338386 DOI: 10.1007/s00210-023-02484-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 03/31/2023] [Indexed: 04/21/2023]
Abstract
Increasing cGMP is a unique therapeutic principle, and drugs inhibiting cGMP-degrading enzymes or stimulating cGMP production are approved for the treatment of various diseases such as erectile dysfunction, coronary artery disease, pulmonary hypertension, chronic heart failure, irritable bowel syndrome, or achondroplasia. In addition, cGMP-increasing therapies are preclinically profiled or in clinical development for quite a broad set of additional indications, e.g., neurodegenerative diseases or different forms of dementias, bone formation disorders, underlining the pivotal role of cGMP signaling pathways. The fundamental understanding of the signaling mediated by nitric oxide-sensitive (soluble) guanylyl cyclase and membrane-associated receptor (particulate) guanylyl cyclase at the molecular and cellular levels, as well as in vivo, especially in disease models, is a key prerequisite to fully exploit treatment opportunities and potential risks that could be associated with an excessive increase in cGMP. Furthermore, human genetic data and the clinical effects of cGMP-increasing drugs allow back-translation into basic research to further learn about signaling and treatment opportunities. The biannual international cGMP conference, launched nearly 20 years ago, brings all these aspects together as an established and important forum for all topics from basic science to clinical research and pivotal clinical trials. This review summarizes the contributions to the "10th cGMP Conference on cGMP Generators, Effectors and Therapeutic Implications," which was held in Augsburg in 2022 but will also provide an overview of recent key achievements and activities in the field of cGMP research.
Collapse
Affiliation(s)
- Andreas Friebe
- Institute of Physiology, University of Würzburg, Röntgenring 9, D-97070 Würzburg, Germany
| | - Jan R. Kraehling
- Pharmaceuticals, Research and Early Development, Pharma Research Center, Bayer AG, Aprather Weg 18a, D-42096 Wuppertal, Germany
| | - Michael Russwurm
- Institute of Pharmacology, Ruhr-University Bochum, Universitätsstr. 150, D-44801 Bochum, Germany
| | - Peter Sandner
- Pharmaceuticals, Research and Early Development, Pharma Research Center, Bayer AG, Aprather Weg 18a, D-42096 Wuppertal, Germany
- Institute of Pharmacology, Hannover Medical School, Carl-Neuberg-Str. 1, D-30625 Hannover, Germany
| | - Achim Schmidtko
- Institute of Pharmacology and Clinical Pharmacy, Goethe University, Max-Von-Laue-Str. 9, D-60438 Frankfurt Am Main, Germany
| |
Collapse
|
21
|
Zuccarini M, Pruccoli L, Balducci M, Giuliani P, Caciagli F, Ciccarelli R, Di Iorio P. Influence of Guanine-Based Purines on the Oxidoreductive Reactions Involved in Normal or Altered Brain Functions. J Clin Med 2023; 12:jcm12031172. [PMID: 36769818 PMCID: PMC9917437 DOI: 10.3390/jcm12031172] [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: 12/05/2022] [Revised: 01/23/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023] Open
Abstract
The production of reactive oxygen species (ROS) in the brain is homeostatically controlled and contributes to normal neural functions. Inefficiency of control mechanisms in brain aging or pathological conditions leads to ROS overproduction with oxidative neural cell damage and degeneration. Among the compounds showing therapeutic potential against neuro-dysfunctions induced by oxidative stress are the guanine-based purines (GBPs), of which the most characterized are the nucleoside guanosine (GUO) and the nucleobase guanine (GUA), which act differently. Indeed, the administration of GUO to in vitro or in vivo models of acute brain injury (ischemia/hypoxia or trauma) or chronic neurological/neurodegenerative disorders, exerts neuroprotective and anti-inflammatory effects, decreasing the production of reactive radicals and improving mitochondrial function via multiple molecular signals. However, GUO administration to rodents also causes an amnesic effect. In contrast, the metabolite, GUA, could be effective in memory-related disorders by transiently increasing ROS production and stimulating the nitric oxide/soluble guanylate cyclase/cGMP/protein kinase G cascade, which has long been recognized as beneficial for cognitive function. Thus, it is worth pursuing further studies to ascertain the therapeutic role of GUO and GUA and to evaluate the pathological brain conditions in which these compounds could be more usefully used.
Collapse
Affiliation(s)
- Mariachiara Zuccarini
- Department of Medical, Oral and Biotechnological Sciences, University of Chieti-Pescara, Via dei Vestini 29, 66100 Chieti, Italy
- Center for Advanced Studies and Technologies (CAST), University of Chieti-Pescara, Via L. Polacchi, 66100 Chieti, Italy
| | - Letizia Pruccoli
- Department for Life Quality Studies, Alma Mater Studiorum-University of Bologna, 47921 Rimini, Italy
| | - Martina Balducci
- Department for Life Quality Studies, Alma Mater Studiorum-University of Bologna, 47921 Rimini, Italy
| | - Patricia Giuliani
- Department of Medical, Oral and Biotechnological Sciences, University of Chieti-Pescara, Via dei Vestini 29, 66100 Chieti, Italy
- Center for Advanced Studies and Technologies (CAST), University of Chieti-Pescara, Via L. Polacchi, 66100 Chieti, Italy
| | - Francesco Caciagli
- Center for Advanced Studies and Technologies (CAST), University of Chieti-Pescara, Via L. Polacchi, 66100 Chieti, Italy
| | - Renata Ciccarelli
- Center for Advanced Studies and Technologies (CAST), University of Chieti-Pescara, Via L. Polacchi, 66100 Chieti, Italy
| | - Patrizia Di Iorio
- Department of Medical, Oral and Biotechnological Sciences, University of Chieti-Pescara, Via dei Vestini 29, 66100 Chieti, Italy
- Center for Advanced Studies and Technologies (CAST), University of Chieti-Pescara, Via L. Polacchi, 66100 Chieti, Italy
- Correspondence:
| |
Collapse
|
22
|
Domiati S, Abd El Galil K, Ragab H. Molecular Mechanics Simulations and Experimental Investigation of the Effect of Tadalafil on Various Inflammatory Pain Mediators. ACS OMEGA 2022; 7:43747-43758. [PMID: 36506154 PMCID: PMC9730770 DOI: 10.1021/acsomega.2c04761] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 11/09/2022] [Indexed: 06/17/2023]
Abstract
PURPOSE Tadalafil's exact analgesic mechanism is still unclear. The current study aimed to elucidate this mechanism in an inflammatory pain model. METHODS Computer-assisted simulation docking experiments were carried out to assess the binding of tadalafil to different ligands. The anti-inflammatory and analgesic effects of tadalafil were evaluated using formalin-induced paw edema and a von Frey filament test, respectively. The plantar paw of the mice was then dissected to quantify iNOS, nNOS, COX-2, TNFα, IL1, and IL10 gene expression levels using a real-time polymerase chain reaction. iNOS, TNFα, and COX-2 inhibition was reassessed in vitro using the ELISA technique. One-way analysis of variance followed by post hoc Tukey test or t-test was used to compare the means. RESULTS Docking analysis showed a superior binding score of tadalafil to COX-2, iNOS, IL-1, and TNF-α compared to that of indomethacin and morphine and a similar binding score to nNOS and IL-10 relative to that of indomethacin. In the in vivo study, tadalafil, after an hour of formalin administration, inhibited significantly paw edema, similar to indomethacin. Furthermore, it significantly increased the withdrawal force in the von Frey filament test as compared to the negative control, which was similar to the effect observed with indomethacin and morphine. The RT-PCR revealed that tadalafil reduced significantly the iNOS, COX-2, and TNF-α gene expressions but had no effect on nNOS, IL 1, and IL10. In vitro ELISA tests confirmed the inhibition of iNOS, COX-2, and TNF-α. CONCLUSION Tadalafil probably exerts its analgesic effect through the simultaneous inhibition of iNOS, COX-2, and TNF-α, which is not the case with other nonsteroidal anti-inflammatory drugs. Nevertheless, further studies are required to confirm its mechanism.
Collapse
Affiliation(s)
- Souraya Domiati
- Department
of Pharmacology and Therapeutics, Faculty of Pharmacy, Beirut Arab University, Beirut1107 2809, Lebanon
| | - Khaled Abd El Galil
- Department
of Pharmaceutical Sciences, Faculty of Pharmacy, Beirut Arab University, Beirut1107 2809, Lebanon
- Department
of Microbiology and Immunology, Faculty of Pharmacy, Mansoura University, Mansoura35516, Egypt
| | - Hanan Ragab
- Department
of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria21521, Egypt
| |
Collapse
|
23
|
Sheng J, Zhang S, Wu L, Kumar G, Liao Y, GK P, Fan H. Inhibition of phosphodiesterase: A novel therapeutic target for the treatment of mild cognitive impairment and Alzheimer's disease. Front Aging Neurosci 2022; 14:1019187. [PMID: 36268188 PMCID: PMC9577554 DOI: 10.3389/fnagi.2022.1019187] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Accepted: 09/05/2022] [Indexed: 11/13/2022] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia and is ranked as the 6th leading cause of death in the US. The prevalence of AD and dementia is steadily increasing and expected cases in USA is 14.8 million by 2050. Neuroinflammation and gradual neurodegeneration occurs in Alzheimer's disease. However, existing medications has limitation to completely abolish, delay, or prevent disease progression. Phosphodiesterases (PDEs) are large family of enzymes to hydrolyze the 3'-phosphodiester links in cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP) in signal-transduction pathways for generation of 5'-cyclic nucleotides. It plays vital role to orchestrate several pharmacological activities for proper cell functioning and regulating the levels of cAMP and cGMP. Several evidence has suggested that abnormal cAMP signaling is linked to cognitive problems in neurodegenerative disorders like AD. Therefore, the PDE family has become a widely accepted and multipotential therapeutic target for neurodegenerative diseases. Notably, modulation of cAMP/cGMP by phytonutrients has a huge potential for the management of AD. Natural compounds have been known to inhibit phosphodiesterase by targeting key enzymes of cGMP synthesis pathway, however, the mechanism of action and their therapeutic efficacy has not been explored extensively. Currently, few PDE inhibitors such as Vinpocetine and Nicergoline have been used for treatment of central nervous system (CNS) disorders. Considering the role of flavonoids to inhibit PDE, this review discussed the therapeutic potential of natural compounds with PDE inhibitory activity for the treatment of AD and related dementia.
Collapse
Affiliation(s)
- Jianwen Sheng
- Department of Gastroenterology, The People’s Hospital of Yichun City, Yichun, China
| | - Shanjin Zhang
- Department of Gastroenterology, The People’s Hospital of Yichun City, Yichun, China
| | - Lule Wu
- Department of Gastroenterology, The People’s Hospital of Yichun City, Yichun, China
| | - Gajendra Kumar
- Department of Neuroscience, City University of Hong Kong, Kowloon Tong, Hong Kong SAR, China
| | - Yuanhang Liao
- Department of Gastroenterology, The People’s Hospital of Yichun City, Yichun, China
| | - Pratap GK
- Department of Biochemistry, Davangere University, Davangere, India
| | - Huizhen Fan
- Department of Gastroenterology, The People’s Hospital of Yichun City, Yichun, China
| |
Collapse
|
24
|
Scaffold Repurposing Reveals New Nanomolar Phosphodiesterase Type 5 (PDE5) Inhibitors Based on Pyridopyrazinone Scaffold: Investigation of In Vitro and In Silico Properties. Pharmaceutics 2022; 14:pharmaceutics14091954. [PMID: 36145702 PMCID: PMC9501832 DOI: 10.3390/pharmaceutics14091954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 09/08/2022] [Accepted: 09/09/2022] [Indexed: 11/17/2022] Open
Abstract
Inhibition of PDE5 results in elevation of cGMP leading to vascular relaxation and reduction in the systemic blood pressure. Therefore, PDE5 inhibitors are used as antihypertensive and antianginal agents in addition to their major use as male erectile dysfunction treatments. Previously, we developed a novel series of 34 pyridopyrazinone derivatives as anticancer agents (series A–H). Herein, a multi-step in silico approach was preliminary conducted to evaluate the predicted PDE5 inhibitory activity, followed by an in vitro biological evaluation over the enzymatic level and a detailed SAR study. The designed 2D-QSAR model which was carried out to predict the IC50 of the tested compounds revealed series B, D, E and G with nanomolar range of IC50 values (6.00–81.56 nM). A further docking simulation model was performed to investigate the binding modes within the active site of PDE5. Interestingly, most of the tested compounds showed almost the same binding modes of that of reported PDE5 inhibitors. To validate the in silico results, an in vitro enzymatic assay over PDE5 enzyme was performed for a number of the promising candidates with different substitutions. Both series E and G exhibited a potent inhibitory activity (IC50 = 18.13–41.41 nM). Compound 11b (series G, oxadiazole-based derivatives with terminal 4-NO2 substituted phenyl ring and rigid linker) was the most potent analogue with IC50 value of 18.13 nM. Structure–activity relationship (SAR) data attained for various substitutions were rationalized. Furthermore, a molecular dynamic simulation gave insights into the inhibitory activity of the most active compound (11b). Accordingly, this report presents a successful scaffold repurposing approach that reveals compound 11b as a highly potent nanomolar PDE5 inhibitor worthy of further investigation.
Collapse
|
25
|
Lee DH, Lee JY, Hong DY, Lee EC, Park SW, Jo YN, Park YJ, Cho JY, Cho YJ, Chae SH, Lee MR, Oh JS. ROCK and PDE-5 Inhibitors for the Treatment of Dementia: Literature Review and Meta-Analysis. Biomedicines 2022; 10:biomedicines10061348. [PMID: 35740369 PMCID: PMC9219677 DOI: 10.3390/biomedicines10061348] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 06/02/2022] [Accepted: 06/05/2022] [Indexed: 12/14/2022] Open
Abstract
Dementia is a disease in which memory, thought, and behavior-related disorders progress gradually due to brain damage caused by injury or disease. It is mainly caused by Alzheimer’s disease or vascular dementia and several other risk factors, including genetic factors. It is difficult to treat as its incidence continues to increase worldwide. Many studies have been performed concerning the treatment of this condition. Rho-associated kinase (ROCK) and phosphodiesterase-5 (PDE-5) are attracting attention as pharmacological treatments to improve the symptoms. This review discusses how ROCK and PDE-5 affect Alzheimer’s disease, vascular restructuring, and exacerbation of neuroinflammation, and how their inhibition helps improve cognitive function. In addition, the results of the animal behavior analysis experiments utilizing the Morris water maze were compared through meta-analysis to analyze the effects of ROCK inhibitors and PDE-5 inhibitors on cognitive function. According to the selection criteria, 997 publications on ROCK and 1772 publications on PDE-5 were screened, and conclusions were drawn through meta-analysis. Both inhibitors showed good improvement in cognitive function tests, and what is expected of the synergy effect of the two drugs was confirmed in this review.
Collapse
Affiliation(s)
- Dong-Hun Lee
- Department of Neurosurgery, College of Medicine, Soonchunhyang University, Cheonan Hospital, Cheonan 31151, Korea; (D.-H.L.); (J.Y.L.); (D.-Y.H.); (E.C.L.); (S.-W.P.)
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soon Chun Hyang University, Cheonan 31151, Korea
| | - Ji Young Lee
- Department of Neurosurgery, College of Medicine, Soonchunhyang University, Cheonan Hospital, Cheonan 31151, Korea; (D.-H.L.); (J.Y.L.); (D.-Y.H.); (E.C.L.); (S.-W.P.)
| | - Dong-Yong Hong
- Department of Neurosurgery, College of Medicine, Soonchunhyang University, Cheonan Hospital, Cheonan 31151, Korea; (D.-H.L.); (J.Y.L.); (D.-Y.H.); (E.C.L.); (S.-W.P.)
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soon Chun Hyang University, Cheonan 31151, Korea
| | - Eun Chae Lee
- Department of Neurosurgery, College of Medicine, Soonchunhyang University, Cheonan Hospital, Cheonan 31151, Korea; (D.-H.L.); (J.Y.L.); (D.-Y.H.); (E.C.L.); (S.-W.P.)
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soon Chun Hyang University, Cheonan 31151, Korea
| | - Sang-Won Park
- Department of Neurosurgery, College of Medicine, Soonchunhyang University, Cheonan Hospital, Cheonan 31151, Korea; (D.-H.L.); (J.Y.L.); (D.-Y.H.); (E.C.L.); (S.-W.P.)
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soon Chun Hyang University, Cheonan 31151, Korea
| | - Yu Na Jo
- Department of Medicine, College of Medicine, Soonchunhyang University, Cheonan 31151, Korea; (Y.N.J.); (Y.J.P.); (J.Y.C.); (Y.J.C.); (S.H.C.)
| | - Yu Jin Park
- Department of Medicine, College of Medicine, Soonchunhyang University, Cheonan 31151, Korea; (Y.N.J.); (Y.J.P.); (J.Y.C.); (Y.J.C.); (S.H.C.)
| | - Jae Young Cho
- Department of Medicine, College of Medicine, Soonchunhyang University, Cheonan 31151, Korea; (Y.N.J.); (Y.J.P.); (J.Y.C.); (Y.J.C.); (S.H.C.)
| | - Yoo Jin Cho
- Department of Medicine, College of Medicine, Soonchunhyang University, Cheonan 31151, Korea; (Y.N.J.); (Y.J.P.); (J.Y.C.); (Y.J.C.); (S.H.C.)
| | - Su Hyun Chae
- Department of Medicine, College of Medicine, Soonchunhyang University, Cheonan 31151, Korea; (Y.N.J.); (Y.J.P.); (J.Y.C.); (Y.J.C.); (S.H.C.)
| | - Man Ryul Lee
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soon Chun Hyang University, Cheonan 31151, Korea
- Correspondence: (M.R.L.); (J.S.O.)
| | - Jae Sang Oh
- Department of Neurosurgery, College of Medicine, Soonchunhyang University, Cheonan Hospital, Cheonan 31151, Korea; (D.-H.L.); (J.Y.L.); (D.-Y.H.); (E.C.L.); (S.-W.P.)
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soon Chun Hyang University, Cheonan 31151, Korea
- Correspondence: (M.R.L.); (J.S.O.)
| |
Collapse
|
26
|
Özkul B, Urfalı FE, Sever İH, Bozkurt MF, Söğüt İ, Elgörmüş ÇS, Erdogan MA, Erbaş O. Demonstration of Ameliorating Effect of Vardenafil Through Its Anti-Inflammatory and Neuroprotective Properties in Autism Spectrum Disorder Induced by Propionic Acid on Rat Model. Int J Neurosci 2022; 132:1150-1164. [PMID: 35584252 DOI: 10.1080/00207454.2022.2079507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Introduction: Autism spectrum disorder (ASD) is a neurodevelopmental disorder with complex etiology. In this study, we aimed to determine the ameliorating effects of vardenafil in the ASD rat model induced by propionic acid (PPA) in terms of neurobehavioral changes and also support these effects with histopathological changes, brain biochemical analysis and magnetic resonance spectroscopy (MRS) findings.Materials and Methods: Twenty-one male rats were randomly assigned into 3 groups. Group 1 (control, 7 rats) did not receive treatment. Rats in groups 2 and 3 were given PPA at the dose of 250 mg/kg/day intraperitoneally for 5 days. After PPA administration, animals in group 2 (PPAS, 7 rats) were given saline and animals in group 3 (PPAV, 7 rats) were given vardenafil. Behavioral tests were performed between the 20th and 24th days of the study. The rats were taken for MRS on the 25th day. At the end of the study, brain levels of interleukin-2 (IL-2), IL-17, tumor necrosis factor-α, nerve growth factor, cGMP and lactate levels were measured. In the cerebellum and the CA1 and CA3 regions of the hippocampus, counts of neurons and Purkinje cells and glial fibrillary acidic protein (associated with gliosis) were evaluated histologically.Results: Three chamber sociability and passive avoiding test, histopathological results, lactate levels derived from MRS, and biochemical biomarkers revealed significant differences among the PPAV and PPAS groups.Conclusion: We concluded that vardenafil improves memory and social behaviors and prevent loss of neuronal and Purkinje cell through its anti-inflammatory and neuroprotective effect.
Collapse
Affiliation(s)
- Bahattin Özkul
- Faculty of Medicine, Department of Radiology, Istanbul Atlas University, Istanbul, Turkey
| | - Furkan Ertürk Urfalı
- Department of Radiology, Faculty of Medicine, Kutahya Saglık Bilimleri, Kutahya, Turkey
| | - İbrahim Halil Sever
- Department of Radiology, Faculty of Medicine, Demiroğlu Bilim University, Istanbul, Turkey
| | - Mehmet Fatih Bozkurt
- Department of Pathology, Faculty of Veterinary, Afyon Kocatepe University, Afyon, Turkey
| | - İbrahim Söğüt
- Department of Biochemistry, Faculty of Medicine, Demiroğlu Bilim University, Istanbul, Turkey
| | - Çağrı Serdar Elgörmüş
- Department of Emergency, Faculty of Medicine, Istanbul Atlas University, Istanbul, Turkey
| | - Mumin Alper Erdogan
- Department of Physiology, Faculty of Medicine, Izmir Katip Celebi University, Izmir, Turkey
| | - Oytun Erbaş
- Department of Physiology, Faculty of Medicine, Demiroğlu Bilim University, Istanbul, Turkey
| |
Collapse
|
27
|
Xiong Y, Wintermark P. The Role of Sildenafil in Treating Brain Injuries in Adults and Neonates. Front Cell Neurosci 2022; 16:879649. [PMID: 35620219 PMCID: PMC9127063 DOI: 10.3389/fncel.2022.879649] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Accepted: 04/04/2022] [Indexed: 12/02/2022] Open
Abstract
Sildenafil is a recognized treatment for patients suffering from erectile dysfunction and pulmonary hypertension. However, new evidence suggests that it may have a neuroprotective and a neurorestorative role in the central nervous system of both adults and neonates. Phosphodiesterase type 5-the target of sildenafil-is distributed in many cells throughout the body, including neurons and glial cells. This study is a comprehensive review of the demonstrated effects of sildenafil on the brain with respect to its function, extent of injury, neurons, neuroinflammation, myelination, and cerebral vessels.
Collapse
Affiliation(s)
- Ying Xiong
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
- Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Pia Wintermark
- Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Division of Newborn Medicine, Department of Pediatrics, Montreal Children’s Hospital, Montreal, QC, Canada
| |
Collapse
|
28
|
Jyoti Dutta B, Singh S, Seksaria S, Das Gupta G, Bodakhe SH, Singh A. Potential role of IP3/Ca 2+ signaling and phosphodiesterases: Relevance to neurodegeneration in Alzheimer's disease and possible therapeutic strategies. Biochem Pharmacol 2022; 201:115071. [PMID: 35525328 DOI: 10.1016/j.bcp.2022.115071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 04/27/2022] [Accepted: 04/28/2022] [Indexed: 11/02/2022]
Abstract
Despite large investments by industry and governments, no disease-modifying medications for the treatment of patients with Alzheimer's disease (AD) have been found. The failures of various clinical trials indicate the need for a more in-depth understanding of the pathophysiology of AD and for innovative therapeutic strategies for its treatment. Here, we review the rational for targeting IP3 signaling, cytosolic calcium dysregulation, phosphodiesterases (PDEs), and secondary messengers like cGMP and cAMP, as well as their correlations with the pathophysiology of AD. Various drugs targeting these signaling cascades are still in pre-clinical and clinical trials which support the ideas presented in this article. Further, we describe different molecular mechanisms and medications currently being used in various pre-clinical and clinical trials involving IP3/Ca+2 signaling. We also highlight various isoforms, as well as the functions and pharmacology of the PDEs broadly expressed in different parts of the brain and attempt to unravel the potential benefits of PDE inhibitors for use as novel medications to alleviate the pathogenesis of AD.
Collapse
Affiliation(s)
- Bhaskar Jyoti Dutta
- Department of Pharmacology, ISF College of Pharmacy, GT Road, Ghal Kalan, Moga-142001, Punjab, India
| | - Shamsher Singh
- Department of Pharmacology, ISF College of Pharmacy, GT Road, Ghal Kalan, Moga-142001, Punjab, India
| | - Sanket Seksaria
- Department of Pharmacology, ISF College of Pharmacy, GT Road, Ghal Kalan, Moga-142001, Punjab, India
| | - Ghanshyam Das Gupta
- Department of Pharmacology, ISF College of Pharmacy, GT Road, Ghal Kalan, Moga-142001, Punjab, India
| | - Surendra H Bodakhe
- Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur - 495009, Chhattisgarh, India
| | - Amrita Singh
- Department of Pharmacology, ISF College of Pharmacy, GT Road, Ghal Kalan, Moga-142001, Punjab, India.
| |
Collapse
|
29
|
Taoro-González L, Cabrera-Pastor A, Sancho-Alonso M, Felipo V. Intracellular and extracelluar cyclic GMP in the brain and the hippocampus. VITAMINS AND HORMONES 2022; 118:247-288. [PMID: 35180929 DOI: 10.1016/bs.vh.2021.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Cyclic Guanosine-Monophosphate (cGMP) is implicated as second messenger in a plethora of pathways and its effects are executed mainly by cGMP-dependent protein kinases (PKG). It is involved in both peripheral (cardiovascular regulation, intestinal secretion, phototransduction, etc.) and brain (hippocampal synaptic plasticity, neuroinflammation, cognitive function, etc.) processes. Stimulation of hippocampal cGMP signaling have been proved to be beneficial in animal models of aging, Alzheimer's disease or hepatic encephalopathy, restoring different cognitive functions such as passive avoidance, object recognition or spatial memory. However, even when some inhibitors of cGMP-degrading enzymes (PDEs) are already used against peripheral pathologies, their utility as neurological treatments is still under clinical investigation. Additionally, it has been demonstrated a list of cGMP roles as not second but first messenger. The role of extracellular cGMP has been specially studied in hippocampal function and cognitive impairment in animal models and it has emerged as an important modulator of neuroinflammation-mediated cognitive alterations and hippocampal synaptic plasticity malfunction. Specifically, it has been demonstrated that extracellular cGMP decreases hippocampal IL-1β levels restoring membrane expression of glutamate receptors in the hippocampus and cognitive function in hyperammonemic rats. The mechanisms implicated are still unclear and might involve complex interactions between hippocampal neurons, astrocytes and microglia. Membrane targets for extracellular cGMP are still poorly understood and must be addressed in future studies.
Collapse
Affiliation(s)
- Lucas Taoro-González
- Department of Clinical Psychology, Psychobiology and Methodology, Area of Psycobiology, University of La Laguna, Tenerife, Spain
| | - Andrea Cabrera-Pastor
- Fundación Investigación Hospital Clínico, Instituto de Investigación Sanitaria (INCLIVA), Valencia, Spain; Laboratory of Neurobiology, Centro de Investigación Príncipe Felipe, Valencia, Spain
| | - María Sancho-Alonso
- Laboratory of Neurobiology, Centro de Investigación Príncipe Felipe, Valencia, Spain
| | - Vicente Felipo
- Laboratory of Neurobiology, Centro de Investigación Príncipe Felipe, Valencia, Spain.
| |
Collapse
|
30
|
Bupivacaine in combination with sildenafil (Viagra) and vitamin D3 have anti-inflammatory effects in osteoarthritic chondrocytes. CURRENT RESEARCH IN PHARMACOLOGY AND DRUG DISCOVERY 2021; 2:100066. [PMID: 34909684 PMCID: PMC8663929 DOI: 10.1016/j.crphar.2021.100066] [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] [Received: 08/01/2021] [Revised: 10/13/2021] [Accepted: 10/18/2021] [Indexed: 11/21/2022] Open
Abstract
Aims To treat osteoarthritic chondrocytes and thereby reduce the inflammation with a drug combination that primarily affects 5-HT- and ATP-evoked Ca2+ signaling. In osteoarthritic chondrocytes, Ca2+ signaling is elevated, resulting in increased production of ATP and inflammatory mediators. The expression of TLR4 and Na+/K+-ATPase was used to evaluate the inflammatory status of the cells. Main methods Equine chondrocytes were collected from joints with mild structural osteoarthritic changes and cultured in monolayers. The cells were treated with a combination of bupivacaine (1 pM) and sildenafil (1 μM) in combination with vitamin D3 (100 nM). A high-throughput screening system, the Flexstation 3 microplate reader, was used to measure intra- and extracellular Ca2+ signaling after exposure to 5-HT, glutamate, or ATP. Expression of inflammatory receptors was assessed by Western blotting. Key findings Drug treatment substantially reduced 5-HT- and ATP-evoked intracellular Ca2+ release and TLR4 expression compared to those in untreated chondrocytes. The combination of sildenafil, vitamin D3 together with metformin, as the ability to take up glucose is limited, increased Na+/K+-ATPase expression. Significance The combination of these three therapeutic substances at concentrations much lower than usually used, reduced expression of the inflammatory receptor TLR4 and increased the cell membrane enzyme Na+/K+-ATPase, which regulates cell volume and reduces increased intracellular Ca2+ concentrations. These remarkable results indicate that this drug combination has disease-modifying osteoarthritis drug (DMOAD) properties and may be a new clinical therapy for osteoarthritis (OA).
Collapse
|
31
|
Pilarzyk K, Farmer R, Porcher L, Kelly MP. The Role of PDE11A4 in Social Isolation-Induced Changes in Intracellular Signaling and Neuroinflammation. Front Pharmacol 2021; 12:749628. [PMID: 34887755 PMCID: PMC8650591 DOI: 10.3389/fphar.2021.749628] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 10/25/2021] [Indexed: 12/29/2022] Open
Abstract
Phosphodiesterase 11A (PDE11A), an enzyme that degrades cyclic nucleotides (cAMP and cGMP), is the only PDE whose mRNA expression in brain is restricted to the hippocampal formation. Previously, we showed that chronic social isolation changes subsequent social behaviors in adult mice by reducing expression of PDE11A4 in the membrane fraction of the ventral hippocampus (VHIPP). Here we seek extend these findings by determining 1) if isolation-induced decreases in PDE11A4 require chronic social isolation or if they occur acutely and are sustained long-term, 2) if isolation-induced decreases occur uniquely in adults (i.e., not adolescents), and 3) how the loss of PDE11 signaling may increase neuroinflammation. Both acute and chronic social isolation decrease PDE11A4 expression in adult but not adolescent mice. This decrease in PDE11A4 is specific to the membrane compartment of the VHIPP, as it occurs neither in the soluble nor nuclear fractions of the VHIPP nor in any compartment of the dorsal HIPP. The effect of social isolation on membrane PDE11A4 is also selective in that PDE2A and PDE10A expression remain unchanged. Isolation-induced decreases in PDE11A4 expression appear to be functional as social isolation elicited changes in PDE11A-relevant signal transduction cascades (i.e., decreased pCamKIIα and pS6-235/236) and behavior (i.e., increased remote long-term memory for social odor recognition). Interestingly, we found that isolation-induced decreases in membrane PDE11A4 correlated with increased expression of interleukin-6 (IL-6) in the soluble fraction, suggesting pro-inflammatory signaling for this cytokine. This effect on IL-6 is consistent with the fact that PDE11A deletion increased microglia activation, although it left astrocytes unchanged. Together, these data suggest that isolation-induced decreases in PDE11A4 may alter subsequent social behavior via increased neuroinflammatory processes in adult mice.
Collapse
Affiliation(s)
- Katy Pilarzyk
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, United States
| | - Reagan Farmer
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, United States
| | - Latarsha Porcher
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, United States
| | - Michy P Kelly
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, United States.,Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, United States.,Center for Aging Research, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
32
|
Analysis of the early response to spinal cord injury identified a key role for mTORC1 signaling in the activation of neural stem progenitor cells. NPJ Regen Med 2021; 6:68. [PMID: 34686684 PMCID: PMC8536777 DOI: 10.1038/s41536-021-00179-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 09/30/2021] [Indexed: 02/07/2023] Open
Abstract
Xenopus laevis are able to regenerate the spinal cord during larvae stages through the activation of neural stem progenitor cells (NSPCs). Here we use high-resolution expression profiling to characterize the early transcriptome changes induced after spinal cord injury, aiming to identify the signals that trigger NSPC proliferation. The analysis delineates a pathway that starts with a rapid and transitory activation of immediate early genes, followed by migration processes and immune response genes, the pervasive increase of NSPC-specific ribosome biogenesis factors, and genes involved in stem cell proliferation. Western blot and immunofluorescence analysis showed that mTORC1 is rapidly and transiently activated after SCI, and its pharmacological inhibition impairs spinal cord regeneration and proliferation of NSPC through the downregulation of genes involved in the G1/S transition of cell cycle, with a strong effect on PCNA. We propose that the mTOR signaling pathway is a key player in the activation of NPSCs during the early steps of spinal cord regeneration.
Collapse
|
33
|
Correia SS, Liu G, Jacobson S, Bernier SG, Tobin JV, Schwartzkopf CD, Atwater E, Lonie E, Rivers S, Carvalho A, Germano P, Tang K, Iyengar RR, Currie MG, Hadcock JR, Winrow CJ, Jones JE. The CNS-penetrant soluble guanylate cyclase stimulator CYR119 attenuates markers of inflammation in the central nervous system. J Neuroinflammation 2021; 18:213. [PMID: 34537066 PMCID: PMC8449877 DOI: 10.1186/s12974-021-02275-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 09/08/2021] [Indexed: 01/05/2023] Open
Abstract
Background Inflammation in the central nervous system (CNS) is observed in many neurological disorders. Nitric oxide-soluble guanylate cyclase-cyclic guanosine monophosphate (NO–sGC–cGMP) signaling plays an essential role in modulating neuroinflammation. CYR119 is a CNS-penetrant sGC stimulator that amplifies endogenous NO–sGC–cGMP signaling. We evaluated target engagement and the effects of CYR119 on markers of neuroinflammation in vitro in mouse microglial cells and in vivo in quinolinic acid (QA)-induced and high-fat diet-induced rodent neuroinflammation models.
Methods Target engagement was verified in human embryonic kidney (HEK) cells, rat primary neurons, mouse SIM-A9 cells, and in rats by measuring changes in cGMP and downstream targets of sGC signaling [phosphorylated vasodilator-stimulated phosphoprotein (pVASP), phosphorylated cAMP-response element binding (pCREB)]. In SIM-A9 cells stimulated with lipopolysaccharides (LPS), markers of inflammation were measured when cells were treated with or without CYR119. In rats, microinjections of QA and vehicle were administered into the right and left hemispheres of striatum, respectively, and then rats were dosed daily with either CYR119 (10 mg/kg) or vehicle for 7 days. The activation of microglia [ionized calcium binding adaptor molecule 1 (Iba1)] and astrocytes [glial fibrillary acidic protein (GFAP)] was measured by immunohistochemistry. Diet-induced obese (DIO) mice were treated daily with CYR119 (10 mg/kg) for 6 weeks, after which inflammatory genetic markers were analyzed in the prefrontal cortex. Results In vitro, CYR119 synergized with exogenous NO to increase the production of cGMP in HEK cells and in primary rat neuronal cell cultures. In primary neurons, CYR119 stimulated sGC, resulting in accumulation of cGMP and phosphorylation of CREB, likely through the activation of protein kinase G (PKG). CYR119 attenuated LPS-induced elevation of interleukin 6 (IL-6) and tumor necrosis factor (TNF) in mouse microglial cells. Following oral dosing in rats, CYR119 crossed the blood–brain barrier (BBB) and stimulated an increase in cGMP levels in the cerebral spinal fluid (CSF). In addition, levels of proinflammatory markers associated with QA administration or high-fat diet feeding were lower in rodents treated with CYR119 than in those treated with vehicle. Conclusions These data suggest that sGC stimulation could provide neuroprotective effects by attenuating inflammatory responses in nonclinical models of neuroinflammation.
Collapse
Affiliation(s)
- Susana S Correia
- Cyclerion Therapeutics, 245 First St., Riverview II, 18th Floor, Cambridge, MA, 02142, USA
| | - Guang Liu
- Cyclerion Therapeutics, 245 First St., Riverview II, 18th Floor, Cambridge, MA, 02142, USA
| | - Sarah Jacobson
- Cyclerion Therapeutics, 245 First St., Riverview II, 18th Floor, Cambridge, MA, 02142, USA
| | - Sylvie G Bernier
- Cyclerion Therapeutics, 245 First St., Riverview II, 18th Floor, Cambridge, MA, 02142, USA
| | - Jenny V Tobin
- Cyclerion Therapeutics, 245 First St., Riverview II, 18th Floor, Cambridge, MA, 02142, USA
| | - Chad D Schwartzkopf
- Cyclerion Therapeutics, 245 First St., Riverview II, 18th Floor, Cambridge, MA, 02142, USA
| | - Emily Atwater
- Cyclerion Therapeutics, 245 First St., Riverview II, 18th Floor, Cambridge, MA, 02142, USA
| | | | - Sam Rivers
- Cyclerion Therapeutics, 245 First St., Riverview II, 18th Floor, Cambridge, MA, 02142, USA
| | - Andrew Carvalho
- Cyclerion Therapeutics, 245 First St., Riverview II, 18th Floor, Cambridge, MA, 02142, USA
| | - Peter Germano
- Cyclerion Therapeutics, 245 First St., Riverview II, 18th Floor, Cambridge, MA, 02142, USA
| | - Kim Tang
- Ironwood Pharmaceuticals, Cambridge, MA, 02142, USA
| | - Rajesh R Iyengar
- Cyclerion Therapeutics, 245 First St., Riverview II, 18th Floor, Cambridge, MA, 02142, USA
| | - Mark G Currie
- Cyclerion Therapeutics, 245 First St., Riverview II, 18th Floor, Cambridge, MA, 02142, USA
| | - John R Hadcock
- Cyclerion Therapeutics, 245 First St., Riverview II, 18th Floor, Cambridge, MA, 02142, USA
| | - Christopher J Winrow
- Cyclerion Therapeutics, 245 First St., Riverview II, 18th Floor, Cambridge, MA, 02142, USA
| | - Juli E Jones
- Cyclerion Therapeutics, 245 First St., Riverview II, 18th Floor, Cambridge, MA, 02142, USA.
| |
Collapse
|
34
|
Kurakula M, Naveen N. R, Patel B, Manne R, Patel DB. Preparation, Optimization and Evaluation of Chitosan-Based Avanafil Nanocomplex Utilizing Antioxidants for Enhanced Neuroprotective Effect on PC12 Cells. Gels 2021; 7:gels7030096. [PMID: 34287358 PMCID: PMC8293062 DOI: 10.3390/gels7030096] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 07/07/2021] [Accepted: 07/08/2021] [Indexed: 12/13/2022] Open
Abstract
(1) Introduction: in recent decades, interdisciplinary research on the utilization of natural products as “active moiety carriers” was focused on due to their superior safety profile, biodegradability, biocompatibility and the ability for sustained or controlled release activity. The nano-based neuroprotective strategy is explored as an imperative treatment for diabetic neuropathy (DN). Avanafil (AV), that selectively inhibits the degradation of cGMP-specific phosphodiesterase, thereby increasing the levels of cGMP, makes a decisive mediator for cytoprotection. (2) Methods: AVnanocomplex formulations were prepared by a modified anti-solvent precipitation method and the method was optimized by Box–Behnken design. An optimized formulation was characterized and evaluated for various in vitro parameters; (3) results:based on the desirability approach, the formulation containing 2.176 g of chitosan, 7.984 g of zein and 90% v/v ethanol concentration can fulfill the prerequisites of optimum formulation (OB-AV-NC).OB-AV-NC was characterized and evaluated for various parameters. The neuroprotective mechanism of AV was evaluated by pretreatment of PC12 cells with plain AV, avanafil nanocomplex (NC) without antioxidants (AV-NC) and with antioxidants (α-Lipoic acid LP; Ellagic Acid EA), AV-LP-EA-Nanocomplex has also shown considerable attenuation in intracellular reactive oxygen species (ROS) and lipid peroxidation with a significant increase in the PC 12 viability under HG conditions in comparison to pure AV; (4) conclusion: the nanocomplex of AV prepared to utilize natural polymers and antioxidants aided for high solubility of AV and exhibited desired neuroprotective activity.This can be one of the promisingstrategy to translate the AV nanocomplex with safety and efficacy in treating DN.
Collapse
Affiliation(s)
- Mallesh Kurakula
- Department of Biomedical Engineering, The University of Memphis, Memphis, TN 38152, USA
- Correspondence: ; Tel.: +1-901-297-7693
| | - Raghavendra Naveen N.
- Department of Pharmaceutics, Sri Adichunchanagiri College of Pharmacy, Adichunchanagiri University, B.G.Nagar 571448, Karnataka, India;
| | - Bhaumik Patel
- Product Development Department, Cure Pharmaceutical Corporation, Los Angeles, CA 90025, USA;
| | - Ravi Manne
- Chemtex Environmental Laboratory, Quality Control, and Assurance Department, Port Arthur, TX 77642, USA;
| | - Devang B. Patel
- Department of Pharmaceutical Sciences, Arnold and Marie Schwartz College of Pharmacy and Health Sciences, Long Island University, Brooklyn, NY 11201, USA;
| |
Collapse
|
35
|
Sun J, Xiao Z, Haider A, Gebhard C, Xu H, Luo HB, Zhang HT, Josephson L, Wang L, Liang SH. Advances in Cyclic Nucleotide Phosphodiesterase-Targeted PET Imaging and Drug Discovery. J Med Chem 2021; 64:7083-7109. [PMID: 34042442 DOI: 10.1021/acs.jmedchem.1c00115] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cyclic nucleotide phosphodiesterases (PDEs) control the intracellular concentrations of cAMP and cGMP in virtually all mammalian cells. Accordingly, the PDE family regulates a myriad of physiological functions, including cell proliferation, differentiation and apoptosis, gene expression, central nervous system function, and muscle contraction. Along this line, dysfunction of PDEs has been implicated in neurodegenerative disorders, coronary artery diseases, chronic obstructive pulmonary disease, and cancer development. To date, 11 PDE families have been identified; however, their distinct roles in the various pathologies are largely unexplored and subject to contemporary research efforts. Indeed, there is growing interest for the development of isoform-selective PDE inhibitors as potential therapeutic agents. Similarly, the evolving knowledge on the various PDE isoforms has channeled the identification of new PET probes, allowing isoform-selective imaging. This review highlights recent advances in PDE-targeted PET tracer development, thereby focusing on efforts to assess disease-related PDE pathophysiology and to support isoform-selective drug discovery.
Collapse
Affiliation(s)
- Jiyun Sun
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, Massachusetts 02114, United States
| | - Zhiwei Xiao
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, Massachusetts 02114, United States
| | - Achi Haider
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, Massachusetts 02114, United States
| | - Catherine Gebhard
- Department of Nuclear Medicine, University Hospital Zurich, Raemistrasse 100, Zurich 8006, Switzerland
- Center for Molecular Cardiology, University of Zurich, Wagistrasse 12, Schlieren 8952, Switzerland
| | - Hao Xu
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University, 613 West Huangpu Road, Tianhe District, Guangzhou 510630, China
| | - Hai-Bin Luo
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Han-Ting Zhang
- Departments of Neuroscience, Behavioral Medicine & Psychiatry, and Physiology & Pharmacology, the Rockefeller Neuroscience Institute, West Virginia University Health Sciences Center, Morgantown, West Virginia 26506, United States
| | - Lee Josephson
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, Massachusetts 02114, United States
| | - Lu Wang
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, Massachusetts 02114, United States
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University, 613 West Huangpu Road, Tianhe District, Guangzhou 510630, China
| | - Steven H Liang
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, Massachusetts 02114, United States
| |
Collapse
|
36
|
Alhowail A. Molecular insights into the benefits of nicotine on memory and cognition (Review). Mol Med Rep 2021; 23:398. [PMID: 33786606 PMCID: PMC8025477 DOI: 10.3892/mmr.2021.12037] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 10/13/2020] [Indexed: 01/19/2023] Open
Abstract
The health risks of nicotine are well known, but there is some evidence of its beneficial effects on cognitive function. The present review focused on the reported benefits of nicotine in the brain and summarizes the associated underlying mechanisms. Nicotine administration can improve cognitive impairment in Alzheimer's disease (AD), and dyskinesia and memory impairment in Parkinson's disease (PD). In terms of its mechanism of action, nicotine slows the progression of PD by inhibiting Sirtuin 6, a stress‑responsive protein deacetylase, thereby decreasing neuronal apoptosis and improving neuronal survival. In AD, nicotine improves cognitive impairment by enhancing protein kinase B (also referred to as Akt) activity and stimulating phosphoinositide 3‑kinase/Akt signaling, which regulates learning and memory processes. Nicotine may also activate thyroid receptor signaling pathways to improve memory impairment caused by hypothyroidism. In healthy individuals, nicotine improves memory impairment caused by sleep deprivation by enhancing the phosphorylation of calmodulin‑dependent protein kinase II, an essential regulator of cell proliferation and synaptic plasticity. Furthermore, nicotine may improve memory function through its effect on chromatin modification via the inhibition of histone deacetylases, which causes transcriptional changes in memory‑related genes. Finally, nicotine administration has been demonstrated to rescue long‑term potentiation in individuals with sleep deprivation, AD, chronic stress and hypothyroidism, primarily by desensitizing α7 nicotinic acetylcholine receptors. To conclude, nicotine has several cognitive benefits in healthy individuals, as well as in those with cognitive dysfunction associated with various diseases. However, further research is required to shed light on the effect of acute and chronic nicotine treatment on memory function.
Collapse
Affiliation(s)
- Ahmad Alhowail
- Department of Pharmacology and Toxicology, College of Pharmacy, Qassim University, Buraydah 52571, Qassim, Kingdom of Saudi Arabia
| |
Collapse
|
37
|
Zou Z, Chai M, Guo F, Fu X, Lan Y, Cao S, Liu J, Tian L, An G. MicroRNA-126 engineered muscle-derived stem cells attenuates cavernosa injury-induced erectile dysfunction in rats. Aging (Albany NY) 2021; 13:14399-14415. [PMID: 34031263 PMCID: PMC8202866 DOI: 10.18632/aging.203057] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 11/25/2020] [Indexed: 11/25/2022]
Abstract
BACKGROUND Cavernosa injury is a common cause of organic erectile dysfunction (ED), which requires safe and effective treatments. In the present study, the therapeutic efficiency of muscle-derived stem cells (MDSCs) modified with microRNA-126 (miR-126) was determined in rats with cavernosa injury. METHODS MDSCs were transfected with miR-126 and then were transplanted into rats with cavernosa injury. Erectile function, vascular function (western blot and immunofluorescence), extraction, and detection of exosomes were then undertaken. RESULTS On the 28th day after transplantation, the highest value of intra-cavernous pressure (ICP)/mean arterial pressure (MAP) in rats of miRNA-126 group (0.84 ± 0.14) was observed (Control: 0.38 ± 0.07; MDSC: 0.54 ± 0.11, Vector: 0.60 ± 0.02; respectively). Treatment of miRNA-126-modified-MDSCs remarkably strengthened vascular structure, supported by hematoxylin-eosin staining. The expression of CD31, von Willebrand Factor and vascular endothelial factors were higher than those in other groups, indicating improved vascular function. In vitro mechanism studies showed that exosomes containing miR-126 isolated from MDSCs promoted angiogenesis and attenuated apoptosis of human umbilical venous endothelial cells. Finally, insulin receptor substrate 1 and Krüppel-like factor 10 were determined as the direct target genes of miR-126. CONCLUSIONS MiR-126 engineered MDSCs notably repaired cavernosa injury in rats via vascular reconstruction by directly targeting IRS1 and KLF10, in which the exosomes secreted by MDSCs played a critical role.
Collapse
Affiliation(s)
- Zihao Zou
- Center for Reproductive Medicine, Key Laboratory for Major Obstetric Diseases of Guangdong Province, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, PR China
| | - Muyuan Chai
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, Guangdong, PR China
| | - Feixiang Guo
- Center for Reproductive Medicine, Key Laboratory for Major Obstetric Diseases of Guangdong Province, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, PR China
| | - Xin Fu
- Center for Reproductive Medicine, Key Laboratory for Major Obstetric Diseases of Guangdong Province, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, PR China
| | - Yu Lan
- Center for Reproductive Medicine, Key Laboratory for Major Obstetric Diseases of Guangdong Province, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, PR China
| | - Shuqi Cao
- Center for Reproductive Medicine, Key Laboratory for Major Obstetric Diseases of Guangdong Province, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, PR China
| | - Jianan Liu
- Center for Reproductive Medicine, Key Laboratory for Major Obstetric Diseases of Guangdong Province, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, PR China
| | - Long Tian
- Beijing Chao-Yang Hospital, Capital Medical University, Beijing, PR China
| | - Geng An
- Center for Reproductive Medicine, Key Laboratory for Major Obstetric Diseases of Guangdong Province, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, PR China
| |
Collapse
|
38
|
Duarte-Silva E, Meiry da Rocha Araújo S, Oliveira WH, Lós DB, Bonfanti AP, Peron G, de Lima Thomaz L, Verinaud L, Peixoto CA. Sildenafil Alleviates Murine Experimental Autoimmune Encephalomyelitis by Triggering Autophagy in the Spinal Cord. Front Immunol 2021; 12:671511. [PMID: 34054847 PMCID: PMC8156813 DOI: 10.3389/fimmu.2021.671511] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 04/19/2021] [Indexed: 12/15/2022] Open
Abstract
Multiple Sclerosis (MS) is a neuroinflammatory and chronic Central Nervous System (CNS) disease that affects millions of people worldwide. The search for more promising drugs for the treatment of MS has led to studies on Sildenafil, a phosphodiesterase type 5 Inhibitor (PDE5I) that has been shown to possess neuroprotective effects in the Experimental Autoimmune Encephalomyelitis (EAE), an animal model of MS. We have previously shown that Sildenafil improves the clinical score of EAE mice via modulation of apoptotic pathways, but other signaling pathways were not previously covered. Therefore, the aim of the present study was to further investigate the effects of Sildenafil treatment on autophagy and nitrosative stress signaling pathways in EAE. 24 female C57BL/6 mice were divided into the following groups: (A) Control - received only water; (B) EAE - EAE untreated mice; (C) SILD - EAE mice treated with 25mg/kg of Sildenafil s.c. The results showed that EAE mice presented a pro-nitrosative profile characterized by high tissue nitrite levels, lowered levels of p-eNOS and high levels of iNOS. Furthermore, decreased levels of LC3, beclin-1 and ATG5, suggests impaired autophagy, and decreased levels of AMPK in the spinal cord were also detected in EAE mice. Surprisingly, treatment with Sildenafil inhibited nitrosative stress and augmented the levels of LC3, beclin-1, ATG5, p-CREB and BDNF and decreased mTOR levels, as well as augmented p-AMPK. In conclusion, we propose that Sildenafil alleviates EAE by activating autophagy via the eNOS-NO-AMPK-mTOR-LC3-beclin1-ATG5 and eNOS-NO-AMPK-mTOR-CREB-BDNF pathways in the spinal cord.
Collapse
Affiliation(s)
- Eduardo Duarte-Silva
- Laboratory of Ultrastructure, Aggeu Magalhães Institute (IAM), Recife, Brazil
- Postgraduate Program in Biosciences and Biotechnology for Health (PPGBBS), Oswaldo Cruz Foundation (FIOCRUZ-PE)/Aggeu Magalhães Institute (IAM), Recife, Brazil
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Recife, Brazil
| | - Shyrlene Meiry da Rocha Araújo
- Laboratory of Ultrastructure, Aggeu Magalhães Institute (IAM), Recife, Brazil
- Postgraduate Program in Biological Sciences/Center of Biosciences, Federal University of Pernambuco (UFPE), Recife, Brazil
| | - Wilma Helena Oliveira
- Laboratory of Ultrastructure, Aggeu Magalhães Institute (IAM), Recife, Brazil
- Postgraduate Program in Biological Sciences/Center of Biosciences, Federal University of Pernambuco (UFPE), Recife, Brazil
| | - Deniele Bezerra Lós
- Postgraduate Program in Biotechnology/Northeast Network in Biotechnology (RENORBIO), Federal University of Pernambuco (UFPE), Recife, Brazil
| | - Amanda Pires Bonfanti
- Department of Structural and Functional Biology, University of Campinas (UNICAMP), Campinas, Brazil
| | - Gabriela Peron
- Department of Structural and Functional Biology, University of Campinas (UNICAMP), Campinas, Brazil
| | - Livia de Lima Thomaz
- Department of Structural and Functional Biology, University of Campinas (UNICAMP), Campinas, Brazil
| | - Liana Verinaud
- Department of Structural and Functional Biology, University of Campinas (UNICAMP), Campinas, Brazil
| | - Christina Alves Peixoto
- Laboratory of Ultrastructure, Aggeu Magalhães Institute (IAM), Recife, Brazil
- National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| |
Collapse
|
39
|
Targeting the crosstalk between canonical Wnt/β-catenin and inflammatory signaling cascades: A novel strategy for cancer prevention and therapy. Pharmacol Ther 2021; 227:107876. [PMID: 33930452 DOI: 10.1016/j.pharmthera.2021.107876] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 04/05/2021] [Indexed: 02/06/2023]
Abstract
Emerging scientific evidence indicates that inflammation is a critical component of tumor promotion and progression. Most cancers originate from sites of chronic irritation, infections and inflammation, underscoring that the tumor microenvironment is largely orchestrated by inflammatory cells and pro-inflammatory molecules. These inflammatory components are intimately involved in neoplastic processes which foster proliferation, survival, invasion, and migration, making inflammation the primary target for cancer prevention and treatment. The influence of inflammation and the immune system on the progression and development of cancer has recently gained immense interest. The Wnt/β-catenin signaling pathway, an evolutionarily conserved signaling strategy, has a critical role in regulating tissue development. It has been implicated as a major player in cancer development and progression with its regulatory role on inflammatory cascades. Many naturally-occurring and small synthetic molecules endowed with inherent anti-inflammatory properties inhibit this aberrant signaling pathway, making them a promising class of compounds in the fight against inflammatory cancers. This article analyzes available scientific evidence and suggests a crosslink between Wnt/β-catenin signaling and inflammatory pathways in inflammatory cancers, especially breast, gastrointestinal, endometrial, and ovarian cancer. We also highlight emerging experimental findings that numerous anti-inflammatory synthetic and natural compounds target the crosslink between Wnt/β-catenin pathway and inflammatory cascades to achieve cancer prevention and intervention. Current challenges, limitations, and future directions of research are also discussed.
Collapse
|
40
|
Sandner P, Zimmer DP, Milne GT, Follmann M, Hobbs A, Stasch JP. Soluble Guanylate Cyclase Stimulators and Activators. Handb Exp Pharmacol 2021; 264:355-394. [PMID: 30689085 DOI: 10.1007/164_2018_197] [Citation(s) in RCA: 98] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
When Furchgott, Murad, and Ignarro were honored with the Nobel prize for the identification of nitric oxide (NO) in 1998, the therapeutic implications of this discovery could not be fully anticipated. This was due to the fact that available therapeutics like NO donors did not allow a constant and long-lasting cyclic guanylyl monophosphate (cGMP) stimulation and had a narrow therapeutic window. Now, 20 years later, the stimulator of soluble guanylate cyclase (sGC), riociguat, is on the market and is the only drug approved for the treatment of two forms of pulmonary hypertension (PAH/CTEPH), and a variety of other sGC stimulators and sGC activators are in preclinical and clinical development for additional indications. The discovery of sGC stimulators and sGC activators is a milestone in the field of NO/sGC/cGMP pharmacology. The sGC stimulators and sGC activators bind directly to reduced, heme-containing and oxidized, heme-free sGC, respectively, which results in an increase in cGMP production. The action of sGC stimulators at the heme-containing enzyme is independent of NO but is enhanced in the presence of NO whereas the sGC activators interact with the heme-free form of sGC. These highly innovative pharmacological principles of sGC stimulation and activation seem to have a very broad therapeutic potential. Therefore, in both academia and industry, intensive research and development efforts have been undertaken to fully exploit the therapeutic benefit of these new compound classes. Here we summarize the discovery of sGC stimulators and sGC activators and the current developments in both compound classes, including the mode of action, the chemical structures, and the genesis of the terminology and nomenclature. In addition, preclinical studies exploring multiple aspects of their in vitro, ex vivo, and in vivo pharmacology are reviewed, providing an overview of multiple potential applications. Finally, the clinical developments, investigating the treatment potential of these compounds in various diseases like heart failure, diabetic kidney disease, fibrotic diseases, and hypertension, are reported. In summary, sGC stimulators and sGC activators have a unique mode of action with a broad treatment potential in cardiovascular diseases and beyond.
Collapse
Affiliation(s)
- Peter Sandner
- Bayer AG, Pharmaceuticals R&D, Pharma Research Center, Wuppertal, Germany. .,Department of Pharmacology, Hannover Medical School, Hannover, Germany.
| | | | | | - Markus Follmann
- Bayer AG, Pharmaceuticals R&D, Pharma Research Center, Wuppertal, Germany
| | - Adrian Hobbs
- Barts and the London School of Medicine and Dentistry QMUL, London, UK
| | - Johannes-Peter Stasch
- Bayer AG, Pharmaceuticals R&D, Pharma Research Center, Wuppertal, Germany.,Institute of Pharmacy, University Halle-Wittenberg, Halle, Germany
| |
Collapse
|
41
|
Sharma VK, Singh TG, Singh S. Cyclic Nucleotides Signaling and Phosphodiesterase Inhibition: Defying Alzheimer's Disease. Curr Drug Targets 2020; 21:1371-1384. [PMID: 32718286 DOI: 10.2174/1389450121666200727104728] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 06/12/2020] [Accepted: 06/13/2020] [Indexed: 12/16/2022]
Abstract
Defects in brain functions associated with aging and neurodegenerative diseases benefit insignificantly from existing options, suggesting that there is a lack of understanding of pathological mechanisms. Alzheimer's disease (AD) is such a nearly untreatable, allied to age neurological deterioration for which only the symptomatic cure is available and the agents able to mould progression of the disease, is still far away. The altered expression of phosphodiesterases (PDE) and deregulated cyclic nucleotide signaling in AD has provoked a new thought of targeting cyclic nucleotide signaling in AD. Targeting cyclic nucleotides as an intracellular messenger seems to be a viable approach for certain biological processes in the brain and controlling substantial. Whereas, the synthesis, execution, and/or degradation of cyclic nucleotides has been closely linked to cognitive deficits. In relation to cognition, the cyclic nucleotides (cAMP and cGMP) have an imperative execution in different phases of memory, including gene transcription, neurogenesis, neuronal circuitry, synaptic plasticity and neuronal survival, etc. AD is witnessed by impairments of these basic processes underlying cognition, suggesting a crucial role of cAMP/cGMP signaling in AD populations. Phosphodiesterase inhibitors are the exclusive set of enzymes to facilitate hydrolysis and degradation of cAMP and cGMP thereby, maintains their optimum levels initiating it as an interesting target to explore. The present work reviews a neuroprotective and substantial influence of PDE inhibition on physiological status, pathological progression and neurobiological markers of AD in consonance with the intensities of cAMP and cGMP.
Collapse
Affiliation(s)
- Vivek K Sharma
- Chitkara College of Pharmacy, Chitkara University, Punjab, India,Govt. College of Pharmacy, Rohru, District Shimla, Himachal Pradesh-171207, India
| | - Thakur G Singh
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Shareen Singh
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| |
Collapse
|
42
|
The Phosphodiesterase-5 Inhibitor Vardenafil Improves the Activation of BMP Signaling in Response to Hydrogen Peroxide. Cardiovasc Drugs Ther 2020; 34:41-52. [PMID: 32096002 DOI: 10.1007/s10557-020-06939-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE The pleiotropic roles of phosphodiesterase-5 inhibitors (PDE5is) in cardiovascular diseases have attracted attention. The effect of vardenafil (a PDE5i) is partly mediated through reduced oxidative stress, but it is unclear whether vardenafil protects against hydrogen peroxide (H2O2)-induced endothelial cell injury, and the molecular mechanisms that are involved remain unknown. We determined the protective role of vardenafil on H2O2-induced endothelial cell injury in cultured human umbilical vein endothelial cells (HUVECs). METHODS AND RESULTS Vardenafil decreased the number of TUNEL-positive cells, increased the Bcl2/Bax ratio, and ameliorated the numbers of BrdU-positive cells in H2O2-treated HUVECs. The bone morphogenetic protein receptor (BMPR)/p-Smad/MSX2 pathway was enhanced in response to H2O2, and vardenafil treatment could normalize this pathway. To determine whether the BMP pathway is involved, we blocked the BMP pathway using dorsomorphin, which abolished the protective effects of vardenafil. We found that vardenafil improved the H2O2-induced downregulation of BMP-binding endothelial regulator protein (BMPER), which possibly intersects with the BMP pathway in the regulation of endothelial cell injury in response to oxidative stress. CONCLUSIONS We demonstrated for the first time that exogenous H2O2 activates BMPR expression and promotes Smad1/5/8 phosphorylation. Additionally, vardenafil can attenuate H2O2-induced endothelial cell injury in HUVECs. Vardenafil decreases apoptosis through an improved Bcl-2/Bax ratio and increases cell proliferation. Vardenafil protects against endothelial cell injury through ameliorating the intracellular oxidative stress level and BMPER expression. The protective role of vardenafil on H2O2-induced endothelial cell injury is mediated through BMPR/p-Smad/MSX2 in HUVECs.
Collapse
|
43
|
Arenas YM, Cabrera-Pastor A, Juciute N, Mora-Navarro E, Felipo V. Blocking glycine receptors reduces neuroinflammation and restores neurotransmission in cerebellum through ADAM17-TNFR1-NF-κβ pathway. J Neuroinflammation 2020; 17:269. [PMID: 32917219 PMCID: PMC7488331 DOI: 10.1186/s12974-020-01941-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 08/24/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Chronic hyperammonemia induces neuroinflammation in cerebellum, with glial activation and enhanced activation of the TNFR1-NF-kB-glutaminase-glutamate-GABA pathway. Hyperammonemia also increases glycinergic neurotransmission. These alterations contribute to cognitive and motor impairment. Activation of glycine receptors is reduced by extracellular cGMP, which levels are reduced in cerebellum of hyperammonemic rats in vivo. We hypothesized that enhanced glycinergic neurotransmission in hyperammonemic rats (1) contributes to induce neuroinflammation and glutamatergic and GABAergic neurotransmission alterations; (2) is a consequence of the reduced extracellular cGMP levels. The aims were to assess, in cerebellum of hyperammonemic rats, (a) whether blocking glycine receptors with the antagonist strychnine reduces neuroinflammation; (b) the cellular localization of glycine receptor; (c) the effects of blocking glycine receptors on the TNFR1-NF-kB-glutaminase-glutamate-GABA pathway and microglia activation; (d) whether adding extracellular cGMP reproduces the effects of strychnine. METHODS We analyzed in freshly isolated cerebellar slices from control or hyperammonemic rats the effects of strychnine on activation of microglia and astrocytes, the content of TNFa and IL1b, the surface expression of ADAM17, TNFR1 and transporters, the phosphorylation levels of ERK, p38 and ADAM17. The cellular localization of glycine receptor was assessed by immunofluorescence. We analyzed the content of TNFa, IL1b, HMGB1, glutaminase, and the level of TNF-a mRNA and NF-κB in Purkinje neurons. Extracellular concentrations of glutamate and GABA were performed by in vivo microdialysis in cerebellum. We tested whether extracellular cGMP reproduces the effects of strychnine in ex vivo cerebellar slices. RESULTS Glycine receptors are expressed mainly in Purkinje cells. In hyperammonemic rats, enhanced glycinergic neurotransmission leads to reduced membrane expression of ADAM17, resulting in increased surface expression and activation of TNFR1 and of the associated NF-kB pathway. This increases the expression in Purkinje neurons of TNFa, IL-1b, HMGB1, and glutaminase. Increased glutaminase activity leads to increased extracellular glutamate, which increases extracellular GABA. Increased extracellular glutamate and HMGB1 potentiate microglial activation. Blocking glycine receptors with strychnine or extracellular cGMP completely prevents the above pathway in hyperammonemic rats. CONCLUSIONS Glycinergic neurotransmission modulates neuroinflammation. Enhanced glycinergic neurotransmission in hyperammonemia would be due to reduced extracellular cGMP. These results shed some light on possible new therapeutic target pathways for pathologies associated to neuroinflammation.
Collapse
Affiliation(s)
- Yaiza M Arenas
- Laboratory of Neurobiology, Príncipe Felipe Research Center Valencia, C/Eduardo Primo Yúfera 3, 46012, Valencia, Spain
| | - Andrea Cabrera-Pastor
- Laboratory of Neurobiology, Príncipe Felipe Research Center Valencia, C/Eduardo Primo Yúfera 3, 46012, Valencia, Spain.
- Laboratory of Neurological Impairment, Health Research Institute INCLIVA, 46010, Valencia, Spain.
| | - Nora Juciute
- Laboratory of Neurobiology, Príncipe Felipe Research Center Valencia, C/Eduardo Primo Yúfera 3, 46012, Valencia, Spain
| | - Eloy Mora-Navarro
- Laboratory of Neurobiology, Príncipe Felipe Research Center Valencia, C/Eduardo Primo Yúfera 3, 46012, Valencia, Spain
| | - Vicente Felipo
- Laboratory of Neurobiology, Príncipe Felipe Research Center Valencia, C/Eduardo Primo Yúfera 3, 46012, Valencia, Spain
| |
Collapse
|
44
|
Hernández RB, Carrascal M, Abian J, Michalke B, Farina M, Gonzalez YR, Iyirhiaro GO, Moteshareie H, Burnside D, Golshani A, Suñol C. Manganese-induced neurotoxicity in cerebellar granule neurons due to perturbation of cell network pathways with potential implications for neurodegenerative disorders. Metallomics 2020; 12:1656-1678. [PMID: 33206086 DOI: 10.1039/d0mt00085j] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Manganese (Mn) is essential for living organisms, playing an important role in nervous system function. Nevertheless, chronic and/or acute exposure to this metal, especially during early life stages, can lead to neurotoxicity and dementia by unclear mechanisms. Thus, based on previous works of our group with yeast and zebrafish, we hypothesized that the mechanisms mediating manganese-induced neurotoxicity can be associated with the alteration of protein metabolism. These mechanisms may also depend on the chemical speciation of manganese. Therefore, the current study aimed at investigating the mechanisms mediating the toxic effects of manganese in primary cultures of cerebellar granule neurons (CGNs). By exposing cultured CGNs to different chemical species of manganese ([[2-[(dithiocarboxy)amino]ethyl]carbamodithioato]](2-)-kS,kS']manganese, named maneb (MB), and [[1,2-ethanediylbis[carbamodithioato]](2-)]manganese mixture with [[1,2-ethanediylbis[carbamodithioato]](2-)]zinc, named mancozeb (MZ), and manganese chloride (MnCl2)), and using the MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay, we observed that both MB and MZ induced similar cytotoxicity (LC50∼ 7-9 μM), which was higher than that of MnCl2 (LC50∼ 27 μM). Subsequently, we applied systems biology approaches, including metallomics, proteomics, gene expression and bioinformatics, and revealed that independent of chemical speciation, for non-cytotoxic concentrations (0.3-3 μM), Mn-induced neurotoxicity in CGNs is associated with metal dyshomeostasis and impaired protein metabolism. In this way, we verified that MB induced more post-translational alterations than MnCl2, which can be a plausible explanation for cytotoxic differences between both chemical species. The metabolism of proteins is one of the most energy consuming cellular processes and its impairment appears to be a key event of some cellular stress processes reported separately in other studies such as cell cycle arrest, energy impairment, cell signaling, excitotoxicity, immune response, potential protein accumulation and apoptosis. Interestingly, we verified that Mn-induced neurotoxicity shares pathways associated with the development of Alzheimer's disease, Amyotrophic Lateral Sclerosis, Huntington's disease, and Parkinson's disease. This has been observed in baker's yeast and zebrafish suggesting that the mode of action of Mn may be evolutionarily conserved.
Collapse
Affiliation(s)
- Raúl Bonne Hernández
- Laboratory of Bioinorganic and Environmental Toxicology - LABITA, Department of Exact and Earth Sciences, Federal University of São Paulo, Rua Prof. Artur Riedel, 275, CEP 09972-270, Diadema, SP, Brazil.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
dos Santos Maia M, Rodrigues GCS, de Sousa NF, Scotti MT, Scotti L, Mendonça-Junior FJB. Identification of New Targets and the Virtual Screening of Lignans against Alzheimer's Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:3098673. [PMID: 32879651 PMCID: PMC7448245 DOI: 10.1155/2020/3098673] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 06/22/2020] [Accepted: 07/17/2020] [Indexed: 02/08/2023]
Abstract
Alzheimer's disease (AD) is characterized by the progressive disturbance in cognition and affects approximately 36 million people, worldwide. However, the drugs used to treat this disease are only moderately effective and do not alter the course of the neurodegenerative process. This is because the pathogenesis of AD is mainly associated with oxidative stress, and current drugs only target two enzymes involved in neurotransmission. Therefore, the present study sought to identify potential multitarget compounds for enzymes that are directly or indirectly involved in the oxidative pathway, with minimal side effects, for AD treatment. A set of 159 lignans were submitted to studies of QSAR and molecular docking. A combined analysis was performed, based on ligand and structure, followed by the prediction of absorption, distribution, metabolism, excretion, and toxicity (ADMET) properties. The results showed that the combined analysis was able to select 139 potentially active and multitarget lignans targeting two or more enzymes, among them are c-Jun N-terminal kinase 3 (JNK-3), protein tyrosine phosphatase 1B (PTP1B), nicotinamide adenine dinucleotide phosphate oxidase 1 (NOX1), NADPH quinone oxidoreductase 1 (NQO1), phosphodiesterase 5 (PDE5), nuclear factor erythroid 2-related factor 2 (Nrf2), cycloxygenase 2 (COX-2), and inducible nitric oxide synthase (iNOS). The authors conclude that compounds (06) austrobailignan 6, (11) anolignan c, (19) 7-epi-virolin, (64) 6-[(2R,3R,4R,5R)-3,4-dimethyl-5-(3,4,5-trimethoxyphenyl)oxolan-2-yl]-4-methoxy-1,3-benzodioxole, (116) ococymosin, and (135) mappiodoinin b have probabilities that confer neuroprotection and antioxidant activity and represent potential alternative AD treatment drugs or prototypes for the development of new drugs with anti-AD properties.
Collapse
Affiliation(s)
- Mayara dos Santos Maia
- Laboratory of Cheminformatics, Program of Natural and Synthetic Bioactive Products (PgPNSB), Health Sciences Center, Federal University of Paraíba, João Pessoa, PB, Brazil
| | - Gabriela Cristina Soares Rodrigues
- Laboratory of Cheminformatics, Program of Natural and Synthetic Bioactive Products (PgPNSB), Health Sciences Center, Federal University of Paraíba, João Pessoa, PB, Brazil
| | - Natália Ferreira de Sousa
- Laboratory of Cheminformatics, Program of Natural and Synthetic Bioactive Products (PgPNSB), Health Sciences Center, Federal University of Paraíba, João Pessoa, PB, Brazil
| | - Marcus Tullius Scotti
- Laboratory of Cheminformatics, Program of Natural and Synthetic Bioactive Products (PgPNSB), Health Sciences Center, Federal University of Paraíba, João Pessoa, PB, Brazil
| | - Luciana Scotti
- Laboratory of Cheminformatics, Program of Natural and Synthetic Bioactive Products (PgPNSB), Health Sciences Center, Federal University of Paraíba, João Pessoa, PB, Brazil
| | | |
Collapse
|
46
|
The Potent PDE10A Inhibitor MP-10 (PF-2545920) Suppresses Microglial Activation in LPS-Induced Neuroinflammation and MPTP-Induced Parkinson’s Disease Mouse Models. J Neuroimmune Pharmacol 2020; 16:470-482. [DOI: 10.1007/s11481-020-09943-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Accepted: 07/08/2020] [Indexed: 12/12/2022]
|
47
|
Effect of sildenafil on neuroinflammation and synaptic plasticity pathways in experimental autoimmune encephalomyelitis. Int Immunopharmacol 2020; 85:106581. [PMID: 32442900 DOI: 10.1016/j.intimp.2020.106581] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 04/17/2020] [Accepted: 05/07/2020] [Indexed: 12/25/2022]
Abstract
Multiple sclerosis (MS) is a chronic immuno-inflammatory disease of the central nervous system characterized by demyelination and axonal damage. Cognitive changes are common in individuals with MS since inflammatory molecules secreted by microglia interfere with the physiological mechanisms of synaptic plasticity. According to previous data, inhibition of PDE5 promotes the accumulation of cGMP, which inhibits neuroinflammation and seems to improve synaptic plasticity and memory. The present study aimed to evaluate the effect of sildenafil on the signaling pathways of neuroinflammation and synaptic plasticity in experimental autoimmune encephalomyelitis (EAE). C57BL/6 mice were divided into three experimental groups (n = 10/group): (a) Control; (b) EAE; (c) EAE + sild (25 mg/kg/21 days). Sildenafil was able to delay the onset and attenuate the severity of the clinical symptoms of EAE. The drug also reduced the infiltration of CD4+ T lymphocytes and their respective IL-17 and TNF-α cytokines. Moreover, sildenafil reduced neuroinflammation in the hippocampus (assessed by the reduction of inflammatory markers IL-1β, pIKBα and pNFkB and reactive gliosis, as well as elevating the inhibitory cytokines TGF-β and IL-10). Moreover, sildenafil induced increased levels of NeuN, BDNF and pCREB, protein kinases (PKA, PKG, and pERK) and synaptophysin, and modulated the expression of the glutamate receptors AMPA and NMDA. The present findings demonstrated that sildenafil has therapeutic potential for cognitive deficit associated with multiple sclerosis.
Collapse
|
48
|
Rubin LH, Xu Y, Norris PJ, Wang X, Dastgheyb R, Fitzgerald KC, Keating SM, Kaplan RC, Maki PM, Anastos K, Springer G, Benning L, Kassaye S, Gustafson DR, Valcour VG, Williams DW. Early Inflammatory Signatures Predict Subsequent Cognition in Long-Term Virally Suppressed Women With HIV. Front Integr Neurosci 2020; 14:20. [PMID: 32390808 PMCID: PMC7193823 DOI: 10.3389/fnint.2020.00020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 03/23/2020] [Indexed: 12/16/2022] Open
Abstract
Immunologic function is an important determinant of cognition. Here we examined the contribution of early immune signatures to cognitive performance among HIV-infected, virally suppressed women (HIV+VS) and in HIV-uninfected (HIV-) women. Specifically, we measured serum inflammatory markers, developed combinatory immune signatures, and evaluated their associations with cognition. Forty-nine HIV+VS women in the Women’s Interagency HIV Study (WIHS) who achieved viral suppression shortly after effective antiretroviral therapy (ART) initiation, and 56 matched HIV− women were selected. Forty-two serum inflammatory markers were measured within 2 years of effective ART initiation for HIV+VS women, and at an initial timepoint for HIV− women. The same inflammatory markers were also measured approximately 1, 7, and 12 years later for all women. Of the 105 women with complete immune data, 83 (34 HIV+VS, 49 HIV−) also had cognitive data available 12 years later at ≥1 time points (median = 3.1). We searched for combinatory immune signatures by adapting a dynamic matrix factorization analytic method that builds upon Tucker decomposition followed by Ingenuity® Pathway Analysis to facilitate data interpretation. Seven combinatory immune signatures emerged based on the Frobenius residual. Three signatures were common between HIV+VS and HIV− women, while four signatures were unique. These inflammatory signatures predicted subsequent cognitive performance in both groups using mixed-effects modeling, but more domain-specific associations were significant in HIV+VS than HIV− women. Leukocyte influx into brain was a major contributor to cognitive function in HIV+VS women, while T cell exhaustion, inflammatory response indicative of depressive/psychiatric disorders, microglial activity, and cytokine signaling predicted both global and domain-specific performance for HIV− women. Our findings suggest that immune signatures may be useful diagnostic, prognostic, and immunotherapeutic targets predictive of subsequent cognitive performance. Importantly, they also provide insight into common and distinct inflammatory mechanisms underlying cognition in HIV− and HIV+VS women.
Collapse
Affiliation(s)
- Leah H Rubin
- Department of Neurology, Johns Hopkins University, Baltimore, MD, United States.,Department of Psychiatry, Johns Hopkins University, Baltimore, MD, United States.,Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, United States
| | - Yanxun Xu
- Department of Applied Mathematics and Statistics, Johns Hopkins University, Baltimore, MD, United States.,Division of Biostatistics and Bioinformatics, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, United States
| | - Philip J Norris
- Department of Laboratory Medicine, Vitalant Research Institute, University of California, San Francisco, San Francisco, CA, United States
| | - Xuzhi Wang
- Department of Applied Mathematics and Statistics, Johns Hopkins University, Baltimore, MD, United States
| | - Raha Dastgheyb
- Department of Neurology, Johns Hopkins University, Baltimore, MD, United States
| | | | - Sheila M Keating
- Department of Laboratory Medicine, Vitalant Research Institute, University of California, San Francisco, San Francisco, CA, United States
| | - Robert C Kaplan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Pauline M Maki
- Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, United States.,Department of Psychology, University of Illinois at Chicago, Chicago, IL, United States
| | - Kathryn Anastos
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, United States.,Department of General Internal Medicine, Albert Einstein College of Medicine, Bronx, NY, United States.,Department of Obstetrics and Gynecology and Women's Health, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Gayle Springer
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, United States
| | - Lorie Benning
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, United States
| | - Seble Kassaye
- Department of Medicine, Georgetown University, Washington, DC, United States
| | - Deborah R Gustafson
- Department of Neurology, SUNY Downstate Health Sciences University, Brooklyn, NY, United States
| | - Victor G Valcour
- Department of Neurology, University of California, San Francisco, San Francisco, CA, United States
| | - Dionna W Williams
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University, Baltimore, MD, United States.,Division of Clinical Pharmacology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
49
|
Kammoun AK, Khedr A, Ahmed OAA. LC-MS/MS determination of avanafil and its metabolites in rat plasma and brain: pharmacokinetic study after oral administration and transdermal film application. RSC Adv 2020; 10:9407-9413. [PMID: 35497213 PMCID: PMC9050051 DOI: 10.1039/d0ra00569j] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Accepted: 02/26/2020] [Indexed: 01/20/2023] Open
Abstract
Avanafil (AVA) has been FDA approved in 2012 as a phosphodiesterase-type five inhibitor drug (PDE-5), for the treatment of erectile dysfunction (ED). It was necessary to study the pharmacokinetics and bioavailability parameters of AVA since it exhibits side effects, a long time from drug administration. As a result of this, we described a sensitive high-performance-liquid chromatography-triple quad-mass spectrometric method (LC-QqQ-MS) for the analysis of AVA in rat plasma and brain. Furthermore, the concentrations of AVA and its primary metabolites were determined in rat brain since it is known that PDE-5 inhibitor drugs are capable of crossing the blood-brain barrier (BBB). The liquid-liquid extraction method was developed, optimized, and applied for maximum recovery of AVA from plasma and brain homogenates. The percentage of recovery was 96.60 ± 2.44% and 94.50 ± 1.86%, in rat plasma and brain homogenate, respectively. The separation was performed on a Nucleodur C18 column, with mobile phase composed of 0.1% formic acid and acetonitrile (29 : 71, v/v), at flow rate 0.5 mL min-1, and monitored with QqQ-MS applying positive multiple reaction monitoring (MRM) mode. The calculated pharmacokinetic parameters, noncompartmental model, were: C max 1503.82 ± 354.11 ng mL-1 with a t 1/2 value of 4.87 ± 0.42 h and C max 141.94 ± 22.57 ng mL-1 with a t 1/2 value of 7.05 ± 1.59 h, for oral AVA suspension and transdermal film, respectively. The average percentage of total metabolites in plasma and brain was 27.1 ± 2.2% and 7.0 ± 1.0%, respectively.
Collapse
Affiliation(s)
- Ahmed K Kammoun
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, King Abdulaziz University Jeddah 21589 P. O. Box 80260 Saudi Arabia
| | - Alaa Khedr
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, King Abdulaziz University Jeddah 21589 P. O. Box 80260 Saudi Arabia
| | - Osama A A Ahmed
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University Jeddah 21589 P. O. Box 80260 Saudi Arabia
- Department of Pharmaceutics and Industrial, Faculty of Pharmacy, Minia University Minia Egypt
| |
Collapse
|
50
|
Duarte-Silva E, Filho AJMC, Barichello T, Quevedo J, Macedo D, Peixoto C. Phosphodiesterase-5 inhibitors: Shedding new light on the darkness of depression? J Affect Disord 2020; 264:138-149. [PMID: 32056743 DOI: 10.1016/j.jad.2019.11.114] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 11/22/2019] [Accepted: 11/26/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND Phosphodiesterase-5 inhibitors (PDE5Is) are used to treat erectile dysfunction (ED). Recently, the antidepressant-like effect of PDE5Is was demonstrated in animal models of depression. In clinical settings, PDE5Is were studied only for ED associated depression. Hence, there are no studies evaluating the effects of PDE5Is for the treatment of major depressive disorder (MDD) without ED. In this review article, we aimed to discuss the use of PDE5Is in the context of MDD, highlighting the roles of PDE genes in the development of MDD, the potential mechanisms by which PDE5Is can be beneficial for MDD and the potentials and limitations of PDE5Is repurposing to treat MDD. METHODS We used PubMed (MEDLINE) database to collect the studies cited in this review. Papers written in English language regardless the year of publication were selected. RESULTS A few preclinical studies support the antidepressant-like activity of PDE5Is. Clinical studies in men with ED and depression suggest that PDE5Is improve depressive symptoms. No clinical studies were conducted in subjects suffering from depression without ED. Antidepressant effect of PDE5Is may be explained by multiple mechanisms including inhibition of brain inflammation and modulation of neuroplasticity. LIMITATIONS The low number of preclinical and absence of clinical studies to support the antidepressant effect of PDE5Is. CONCLUSIONS No clinical trial was conducted to date evaluating PDE5Is in depressed patients without ED. PDE5Is' anti-inflammatory and neuroplasticity mechanisms may justify the potential antidepressant effect of these drugs. Despite this, clinical trials evaluating their efficacy in depressed patients need to be conducted.
Collapse
Affiliation(s)
- Eduardo Duarte-Silva
- Laboratory of Ultrastructure, Aggeu Magalhães Institute (IAM), Oswaldo Cruz Foundation (FIOCRUZ-PE), Recife, PE, Brazil; Graduate Program in Biosciences and Biotechnology for Health (PPGBBS), Aggeu Magalhães Institute (IAM), Recife, PE, Brazil.
| | - Adriano José Maia Chaves Filho
- Neuropsychopharmacology Laboratory, Drug Research and Development Center, Faculty of Medicine, Universidade Federal do Ceará, Fortaleza, CE, Brazil
| | - Tatiana Barichello
- Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, 1941 East Road, Houston, TX 77054, United States; Laboratory of Neurosciences, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina-UNESC, Criciúma, SC, Brazil; Center of Excellence on Mood Disorders, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States.
| | - João Quevedo
- Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, 1941 East Road, Houston, TX 77054, United States; Laboratory of Neurosciences, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina-UNESC, Criciúma, SC, Brazil; Center of Excellence on Mood Disorders, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States.
| | - Danielle Macedo
- Neuropsychopharmacology Laboratory, Drug Research and Development Center, Faculty of Medicine, Universidade Federal do Ceará, Fortaleza, CE, Brazil; Department of Physiology and Pharmacology, Faculty of Medicine, Universidade Federal do Ceará, Fortaleza, CE, Brazil; National Institute for Translational Medicine (INCT-TM, CNPq), Ribeirão Preto, Brazil
| | - Christina Peixoto
- Laboratory of Ultrastructure, Aggeu Magalhães Institute (IAM), Oswaldo Cruz Foundation (FIOCRUZ-PE), Recife, PE, Brazil; National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil.
| |
Collapse
|