1
|
Banerjee A, Thekkekkara D, Manjula SN, Nair SP, Lalitha MS. Correlation of autophagy and Alzheimer's disease with special emphasis on the role of phosphodiesterase-4. 3 Biotech 2025; 15:139. [PMID: 40292249 PMCID: PMC12018668 DOI: 10.1007/s13205-025-04306-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Accepted: 04/02/2025] [Indexed: 04/30/2025] Open
Abstract
Autophagy disruption is important in Alzheimer's disease (AD) as it prevents misfolded proteins from being removed, which leads to the accumulation of amyloid plaques and neurofibrillary tangles (NFTs). Restoring autophagy improves neuronal survival and cognitive function, according to experimental models. In AD models, mTOR inhibition and AMPK activation enhance synaptic plasticity and lessen learning deficits. Inhibitors of phosphodiesterase-4 (PDE4) improve cognition and reduce neuroinflammation via altering cyclic adenosine monophosphate (cAMP) transmission. Furthermore, autophagic-lysosomal clearance is encouraged by upregulating transcription factor EB (TFEB), which lessens the pathogenic damage linked to AD. These results point to autophagy modification as a promising therapeutic approach, with the mTOR, AMPK, cAMP, and TFEB pathways being possible targets for drugs. Though much evidence is based on animal studies, these findings provide valuable insights into autophagy's role in AD pathology, offering promising directions for future research and drug development.
Collapse
Affiliation(s)
- Aniruddha Banerjee
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysore, Karnataka 570 015 India
| | - Dithu Thekkekkara
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysore, Karnataka 570 015 India
| | - S. N. Manjula
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysore, Karnataka 570 015 India
| | - Salini P. Nair
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysore, Karnataka 570 015 India
| | - Mankala Sree Lalitha
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysore, Karnataka 570 015 India
| |
Collapse
|
2
|
Monaco M, Torazza C, Fedele E, Grilli M. The Impact of the Exposome on Alzheimer's Disease: The Influence of Nutrition. Int J Mol Sci 2025; 26:3015. [PMID: 40243652 PMCID: PMC11988514 DOI: 10.3390/ijms26073015] [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: 02/14/2025] [Revised: 03/18/2025] [Accepted: 03/24/2025] [Indexed: 04/18/2025] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease characterized by cognitive decline, memory loss, and behavioural changes. While genetic predispositions and pathological processes have been the traditional focus, this review highlights the fundamental role of environmental factors, particularly nutrition, within the exposome framework in modulating the risk and progression of AD. The exposome, which includes the totality of environmental exposures in an individual's lifetime, provides a comprehensive approach to understanding the complex aetiology of AD. In this review, we explore the impact of dietary factors and cyclic nucleotide pathways (cAMP/cGMP) on AD, emphasizing the potential of dietary interventions as therapeutic strategies. We investigate key aspects of how nutrition affects the accumulation of β-amyloid, the aggregation of tau proteins, and neuroinflammation. We also examine the impact of specific nutrients on cognitive performance and the risk of AD. Additionally, we discuss the potential of nutraceuticals with anti-phosphodiesterase activity and the role of various animal models of AD (such as 5xFAD, 3xTg-AD, Tg2576, and APP/PS1 mice) in demonstrating the effects of dietary interventions on disease onset and progression.
Collapse
Affiliation(s)
- Martina Monaco
- Pharmacology and Toxicology Unit, Department of Pharmacy, School of Medical and Pharmaceutical Sciences, University of Genoa, Viale Cembrano 4, 16148 Genoa, Italy; (M.M.); (C.T.); (E.F.)
| | - Carola Torazza
- Pharmacology and Toxicology Unit, Department of Pharmacy, School of Medical and Pharmaceutical Sciences, University of Genoa, Viale Cembrano 4, 16148 Genoa, Italy; (M.M.); (C.T.); (E.F.)
| | - Ernesto Fedele
- Pharmacology and Toxicology Unit, Department of Pharmacy, School of Medical and Pharmaceutical Sciences, University of Genoa, Viale Cembrano 4, 16148 Genoa, Italy; (M.M.); (C.T.); (E.F.)
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Massimo Grilli
- Pharmacology and Toxicology Unit, Department of Pharmacy, School of Medical and Pharmaceutical Sciences, University of Genoa, Viale Cembrano 4, 16148 Genoa, Italy; (M.M.); (C.T.); (E.F.)
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| |
Collapse
|
3
|
Sun R, Han M, Lin Y, Ma S, Tu H, Yang X, Zhang F, Zhang HT. Inhibition of PDE4B ameliorates cognitive defects in the model of alcoholic dementia in 3xTg-AD mice via PDE4B/cAMP/PKA signaling. Int J Neuropsychopharmacol 2025; 28:pyaf009. [PMID: 39921664 PMCID: PMC11923544 DOI: 10.1093/ijnp/pyaf009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 02/07/2025] [Indexed: 02/10/2025] Open
Abstract
BACKGROUND Chronic, heavy alcohol use may lead to permanent brain damage, cognitive impairment, and dementia. One of the most serious consequences is alcoholic dementia (AlD). Phosphodiesterase-4 (PDE4) inhibitors have been shown to exhibit beneficial effects on cognition deficits and alcoholism. However, it is not known whether PDE4 inhibitors can be used to treat AlD. A33, a relatively selective PDE4B inhibitor, is absent of the emetic effect associated with PDE4D. The effect of A33 on memory and cognition in AlD remains unclear. METHODS We investigated the effects of A33 and the PDE4 inhibitor rolipram on memory and cognition using an AlD animal model, that is, APP/PS1/Tau mice drinking alcohol in the 2-bottle choice test, with or without A33 or rolipram treatment for 3 weeks. The animal groups were compared in behavioral tests related to learning and memory. Neurochemical measures were conducted to explore the underlying mechanism of A33. RESULTS Compared to wild-type controls, AlD mice showed impairments of learning ability and memory in the behavior tests; this was attenuated by treatment of rolipram or A33. In addition, administration of rolipram or A33 in AlD mice further alleviated neuropathological alterations in the hippocampus, including Aβ expression and deposition; rolipram or A33 also decreased the levels of inflammatory cytokines, including interleukin-1β (IL-1β), interleukin-6 (IL-6), and tumor necrosis factor-α (TNF-α), as well as nuclear factor kappa-B (NF-κB). Further, rolipram or A33 decreased the activation of microglia while increased cyclic adenosine monophosphate (cAMP) levels in the hippocampus of AlD mice. CONCLUSIONS These results revealed that the alleviation of the cognitive impairment of AlD in APP/PS1/Tau triple transgenic mice by rolipram or A33 was linked to the action of the PDE4B/cAMP/PKA signaling pathway. A33 can be a promising therapeutic agent for AlD-related cognitive dysfunction.
Collapse
Affiliation(s)
- Rongzhen Sun
- Department of Pharmacology, Qingdao University School of Pharmacy, Qingdao, China
| | - Mei Han
- Department of Pharmacology, Qingdao University School of Pharmacy, Qingdao, China
| | - Yuanyuan Lin
- Department of Pharmacology, Qingdao University School of Pharmacy, Qingdao, China
| | - Shengyao Ma
- Department of Pharmacology, Qingdao University School of Pharmacy, Qingdao, China
| | - Huan Tu
- Department of Pharmacology, Qingdao University School of Pharmacy, Qingdao, China
| | - Xueliang Yang
- Department of Pharmacology, Qingdao University School of Pharmacy, Qingdao, China
| | - Fang Zhang
- Department of Pharmacology, Qingdao University School of Pharmacy, Qingdao, China
| | - Han-Ting Zhang
- Department of Pharmacology, Qingdao University School of Pharmacy, Qingdao, China
| |
Collapse
|
4
|
Kumari S, Bagri K, Deshmukh R. Connecting dots: Preclinical foundations to clinical realities of PDE4 inhibitors in Alzheimer's disease. Inflammopharmacology 2025; 33:593-603. [PMID: 39808238 DOI: 10.1007/s10787-024-01638-1] [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: 12/04/2024] [Accepted: 12/23/2024] [Indexed: 01/16/2025]
Abstract
Alzheimer's Disease (AD), a progressive and age-associated neurodegenerative disorder, is primarily characterized by amyloid-beta (Aβ) plaques and neurofibrillary tangles. Despite advances in targeting Aβ-mediated neuronal damage with anti-Aβ antibodies, these treatments provide only symptomatic relief and fail to address the multifactorial pathology of the disease. This necessitates the exploration of novel therapeutic approaches and a deeper understanding of molecular signaling mechanisms underlying AD. Phosphodiesterases (PDEs), particularly Phosphodiesterase 4 (PDE4), play a pivotal role in regulating cyclic adenosine monophosphate (cAMP), a key molecule involved in memory consolidation and cognitive function. PDE4 inhibitors have demonstrated potential in enhancing memory and cognition in preclinical models of AD by modulating cAMP signaling. However, their clinical translation has been limited due to challenges such as adverse effects, narrow therapeutic windows, and low specificity in mechanism of action. This review bridges the gap between preclinical discoveries and clinical applications of PDE4 inhibitors in AD. It highlights preclinical evidence supporting the neuroprotective and anti-inflammatory effects of PDE4 inhibitors while addressing challenges in their clinical development, including issues of safety, efficacy, and disease-specific targeting. By integrating findings from both preclinical and clinical studies, we provide a comprehensive understanding of the therapeutic potential of PDE4 inhibitors in AD. Furthermore, this review outlines future research directions aimed at optimizing PDE4 inhibition strategies for AD treatment, offering a roadmap to translate foundational insights into clinical realities.
Collapse
Affiliation(s)
- Shilpa Kumari
- Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab Technical University, Bathinda, 151001, Punjab, India
- Department of Pharmacology, Central University of Punjab, Bathinda, 151001, Punjab, India
| | - Kajal Bagri
- Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab Technical University, Bathinda, 151001, Punjab, India
- Department of Pharmacology, Central University of Punjab, Bathinda, 151001, Punjab, India
| | - Rahul Deshmukh
- Department of Pharmacology, Central University of Punjab, Bathinda, 151001, Punjab, India.
| |
Collapse
|
5
|
Zhong J, Yu X, Lin Z. Phosphodiesterase 4 inhibition as a novel treatment for stroke. PeerJ 2025; 13:e18905. [PMID: 39897494 PMCID: PMC11786714 DOI: 10.7717/peerj.18905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 01/06/2025] [Indexed: 02/04/2025] Open
Abstract
The incidence of stroke ranks third among the leading causes of mortality worldwide. It has the characteristics of high morbidity, high disability rate and high recurrence rate. The current risk associated with stroke surgery is exceedingly high. It may potentially outweigh the benefits and fail to ameliorate the cerebral tissue damage following ischemia. Therefore, pharmacological intervention assumes paramount importance. The use of thrombolytic drugs is most common in the treatment of stroke; however, its efficacy is limited due to its time-sensitive nature and propensity for increased bleeding. Over the past few years, the treatment of stroke has witnessed a surge in interest towards neuroprotective drugs that possess the potential to enhance neurological function. The PDE4D gene has been demonstrated to have a positive correlation with the risk of ischemic stroke. Additionally, the utilization of phosphodiesterase 4 inhibitors can enhance synaptic plasticity within the neural circuitry, regulate cellular metabolism, and prevent secondary brain injury caused by impaired blood flow. These mechanisms collectively facilitate the recovery of functional neurons, thereby serving as potential therapeutic interventions. Therefore, the comprehensive investigation of phosphodiesterase 4 as an innovative pharmacological target for stroke injury provides valuable insights into the development of therapeutic interventions in stroke treatment. This review is intended for, but not limited to, pharmacological researchers, drug target researchers, neurologists, neuromedical researchers, and behavioral scientists.
Collapse
Affiliation(s)
- Jiahong Zhong
- Department of Clinical Pharmacy, Meizhou People’s Hospital, Meizhou, Guangdong, China
| | - Xihui Yu
- Department of Pharmacy, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Zhuomiao Lin
- Department of Clinical Pharmacy, Meizhou People’s Hospital, Meizhou, Guangdong, China
| |
Collapse
|
6
|
Kurakin S, Ivankov O, Dushanov E, Murugova T, Ermakova E, Efimov S, Mukhametzyanov T, Smerdova S, Klochkov V, Kuklin A, Kučerka N. Calcium ions do not influence the Aβ(25-35) triggered morphological changes of lipid membranes. Biophys Chem 2024; 313:107292. [PMID: 39018778 DOI: 10.1016/j.bpc.2024.107292] [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: 03/06/2024] [Revised: 07/04/2024] [Accepted: 07/09/2024] [Indexed: 07/19/2024]
Abstract
We have studied the effect of calcium ions (Ca2+) at various concentrations on the structure of lipid vesicles in the presence of amyloid-beta peptide Aβ(25-35). In particular, we have investigated the influence of calcium ions on the formation of recently documented bicelle-like structures (BLSs) emerged as a result of Aβ(25-35) triggered membrane disintegration. First, we have shown by using small-angle X-ray and neutron scattering that peptide molecules rigidify the lipid bilayer of gel phase DPPC unilamellar vesicles (ULVs), while addition of the calcium ions to the system hinders this effect of Aβ(25-35). Secondly, the Aβ(25-35) demonstrates a critical peptide concentration at which the BLSs reorganize from ULVs due to heating and cooling the samples through the lipid main phase transition temperature (Tm). However, addition of calcium ions does not affect noticeably the Aβ-induced formation of BLSs and their structural parameters, though the changes in peptide's secondary structure, e.g. the increased α-helix fraction, has been registered by circular dichroism spectroscopy. Finally, according to 31P nuclear magnetic resonance (NMR) measurements, calcium ions do not affect the lipid-peptide arrangement in BLSs and their ability to align in the magnetic field of NMR spectrometer. The influences of various concentrations of calcium ions on the lipid-peptide interactions may prove biologically important because their local concentrations vary widely in in-vivo conditions. In the present work, calcium ions were investigated as a possible tool aimed at regulating the lipid-peptide interactions that demonstrated the disruptive effect of Aβ(25-35) on lipid membranes.
Collapse
Affiliation(s)
- Sergei Kurakin
- Frank Laboratory of Neutron Physics, Joint Institute for Nuclear Research, Joliot-Curie 6, Dubna, Moscow Region 141980, Russia; Institute of Physics, Kazan Federal University, Kremlevskaya 18, Kazan 420008, Russia.
| | - Oleksandr Ivankov
- Frank Laboratory of Neutron Physics, Joint Institute for Nuclear Research, Joliot-Curie 6, Dubna, Moscow Region 141980, Russia
| | - Ermuhammad Dushanov
- Laboratory of Radiation Biology, Joint Institute for Nuclear Research, Joliot-Curie 6, Dubna, Moscow Region 141980, Russia; Department of Biophysics, Dubna State University, Universitetskaya 19, Dubna, Moscow Region 141982, Russia
| | - Tatiana Murugova
- Frank Laboratory of Neutron Physics, Joint Institute for Nuclear Research, Joliot-Curie 6, Dubna, Moscow Region 141980, Russia
| | - Elena Ermakova
- Frank Laboratory of Neutron Physics, Joint Institute for Nuclear Research, Joliot-Curie 6, Dubna, Moscow Region 141980, Russia
| | - Sergey Efimov
- Institute of Physics, Kazan Federal University, Kremlevskaya 18, Kazan 420008, Russia
| | - Timur Mukhametzyanov
- Butlerov Chemistry Institute, Kazan Federal University, Kremlevskaya 18, Kazan 420008, Russia
| | - Svetlana Smerdova
- Kazan National Research Technological University, Karl Marx 68, Kazan 420015, Russia
| | - Vladimir Klochkov
- Institute of Physics, Kazan Federal University, Kremlevskaya 18, Kazan 420008, Russia
| | - Alexander Kuklin
- Frank Laboratory of Neutron Physics, Joint Institute for Nuclear Research, Joliot-Curie 6, Dubna, Moscow Region 141980, Russia; Moscow Institute of Physics and Technology, Instytutskiy Pereulok 9, Dolgoprudny, Moscow Region 141701, Russia
| | - Norbert Kučerka
- Frank Laboratory of Neutron Physics, Joint Institute for Nuclear Research, Joliot-Curie 6, Dubna, Moscow Region 141980, Russia; Department of Physical Chemistry of Drugs, Faculty of Pharmacy, Comenius University Bratislava, Odbojárov 10, Bratislava 832 32, Slovakia.
| |
Collapse
|
7
|
Gallo LH, Akanda N, Autar K, Patel A, Cox I, Powell HA, Grillo M, Barakat N, Morgan D, Guo X, Hickman JJ. A functional aged human iPSC-cortical neuron model recapitulates Alzheimer's disease, senescence, and the response to therapeutics. Alzheimers Dement 2024; 20:5940-5960. [PMID: 39077965 PMCID: PMC11633364 DOI: 10.1002/alz.14044] [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: 03/04/2024] [Revised: 05/01/2024] [Accepted: 05/13/2024] [Indexed: 07/31/2024]
Abstract
INTRODUCTION The degeneration of cortical layers is associated with cognitive decline in Alzheimer's disease (AD). Current therapies for AD are not disease-modifying, and, despite substantial efforts, research and development for AD has faced formidable challenges. In addition, cellular senescence has emerged as a significant contributor to therapy resistance. METHODS Human iPSC-derived cortical neurons were cultured on microelectrode arrays to measure long-term potentiation (LTP) noninvasively. Neurons were treated with pathogenic amyloid-β (Aβ) to analyze senescence and response to therapeutic molecules. RESULTS Microphysiological recordings revealed Aβ dampened cortical LTP activity and accelerated neuronal senescence. Aging neurons secreted inflammatory factors previously detected in brain, plasma, and cerebral spinal fluid of AD patients, in which drugs modulated senescence-related factors. DISCUSSION This platform measures and records neuronal LTP activity in response to Aβ and therapeutic molecules in real-time. Efficacy data from similar platforms have been accepted by the FDA for neurodegenerative diseases, expediting regulatory submissions. HIGHLIGHTS This work developed a progerontic model of amyloid-β (Aβ)-driven cortical degeneration. This work measured neuronal LTP and correlated function with aging biomarkers. Aβ is a driver of neuronal senescence and cortical degeneration. Molecules rescued neuronal function but did not halt Aβ-driven senescence. Therapeutic molecules modulated secretion of inflammatory factors by aging neurons.
Collapse
Affiliation(s)
- Leandro H. Gallo
- Nanoscience Technology CenterUniversity of Central FloridaOrlandoFloridaUSA
| | - Nesar Akanda
- Nanoscience Technology CenterUniversity of Central FloridaOrlandoFloridaUSA
| | - Kaveena Autar
- Nanoscience Technology CenterUniversity of Central FloridaOrlandoFloridaUSA
- Hesperos Inc.OrlandoFloridaUSA
| | - Aakash Patel
- Nanoscience Technology CenterUniversity of Central FloridaOrlandoFloridaUSA
- Hesperos Inc.OrlandoFloridaUSA
| | - Ian Cox
- Nanoscience Technology CenterUniversity of Central FloridaOrlandoFloridaUSA
| | - Haley A. Powell
- Nanoscience Technology CenterUniversity of Central FloridaOrlandoFloridaUSA
| | - Marcella Grillo
- Nanoscience Technology CenterUniversity of Central FloridaOrlandoFloridaUSA
- Hesperos Inc.OrlandoFloridaUSA
| | - Natali Barakat
- Nanoscience Technology CenterUniversity of Central FloridaOrlandoFloridaUSA
| | - Dave Morgan
- Department of Translational NeuroscienceMichigan State University College of Human MedicineGrand Rapids Research CenterGrand RapidsMichiganUSA
| | - Xiufang Guo
- Nanoscience Technology CenterUniversity of Central FloridaOrlandoFloridaUSA
| | - James J. Hickman
- Nanoscience Technology CenterUniversity of Central FloridaOrlandoFloridaUSA
- Hesperos Inc.OrlandoFloridaUSA
| |
Collapse
|
8
|
Zahra N, Rafique S, Naveed Z, Nadeem J, Waqas M, Ali A, Shah M, Idrees M. Regulatory pathways and therapeutic potential of PDE4 in liver pathophysiology. Life Sci 2024; 345:122565. [PMID: 38521388 DOI: 10.1016/j.lfs.2024.122565] [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: 12/07/2023] [Revised: 03/11/2024] [Accepted: 03/13/2024] [Indexed: 03/25/2024]
Abstract
Phosphodiesterase 4 (PDE4), crucial in regulating the cyclic adenosine monophosphate (cAMP) signaling pathway, significantly impacts liver pathophysiology. This article highlights the comprehensive effects of PDE4 on liver health and disease, and its potential as a therapeutic agent. PDE4's role in degrading cAMP disrupts intracellular signaling, increasing pro-inflammatory cytokines like tumor necrosis factor-alpha (TNF-α) and interleukin-6 (IL-6). This contributes to liver inflammation in conditions such as hepatitis and non-alcoholic steatohepatitis (NASH). Additionally, PDE4 is a key factor in liver fibrosis, characterized by excessive extracellular matrix deposition. Inhibiting PDE4 shows promise in reducing liver fibrosis by decreasing the activation of hepatic stellate cells, which is pivotal in fibrogenesis. PDE4 also influences hepatocyte apoptosis a common feature of liver diseases. PDE4 inhibitors protect against hepatocyte apoptosis by raising intracellular cAMP levels, thus activating anti-apoptotic pathways. This suggests potential in targeting PDE4 to prevent hepatocyte loss. Moreover, PDE4 regulates hepatic glucose production and lipid metabolism, essential for liver function. Altering cAMP levels through PDE4 affects enzymes in these metabolic pathways, making PDE4 a target for metabolic disorders like type 2 diabetes and non-alcoholic fatty liver disease (NAFLD). Since PDE4 plays a multifaceted role in liver pathophysiology, influencing PDE4's mechanisms in liver diseases could lead to novel therapeutic strategies. Still, extensive research is required to explore the molecular mechanisms and clinical potential of targeting PDE4 in liver pathologies.
Collapse
Affiliation(s)
- Noureen Zahra
- National Centre of Excellence in Molecular Biology (CEMB), University of the Punjab, Lahore, Pakistan
| | - Shazia Rafique
- National Centre of Excellence in Molecular Biology (CEMB), University of the Punjab, Lahore, Pakistan.
| | - Zoya Naveed
- National Centre of Excellence in Molecular Biology (CEMB), University of the Punjab, Lahore, Pakistan
| | - Jannat Nadeem
- National Centre of Excellence in Molecular Biology (CEMB), University of the Punjab, Lahore, Pakistan
| | - Muhammad Waqas
- Department of Biotechnology and Genetic Engineering, Hazara University Mansehra, Pakistan
| | - Amjad Ali
- Department of Biotechnology and Genetic Engineering, Hazara University Mansehra, Pakistan
| | - Masaud Shah
- Department of Physiology Ajou University, South Korea
| | - Muhammad Idrees
- National Centre of Excellence in Molecular Biology (CEMB), University of the Punjab, Lahore, Pakistan
| |
Collapse
|
9
|
Jino K, Miyamoto K, Kanbara T, Unemura C, Horiguchi N, Ago Y. Allosteric inhibition of phosphodiesterase 4D induces biphasic memory-enhancing effects associated with learning-activated signaling pathways. Psychopharmacology (Berl) 2024; 241:805-816. [PMID: 38114603 DOI: 10.1007/s00213-023-06510-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 11/20/2023] [Indexed: 12/21/2023]
Abstract
RATIONALE Phosphodiesterase 4D negative allosteric modulators (PDE4D NAMs) enhance memory and cognitive function in animal models without emetic-like side effects. However, the relationship between increased cyclic adenosine monophosphate (cAMP) signaling and the effects of PDE4D NAM remains elusive. OBJECTIVE To investigate the roles of hippocampal cAMP metabolism and synaptic activation in the effects of D159687, a PDE4D NAM, under baseline and learning-stimulated conditions. RESULTS At 3 mg/kg, D159687 enhanced memory formation and consolidation in contextual fear conditioning; however, neither lower (0.3 mg/kg) nor higher (30 mg/kg) doses induced memory-enhancing effects. A biphasic (bell-shaped) dose-response effect was also observed in a scopolamine-induced model of amnesia in the Y-maze, whereas D159687 dose-dependently caused an emetic-like effect in the xylazine/ketamine anesthesia test. At 3 mg/kg, D159687 increased cAMP levels in the hippocampal CA1 region after conditioning in the fear conditioning test, but not in the home-cage or conditioning cage (i.e., context only). By contrast, 30 mg/kg of D159687 increased hippocampal cAMP levels under all conditions. Although both 3 and 30 mg/kg of D159687 upregulated learning-induced Fos expression in the hippocampal CA1 30 min after conditioning, 3 mg/kg, but not 30 mg/kg, of D159687 induced phosphorylation of synaptic plasticity-related proteins such as cAMP-responsive element-binding protein, synaptosomal-associated protein 25 kDa, and the N-methyl-D-aspartate receptor subunit NR2A. CONCLUSIONS Our findings suggest that learning-stimulated conditions can alter the effects of a PDE4D NAM on hippocampal cAMP levels and imply that a PDE4D NAM exerts biphasic memory-enhancing effects associated with synaptic plasticity-related signaling activation.
Collapse
Affiliation(s)
- Kohei Jino
- Laboratory for Drug Discovery and Disease Research, Shionogi Pharmaceutical Research Center, Shionogi & Co., Ltd., 3-1-1 Futaba-cho, Toyonaka-shi, Osaka, 561-0825, Japan
- Department of Cellular and Molecular Pharmacology, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, Hiroshima, 734-8553, Japan
| | - Keisuke Miyamoto
- Laboratory for Drug Discovery and Disease Research, Shionogi Pharmaceutical Research Center, Shionogi & Co., Ltd., 3-1-1 Futaba-cho, Toyonaka-shi, Osaka, 561-0825, Japan
| | - Tomoe Kanbara
- Laboratory for Drug Discovery and Disease Research, Shionogi Pharmaceutical Research Center, Shionogi & Co., Ltd., 3-1-1 Futaba-cho, Toyonaka-shi, Osaka, 561-0825, Japan
| | - Chie Unemura
- Laboratory for Drug Discovery and Disease Research, Shionogi Pharmaceutical Research Center, Shionogi & Co., Ltd., 3-1-1 Futaba-cho, Toyonaka-shi, Osaka, 561-0825, Japan
| | - Naotaka Horiguchi
- Laboratory for Drug Discovery and Disease Research, Shionogi Pharmaceutical Research Center, Shionogi & Co., Ltd., 3-1-1 Futaba-cho, Toyonaka-shi, Osaka, 561-0825, Japan.
| | - Yukio Ago
- Department of Cellular and Molecular Pharmacology, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, Hiroshima, 734-8553, Japan.
| |
Collapse
|
10
|
Kurakin S, Badreeva D, Dushanov E, Shutikov A, Efimov S, Timerova A, Mukhametzyanov T, Murugova T, Ivankov O, Mamatkulov K, Arzumanyan G, Klochkov V, Kučerka N. Arrangement of lipid vesicles and bicelle-like structures formed in the presence of Aβ(25-35) peptide. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2024; 1866:184237. [PMID: 37820938 DOI: 10.1016/j.bbamem.2023.184237] [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: 04/17/2023] [Revised: 08/31/2023] [Accepted: 10/03/2023] [Indexed: 10/13/2023]
Abstract
Our complementary experimental data and molecular dynamics (MD) simulations results reveal the structure of previously observed lipid bicelle-like structures (BLSs) formed in the presence of amyloid-beta peptide Aβ(25-35) below the main phase transition temperature (Tm) of saturated phosphatidylcholine lipids and small unilamellar vesicles (SUVs) above this temperature. First, we show by using solid-state 31P nuclear magnetic resonance (NMR) spectroscopy that our BLSs being in the lipid gel phase demonstrate magnetic alignment along the magnetic field of NMR spectrometer and undergo a transition to SUVs in the lipid fluid phase when heated through the Tm. Secondly, thanks to the BLS alignment we present their lipid structure. Lipids are found located not only in the flat bilayered part but also around its perimeter, which is corroborated by the results of coarse-grained (CG) MD simulations. Finally, peptides appear to mix randomly with lipids in SUVs while assuming predominantly unordered secondary structures revealed by circular dichroism (CD), Raman spectroscopy, and all-atom MD simulations. Importantly, the former is changing little when the system undergoes morphological transitions between BLSs and SUVs. Our structural results then offer a platform for studying and understanding mechanisms of morphological transformations caused by the disruptive effect of amyloid-beta peptides on the lipid bilayer.
Collapse
Affiliation(s)
- Sergei Kurakin
- Frank Laboratory of Neutron Physics, Joint Institute for Nuclear Research, Joliot-Curie 6, Dubna, Moscow Region 141980, Russia; Institute of Physics, Kazan Federal University, Kremlevskaya 18, Kazan 420008, Russia.
| | - Dina Badreeva
- Meshcheryakov Laboratory of Information Technologies, Joint Institute for Nuclear Research, Joliot-Curie 6, Dubna, Moscow Region 141980, Russia
| | - Ermuhammad Dushanov
- Laboratory of Radiation Biology, Joint Institute for Nuclear Research, Joliot-Curie 6, Dubna, Moscow Region 141980, Russia
| | - Artyom Shutikov
- Frank Laboratory of Neutron Physics, Joint Institute for Nuclear Research, Joliot-Curie 6, Dubna, Moscow Region 141980, Russia
| | - Sergey Efimov
- Institute of Physics, Kazan Federal University, Kremlevskaya 18, Kazan 420008, Russia
| | - Ayzira Timerova
- Institute of Physics, Kazan Federal University, Kremlevskaya 18, Kazan 420008, Russia
| | - Timur Mukhametzyanov
- Butlerov Chemistry Institute, Kazan Federal University, Kremlevskaya 18, Kazan 420008, Russia
| | - Tatiana Murugova
- Frank Laboratory of Neutron Physics, Joint Institute for Nuclear Research, Joliot-Curie 6, Dubna, Moscow Region 141980, Russia
| | - Oleksandr Ivankov
- Frank Laboratory of Neutron Physics, Joint Institute for Nuclear Research, Joliot-Curie 6, Dubna, Moscow Region 141980, Russia
| | - Kahramon Mamatkulov
- Frank Laboratory of Neutron Physics, Joint Institute for Nuclear Research, Joliot-Curie 6, Dubna, Moscow Region 141980, Russia
| | - Grigory Arzumanyan
- Frank Laboratory of Neutron Physics, Joint Institute for Nuclear Research, Joliot-Curie 6, Dubna, Moscow Region 141980, Russia
| | - Vladimir Klochkov
- Institute of Physics, Kazan Federal University, Kremlevskaya 18, Kazan 420008, Russia
| | - Norbert Kučerka
- Frank Laboratory of Neutron Physics, Joint Institute for Nuclear Research, Joliot-Curie 6, Dubna, Moscow Region 141980, Russia; Department of Physical Chemistry of Drugs, Faculty of Pharmacy, Comenius University Bratislava, Odbojárov 10, Bratislava 832 32, Slovakia.
| |
Collapse
|
11
|
Jin M, Wei Z, Ramalingam N, Xiao M, Xu A, Yu X, Song Q, Liu W, Zhao J, Zhang D, Selkoe DJ, Li S. Activation of β 2-adrenergic receptors prevents AD-type synaptotoxicity via epigenetic mechanisms. Mol Psychiatry 2023; 28:4877-4888. [PMID: 37365243 DOI: 10.1038/s41380-023-02145-5] [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] [Indexed: 06/28/2023]
Abstract
We previously reported that prolonged exposure to an enriched environment (EE) enhances hippocampal synaptic plasticity, with one of the significant mechanistic pathways being activation of β2-adrenergic receptor (β2-AR) signaling, thereby mitigating the synaptotoxic effects of soluble oligomers of amyloid β-protein (oAβ). However, the detailed mechanism remained elusive. In this work, we recorded field excitatory postsynaptic potentials (fEPSP) in the CA1 region of mouse hippocampal slices treated with or without toxic Aβ-species. We found that pharmacological activation of β2-AR, but not β1-AR, selectively mimicked the effects of EE in enhancing LTP and preventing oAβ-induced synaptic dysfunction. Mechanistic analyses showed that certain histone deacetylase (HDAC) inhibitors mimicked the benefits of EE, but this was not seen in β2-AR knockout mice, suggesting that activating β2-AR prevents oAβ-mediated synaptic dysfunction via changes in histone acetylation. EE or activation of β-ARs each decreased HDAC2, whereas Aβ oligomers increased HDAC2 levels in the hippocampus. Further, oAβ-induced inflammatory effects and neurite degeneration were prevented by either β2-AR agonists or certain specific HDAC inhibitors. These preclinical results suggest that activation of β2-AR is a novel potential therapeutic strategy to mitigate oAβ-mediated features of AD.
Collapse
Affiliation(s)
- Ming Jin
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Zhiyun Wei
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Nagendran Ramalingam
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Meng Xiao
- Department of Neurology, Xinxiang Medical University, Xinxiang, 453100, China
- Shenzhen Hospital, Beijing University of Chinese Medicine, Shenzhen, 518172, China
| | - Anqi Xu
- Department of Neurology, Xinxiang Medical University, Xinxiang, 453100, China
| | - Xiaohan Yu
- Department of Neurology, Xinxiang Medical University, Xinxiang, 453100, China
| | - Qingyang Song
- Department of Neurology, Xinxiang Medical University, Xinxiang, 453100, China
| | - Wen Liu
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Jianhua Zhao
- Department of Neurology, Xinxiang Medical University, Xinxiang, 453100, China
- Henan Key Laboratory of Neurorestoratology, Xinxiang, Henan, 453100, China
| | - Dainan Zhang
- Department of Neurology, Xinxiang Medical University, Xinxiang, 453100, China
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| | - Dennis J Selkoe
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Shaomin Li
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
12
|
Dastgheib M, Falak R, Moghaddam MV, Hassanzadeh G, Safa M, Hosseini A. Rolipram and pentoxifylline combination ameliorates the morphological abnormalities of dorsal root ganglion neurons in experimental diabetic neuropathy by reducing mitochondrial dysfunction and apoptosis. J Biochem Mol Toxicol 2023; 37:e23459. [PMID: 37431890 DOI: 10.1002/jbt.23459] [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: 02/22/2023] [Revised: 06/20/2023] [Accepted: 07/04/2023] [Indexed: 07/12/2023]
Abstract
Diabetic neuropathy (DN) is the most prevalent complication of diabetes. Pharmacological treatments for DN are often limited in efficacy, so the development of new agents to alleviate DN is essential. The aim of this study was to evaluate the effects of rolipram, a selective phosphodiesterase-4 inhibitor (PDE-4I), and pentoxifylline, a general PDE inhibitor, using a rat model of DN. In this study, a diabetic rat model was established by i.p. injection of STZ (55 mg/kg). Rats were treated with rolipram (1 mg/kg), pentoxifylline (100 mg/kg), and combination of rolipram (0.5 mg/kg) and pentoxifylline (50 mg/kg), orally for 5 weeks. After treatments, sensory function was assessed by hot plate test. Then rats were anesthetized and dorsal root ganglion (DRG) neurons isolated. Cyclic adenosine monophosphate (cAMP), adenosine triphosphate (ATP, adenosine diphosphate and mitochondrial membrane potential (MMP) levels, Cytochrome c release, Bax, Bcl-2, caspase-3 proteins expression in DRG neurons were assessed by biochemical and ELISA methods, and western blot analysis. DRG neurons were histologically examined using hematoxylin and eosin (H&E) staining method. Rolipram and/or pentoxifylline significantly attenuated sensory dysfunction by modulating nociceptive threshold. Rolipram and/or pentoxifylline treatment dramatically increased the cAMP level, prevented mitochondrial dysfunction, apoptosis and degeneration of DRG neurons, which appears to be mediated by inducing ATP and MMP, improving cytochrome c release, as well as regulating the expression of Bax, Bcl-2, and caspase-3 proteins, and improving morphological abnormalities of DRG neurons. We found maximum effectiveness with rolipram and pentoxifylline combination on mentioned factors. These findings encourage the use of rolipram and pentoxifylline combination as a novel experimental evidence for further clinical investigations in the treatment of DN.
Collapse
Affiliation(s)
- Mona Dastgheib
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Reza Falak
- Department of Immunology, Iran University of Medical Sciences, Tehran, Iran
| | | | | | - Majid Safa
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Asieh Hosseini
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
13
|
Thwe PN, Yeong KY, Choo WS. Anti-Amyloid β Aggregation Activity and Cell Viability Effect of Betacyanins from Red Pitahaya (Hylocereus polyrhizus) for Alzheimer's Disease. PLANT FOODS FOR HUMAN NUTRITION (DORDRECHT, NETHERLANDS) 2023; 78:613-619. [PMID: 37466824 DOI: 10.1007/s11130-023-01081-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 07/06/2023] [Indexed: 07/20/2023]
Abstract
Betacyanin-rich extract from red beet (Beta vulgaris) was recently reported to inhibit amyloid β (Aβ) aggregation, a main pathological event in Alzheimer's disease. However, the anti-Aβ aggregation effect of individual betacyanin isolates has not been reported before. This study investigated the anti-Aβ aggregation activity and cytotoxicity of betacyanins from red pitahaya or red dragon fruit (Hylocereus polyrhizus). Betacyanin fraction (IC50 = 16.02 ± 1.15 µg/mL) and individual betacyanin isolates exhibited anti-Aβ aggregation activity in a concentration-dependent manner using a thioflavin T fluorescence assay. The highest to lowest IC50 was in the order of betanin (426.30 ± 29.55 µM), phyllocactin (175.22 ± 1.52 µM), and hylocerenin (131.73 ± 5.58 µM), following a trend of increase in functional groups of carboxyl, hydroxyl, and/or carbonyl. Further, the betacyanin fraction of 135.78 µg/mL and below, which were concentrations with an anti-Aβ aggregation effect, were validated as non-neurotoxic based on an in vitro cytotoxicity assay using human neuroblastoma (SH-SY5Y) cells. These findings highlight the potential neuroprotective activity of betacyanins for Alzheimer's disease.
Collapse
Affiliation(s)
- Pan Nu Thwe
- School of Science, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, Selangor, 47500, Malaysia
| | - Keng Yoon Yeong
- School of Science, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, Selangor, 47500, Malaysia
| | - Wee Sim Choo
- School of Science, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, Selangor, 47500, Malaysia.
| |
Collapse
|
14
|
Zhao F, Zhong L, Wang Y, Wang R, Yang Z, Luo Y, Zheng Y. Untargeted metabolomics uncovering neuroprotective effect of Dl-3-n-butylphthalide on improving cognitive impairment induced by chronic cerebral hypoperfusion in rats. Int Immunopharmacol 2023; 119:110271. [PMID: 37172424 DOI: 10.1016/j.intimp.2023.110271] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 04/28/2023] [Accepted: 04/29/2023] [Indexed: 05/15/2023]
Abstract
Chronic cerebral hypoperfusion (CCH) can cause cognitive impairments. Dl-3-n-butylphthalide (NBP) is widely used in neurological disorders; but, the role of NBP in CCH remains unclear. This study aimed to investigate the potential mechanism of NBP on CCH through untargeted metabolomics. Animals were divided into CCH, Sham, and NBP groups. A rat model of bilateral carotid artery ligation was used to simulate CCH. Cognitive function of the rats was assessed using the Morris water maze test. Additionally, we used LC-MS/MS to detect ionic intensities of metabolites between the three groups for off-target metabolism analysis and to screen for differential metabolites. The analysis showed an improvement in cognitive function in rats after NBP treatment. Moreover, metabolomic studies showed that the serum metabolic profiles of the Sham and CCH groups were significantly altered, and 33 metabolites were identified as potential biomarkers associated with the effects of NBP. These metabolites were enriched in 24 metabolic pathways.And the pathway of differential metabolite enrichment was further verified by immunofluorescence. Thus, the study provides a theoretical basis for the pathogenesis of CCH and the treatment of CCH by NBP, and supports a wider application of NBP drugs.
Collapse
Affiliation(s)
- Fangfang Zhao
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China; Beijing Geriatric Medical Research Center and Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing, China
| | - Liyuan Zhong
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China; Beijing Geriatric Medical Research Center and Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing, China
| | - Yilin Wang
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China; Beijing Geriatric Medical Research Center and Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing, China
| | - Rongliang Wang
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China; Beijing Geriatric Medical Research Center and Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing, China
| | - Zhenhong Yang
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China; Beijing Geriatric Medical Research Center and Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing, China
| | - Yumin Luo
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China; Beijing Geriatric Medical Research Center and Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing, China; Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China.
| | - Yangmin Zheng
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China; Beijing Geriatric Medical Research Center and Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing, China.
| |
Collapse
|
15
|
Yanai S, Tago T, Toyohara J, Arasaki T, Endo S. Reversal of spatial memory impairment by phosphodiesterase 3 inhibitor cilostazol is associated with reduced neuroinflammation and increased cerebral glucose uptake in aged male mice. Front Pharmacol 2022; 13:1031637. [PMID: 36618932 PMCID: PMC9810637 DOI: 10.3389/fphar.2022.1031637] [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: 08/30/2022] [Accepted: 11/30/2022] [Indexed: 12/24/2022] Open
Abstract
The nucleotide second messenger 3', 5'-cyclic adenosine monophosphate (cAMP) and 3', 5'-cyclic guanosine monophosphate (cGMP) mediate fundamental functions of the brain, including learning and memory. Phosphodiesterase 3 (PDE3) can hydrolyze both cAMP and cGMP and appears to be involved in the regulation of their contents in cells. We previously demonstrated that long-term administration of cilostazol, a PDE3 inhibitor, maintained good memory performance in aging mice. Here, we report on studies aimed at determining whether cilostazol also reverses already-impaired memory in aged male mice. One month of oral 1.5% cilostazol administration in 22-month-old mice reversed age-related declines in hippocampus-dependent memory tasks, including the object recognition and the Morris water maze. Furthermore, cilostazol reduced neuroinflammation, as evidenced by immunohistochemical staining, and increased glucose uptake in the brain, as evidence by positron emission tomography (PET) with 2-deoxy-2-[18F]fluoro-d-glucose ([18F]FDG). These results suggest that already-expressed memory impairment in aged male mice that depend on cyclic nucleotide signaling can be reversed by inhibition of PDE3. The reversal of age-related memory impairments may occur in the central nervous system, either through cilostazol-enhanced recall or strengthening of weak memories that otherwise may be resistant to recall.
Collapse
Affiliation(s)
- Shuichi Yanai
- Aging Neuroscience Research Team, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Tetsuro Tago
- Research Team for Neuroimaging, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Jun Toyohara
- Research Team for Neuroimaging, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Tomoko Arasaki
- Aging Neuroscience Research Team, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Shogo Endo
- Aging Neuroscience Research Team, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan,*Correspondence: Shogo Endo,
| |
Collapse
|
16
|
Emerging Potential of the Phosphodiesterase (PDE) Inhibitor Ibudilast for Neurodegenerative Diseases: An Update on Preclinical and Clinical Evidence. Molecules 2022; 27:molecules27238448. [PMID: 36500540 PMCID: PMC9737612 DOI: 10.3390/molecules27238448] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 11/28/2022] [Accepted: 11/29/2022] [Indexed: 12/11/2022] Open
Abstract
Neurodegenerative diseases constitute a broad range of central nervous system disorders, characterized by neuronal degeneration. Alzheimer's disease, Parkinson's disease, amyolotrophic lateral sclerosis (ALS), and progressive forms of multiple sclerosis (MS) are some of the most frequent neurodegenerative diseases. Despite their diversity, these diseases share some common pathophysiological mechanisms: the abnormal aggregation of disease-related misfolded proteins, autophagosome-lysosome pathway dysregulation, impaired ubiquitin-proteasome system, oxidative damage, mitochondrial dysfunction and excessive neuroinflammation. There is still no effective drug that could halt the progression of neurodegenerative diseases, and the current treatments are mainly symptomatic. In this regard, the development of novel multi-target pharmaceutical approaches presents an attractive therapeutic strategy. Ibudilast, an anti-inflammatory drug firstly developed as an asthma treatment, is a cyclic nucleotide phosphodiesterases (PDEs) inhibitor, which mainly acts by increasing the amount of cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP), while downregulating the pro-inflammatory factors, such as tumor necrosis factor-α (TNF-α), macrophage migration inhibitory factor (MIF) and Toll-like receptor 4 (TLR-4). The preclinical evidence shows that ibudilast may act neuroprotectively in neurodegenerative diseases, by suppressing neuroinflammation, inhibiting apoptosis, regulating the mitochondrial function and by affecting the ubiquitin-proteasome and autophagosome-lysosome pathways, as well as by attenuating oxidative stress. The clinical trials in ALS and progressive MS also show some promising results. Herein, we aim to provide an update on the emerging preclinical and clinical evidence on the therapeutic potential of ibudilast in these disorders, discuss the potential challenges and suggest the future directions.
Collapse
|
17
|
Crocetti L, Floresta G, Cilibrizzi A, Giovannoni MP. An Overview of PDE4 Inhibitors in Clinical Trials: 2010 to Early 2022. Molecules 2022; 27:4964. [PMID: 35956914 PMCID: PMC9370432 DOI: 10.3390/molecules27154964] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 08/01/2022] [Accepted: 08/02/2022] [Indexed: 11/16/2022] Open
Abstract
Since the early 1980s, phosphodiesterase 4 (PDE4) has been an attractive target for the treatment of inflammation-based diseases. Several scientific advancements, by both academia and pharmaceutical companies, have enabled the identification of many synthetic ligands for this target, along with the acquisition of precise information on biological requirements and linked therapeutic opportunities. The transition from pre-clinical to clinical phase was not easy for the majority of these compounds, mainly due to their significant side effects, and it took almost thirty years for a PDE4 inhibitor to become a drug i.e., Roflumilast, used in the clinics for the treatment of chronic obstructive pulmonary disease. Since then, three additional compounds have reached the market a few years later: Crisaborole for atopic dermatitis, Apremilast for psoriatic arthritis and Ibudilast for Krabbe disease. The aim of this review is to provide an overview of the compounds that have reached clinical trials in the last ten years, with a focus on those most recently developed for respiratory, skin and neurological disorders.
Collapse
Affiliation(s)
- Letizia Crocetti
- NEUROFARBA, Pharmaceutical and Nutraceutical Section, University of Florence, Via Ugo Schiff 6, 50019 Sesto Fiorentino, Italy
| | - Giuseppe Floresta
- Department of Drug and Health Sciences, University of Catania, Viale A. Doria 6, 95125 Catania, Italy
| | - Agostino Cilibrizzi
- Institute of Pharmaceutical Science, King’s College London, Stamford Street, London SE1 9NH, UK
| | - Maria Paola Giovannoni
- NEUROFARBA, Pharmaceutical and Nutraceutical Section, University of Florence, Via Ugo Schiff 6, 50019 Sesto Fiorentino, Italy
| |
Collapse
|
18
|
Roflumilast, a Phosphodiesterase-4 Inhibitor, Ameliorates Sleep Deprivation-Induced Cognitive Dysfunction in C57BL/6J Mice. ACS Chem Neurosci 2022; 13:1938-1947. [PMID: 35736514 DOI: 10.1021/acschemneuro.2c00127] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Sleep deprivation (SD) interferes with long-term memory and cognitive functions by overactivation of phosphodiesterase (PDEs) enzymes. PDE4, a nonredundant regulator of the cyclic nucleotides (cAMP), is densely expressed in the hippocampus and is involved in learning and memory processes. In the present study, we investigated the effects of Roflumilast (ROF), a PDE4B inhibitor, on sleep deprivation-induced cognitive dysfunction in a mouse model. Memory assessment was performed using a novel object recognition task, and the hippocampal cAMP level was estimated by the ELISA method. The alterations in the expressions of PDE4B, amyloid-β (Aβ), CREB, BDNF, and synaptic proteins (Synapsin I, SAP 97, PSD 95) were assessed to gain insights into the possible mechanisms of action of ROF using the Western blot technique. Results show that ROF reversed SD-induced cognitive decline in mice. ROF downregulated PDE4B and Aβ expressions in the brain. Additionally, ROF improved the cAMP level and the protein expressions of synapsin I, SAP 97, and PSD 95 in the hippocampal region of SD mice. Taken together, these results suggest that ROF can suppress the deleterious effects of SD-induced cognitive dysfunction via the PDE4B-mediated cAMP/CREB/BDNF signaling cascade.
Collapse
|
19
|
Chang B, Liu Y, Hu J, Tang Z, Qiu Z, Song Z, Jia A, Zhang Y. Bupleurum chinense DC improves CUMS-induced depressive symptoms in rats through upregulation of the cAMP/PKA/CREB signalling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2022; 289:115034. [PMID: 35092825 DOI: 10.1016/j.jep.2022.115034] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 01/17/2022] [Accepted: 01/21/2022] [Indexed: 05/15/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Bupleurum chinense DC. (B. chinense) is the dried root of B. chinense, belonging to the Umbelliferae family. B. chinense has been reported since ancient times for its effect of soothing the liver and relieving depression. Additionally, its important role in treating depression, depressed mood disorders and anti-inflammation has been proven in previous studies. However, its specific mechanism of action remains unknown. AIM OF THE STUDY The key targets and metabolites of the antidepressant effect of B. chinense were investigated based on the cAMP signalling pathway. The study examined the mechanism for the antidepressant effect of B. chinense by target prediction, analysis of related metabolites and potential metabolic pathways. MATERIALS AND METHODS A network pharmacology approach was used to predict the antidepressant targets and pathways of B. chinense. A depression rat model was established through the CUMS (chronic unpredictable mild stress) procedure. The depression model was assessed by body weight, sugar-water preference, water maze and enzyme-linked immunosorbent assay (ELISA) indicators (5hydroxytryptamine, etc.). The key metabolic pathways were screened by correlations between metabolites and key targets. Finally, a quantitative analysis of key targets and metabolites was experimentally validated. RESULTS B. chinense significantly ameliorated the reduction in body weight, sugar-water preference rate and cognitive performance in the water maze experiment in rats with depression induced by CUMS. ELISA, Western blotting (WB) and reverse transcription-polymerase chain reaction (RT-PCR) assays showed that B. chinense significantly improves the expression of protein kinase cyclic adenylic acid (cAMP)-activated catalytic subunit alpha (PRKACA), cAMP-response element-binding protein (CREB) and cAMP activation in the rat brain induced by CUMS. According to metabolic pathway analysis, B. chinense shows an antidepressant effect primarily by regulating the cAMP metabolic pathway. CONCLUSION B. chinense upregulated PRKACA and CREB expression and the level of the key metabolite cAMP in the cAMP/PKA/CREB pathway while reducing the inflammatory response to depression treatment. These new findings support future research on the antidepressant effects of B. chinense.
Collapse
Affiliation(s)
- Baijin Chang
- Changchun University of Chinese Medicine, 130117, Chang Chun, PR China; Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi Collaborative Innovation Center Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, 712083, Xianyang, PR China
| | - Yanru Liu
- Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi Collaborative Innovation Center Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, 712083, Xianyang, PR China.
| | - Jingting Hu
- The Third Clinical Affiliated Hospital of Changchun University of Chinese Medicine, 130117, Chang chun, PR China
| | - Zhishu Tang
- Changchun University of Chinese Medicine, 130117, Chang Chun, PR China; Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi Collaborative Innovation Center Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, 712083, Xianyang, PR China; China Academy of Chinese Medical Sciences, 100700, Beijing, PR China.
| | - Zhidong Qiu
- Changchun University of Chinese Medicine, 130117, Chang Chun, PR China
| | - Zhongxing Song
- Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi Collaborative Innovation Center Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, 712083, Xianyang, PR China
| | - Ailing Jia
- Changchun University of Chinese Medicine, 130117, Chang Chun, PR China
| | - Yuru Zhang
- Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi Collaborative Innovation Center Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, 712083, Xianyang, PR China
| |
Collapse
|
20
|
Baicalin attenuates amyloid β oligomers induced memory deficits and mitochondria fragmentation through regulation of PDE-PKA-Drp1 signalling. Psychopharmacology (Berl) 2022; 239:851-865. [PMID: 35103832 DOI: 10.1007/s00213-022-06076-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 01/24/2022] [Indexed: 12/13/2022]
Abstract
RATIONALE Mitochondrial fragmentation contributes to the initiation of Alzheimer's disease (AD) pathology. Baicalin plays a significant role in rescuing mitochondrial dysfunction. However, the effect of baicalin treatment on the modulation of mitochondrial fragmentation has not yet been assessed. OBJECTIVES The present study was designed to evaluate the effect of baicalin on memory and understand its mechanism of action. RESULTS Baicalin treatment significantly reversed the altered learning and memory behaviours in AD mouse model. We found that baicalin treatment significantly improved the levels of microtubule association protein-2 and enhanced the expression of synaptophysin and postsynaptic density protein 95 (PSD95). Moreover, treatment with baicalin reversed amyloid-β oligomer (AβO)-induced abnormalities in the succinate dehydrogenase complex iron sulphur subunit B (SDHB) and cytochrome c oxidase components I (COXI) and mitochondrial fragmentation in the hippocampus. Further, we found that baicalin decreased the PDE4 levels and upregulated the levels of phosphorylated Ser157 site of vasodilator-stimulated phosphoprotein (pVASPs157) and phosphorylated Ser637 site of mitochondrial dynamin-related protein 1 (pDrp1S637). Moreover, in AβO-treated HT-22 cells, H89 inhibited the effect of baicalin on PSD95, mitochondrial fragmentation, SDHB and COXI, PDE4, pVASPs157, and pDrp1S637. CONCLUSION The effect of baicalin on memory improvement may be due to improved synaptic plasticity, mitochondrial fragmentation, and rescue of dysfunction via the inhibition of PDE4, which leads to activation of pDrp1S637 in the AβO-induced model.
Collapse
|
21
|
Wang X, Wang K, Wu X, Huang W, Yang L. Role of the cAMP-PKA-CREB-BDNF pathway in abnormal behaviours of serotonin transporter knockout mice. Behav Brain Res 2022; 419:113681. [PMID: 34838579 DOI: 10.1016/j.bbr.2021.113681] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 11/16/2021] [Accepted: 11/19/2021] [Indexed: 11/02/2022]
Abstract
Serotonin transporter gene-linked polymorphic region polymorphisms are associated with anxiety, neuroticism, affective disorders and vulnerability to stressful life events; however, the relevant physiological mechanisms are not well understood. Serotonin transporter knockout mice have been widely used as a model of allelic variation of serotonin transporter function in humans; herein, wild-type mice and heterozygous and homozygous knockout mice models were established to explore the behavioural changes related to different genotypes and the possible physiological mechanisms. Behavioural changes were assessed using behavioural tests, namely, elevated plus maze, open field, Morris water maze and rotarod tests. Serum indicators were detected using the enzyme-linked immunosorbent assay. Compared with wild-type mice, homozygous mice showed significant anxiety-like behaviours in the plus maze and open field tests; conversely, anxiety-like behaviours in heterozygous mice were less pronounced. Homozygous mice also showed cognitive impairment and motor inhibition in the Morris water maze and rotarod tests. Serotonin levels decreased in both heterozygous and homozygous mice, and 5-hydroxytryptophan, protein kinase A, adenylyl cyclase, cyclic adenosine monophosphate response element-binding protein and brain-derived neurotrophic factor levels were lower in homozygous mice than in wild-type and heterozygous mice, whereas no statistical differences were found between wild-type and heterozygous mice. Additionally, there was a correlation between serological and behavioural indicators. This study provided experimental evidence that the cyclic adenosine monophosphate-protein kinase A-cyclic adenosine monophosphate response element-binding protein-brain-derived neurotrophic factor pathway may be involved in the regulation of polymorphism to stress and enriched the behavioural and physiological characteristics of serotonin transporter knockout mice.
Collapse
Affiliation(s)
- Xiaomin Wang
- Department of Epidemiology and Health Statistics, School of Public Health, Guangxi Medical University, Nanning, PR China
| | - Ke Wang
- Department of Occupational and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, PR China
| | - Xiangmin Wu
- Department of Occupational and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, PR China
| | - Wenxiu Huang
- Department of Occupational and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, PR China
| | - Li Yang
- Department of Occupational and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, PR China.
| |
Collapse
|
22
|
Fessel J. Reversing Alzheimer's disease dementia with clemastine, fingolimod, or rolipram, plus anti-amyloid therapy. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2022; 8:e12242. [PMID: 35128031 PMCID: PMC8804619 DOI: 10.1002/trc2.12242] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 11/09/2021] [Accepted: 12/13/2021] [Indexed: 12/17/2022]
Abstract
A few anti-amyloid trials offer a slight possibility of preventing progression of cognitive loss, but none has reversed the process. A possible reason is that amyloid may be necessary but insufficient in the pathogenesis of AD, and other causal factors may need addressing in addition to amyloid. It is argued here that drugs addressing myelination and synaptogenesis are the optimum partners for anti-amyloid drugs, since there is much evidence that early in the process that leads to AD, both neural circuits and synaptic activity are dysfunctional. Evidence to support this argument is presented. Evidence is also presented that clemastine, fingolimod, and rolipram, benefit both myelination and synaptogenesis. It is suggested that a regimen that includes one of them plus an anti-amyloid drug, could reverse AD.
Collapse
Affiliation(s)
- Jeffrey Fessel
- Professor of Clinical Medicine, Emeritus, Department of MedicineUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| |
Collapse
|
23
|
The Role of Melatonin on Behavioral Changes and Concomitant Oxidative Stress in icvAβ 1-42 Rat Model with Pinealectomy. Int J Mol Sci 2021; 22:ijms222312763. [PMID: 34884567 PMCID: PMC8657444 DOI: 10.3390/ijms222312763] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/16/2021] [Accepted: 11/22/2021] [Indexed: 02/06/2023] Open
Abstract
One of the pathological hallmarks of Alzheimer’s disease (AD) associated with its progression that contributes to β-amyloid (Aβ) generation is oxidative stress (OS). Clinical data suggest that melatonin is a potent antioxidant that might be effective in the adjunctive therapy of this neurodegenerative disease. The present study aimed to explore the role of melatonin on behavioral changes and markers of OS in three rat models, namely, pinealectomy (pin) model of melatonin deficit, intracerebroventricular (icv)Aβ1-42 model of AD, and combination of both pin and Aβ1-42 model (pin+icvAβ1-42). The chronic injection with vehicle/melatonin (50 mg/kg, i.p. for 40 days) started on the same day of sham/pin and icv vehicle/Aβ1-42 infusion procedures. Anxiety in the open field and the elevated plus-maze test and cognitive responses in the object recognition test were tested between the 30th–35th day after the surgical procedures. Markers of OS in the frontal cortex (FC) and hippocampus were detected by the ELISA method. Melatonin treatment corrected the exacerbated anxiety response only in the pin+icvAβ1-42 model while it alleviated the cognitive impairment in the three models. Pinealectomy disturbed the antioxidant system via enhanced SOD activity and decreased GSH levels both in the FC and hippocampus. The Aβ1-42 model decreased the SOD activity in the FC and elevated the MDA level in the two brain structures. The pin+icvAβ1-42 model impaired the antioxidant system and elevated lipid peroxidation. Melatonin supplementation restored only the elevated MDA level of icvAβ1-42 and pin+icvAβ1-42 model in the hippocampus. In conclusion, our study reveals that the pin+icvAβ1-42 rat model triggers more pronounced anxiety and alterations in markers of OS that may be associated with melatonin deficit concomitant to icvAβ1-42-induced AD pathology.
Collapse
|
24
|
Dong XL, Wang YH, Xu J, Zhang N. The protective effect of the PDE-4 inhibitor rolipram on intracerebral haemorrhage is associated with the cAMP/AMPK/SIRT1 pathway. Sci Rep 2021; 11:19737. [PMID: 34611179 PMCID: PMC8492710 DOI: 10.1038/s41598-021-98743-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 09/13/2021] [Indexed: 12/22/2022] Open
Abstract
Rolipram specifically inhibits phosphodiesterase (PDE) 4, thereby preventing inactivation of the intracellular second messenger cyclic adenosine monophosphate (cAMP). Rolipram has been shown to play a neuroprotective role in some central nervous system (CNS) diseases. However, the role of PDE4 and the potential protective effect of rolipram on the pathophysiological process of intracerebral haemorrhage (ICH) are still not entirely clear. In this study, a mouse model of ICH was established by the collagenase method. Rolipram reduced brain oedema, blood–brain barrier (BBB) leakage, neuronal apoptosis and inflammatory cytokine release and improved neurological function in our mouse model of ICH. Moreover, rolipram increased the levels of cAMP and silent information regulator 1 (SIRT1) and upregulated the phosphorylation of AMP-activated protein kinase (AMPK). Furthermore, these effects of rolipram could be reversed by the SIRT1 inhibitor sirtinol. In conclusion, rolipram can play a neuroprotective role in the pathological process of ICH by activating the cAMP/AMPK/SIRT1 pathway.
Collapse
Affiliation(s)
- Xiao-Liu Dong
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, 300052, China.,Department of Neurorehabilitation, Tangshan People's Hospital, Tangshan, 063000, China
| | - Yan-Hui Wang
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Jing Xu
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, 300052, China.
| | - Nan Zhang
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, 300052, China.
| |
Collapse
|
25
|
Shi Y, Dai Q, Ji B, Huang L, Zhuang X, Mo Y, Wang J. Electroacupuncture Pretreatment Prevents Cognitive Impairment Induced by Cerebral Ischemia-Reperfusion via Adenosine A1 Receptors in Rats. Front Aging Neurosci 2021; 13:680706. [PMID: 34413765 PMCID: PMC8369428 DOI: 10.3389/fnagi.2021.680706] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 06/07/2021] [Indexed: 12/20/2022] Open
Abstract
A previous study has demonstrated that pretreatment with electroacupuncture (EA) induces rapid tolerance to focal cerebral ischemia. In the present study, we investigated whether adenosine receptor 1 (A1 R) is involved in EA pretreatment-induced cognitive impairment after focal cerebral ischemia in rats. Two hours after EA pretreatment, focal cerebral ischemia was induced by middle cerebral artery occlusion for 120 min in male Sprague-Dawley rats. The neurobehavioral score, cognitive function [as determined by the Morris water maze (MWM) test], neuronal number, and the Bax/Bcl-2 ratio was evaluated at 24 h after reperfusion in the presence or absence of CCPA (a selective A1 receptor agonist), DPCPX (a selective A1 receptor antagonist) into left lateral ventricle, or A1 short interfering RNA into the hippocampus area. The expression of the A1 receptor in the hippocampus was also investigated. The result showed that EA pretreatment upregulated the neuronal expression of the A1 receptor in the rat hippocampus at 90 min. And EA pretreatment reversed cognitive impairment, improved neurological outcome, and inhibited apoptosis at 24 h after reperfusion. Pretreatment with CCPA could imitate the beneficial effects of EA pretreatment. But the EA pretreatment effects were abolished by DPCPX. Furthermore, A1 receptor protein was reduced by A1 short interfering RNA which attenuated EA pretreatment-induced cognitive impairment.
Collapse
Affiliation(s)
- Yiyi Shi
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Qinxue Dai
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Binbin Ji
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Luping Huang
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiuxiu Zhuang
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yunchang Mo
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Junlu Wang
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
26
|
Thapa K, Singh TG, Kaur A. Cyclic nucleotide phosphodiesterase inhibition as a potential therapeutic target in renal ischemia reperfusion injury. Life Sci 2021; 282:119843. [PMID: 34298037 DOI: 10.1016/j.lfs.2021.119843] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 07/10/2021] [Accepted: 07/13/2021] [Indexed: 12/19/2022]
Abstract
AIMS Ischemia/reperfusion (I/R) occurs in renal artery stenosis, partial nephrectomy and most commonly during kidney transplantation. It brings serious consequences such as DGF (Delayed Graft Function) or organ dysfunction leading to renal failure and ultimate death. There is no effective therapy to handle the consequences of Renal Ischemia/Reperfusion (I/R) injury. Cyclic nucleotides, cAMP and cGMP are the important second messengers that stimulate intracellular signal transduction for cell survival in response to growth factors and peptide hormones in normal tissues and in kidneys plays significant role that involves vascular tone regulation, inflammation and proliferation of parenchymal cells. Renal ischemia and subsequent reperfusion injury stimulate signal transduction pathways involved in oxidative stress, inflammation, alteration in renal blood flow leading to necrosis and apoptosis of renal cell. MATERIALS AND METHODS An extensive literature review of various search engines like PubMed, Medline, Bentham, Scopus, and EMBASE (Elsevier) databases was carried out. To understand the functioning of Phosphodiesterases (PDEs) and its pharmacological modulation in Renal Ischemia-Reperfusion Injury. KEY FINDINGS Current therapeutic options may not be enough to treat renal I/R injury in group of patients and therefore, the current review has discussed the general characteristics and physiology of PDEs and preclinical-studies defining the relationship between PDEs expression in renal injury due to I/R and its outcome on renal function. SIGNIFICANCE The role of PDE inhibitors in renal I/R injury and the clinical status of drugs for various renal diseases have been summarized in this review.
Collapse
Affiliation(s)
- Komal Thapa
- Chitkara College of Pharmacy, Chitkara University, 140401 Punjab, India; School of Pharmacy, Himachal Pradesh, India
| | | | - Amarjot Kaur
- Chitkara College of Pharmacy, Chitkara University, 140401 Punjab, India
| |
Collapse
|
27
|
Ponsaerts L, Alders L, Schepers M, de Oliveira RMW, Prickaerts J, Vanmierlo T, Bronckaers A. Neuroinflammation in Ischemic Stroke: Inhibition of cAMP-Specific Phosphodiesterases (PDEs) to the Rescue. Biomedicines 2021; 9:703. [PMID: 34206420 PMCID: PMC8301462 DOI: 10.3390/biomedicines9070703] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 06/15/2021] [Accepted: 06/16/2021] [Indexed: 12/27/2022] Open
Abstract
Ischemic stroke is caused by a thromboembolic occlusion of a major cerebral artery, with the impaired blood flow triggering neuroinflammation and subsequent neuronal damage. Both the innate immune system (e.g., neutrophils, monocytes/macrophages) in the acute ischemic stroke phase and the adaptive immune system (e.g., T cells, B cells) in the chronic phase contribute to this neuroinflammatory process. Considering that the available therapeutic strategies are insufficiently successful, there is an urgent need for novel treatment options. It has been shown that increasing cAMP levels lowers neuroinflammation. By inhibiting cAMP-specific phosphodiesterases (PDEs), i.e., PDE4, 7, and 8, neuroinflammation can be tempered through elevating cAMP levels and, thereby, this can induce an improved functional recovery. This review discusses recent preclinical findings, clinical implications, and future perspectives of cAMP-specific PDE inhibition as a novel research interest for the treatment of ischemic stroke. In particular, PDE4 inhibition has been extensively studied, and is promising for the treatment of acute neuroinflammation following a stroke, whereas PDE7 and 8 inhibition more target the T cell component. In addition, more targeted PDE4 gene inhibition, or combined PDE4 and PDE7 or 8 inhibition, requires more extensive research.
Collapse
Affiliation(s)
- Laura Ponsaerts
- Biomedical Research Institute, Hasselt University, 3500 Hasselt, Belgium; (L.P.); (L.A.); (M.S.)
- European Graduate School of Neuroscience (EURON), Universiteitssingel 40, 6229 ER Maastricht, The Netherlands;
| | - Lotte Alders
- Biomedical Research Institute, Hasselt University, 3500 Hasselt, Belgium; (L.P.); (L.A.); (M.S.)
- European Graduate School of Neuroscience (EURON), Universiteitssingel 40, 6229 ER Maastricht, The Netherlands;
| | - Melissa Schepers
- Biomedical Research Institute, Hasselt University, 3500 Hasselt, Belgium; (L.P.); (L.A.); (M.S.)
- European Graduate School of Neuroscience (EURON), Universiteitssingel 40, 6229 ER Maastricht, The Netherlands;
- Department Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, European Graduate School of Neuroscience, Maastricht University, 6200 MD Maastricht, The Netherlands
| | | | - Jos Prickaerts
- European Graduate School of Neuroscience (EURON), Universiteitssingel 40, 6229 ER Maastricht, The Netherlands;
- Department Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, European Graduate School of Neuroscience, Maastricht University, 6200 MD Maastricht, The Netherlands
| | - Tim Vanmierlo
- Biomedical Research Institute, Hasselt University, 3500 Hasselt, Belgium; (L.P.); (L.A.); (M.S.)
- European Graduate School of Neuroscience (EURON), Universiteitssingel 40, 6229 ER Maastricht, The Netherlands;
- Department Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, European Graduate School of Neuroscience, Maastricht University, 6200 MD Maastricht, The Netherlands
| | - Annelies Bronckaers
- Biomedical Research Institute, Hasselt University, 3500 Hasselt, Belgium; (L.P.); (L.A.); (M.S.)
- European Graduate School of Neuroscience (EURON), Universiteitssingel 40, 6229 ER Maastricht, The Netherlands;
| |
Collapse
|
28
|
Saroj P, Bansal Y, Singh R, Akhtar A, Sodhi RK, Bishnoi M, Sah SP, Kuhad A. Neuroprotective effects of roflumilast against quinolinic acid-induced rat model of Huntington's disease through inhibition of NF-κB mediated neuroinflammatory markers and activation of cAMP/CREB/BDNF signaling pathway. Inflammopharmacology 2021; 29:499-511. [PMID: 33517508 DOI: 10.1007/s10787-020-00787-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 12/29/2020] [Indexed: 01/23/2023]
Abstract
Huntington's disease (HD) is a progressive neurodegenerative and hyperkinetic movement disorder. Decreased activity of cAMP-responsive element-binding protein (CREB) is thought to contribute to the death of striatal medium spiny neurons in HD. The present study has been designed to explore the possible role of roflumilast against qunilonic acid (QA) induced neurotoxicity in rats intending to investigate whether it inhibits the neuroinflammatory response through activation of the cAMP/CREB/BDNF signaling pathway. QA was microinjected (200 nmol/2 µl, bilaterally) through the intrastriatal route in the stereotaxic apparatus. Roflumilast (0.5, 1, and 2 mg/kg, orally) once-daily treatment for 21 days significantly improved locomotor activity in actophotometer, motor coordination in rotarod, and impaired gait performance in narrow beam walk test. Moreover, roflumilast treatment significantly attenuated oxidative and nitrosative stress (p < 0.05) through attenuating lipid peroxidation nitrite concentration and enhancing reduced glutathione, superoxide dismutase, and catalase levels. Furthermore, roflumilast also significantly decreased elevated pro-inflammatory cytokines like TNF-α (p < 0.01), IL-6 (p < 0.01), IFN-γ (p < 0.05), NF-κB (p < 0.05) and significantly increased BDNF(p < 0.05) in the striatum and cortex of rat brain. The results further demonstrated that roflumilast effectively increased the gene expression of cAMP(p < 0.05), CREB(p < 0.05) and decreased the gene expression of PDE4 (p < 0.05) in qRT-PCR. These results conclusively depicted that roflumilast could be a potential candidate as an effective therapeutic agent in the management of HD through the cAMP/CREB/BDNF signaling pathway.
Collapse
Affiliation(s)
- Priyanka Saroj
- Pharmacology Division, University Institute of Pharmaceutical Sciences (UIPS), UGC Centre of Advanced Study (UGC-CAS), Panjab University, Chandigarh, 160014, India
| | - Yashika Bansal
- Pharmacology Division, University Institute of Pharmaceutical Sciences (UIPS), UGC Centre of Advanced Study (UGC-CAS), Panjab University, Chandigarh, 160014, India
| | - Raghunath Singh
- Pharmacology Division, University Institute of Pharmaceutical Sciences (UIPS), UGC Centre of Advanced Study (UGC-CAS), Panjab University, Chandigarh, 160014, India
| | - Ansab Akhtar
- Pharmacology Division, University Institute of Pharmaceutical Sciences (UIPS), UGC Centre of Advanced Study (UGC-CAS), Panjab University, Chandigarh, 160014, India
| | - Rupinder Kaur Sodhi
- Pharmacology Division, University Institute of Pharmaceutical Sciences (UIPS), UGC Centre of Advanced Study (UGC-CAS), Panjab University, Chandigarh, 160014, India
| | - Mahendra Bishnoi
- National Agri-Food Biotechnology Institue (NABI), Sector 81, SAS Nagar, Mohali, Punjab, 140306, India
| | - Sangeeta Pilkhwal Sah
- Pharmacology Division, University Institute of Pharmaceutical Sciences (UIPS), UGC Centre of Advanced Study (UGC-CAS), Panjab University, Chandigarh, 160014, India.
- Department of Physiology, University Institute of Pharmaceutical Science (UIPS), UGC Center of Advanced Study (UGC-CAS), Panjab University, Chandigarh, 160014, India.
| | - Anurag Kuhad
- Pharmacology Division, University Institute of Pharmaceutical Sciences (UIPS), UGC Centre of Advanced Study (UGC-CAS), Panjab University, Chandigarh, 160014, India.
| |
Collapse
|
29
|
Gorny N, Kelly MP. Alterations in cyclic nucleotide signaling are implicated in healthy aging and age-related pathologies of the brain. VITAMINS AND HORMONES 2021; 115:265-316. [PMID: 33706951 DOI: 10.1016/bs.vh.2020.12.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
It is not only important to consider how hormones may change with age, but also how downstream signaling pathways that couple to hormone receptors may change. Among these hormone-coupled signaling pathways are the 3',5'-cyclic guanosine monophosphate (cGMP) and 3',5'-cyclic adenosine monophosphate (cAMP) intracellular second messenger cascades. Here, we test the hypothesis that dysfunction of cAMP and/or cGMP synthesis, execution, and/or degradation occurs in the brain during healthy and pathological diseases such as Alzheimer's disease, Parkinson's disease, and Huntington's disease. Although most studies report lower cyclic nucleotide signaling in the aged brain, with further reductions noted in the context of age-related diseases, there are select examples where cAMP signaling may be elevated in select tissues. Thus, therapeutics would need to target cAMP/cGMP in a tissue-specific manner if efficacy for select symptoms is to be achieved without worsening others.
Collapse
Affiliation(s)
- Nicole Gorny
- Department of Anatomy & Neurobiology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Michy P Kelly
- Department of Anatomy & Neurobiology, University of Maryland School of Medicine, Baltimore, MD, United States.
| |
Collapse
|
30
|
Tang C, Liu D, Fan Y, Yu J, Li C, Su J, Wang C. Visualization and bibliometric analysis of cAMP signaling system research trends and hotspots in cancer. J Cancer 2021; 12:358-370. [PMID: 33391432 PMCID: PMC7738981 DOI: 10.7150/jca.47158] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 10/02/2020] [Indexed: 12/13/2022] Open
Abstract
Cyclic adenosine monophosphate (cAMP) is an essential second messenger that widely distributed among prokaryotic and eukaryotic organisms. cAMP can regulate various biological processes, including cell proliferation, differentiation, apoptosis and immune functions. Any dysregulation or alteration of cAMP signaling may cause cell metabolic disorder, immune dysfunction and lead to disease or cancer. This study aimed to conduct a scientometric analysis of cAMP signaling system in cancer field, and explored the research trend, hotspots and frontiers from the past decade. Relevant literatures published from 2009 to 2019 were collected in the Web of Science Core Collection database. EndNote X9 was used to remove duplicate articles, and irrelevant articles were manually filtered. Bibliometric analyses were completed by CiteSpace V. A total of 4306 articles were included in this study. The number of related literatures published each year is gradually increasing. Most of them belong to “Biochemistry & Molecular Biology”, “Oncology”, “Cell Biology”, “Pharmacology & Pharmacy” and “Endocrinology & Metabolism” areas. In the past decade, USA, China, and Japan contributed the most to the research of cAMP signaling system in cancer. The frontiers and hotspots of cAMP signaling pathway system related to cancer fields mainly focused on cancer cell apoptosis, metastasis, and multiple tumors occurrence in patients with Carney complex. Intervention of the cAMP metabolic pathway may be a potential and promising therapeutic strategy for controlling clinical cancer and tumor diseases.
Collapse
Affiliation(s)
- Caoli Tang
- Department of Preventive Medicine, School of Public Health, Wuhan University, Donghu Road 115, Wuhan 430071, Hubei, China
| | - Duanya Liu
- Department of Preventive Medicine, School of Public Health, Wuhan University, Donghu Road 115, Wuhan 430071, Hubei, China
| | - Yongsheng Fan
- Department of Preventive Medicine, School of Public Health, Wuhan University, Donghu Road 115, Wuhan 430071, Hubei, China
| | - Jun Yu
- Department of Preventive Medicine, School of Public Health, Wuhan University, Donghu Road 115, Wuhan 430071, Hubei, China
| | - Cong Li
- Department of Preventive Medicine, School of Public Health, Wuhan University, Donghu Road 115, Wuhan 430071, Hubei, China
| | - Jianmei Su
- Department of Preventive Medicine, School of Public Health, Wuhan University, Donghu Road 115, Wuhan 430071, Hubei, China.,Key Laboratory of Regional Development and Environmental Response, Faculty of Resources and Environmental Science, Hubei University, Friendship Avenue 368, Wuhan 430062, Hubei, China
| | - Chunhong Wang
- Department of Preventive Medicine, School of Public Health, Wuhan University, Donghu Road 115, Wuhan 430071, Hubei, China
| |
Collapse
|
31
|
Lv J, Chen L, Zhu N, Sun Y, Pan J, Gao J, Liu J, Liu G, Tao Y. Beta amyloid-induced time-dependent learning and memory impairment: involvement of HPA axis dysfunction. Metab Brain Dis 2020; 35:1385-1394. [PMID: 32860609 DOI: 10.1007/s11011-020-00613-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 08/24/2020] [Indexed: 12/28/2022]
Abstract
Aβ aggregation is one of the pathological biomarkers of Alzheimer's disease (AD). However, the possible mechanism related to Aβ-induced pathological signaling pathway is still unknown. In the present study, Aβ1-42-induced time-dependent memory impairment and its possible relationship to hypothalamic-pituitary-adrenal (HPA) axis hyperactivity were examined. Aβ1-42-treated mice significantly impaired acquisition activity in the learning curve at 10 days, 1 and 4 months in the Morris water-maze (MWM) task. This learning activity was back to normal at 8 months after Aβ1-42 treatment. In the probe trial test, Aβ1-42-treated mice needed longer latencies to touch the precious platform location and fewer numbers of crossing from 10 days to 4 months after microinjection. This Aβ1-42 induced memory loss was consistent with the results of the step-down passive avoidance test. The HPA axis related parameters, such as corticosterone (CORT) level in the serum, glucocorticoid receptor (GR) and corticotropin-releasing factor receptor (CRF-R) expression in the frontal cortex and hippocampus increased in Aβ1-42-treated mice from 10 days to 4 months. While the downstream molecules phosphorylation of cyclic AMP response element binding (pCREB) and brain-derived neurotrophic factor (BDNF) expression decreased during this time. These effects were back to normal 8 months after treatment with Aβ1-42. Altogether, our results suggested that Aβ1-42 induced significant learning and memory impairment, which is involved in HPA axis dysfunction.
Collapse
Affiliation(s)
- Jinpeng Lv
- College of Pharmaceutical Engineering and Life Sciences, Changzhou University, Changzhou, 213000, China
| | - Ling Chen
- Department of Clinical Pharmacology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, People's Republic of China
| | - Naping Zhu
- Brain Institute, School of Pharmacy, Wenzhou Medical University, Wenzhou, 325035, China
| | - Yindi Sun
- Department of Traditional Medical Orthopedics, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054, Shaanxi, China
| | - Jianchun Pan
- Brain Institute, School of Pharmacy, Wenzhou Medical University, Wenzhou, 325035, China
| | - Jinsheng Gao
- Department of Oncology, Shanxi Province Research Institute of Traditional Chinese Medicine, Taiyuan, 030000, China
| | - Jianwu Liu
- College of Pharmaceutical Engineering and Life Sciences, Changzhou University, Changzhou, 213000, China
| | - Guangjun Liu
- The Second People's Hospital of Changzhou, Affiliate Hospital of NanJing Medical University, Changzhou, 213000, China.
| | - Yuanxiang Tao
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, 07103, USA.
| |
Collapse
|
32
|
Chang YF, Zhang D, Hu WM, Liu DX, Li L. Semaglutide-mediated protection against Aβ correlated with enhancement of autophagy and inhibition of apotosis. J Clin Neurosci 2020; 81:234-239. [PMID: 33222922 DOI: 10.1016/j.jocn.2020.09.054] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 08/11/2020] [Accepted: 09/28/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND Semaglutide, a glucagon-like peptide-1 (GLP-1) analogue with an extended half-life of approximately 1 week has being come into clinic trial to treat parkingson's disease but little is known about its effect to prevent against Alzheimer's disease (AD). The goal of the present study was to explore the potential mechanisms of semaglutide to protect against AD. METHODS We treated SH-SY5Y cell line with Aβ25-35 as an AD model. Further, SH-SY5Y cells damaged by Aβ25-35 were treated by semaglutide. Autophagy-related proteins and apoptosis-related proteins were measured to explore molecular mechanisms for semaglutide to protect against Aβ25-35. RESULTS Semaglutide enhanced autophagy by increasing the expression of LC3II, Atg7, Beclin-1 and P62 which were inhibited by Aβ25-35. Further we showed that semaglutide inhibited apoptosis by inhibiting the expression of Bax induced by Aβ25-35 and increasing the expression of Bcl2 inhibited by Aβ25-35. CONCLUSION Our results provide a clue for the hypothesis that autophagy enhancement and apoptosis inhibition may be involved in the effect of semaglutide to protect against Aβ 25-35.
Collapse
Affiliation(s)
- Yan-Fang Chang
- Key Laboratory of Cellular Physiology, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Di Zhang
- Chemistry Department, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Wei-Min Hu
- Neurology Department, Second Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Dong-Xing Liu
- Key Laboratory of Cellular Physiology, Shanxi Medical University, Taiyuan, Shanxi, China; Neurology Department, Shanxi Cardiovascular Hospital, Taiyuan, Shanxi, China
| | - Lin Li
- Key Laboratory of Cellular Physiology, Shanxi Medical University, Taiyuan, Shanxi, China.
| |
Collapse
|
33
|
Roflumilast: A potential drug for the treatment of cognitive impairment? Neurosci Lett 2020; 736:135281. [DOI: 10.1016/j.neulet.2020.135281] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 07/19/2020] [Accepted: 07/25/2020] [Indexed: 12/16/2022]
|
34
|
Dominant-Negative Attenuation of cAMP-Selective Phosphodiesterase PDE4D Action Affects Learning and Behavior. Int J Mol Sci 2020; 21:ijms21165704. [PMID: 32784895 PMCID: PMC7460819 DOI: 10.3390/ijms21165704] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 07/26/2020] [Accepted: 08/06/2020] [Indexed: 12/22/2022] Open
Abstract
PDE4 cyclic nucleotide phosphodiesterases reduce 3′, 5′ cAMP levels in the CNS and thereby regulate PKA activity and the phosphorylation of CREB, fundamental to depression, cognition, and learning and memory. The PDE4 isoform PDE4D5 interacts with the signaling proteins β-arrestin2 and RACK1, regulators of β2-adrenergic and other signal transduction pathways. Mutations in PDE4D in humans predispose to acrodysostosis, associated with cognitive and behavioral deficits. To target PDE4D5, we developed mice that express a PDE4D5-D556A dominant-negative transgene in the brain. Male transgenic mice demonstrated significant deficits in hippocampus-dependent spatial learning, as assayed in the Morris water maze. In contrast, associative learning, as assayed in a fear conditioning assay, appeared to be unaffected. Male transgenic mice showed augmented activity in prolonged (2 h) open field testing, while female transgenic mice showed reduced activity in the same assay. Transgenic mice showed no demonstrable abnormalities in prepulse inhibition. There was also no detectable difference in anxiety-like behavior, as measured in the elevated plus-maze. These data support the use of a dominant-negative approach to the study of PDE4D5 function in the CNS and specifically in learning and memory.
Collapse
|
35
|
Bhat A, Ray B, Mahalakshmi AM, Tuladhar S, Nandakumar DN, Srinivasan M, Essa MM, Chidambaram SB, Guillemin GJ, Sakharkar MK. Phosphodiesterase-4 enzyme as a therapeutic target in neurological disorders. Pharmacol Res 2020; 160:105078. [PMID: 32673703 DOI: 10.1016/j.phrs.2020.105078] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 07/09/2020] [Accepted: 07/10/2020] [Indexed: 02/08/2023]
Abstract
Phosphodiesterases (PDE) are a diverse family of enzymes (11 isoforms so far identified) responsible for the degradation of cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP) which are involved in several cellular and biochemical functions. Phosphodiesterase 4 (PDE4) is the major isoform within this group and is highly expressed in the mammalian brain. An inverse association between PDE4 and cAMP levels is the key mechanism in various pathophysiological conditions like airway inflammatory diseases-chronic obstruction pulmonary disease (COPD), asthma, psoriasis, rheumatoid arthritis, and neurological disorders etc. In 2011, roflumilast, a PDE4 inhibitor (PDE4I) was approved for the treatment of COPD. Subsequently, other PDE4 inhibitors (PDE4Is) like apremilast and crisaborole were approved by the Food and Drug Administration (FDA) for psoriasis, atopic dermatitis etc. Due to the adverse effects like unbearable nausea and vomiting, dose intolerance and diarrhoea, PDE4 inhibitors have very less clinical compliance. Efforts are being made to develop allosteric modulation with high specificity to PDE4 isoforms having better efficacy and lesser adverse effects. Interestingly, repositioning PDE4Is towards neurological disorders including Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), multiple sclerosis (MS) and sleep disorders, is gaining attention. This review is an attempt to summarize the data on the effects of PDE4 overexpression in neurological disorders and the use of PDE4Is and newer allosteric modulators as therapeutic options. We have also compiled a list of on-going clinical trials on PDE4 inhibitors in neurological disorders.
Collapse
Affiliation(s)
- Abid Bhat
- Dept. of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, India
| | - Bipul Ray
- Dept. of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, India
| | | | - Sunanda Tuladhar
- Dept. of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, India
| | - D N Nandakumar
- Department of Neurochemistry, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, 560029, India
| | - Malathi Srinivasan
- Department of Lipid Science, CSIR - Central Food Technological Research Institute (CFTRI), CFTRI Campus, Mysuru, 570020, India
| | - Musthafa Mohamed Essa
- Ageing and Dementia Research Group, Sultan Qaboos University, Muscat, Oman; Department of Food Science and Nutrition, CAMS, Sultan Qaboos University, Muscat, Oman.
| | - Saravana Babu Chidambaram
- Dept. of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, India; Centre for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education & Research, Mysuru, India.
| | - Gilles J Guillemin
- Neuroinflammation group, Faculty of Medicine and Health Sciences, Macquarie University, NSW, 2109, Australia.
| | - Meena Kishore Sakharkar
- College of Pharmacy and Nutrition, University of Saskatchewan, 107, Wiggins Road, Saskatoon, SK, S7N 5C9, Canada
| |
Collapse
|
36
|
Wang H, Zhang FF, Xu Y, Fu HR, Wang XD, Wang L, Chen W, Xu XY, Gao YF, Zhang JG, Zhang HT. The Phosphodiesterase-4 Inhibitor Roflumilast, a Potential Treatment for the Comorbidity of Memory Loss and Depression in Alzheimer's Disease: A Preclinical Study in APP/PS1 Transgenic Mice. Int J Neuropsychopharmacol 2020; 23:700-711. [PMID: 32645141 PMCID: PMC7727475 DOI: 10.1093/ijnp/pyaa048] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Revised: 06/24/2020] [Accepted: 07/07/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Depression is highly related to Alzheimer's disease (AD), yet no effective treatment is available. Phosphodiesterase-4 (PDE4) has been considered a promising target for treatment of AD and depression. Roflumilast, the first PDE4 inhibitor approved for clinical use, improves cognition at doses that do not cause side effects such as emesis. METHODS Here we examined the effects of roflumilast on behavioral dysfunction and the related mechanisms in APPswe/PS1dE9 transgenic mice, a widely used model of AD. Mice at 10 months of age were examined for memory in the novel object recognition and Morris water-maze tests and depression-like behavior in the tail-suspension test and forced swimming test before killing for neurochemical assays. RESULTS In the novel object recognition and Morris water-maze, APPswe/PS1dE9 mice showed significant cognitive declines, which were reversed by roflumilast at 5 and 10 mg/kg orally once per day. In the tail-suspension test and forced swimming test, the AD mice showed prolonged immobility time, which was also reversed by roflumilast. In addition, the staining of hematoxylin-eosin and Nissl showed that roflumilast relieved the neuronal cell injuries, while terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick-end labelling analysis indicated that roflumilast ameliorated cell apoptosis in AD mice. Further, roflumilast reversed the decreased ratio of B-cell lymphoma-2/Bcl-2-associated X protein and the increased expression of PDE4B and PDE4D in the cerebral cortex and hippocampus of AD mice. Finally, roflumilast reversed the decreased levels of cyclic AMP (cAMP) and expression of phosphorylated cAMP response element-binding protein and brain derived neurotrophic factor in AD mice. CONCLUSIONS Together, these results suggest that roflumilast not only improves learning and memory but also attenuates depression-like behavior in AD mice, likely via PDE4B/PDE4D-mediated cAMP/cAMP response element-binding protein/brain derived neurotrophic factor signaling. Roflumilast can be a therapeutic agent for AD, in particular the comorbidity of memory loss and depression.
Collapse
Affiliation(s)
- Hao Wang
- Institute of Pharmacology, Shandong First Medical University and Shandong Academy of Medical Sciences, Tai’an, China
| | - Fang-fang Zhang
- Institute of Pharmacology, Shandong First Medical University and Shandong Academy of Medical Sciences, Tai’an, China
| | - Yong Xu
- Institute of Pharmacology, Shandong First Medical University and Shandong Academy of Medical Sciences, Tai’an, China
| | - Hua-rong Fu
- Institute of Pharmacology, Shandong First Medical University and Shandong Academy of Medical Sciences, Tai’an, China
| | - Xiao-dan Wang
- Institute of Pharmacology, Shandong First Medical University and Shandong Academy of Medical Sciences, Tai’an, China
| | - Lei Wang
- Institute of Pharmacology, Shandong First Medical University and Shandong Academy of Medical Sciences, Tai’an, China
| | - Wei Chen
- Institute of Pharmacology, Shandong First Medical University and Shandong Academy of Medical Sciences, Tai’an, China
| | - Xiao-yan Xu
- Institute of Pharmacology, Shandong First Medical University and Shandong Academy of Medical Sciences, Tai’an, China
| | - Yong-feng Gao
- Institute of Pharmacology, Shandong First Medical University and Shandong Academy of Medical Sciences, Tai’an, China
| | - Ji-guo Zhang
- Institute of Pharmacology, Shandong First Medical University and Shandong Academy of Medical Sciences, Tai’an, China
| | - Han-Ting Zhang
- Departments of Neuroscience and Behavioral Medicine & Psychiatry, the Rockefeller Neuroscience Institute, West Virginia University Health Sciences Center, Morgantown, West Virginia,Correspondence: Han-Ting Zhang, MD, PhD, Department of Neuroscience, the Rockefeller Neurosciences Institute, West Virginia University Health Sciences Center, Morgantown, WV 26506 ()
| |
Collapse
|
37
|
Sanders O, Rajagopal L. Phosphodiesterase Inhibitors for Alzheimer's Disease: A Systematic Review of Clinical Trials and Epidemiology with a Mechanistic Rationale. J Alzheimers Dis Rep 2020; 4:185-215. [PMID: 32715279 PMCID: PMC7369141 DOI: 10.3233/adr-200191] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/17/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Preclinical studies, clinical trials, and reviews suggest increasing 3',5'-cyclic adenosine monophosphate (cAMP) and 3',5'-cyclic guanosine monophosphate (cGMP) with phosphodiesterase inhibitors is disease-modifying in Alzheimer's disease (AD). cAMP/protein kinase A (PKA) and cGMP/protein kinase G (PKG) signaling are disrupted in AD. cAMP/PKA and cGMP/PKG activate cAMP response element binding protein (CREB). CREB binds mitochondrial and nuclear DNA, inducing synaptogenesis, memory, and neuronal survival gene (e.g., brain-derived neurotrophic factor) and peroxisome proliferator-activated receptor-γ coactivator-1α (PGC1α). cAMP/PKA and cGMP/PKG activate Sirtuin-1, which activates PGC1α. PGC1α induces mitochondrial biogenesis and antioxidant genes (e.g.,Nrf2) and represses BACE1. cAMP and cGMP inhibit BACE1-inducing NFκB and tau-phosphorylating GSK3β. OBJECTIVE AND METHODS We review efficacy-testing clinical trials, epidemiology, and meta-analyses to critically investigate whether phosphodiesteraseinhibitors prevent or treat AD. RESULTS Caffeine and cilostazol may lower AD risk. Denbufylline and sildenafil clinical trials are promising but preliminary and inconclusive. PF-04447943 and BI 409,306 are ineffective. Vinpocetine, cilostazol, and nicergoline trials are mixed. Deprenyl/selegiline trials show only short-term benefits. Broad-spectrum phosphodiesterase inhibitor propentofylline has been shown in five phase III trials to improve cognition, dementia severity, activities of daily living, and global assessment in mild-to-moderate AD patients on multiple scales, including the ADAS-Cogand the CIBIC-Plus in an 18-month phase III clinical trial. However, two books claimed based on a MedScape article an 18-month phase III trial failed, so propentofylline was discontinued. Now, propentofylline is used to treat canine cognitive dysfunction, which, like AD, involves age-associated wild-type Aβ deposition. CONCLUSION Phosphodiesterase inhibitors may prevent and treat AD.
Collapse
|
38
|
Ooi TC, Ahmad Munawar M, Mohd Rosli NH, Abdul Malek SNA, Rosli H, Ibrahim FW, Azmi N, Haron H, Sharif R, Shahar S, Rajab NF. Neuroprotection of Tropical Fruit Juice Mixture via the Reduction of iNOS Expression and CRH Level in β-Amyloid-Induced Rats Model of Alzheimer's Disease. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2020; 2020:5126457. [PMID: 32382294 PMCID: PMC7180421 DOI: 10.1155/2020/5126457] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 03/16/2020] [Accepted: 03/21/2020] [Indexed: 01/08/2023]
Abstract
This study aimed to determine the effects of tropical fruit juice mixture (pomegranate, white guava, and Roselle) on biochemical, behavioral, and histopathological changes of β-amyloid- (Aβ-) induced rats. Formulation 8 (F8) of tropical fruit juice mixture was chosen for this present study due to its high phenolic content and antioxidant capacity. Forty Wistar male rats were divided into five groups: dPBS (sham-operated control), dAβ (Aβ control), JPBS (F8 and PBS), JAβ (F8 and Aβ), and IBFAβ (ibuprofen and Aβ). F8 (5 ml/kg BW), and ibuprofen (10 ml/kg BW) was given orally daily for four weeks before the intracerebroventricular infusion of Aβ for two weeks. Histological analysis and neuronal count of hippocampus tissue in the Cornu Ammonis (CA1) region showed that supplementation with F8 was able to prevent Aβ-induced tissue damage and neuronal shrinkage. However, no significant difference in locomotor activity and novel object recognition (NOR) percentage was detected among different groups at day 7 and day 14 following Aβ infusion. Only effect of time differences (main effect of day) was observed at day 7 as compared to day 14, where reduction in locomotor activity and NOR percentage was observed in all groups, with F (1, 7) = 6.940, p < 0.05 and F (1, 7) = 7.152, p < 0.05, respectively. Besides, the MDA level of the JAβ group was significantly lower (p < 0.01) than that of the dPBS group. However, no significant changes in SOD activity were detected among different groups. Significant reduction in plasma CRH level (p < 0.05) and iNOS expression (p < 0.01) in the brain was detected in the JAβ group as compared to the dAβ group. Hence, our current findings suggest that the tropical fruit juice mixture (F8) has the potential to protect the rats from Aβ-induced neurotoxicity in brain hippocampus tissue possibly via its antioxidant properties and the suppression of iNOS expression and CRH production.
Collapse
Affiliation(s)
- Theng Choon Ooi
- Center for Healthy Ageing & Wellness, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, 50300 Kuala Lumpur, Malaysia
| | - Munirah Ahmad Munawar
- Biomedical Sciences Programme, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, 50300 Kuala Lumpur, Malaysia
| | - Nur Hasnieza Mohd Rosli
- Biomedical Sciences Programme, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, 50300 Kuala Lumpur, Malaysia
| | - Siti Nur Aqilah Abdul Malek
- Center for Healthy Ageing & Wellness, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, 50300 Kuala Lumpur, Malaysia
| | - Hanisah Rosli
- Center for Healthy Ageing & Wellness, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, 50300 Kuala Lumpur, Malaysia
| | - Farah Wahida Ibrahim
- Biomedical Sciences Programme, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, 50300 Kuala Lumpur, Malaysia
- Center for Toxicology and Health Risk Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, 50300 Kuala Lumpur, Malaysia
| | - Norazrina Azmi
- Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, 50300 Kuala Lumpur, Malaysia
| | - Hasnah Haron
- Center for Healthy Ageing & Wellness, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, 50300 Kuala Lumpur, Malaysia
| | - Razinah Sharif
- Center for Healthy Ageing & Wellness, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, 50300 Kuala Lumpur, Malaysia
| | - Suzana Shahar
- Center for Healthy Ageing & Wellness, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, 50300 Kuala Lumpur, Malaysia
| | - Nor Fadilah Rajab
- Center for Healthy Ageing & Wellness, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, 50300 Kuala Lumpur, Malaysia
- Biomedical Sciences Programme, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, 50300 Kuala Lumpur, Malaysia
| |
Collapse
|
39
|
Hai-Na Z, Xu-Ben Y, Cong-Rong T, Yan-Cheng C, Fan Y, Lei-Mei X, Ruo-Lan S, Ye-Zi, Ye-Xuan W, Jing L. Atorvastatin ameliorates depressive behaviors and neuroinflammatory in streptozotocin-induced diabetic mice. Psychopharmacology (Berl) 2020; 237:695-705. [PMID: 31786648 DOI: 10.1007/s00213-019-05406-w] [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: 01/26/2019] [Accepted: 11/18/2019] [Indexed: 12/13/2022]
Abstract
Depression is a chronic and progressive syndrome and commonly associated with several neuropsychiatric comorbidities, of which depression is the most studied. It has been demonstrated that statins also have anti-inflammatory and immunomodulatory properties, which being explored for potential benefits in depression. However, the role of statins in the treatment of diabetes-related depression has not been well examined. Herein, we investigated the effects of atorvastatin on depressive behaviors and neuroinflammation in streptozotocin-induced diabetic mice. Our data indicated that oral administration of atorvastatin at 10 or 20 mg/kg for 3 weeks markedly ameliorated diabetes-associated depressive behaviors reflected by better performance in sucrose preference test (SPT), tail suspension test (TST), and novelty-suppressed feeding test (NSFT). The study further showed that atrovastatin decreased the expression of nucleus NF-κB p65 expression and ameliorated neuroinflammatory responses in prefrontal cortex as evidenced by less Iba-1-positive cells and lower inflammatory mediators including IL-1β and TNF-α. As expected, atorvastatin-treated diabetic mice exhibited significant improvement of hyperlipidemia rather than hyperglycemia. These results suggest that atorvastatin has the potential to be employed as a therapy for diabetes-related depression.
Collapse
Affiliation(s)
- Zhang Hai-Na
- Department of Pharmacy, Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China.,Department of Pharmacy, Wenzhou Medical University, Wenzhou, 325000, China
| | - Yu Xu-Ben
- Department of Pharmacy, Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China.,Department of Pharmacy, Wenzhou Medical University, Wenzhou, 325000, China
| | - Tang Cong-Rong
- Department of Pharmacy, Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China.,Department of Pharmacy, Wenzhou Medical University, Wenzhou, 325000, China
| | - Cao Yan-Cheng
- Department of Pharmacy, Wenzhou Medical University, Wenzhou, 325000, China
| | - Yang Fan
- Department of Pharmacy, Wenzhou Medical University, Wenzhou, 325000, China
| | - Xu Lei-Mei
- Department of Pharmacy, Wenzhou Medical University, Wenzhou, 325000, China
| | - Sun Ruo-Lan
- Department of Pharmacy, Wenzhou Medical University, Wenzhou, 325000, China
| | - Ye-Zi
- Department of Pharmacy, Wenzhou Medical University, Wenzhou, 325000, China
| | - Wang Ye-Xuan
- Department of Pharmacy, Wenzhou Medical University, Wenzhou, 325000, China
| | - Liang Jing
- Department of Pharmacy, Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China. .,Department of Pharmacy, Wenzhou Medical University, Wenzhou, 325000, China.
| |
Collapse
|
40
|
Understanding PDE4's function in Alzheimer's disease; a target for novel therapeutic approaches. Biochem Soc Trans 2020; 47:1557-1565. [PMID: 31642904 PMCID: PMC6824677 DOI: 10.1042/bst20190763] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 09/25/2019] [Accepted: 09/27/2019] [Indexed: 12/22/2022]
Abstract
Phosphodiesterases (PDEs) have long been considered as targets for the treatment of Alzheimer's disease (AD) and a substantial body of evidence suggests that one sub-family from the super-family of PDEs, namely PDE4D, has particular significance in this context. This review discusses the role of PDE4 in the orchestration of cAMP response element binding signaling in AD and outlines the benefits of targeting PDE4D specifically. We examine the limited available literature that suggests PDE4 expression does not change in AD brains together with reports that show PDE4 inhibition as an effective treatment in this age-related neurodegenerative disease. Actually, aging induces changes in PDE4 expression/activity in an isoform and brain-region specific manner that proposes a similar complexity in AD brains. Therefore, a more detailed account of AD-related alterations in cellular/tissue location and the activation status of PDE4 is required before novel therapies can be developed to target cAMP signaling in this disease.
Collapse
|
41
|
Mehri N, Haddadi R, Ganji M, Shahidi S, Soleimani Asl S, Taheri Azandariani M, Ranjbar A. Effects of vitamin D in an animal model of Alzheimer's disease: behavioral assessment with biochemical investigation of Hippocampus and serum. Metab Brain Dis 2020; 35:263-274. [PMID: 31853828 DOI: 10.1007/s11011-019-00529-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 12/09/2019] [Indexed: 02/06/2023]
Abstract
Regulatory role of vitamin D (VitD) in cognitive memory and learning has been proposed. Here, we examine the behavioral and biochemical effects of VitD in Alzheimer's disease (AD), as the most common form of dementia, in male Wistar rats. Animals (n = 48) were randomly divided into six groups: control, sham solvent, sham surgery, VitD (by intraperitoneal injection), AD (receiving intrahippocampal injection of amyloid-beta peptide, Aβ), and combination of VitD and Aβ. Learning and memory functions were investigated through the passive avoidance and the Morris water maze (MWM) tasks. Moreover, oxidative stress biomarkers including total antioxidant capacity (TAC), total thiol groups (TTG), lipid peroxidation (LPO), and DNA damage were assessed in hippocampus and serum. In passive avoidance task, Aβ significantly impaired the step-through latency and time in dark compartment. It also increased escape latency and time spent in the target quadrant in the MWM. VitD administration attenuated the Aβ-induced memory impairment in passive avoidance and MWM tests. Furthermore, VitD reduced deleterious biochemical effect of Aβ by enhancing the levels of TAC and TTG in addition to decreasing LPO and DNA damage levels in both hippocampus and serum. We showed, for the first time, that VitD administration improves the impaired Aβ-induced memory and that, by acting as a strong antioxidant, it can attenuate the stress oxidative biomarkers in hippocampus and serum of rats with AD. Altogether, our results provide evidence for further application of VitD in neurodegenerative disorders such as AD to enlighten the involved mechanisms.
Collapse
Affiliation(s)
- Negar Mehri
- Department of Toxicology and Pharmacology, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Rasool Haddadi
- Department of Toxicology and Pharmacology, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Maziar Ganji
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Siamak Shahidi
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Sara Soleimani Asl
- Department of Anatomy, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
- Endometrium and Endometriosis Research Centre, Hamadan University of Medical Sciences, Hamadan, Iran
| | | | - Akram Ranjbar
- Department of Toxicology and Pharmacology, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, Iran.
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran.
| |
Collapse
|
42
|
Tancheva LP, Lazarova MI, Alexandrova AV, Dragomanova ST, Nicoletti F, Tzvetanova ER, Hodzhev YK, Kalfin RE, Miteva SA, Mazzon E, Tzvetkov NT, Atanasov AG. Neuroprotective Mechanisms of Three Natural Antioxidants on a Rat Model of Parkinson's Disease: A Comparative Study. Antioxidants (Basel) 2020; 9:antiox9010049. [PMID: 31935828 PMCID: PMC7022962 DOI: 10.3390/antiox9010049] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Revised: 12/28/2019] [Accepted: 12/30/2019] [Indexed: 02/07/2023] Open
Abstract
We compared the neuroprotective action of three natural bio-antioxidants (AOs): ellagic acid (EA), α-lipoic acid (LA), and myrtenal (Myrt) in an experimental model of Parkinson’s disease (PD) that was induced in male Wistar rats through an intrastriatal injection of 6-hydroxydopamine (6-OHDA). The animals were divided into five groups: the sham-operated (SO) control group; striatal 6-OHDA-lesioned control group; and three groups of 6-OHDA-lesioned rats pre-treated for five days with EA, LA, and Myrt (50 mg/kg; intraperitoneally- i.p.), respectively. On the 2nd and the 3rd week post lesion, the animals were subjected to several behavioral tests: apomorphine-induced rotation; rotarod; and the passive avoidance test. Biochemical evaluation included assessment of main oxidative stress parameters as well as dopamine (DA) levels in brain homogenates. The results showed that all three test compounds improved learning and memory performance as well as neuromuscular coordination. Biochemical assays showed that all three compounds substantially decreased lipid peroxidation (LPO) levels, and restored catalase (CAT) activity and DA levels that were impaired by the challenge with 6-OHDA. Based on these results, we can conclude that the studied AOs demonstrate properties that are consistent with significant antiparkinsonian effects. The most powerful neuroprotective effect was observed with Myrt, and this work represents the first demonstration of its anti-Parkinsonian impact.
Collapse
Affiliation(s)
- Lyubka P. Tancheva
- Department of Behavior Neurobiology, Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia 1113, Bulgaria; (S.T.D.); (S.A.M.)
- Correspondence: (L.P.T.); (A.G.A.); Tel.: +359-2979-2175 (L.P.T.); +48-227-367-022 (A.G.A.)
| | - Maria I. Lazarova
- Department of Synaptic Signaling and Communications, Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia 1113, Bulgaria; (M.I.L.); (R.E.K.)
| | - Albena V. Alexandrova
- Department Biological Effects of Natural and Synthetic Substances, Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia 1113, Bulgaria; (A.V.A.); (E.R.T.)
| | - Stela T. Dragomanova
- Department of Behavior Neurobiology, Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia 1113, Bulgaria; (S.T.D.); (S.A.M.)
- Department of Pharmacology, Toxicology and Pharmacotherapy, Faculty of Pharmacy, Medical University, Varna 9002, Bulgaria
| | - Ferdinando Nicoletti
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 89, 95123 Catania, Italy;
| | - Elina R. Tzvetanova
- Department Biological Effects of Natural and Synthetic Substances, Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia 1113, Bulgaria; (A.V.A.); (E.R.T.)
| | - Yordan K. Hodzhev
- Department of Sensory Neurobiology, Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia 1113, Bulgaria;
| | - Reni E. Kalfin
- Department of Synaptic Signaling and Communications, Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia 1113, Bulgaria; (M.I.L.); (R.E.K.)
| | - Simona A. Miteva
- Department of Behavior Neurobiology, Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia 1113, Bulgaria; (S.T.D.); (S.A.M.)
| | - Emanuela Mazzon
- IRCCS Centro Neurolesi “Bonino-Pulejo”, Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy;
| | - Nikolay T. Tzvetkov
- Department of Biochemical Pharmacology and Drug Design, Institute of Molecular Biology, Bulgarian Academy of Sciences, Sofia 1113, Bulgaria;
| | - Atanas G. Atanasov
- Department of Synaptic Signaling and Communications, Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia 1113, Bulgaria; (M.I.L.); (R.E.K.)
- Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzebiec, 05-552 Magdalenka, Poland
- Department of Pharmacognosy, University of Vienna, 1090 Vienna, Austria
- Ludwig Boltzmann Institute for Digital Health and Patient Safety, Medical University of Vienna, Spitalgasse 23, 1090 Vienna, Austria
- Correspondence: (L.P.T.); (A.G.A.); Tel.: +359-2979-2175 (L.P.T.); +48-227-367-022 (A.G.A.)
| |
Collapse
|
43
|
Cui SY, Yang MX, Zhang YH, Zheng V, Zhang HT, Gurney ME, Xu Y, O'Donnell JM. Protection from Amyloid β Peptide-Induced Memory, Biochemical, and Morphological Deficits by a Phosphodiesterase-4D Allosteric Inhibitor. J Pharmacol Exp Ther 2019; 371:250-259. [PMID: 31488603 PMCID: PMC6815937 DOI: 10.1124/jpet.119.259986] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 08/26/2019] [Indexed: 11/22/2022] Open
Abstract
Recent imaging studies of amyloid and tau in cognitively normal elderly subjects imply that Alzheimer's pathology can be tolerated by the brain to some extent due to compensatory mechanisms operating at the cellular and synaptic levels. The present study investigated the effects of an allosteric inhibitor of phosphodiesterase-4D (PDE4D), known as BPN14770 (2-(4-((2-(3-Chlorophenyl)-6-(trifluoromethyl)pyridin-4-yl)methyl)phenyl)acetic Acid), on impairment of memory, dendritic structure, and synaptic proteins induced by bilateral microinjection of oligomeric amyloid beta (Aβ 1-42 into the hippocampus of humanized PDE4D (hPDE4D) mice. The hPDE4D mice provide a unique and powerful genetic tool for assessing PDE4D target engagement. Behavioral studies showed that treatment with BPN14770 significantly improved memory acquisition and retrieval in the Morris water maze test and the percentage of alternations in the Y-maze test in the model of Aβ impairment. Microinjection of oligomeric Aβ 1-42 caused decreases in the number of dendrites, dendritic length, and spine density of pyramid neurons in the hippocampus. These changes were prevented by BPN14770 in a dose-dependent manner. Furthermore, molecular studies showed that BPN14770 prevented Aβ-induced decreases in synaptophysin, postsynaptic density protein 95, phosphorylated cAMP-response element binding protein (CREB)/CREB, brain-derived neurotrophic factor, and nerve growth factor inducible protein levels in the hippocampus. The protective effects of BPN14770 against Aβ-induced memory deficits, synaptic damage, and the alteration in the cAMP-meditated cell signaling cascade were blocked by H-89 (N-[2-(p-Bromocinnamylamino)ethyl]-5-isoquinolinesulfonamide dihydrochloride), an inhibitor of protein kinase A. These results suggest that BPN14770 may activate compensatory mechanisms that support synaptic health even with the onset of amyloid pathology in Alzheimer's disease. SIGNIFICANCE STATEMENT: This study demonstrates that a phosphodiesterase-4D allosteric inhibitor, BPN14770, protects against memory loss and neuronal atrophy induced by oligomeric Aβ 1-42. The study provides useful insight into the potential role of compensatory mechanisms in Alzheimer's disease in a model of oligomeric Aβ 1-42 neurotoxicity.
Collapse
Affiliation(s)
- Su-Ying Cui
- Department of Pharmacology, School of Basic Medical Science, Peking University, Beijing, China (S.-Y.C., Y.-H.Z.); Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, State University of New York at Buffalo, Buffalo, New York (S.-Y.C., M.-X.Y., V.Z., Y.X., J.M.O.); Departments of Behavioral Medicine and Psychiatry, Physiology and Pharmacology, and Neuroscience, The Rockefeller Neurosciences Institute, West Virginia University Health Sciences Center, Morgantown, West Virginia (H.-T.Z.); and Tetra Discovery Partners Inc., Grand Rapids, Michigan (M.E.G.)
| | - Ming-Xin Yang
- Department of Pharmacology, School of Basic Medical Science, Peking University, Beijing, China (S.-Y.C., Y.-H.Z.); Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, State University of New York at Buffalo, Buffalo, New York (S.-Y.C., M.-X.Y., V.Z., Y.X., J.M.O.); Departments of Behavioral Medicine and Psychiatry, Physiology and Pharmacology, and Neuroscience, The Rockefeller Neurosciences Institute, West Virginia University Health Sciences Center, Morgantown, West Virginia (H.-T.Z.); and Tetra Discovery Partners Inc., Grand Rapids, Michigan (M.E.G.)
| | - Yong-He Zhang
- Department of Pharmacology, School of Basic Medical Science, Peking University, Beijing, China (S.-Y.C., Y.-H.Z.); Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, State University of New York at Buffalo, Buffalo, New York (S.-Y.C., M.-X.Y., V.Z., Y.X., J.M.O.); Departments of Behavioral Medicine and Psychiatry, Physiology and Pharmacology, and Neuroscience, The Rockefeller Neurosciences Institute, West Virginia University Health Sciences Center, Morgantown, West Virginia (H.-T.Z.); and Tetra Discovery Partners Inc., Grand Rapids, Michigan (M.E.G.)
| | - Victor Zheng
- Department of Pharmacology, School of Basic Medical Science, Peking University, Beijing, China (S.-Y.C., Y.-H.Z.); Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, State University of New York at Buffalo, Buffalo, New York (S.-Y.C., M.-X.Y., V.Z., Y.X., J.M.O.); Departments of Behavioral Medicine and Psychiatry, Physiology and Pharmacology, and Neuroscience, The Rockefeller Neurosciences Institute, West Virginia University Health Sciences Center, Morgantown, West Virginia (H.-T.Z.); and Tetra Discovery Partners Inc., Grand Rapids, Michigan (M.E.G.)
| | - Han-Ting Zhang
- Department of Pharmacology, School of Basic Medical Science, Peking University, Beijing, China (S.-Y.C., Y.-H.Z.); Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, State University of New York at Buffalo, Buffalo, New York (S.-Y.C., M.-X.Y., V.Z., Y.X., J.M.O.); Departments of Behavioral Medicine and Psychiatry, Physiology and Pharmacology, and Neuroscience, The Rockefeller Neurosciences Institute, West Virginia University Health Sciences Center, Morgantown, West Virginia (H.-T.Z.); and Tetra Discovery Partners Inc., Grand Rapids, Michigan (M.E.G.)
| | - Mark E Gurney
- Department of Pharmacology, School of Basic Medical Science, Peking University, Beijing, China (S.-Y.C., Y.-H.Z.); Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, State University of New York at Buffalo, Buffalo, New York (S.-Y.C., M.-X.Y., V.Z., Y.X., J.M.O.); Departments of Behavioral Medicine and Psychiatry, Physiology and Pharmacology, and Neuroscience, The Rockefeller Neurosciences Institute, West Virginia University Health Sciences Center, Morgantown, West Virginia (H.-T.Z.); and Tetra Discovery Partners Inc., Grand Rapids, Michigan (M.E.G.)
| | - Ying Xu
- Department of Pharmacology, School of Basic Medical Science, Peking University, Beijing, China (S.-Y.C., Y.-H.Z.); Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, State University of New York at Buffalo, Buffalo, New York (S.-Y.C., M.-X.Y., V.Z., Y.X., J.M.O.); Departments of Behavioral Medicine and Psychiatry, Physiology and Pharmacology, and Neuroscience, The Rockefeller Neurosciences Institute, West Virginia University Health Sciences Center, Morgantown, West Virginia (H.-T.Z.); and Tetra Discovery Partners Inc., Grand Rapids, Michigan (M.E.G.)
| | - James M O'Donnell
- Department of Pharmacology, School of Basic Medical Science, Peking University, Beijing, China (S.-Y.C., Y.-H.Z.); Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, State University of New York at Buffalo, Buffalo, New York (S.-Y.C., M.-X.Y., V.Z., Y.X., J.M.O.); Departments of Behavioral Medicine and Psychiatry, Physiology and Pharmacology, and Neuroscience, The Rockefeller Neurosciences Institute, West Virginia University Health Sciences Center, Morgantown, West Virginia (H.-T.Z.); and Tetra Discovery Partners Inc., Grand Rapids, Michigan (M.E.G.)
| |
Collapse
|
44
|
Ruan L, Du K, Tao M, Shan C, Ye R, Tang Y, Pan H, Lv J, Zhang M, Pan J. Phosphodiesterase-2 Inhibitor Bay 60-7550 Ameliorates Aβ-Induced Cognitive and Memory Impairment via Regulation of the HPA Axis. Front Cell Neurosci 2019; 13:432. [PMID: 31632240 PMCID: PMC6783519 DOI: 10.3389/fncel.2019.00432] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 09/09/2019] [Indexed: 01/01/2023] Open
Abstract
The dysfunction of the hypothalamus-pituitary-adrenal (HPA) axis is often seen in Alzheimer's disease (AD) patients with cognitive deficits. Selective inhibition of phosphodiesterase (PDE) 4 and 5 has already proven to be effective in reducing beta-amyloid 1-42 (Aβ1-42)-mediated pathology by regulating corticotropin-releasing factor (CRF) and glucocorticoid receptor (GR) expression, suggesting that PDE-dependent signaling is involved in Aβ1-42-induced HPA axis dysfunction. However, nausea and vomiting are the side effects of some PDE4 inhibitors, which turn our attention to other PDEs. PDE2 are highly expressed in the hippocampus and cortex, which associate with learning and memory, but not in the area postrema that would cause vomiting. The present study suggested that microinjection of Aβ1-42 to the intracerebroventricle induced learning and memory impairments and dysregulation of the HPA axis by increased expression of CRF and GR. However, the PDE2 inhibitor Bay 60-7550 significantly ameliorated the learning and memory impairment in the Morris water maze (MWM) and step-down passive avoidance tests. The Aβ1-42-induced increased CRF and GR levels were also reversed by the treatment with Bay 60-7550. These Bay 60-7550's effects were prevented by pretreatment with the PKG inhibitor KT5823. Moreover, the Bay 60-7550-induced downstream phosphorylation of cyclic AMP response element binding (pCREB) and brain-derived neurotrophic factor (BDNF) expression was also prevented (or partially prevented) by KT5823 or the PKA inhibitor H89. These results may lead to the discovery of novel strategies for the treatment of age-related cognitive disorders, such as AD, which affects approximately 44 million people worldwide.
Collapse
Affiliation(s)
- Lina Ruan
- Brain Institute, School of Pharmacy, Wenzhou Medical University, Wenzhou, China
| | - Kai Du
- Brain Institute, School of Pharmacy, Wenzhou Medical University, Wenzhou, China
| | - Mengjia Tao
- Brain Institute, School of Pharmacy, Wenzhou Medical University, Wenzhou, China
| | - Chunyan Shan
- Brain Institute, School of Pharmacy, Wenzhou Medical University, Wenzhou, China
| | - Ruixuan Ye
- Brain Institute, School of Pharmacy, Wenzhou Medical University, Wenzhou, China
| | - Yali Tang
- Brain Institute, School of Pharmacy, Wenzhou Medical University, Wenzhou, China
| | - Hanbo Pan
- Ningbo Key Laboratory of Behavioral Neuroscience, Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, China
| | - Jinpeng Lv
- College of Pharmaceutical Engineering and Life Sciences, Changzhou University, Changzhou, China
| | - Meixi Zhang
- Brain Institute, School of Pharmacy, Wenzhou Medical University, Wenzhou, China.,Pingyang County Hospital of Traditional Chinese Medicine, Pingyang County, China
| | - Jianchun Pan
- Brain Institute, School of Pharmacy, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
45
|
Nabavi SM, Talarek S, Listos J, Nabavi SF, Devi KP, Roberto de Oliveira M, Tewari D, Argüelles S, Mehrzadi S, Hosseinzadeh A, D'onofrio G, Orhan IE, Sureda A, Xu S, Momtaz S, Farzaei MH. Phosphodiesterase inhibitors say NO to Alzheimer's disease. Food Chem Toxicol 2019; 134:110822. [PMID: 31536753 DOI: 10.1016/j.fct.2019.110822] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Revised: 09/13/2019] [Accepted: 09/14/2019] [Indexed: 12/18/2022]
Abstract
Phosphodiesterases (PDEs) consisted of 11 subtypes (PDE1 to PDE11) and over 40 isoforms that regulate levels of cyclic guanosine monophosphate (cGMP) and cyclic adenosine monophosphate (cAMP), the second messengers in cell functions. PDE inhibitors (PDEIs) have been attractive therapeutic targets due to their involvement in diverse medical conditions, e.g. cardiovascular diseases, autoimmune diseases, Alzheimer's disease (AD), etc. Among them; AD with a complex pathology is a progressive neurodegenerative disorder which affect mostly senile people in the world and only symptomatic treatment particularly using cholinesterase inhibitors in clinic is available at the moment for AD. Consequently, novel treatment strategies towards AD are still searched extensively. Since PDEs are broadly expressed in the brain, PDEIs are considered to modulate neurodegenerative conditions through regulating cAMP and cGMP in the brain. In this sense, several synthetic or natural molecules inhibiting various PDE subtypes such as rolipram and roflumilast (PDE4 inhibitors), vinpocetine (PDE1 inhibitor), cilostazol and milrinone (PDE3 inhibitors), sildenafil and tadalafil (PDE5 inhibitors), etc have been reported showing encouraging results for the treatment of AD. In this review, PDE superfamily will be scrutinized from the view point of structural features, isoforms, functions and pharmacology particularly attributed to PDEs as target for AD therapy.
Collapse
Affiliation(s)
- Seyed Mohammad Nabavi
- Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Sylwia Talarek
- Department of Pharmacology and Pharmacodynamics, Medical University of Lublin, Chodźki 4a St, 20-093, Lublin, Poland.
| | - Joanna Listos
- Department of Pharmacology and Pharmacodynamics, Medical University of Lublin, Chodźki 4a St, 20-093, Lublin, Poland.
| | - Seyed Fazel Nabavi
- Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Kasi Pandima Devi
- Department of Biotechnology, Alagappa University, Karaikudi, 630003, Tamil Nadu, India.
| | - Marcos Roberto de Oliveira
- Departamento de Química (DQ), Instituto de Ciências Exatas e da Terra (ICET), Universidade Federal de Mato Grosso (UFMT), Cuiabá, Brazil.
| | - Devesh Tewari
- Department of Pharmacognosy, School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, 144411, Punjab, India.
| | - Sandro Argüelles
- Department of Physiology, Faculty of Pharmacy, University of Seville, Seville, Spain.
| | - Saeed Mehrzadi
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran.
| | - Azam Hosseinzadeh
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Grazia D'onofrio
- Geriatric Unit and Gerontology-Geriatrics Research Laboratory, Department of Medical Sciences, IRCCS "Casa Sollievo della Sofferenza", Viale Cappuccini 1, 71013, San Giovanni Rotondo, FG, Italy.
| | - Ilkay Erdogan Orhan
- Department of Pharmacognosy, Faculty of Pharmacy, Gazi University, 06330, Ankara, Turkey.
| | - Antoni Sureda
- Research Group on Community Nutrition and Oxidative Stress, University of Balearic Islands, CIBEROBN (Physiopathology of Obesity and Nutrition), E-07122, Palma de Mallorca, Balearic Islands, Spain.
| | - Suowen Xu
- Aab Cardiovascular Research Institute, University of Rochester, Rochester, NY, 14623, USA.
| | - Saeedeh Momtaz
- Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, Karaj, Iran; Toxicology and Diseases Group, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Hosein Farzaei
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
46
|
Alzoubi KH, Al Subeh ZY, Khabour OF. Molecular targets for the interactive effect of etazolate during post-traumatic stress disorder: Role of oxidative stress, BDNF and histones. Behav Brain Res 2019; 369:111930. [PMID: 31047921 DOI: 10.1016/j.bbr.2019.111930] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Revised: 04/19/2019] [Accepted: 04/28/2019] [Indexed: 12/19/2022]
Abstract
Post-traumatic stress disorder (PTSD) develops in individuals after exposure to severe, life-threatening traumatic event. Etazolate is a selective phosphodiesterase IV inhibitor that is highly specific for cAMP, which has anxiolytic and antidepressant effects. We have previously shown that PTSD induced-memory impairment, anxiety and depression were prevented via the administration of etazolate. In the current study, the effect of etazolate on oxidative stress parameters, BDNF, and histone acetylation in the hippocampus were evaluated in a rat model of PTSD. The PTSD was induced by single prolonged stress (SPS) model. Etazolate was administered orally at a dose of 1 mg/kg/day for one month. At the end of the treatment period, the hippocampus was dissected and oxidative stress biomarkers (GSH, GSSG, GPx and TBARS), BDNF protein level, and histone acetylation were assessed. Results revealed that PTSD potentiated oxidative stress in the hippocampus and induced significant reductions in BDNF level and histones acetylation (P < 0.05). Etazolate treatment, on the other hand, led to prevention of changes in these oxidative stress biomarkers (GSH, GSSG, GPx and TBARS), BDNF levels, and histones acetylation. In conclusion, oxidative stress and modulation of BDNF and histones acetylation induced by PTSD can be prevented by treatment with etazolate.
Collapse
Affiliation(s)
- Karem H Alzoubi
- Department of Clinical Pharmacy, Jordan University of Science and Technology, Irbid, Jordan.
| | - Zeinab Y Al Subeh
- Department of Clinical Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| | - Omar F Khabour
- Department of Medical Laboratory Sciences, Jordan University of Science and Technology, Irbid, Jordan
| |
Collapse
|
47
|
Feng H, Wang C, He W, Wu X, Li S, Zeng Z, Wei M, He B. Roflumilast ameliorates cognitive impairment in APP/PS1 mice via cAMP/CREB/BDNF signaling and anti-neuroinflammatory effects. Metab Brain Dis 2019; 34:583-591. [PMID: 30610438 DOI: 10.1007/s11011-018-0374-4] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 12/19/2018] [Indexed: 12/20/2022]
Abstract
Phosphodiesterase type 4 (PDE4) inhibitors can prevent the breakdown of the second messenger cyclic adenosine monophosphate (cAMP) and improve cognitive performances in several animal models of cognition. However, the clinical development of PDE4 inhibitors has been seriously hampered by severe side effects, such as vomiting and nausea. In this study, we investigated the effect and mechanism of roflumilast, an FDA-approved PDE4 inhibitor for treatment of chronic obstructive pulmonary disease (COPD), on learning and memory abilities in the APP/PS1 mouse model of Alzheimer's disease (AD). APP/PS1 transgenic mice received 3 intragastric doses of roflumilast (0.1, 0.2 and 0.4 mg/kg) daily for 3 weeks followed by behavioral tests. Chronic administration of roflumilast significantly improved the learning and memory abilities of APP/PS1 transgenic mice in the novel object recognition task, Morris water maze, and the step-down passive avoidance task. In addition, roflumilast increased the cAMP, phosphorylated cAMP response-element binding protein (p-CREB) and brain-derived neurotrophic factor (BDNF) levels, and reduced the nuclear translocation of nuclear factor-kappa B (NF-κB) p65, and proinflammatory cytokine (IL-6, TNF-a and IL-1β) levels in the hippocampus of APP/PS1 transgenic mice. In conclusion, these findings suggest that roflumilast can enhance cognitive function in APP/PS1 transgenic mice, which may be related to its stimulation of the cAMP/CREB/BDNF pathway and anti-neuroinflammatory effects.
Collapse
Affiliation(s)
- Huancun Feng
- Department of Pharmacy, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510000, Guangdong, China
| | - Canmao Wang
- Department of Pharmacy, Shenzhen University General Hospital, Shenzhen, 518000, Guangdong, China
| | - Wei He
- Department of Pharmacy, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510000, Guangdong, China
| | - Xinjun Wu
- Department of Biology, East Carolina University, Greenville, NC, 27858, USA
| | - Shujie Li
- Department of Pharmacy, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510000, Guangdong, China
| | - Zhenkun Zeng
- Department of Pharmacy, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510000, Guangdong, China
| | - Meidan Wei
- Department of Pharmacy, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510000, Guangdong, China
| | - Binghong He
- Department of Pharmacy, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510000, Guangdong, China.
| |
Collapse
|
48
|
Watremez W, Jackson J, Almari B, McLean SL, Grayson B, Neill JC, Fischer N, Allouche A, Koziel V, Pillot T, Harte MK. Stabilized Low-n Amyloid-β Oligomers Induce Robust Novel Object Recognition Deficits Associated with Inflammatory, Synaptic, and GABAergic Dysfunction in the Rat. J Alzheimers Dis 2019; 62:213-226. [PMID: 29439327 DOI: 10.3233/jad-170489] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND With current treatments for Alzheimer's disease (AD) only providing temporary symptomatic benefits, disease modifying drugs are urgently required. This approach relies on improved understanding of the early pathophysiology of AD. A new hypothesis has emerged, in which early memory loss is considered a synapse failure caused by soluble amyloid-β oligomers (Aβo). These small soluble Aβo, which precede the formation of larger fibrillar assemblies, may be the main cause of early AD pathologies. OBJECTIVE The aim of the current study was to investigate the effect of acute administration of stabilized low-n amyloid-β1-42 oligomers (Aβo1-42) on cognitive, inflammatory, synaptic, and neuronal markers in the rat. METHODS Female and male Lister Hooded rats received acute intracerebroventricular (ICV) administration of either vehicle or 5 nmol of Aβo1-42 (10μL). Cognition was assessed in the novel object recognition (NOR) paradigm at different time points. Levels of inflammatory (IL-1β, IL-6, TNF-α), synaptic (PSD-95, SNAP-25), and neuronal (n-acetylaspartate, parvalbumin-positive cells) markers were investigated in different brain regions (prefrontal and frontal cortex, striatum, dorsal and ventral hippocampus). RESULTS Acute ICV administration of Aβo1-42 induced robust and enduring NOR deficits. These deficits were reversed by acute administration of donepezil and rolipram but not risperidone. Postmortem analysis revealed an increase in inflammatory markers, a decrease in synaptic markers and parvalbumin containing interneurons in the frontal cortex, with no evidence of widespread neuronal loss. CONCLUSION Taken together the results suggest that acute administration of soluble low-n Aβo may be a useful model to study the early mechanisms involved in AD and provide us with a platform for testing novel therapeutic approaches that target the early underlying synaptic pathology.
Collapse
Affiliation(s)
- William Watremez
- Division of Pharmacy and Optometry, University of Manchester, Manchester, UK
| | - Joshua Jackson
- Division of Pharmacy and Optometry, University of Manchester, Manchester, UK
| | - Bushra Almari
- Division of Pharmacy and Optometry, University of Manchester, Manchester, UK
| | - Samantha L McLean
- School of Pharmacy and Medical Sciences, University of Bradford, Bradford, UK
| | - Ben Grayson
- Division of Pharmacy and Optometry, University of Manchester, Manchester, UK
| | - Joanna C Neill
- Division of Pharmacy and Optometry, University of Manchester, Manchester, UK
| | - Nicolas Fischer
- SynAging, Institut Polytechnique National de Lorraine, Vandoeuvre-lès-Nancy, France
| | - Ahmad Allouche
- SynAging, Institut Polytechnique National de Lorraine, Vandoeuvre-lès-Nancy, France
| | - Violette Koziel
- SynAging, Institut Polytechnique National de Lorraine, Vandoeuvre-lès-Nancy, France
| | - Thierry Pillot
- SynAging, Institut Polytechnique National de Lorraine, Vandoeuvre-lès-Nancy, France
| | - Michael K Harte
- Division of Pharmacy and Optometry, University of Manchester, Manchester, UK
| |
Collapse
|
49
|
FCPR16, a novel phosphodiesterase 4 inhibitor, produces an antidepressant-like effect in mice exposed to chronic unpredictable mild stress. Prog Neuropsychopharmacol Biol Psychiatry 2019; 90:62-75. [PMID: 30391306 DOI: 10.1016/j.pnpbp.2018.10.017] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 09/28/2018] [Accepted: 10/30/2018] [Indexed: 12/29/2022]
Abstract
The canonical phosphodiesterase 4 (PDE4) inhibitors produce antidepressant-like effects in a variety of animal models. However, severe side effects, particularly vomiting and nausea, limit their clinical application. FCPR16 is a novel PDE4 inhibitor with less vomiting potential. However, whether it will exert an antidepressant-like effect remains unclear. Here, we aimed to evaluate the effect of FCPR16 in mice subjected to chronic unpredictable mild stress (CUMS). Our results showed that FCPR16 produced antidepressant-like effects in multiple behavioral tests, including a forced swimming test, tail suspension test, sucrose preference test and novelty suppression feeding test. Simultaneously, data indicated that FCPR16 enhanced the levels of several proteins, including cAMP, brain derived neurotrophic factor, exchange protein directly activated by cAMP 2 (EPAC-2), synapsin1, postsynaptic density protein 95, phosphorylated cAMP response element binding protein and extracellular regulated protein kinases 1/2, which were downregulated by CUMS in both the cerebral cortex and hippocampus. The number of DCX+ cells in the hippocampus of CUMS mice was increased after FCPR16 treatment. Moreover, treatment with FCPR16 resulted in decreased expression of pro-inflammatory cytokines (TNF-α, IL-6, and IL-1β) and increased expression of anti-inflammatory cytokines (IL-10) in mice challenged with CUMS. Consistently, the mRNA levels of microglial M1 markers (iNOS and TNF-α) were downregulated, while M2 markers (Arginase 1 and CD206) were upregulated in CUMS-exposed mice after FCPR16 treatment. Immunofluorescence analysis showed that FCPR16 inhibited the activation of microglial cells and increased the number of CD206+ in CUMS-exposed mice. Collectively, these results suggested that FCPR16 is a potential compound with effects against depressive-like behaviors, and the antidepressant-like effect of FCPR16 is possibly mediated through activation of the cAMP-mediated signaling pathways and inhibition of neuroinflammation in both the cerebral cortex and hippocampus.
Collapse
|
50
|
Structure of amyloid β 25-35 in lipid environment and cholesterol-dependent membrane pore formation. Sci Rep 2019; 9:2689. [PMID: 30804528 PMCID: PMC6389947 DOI: 10.1038/s41598-019-38749-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 01/08/2019] [Indexed: 01/14/2023] Open
Abstract
The amyloid β (Aβ) peptide and its shorter variants, including a highly cytotoxic Aβ25–35 peptide, exert their neurotoxic effect during Alzheimer’s disease by various mechanisms, including cellular membrane permeabilization. The intrinsic polymorphism of Aβ has prevented the identification of the molecular basis of Aβ pore formation by direct structural methods, and computational studies have led to highly divergent pore models. Here, we have employed a set of biophysical techniques to directly monitor Ca2+-transporting Aβ25–35 pores in lipid membranes, to quantitatively characterize pore formation, and to identify the key structural features of the pore. Moreover, the effect of membrane cholesterol on pore formation and the structure of Aβ25–35 has been elucidated. The data suggest that the membrane-embedded peptide forms 6- or 8-stranded β-barrel like structures. The 8-stranded barrels may conduct Ca2+ ions through an inner cavity, whereas the tightly packed 6-stranded barrels need to assemble into supramolecular structures to form a central pore. Cholesterol affects Aβ25–35 pore formation by a dual mechanism, i.e., by direct interaction with the peptide and by affecting membrane structure. Collectively, our data illuminate the molecular basis of Aβ membrane pore formation, which should advance both basic and clinical research on Alzheimer’s disease and membrane-associated pathologies in general.
Collapse
|