1
|
Maded ZK, Sfar S, Taqa GAA, Lassoued MA, Ben Hadj Ayed O, Fawzi HA. Development and Optimization of Dipyridamole- and Roflumilast-Loaded Nanoemulsion and Nanoemulgel for Enhanced Skin Permeation: Formulation, Characterization, and In Vitro Assessment. Pharmaceuticals (Basel) 2024; 17:803. [PMID: 38931470 PMCID: PMC11207013 DOI: 10.3390/ph17060803] [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: 05/20/2024] [Revised: 06/14/2024] [Accepted: 06/17/2024] [Indexed: 06/28/2024] Open
Abstract
This study explores developing and optimizing a nanoemulsion (NE) system loaded with dipyridamole and roflumilast, aiming to improve skin penetration and retention. The NE formulation was further transformed into a nanoemulgel to enhance its application as a topical treatment for psoriasis. Solubility studies were conducted to select the oil, surfactant, and co-surfactant. Phase diagrams were constructed using the aqueous phase titration method. All the formulations were in nanoscale, and Formula (F2) (which contains oleic acid oil as the oil phase, a mixture of Surfactant Tween 80 and co-surfactant (ethanol) at a ratio of 1:2 in addition to distilled water as an aqueous phase in a ratio of 1:5:4, respectively) was the selected formula depending on the particle size, PDI, and zeta potential. Formula (F2) has the best ratio because it gives the smallest nanoemulsion globule size (particle size average of 167.1 nm), the best homogenicity (lowest PDI of 0.195), and the highest stability (higher zeta potential of -32.22). The selected formula was converted into a nanoemulgel by the addition of 0.5% (w/w) xanthan gum (average particle size of 172.7 nm) and the best homogenicity (lowest PDI of 0.121%) and highest stability (higher zeta potential of -28.31). In conclusion, the selected formula has accepted physical and chemical properties, which enhanced skin penetration.
Collapse
Affiliation(s)
- Zeyad Khalaf Maded
- Laboratory of Pharmaceutical, Chemical, and Pharmacological Drug Development LR12ES09, Faculty of Pharmacy, University of Monastir, Monastir 5000, Tunisia; (Z.K.M.); (M.A.L.); (O.B.H.A.)
| | - Souad Sfar
- Laboratory of Chemical, Galenic and Pharmacological Development of Medicines (LR12ES09), Faculty of Pharmacy of Monastir, University of Monastir, Monastir 5000, Tunisia;
| | - Ghada Abd Alrhman Taqa
- Department of Dental Basic Sciences, College of Dentistry, University of Mosul, Mosul 41002, Iraq;
| | - Mohamed Ali Lassoued
- Laboratory of Pharmaceutical, Chemical, and Pharmacological Drug Development LR12ES09, Faculty of Pharmacy, University of Monastir, Monastir 5000, Tunisia; (Z.K.M.); (M.A.L.); (O.B.H.A.)
| | - Olfa Ben Hadj Ayed
- Laboratory of Pharmaceutical, Chemical, and Pharmacological Drug Development LR12ES09, Faculty of Pharmacy, University of Monastir, Monastir 5000, Tunisia; (Z.K.M.); (M.A.L.); (O.B.H.A.)
| | - Hayder Adnan Fawzi
- Department of Pharmacy, Al Mustafa University College, Baghdad 10064, Iraq
| |
Collapse
|
2
|
Hencz A, Magony A, Thomas C, Kovacs K, Szilagyi G, Pal J, Sik A. Mild hypoxia-induced structural and functional changes of the hippocampal network. Front Cell Neurosci 2023; 17:1277375. [PMID: 37841285 PMCID: PMC10576450 DOI: 10.3389/fncel.2023.1277375] [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: 08/14/2023] [Accepted: 09/15/2023] [Indexed: 10/17/2023] Open
Abstract
Hypoxia causes structural and functional changes in several brain regions, including the oxygen-concentration-sensitive hippocampus. We investigated the consequences of mild short-term hypoxia on rat hippocampus in vivo. The hypoxic group was treated with 16% O2 for 1 h, and the control group with 21% O2. Using a combination of Gallyas silver impregnation histochemistry revealing damaged neurons and interneuron-specific immunohistochemistry, we found that somatostatin-expressing inhibitory neurons in the hilus were injured. We used 32-channel silicon probe arrays to record network oscillations and unit activity from the hippocampal layers under anaesthesia. There were no changes in the frequency power of slow, theta, beta, or gamma bands, but we found a significant increase in the frequency of slow oscillation (2.1-2.2 Hz) at 16% O2 compared to 21% O2. In the hilus region, the firing frequency of unidentified interneurons decreased. In the CA3 region, the firing frequency of some unidentified interneurons decreased while the activity of other interneurons increased. The activity of pyramidal cells increased both in the CA1 and CA3 regions. In addition, the regularity of CA1, CA3 pyramidal cells' and CA3 type II and hilar interneuron activity has significantly changed in hypoxic conditions. In summary, a low O2 environment caused profound changes in the state of hippocampal excitatory and inhibitory neurons and network activity, indicating potential changes in information processing caused by mild short-term hypoxia.
Collapse
Affiliation(s)
- Alexandra Hencz
- Institute of Physiology, Medical School, University of Pecs, Pecs, Hungary
| | - Andor Magony
- Institute of Physiology, Medical School, University of Pecs, Pecs, Hungary
- Institute of Transdisciplinary Discoveries, Medical School, University of Pecs, Pecs, Hungary
| | - Chloe Thomas
- Institute of Clinical Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Krisztina Kovacs
- Institute of Clinical Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Gabor Szilagyi
- Institute of Biochemistry and Medical Chemistry, Medical School, University of Pecs, Pecs, Hungary
| | - Jozsef Pal
- Institute of Physiology, Medical School, University of Pecs, Pecs, Hungary
| | - Attila Sik
- Institute of Physiology, Medical School, University of Pecs, Pecs, Hungary
- Institute of Transdisciplinary Discoveries, Medical School, University of Pecs, Pecs, Hungary
- Institute of Clinical Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
3
|
Cytotoxic and genotoxic evaluation of dipyridamole and its alternative therapeutic potential in cancer therapy: an in vitro and in vivo approach. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2021.131626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
4
|
Wang J, Yue B, Zhang X, Guo X, Sun Z, Niu R. Effect of exercise on microglial activation and transcriptome of hippocampus in fluorosis mice. THE SCIENCE OF THE TOTAL ENVIRONMENT 2021; 760:143376. [PMID: 33172640 DOI: 10.1016/j.scitotenv.2020.143376] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 10/02/2020] [Accepted: 10/21/2020] [Indexed: 06/11/2023]
Abstract
Fluorosis is a widespread endemic disease. Reports have shown that high fluoride causes the dysfunction of central nervous system (CNS) in animals. The neurotoxicity of fluoride may be related to the activation of microglia. Moreover, numerous studies have found that exercise facilitates the plasticity of structure and function in CNS, partly owing to the regulation of microglia activation. The present study was conducted to explore the effect of exercise on the microglial activation of hippocampus in fluorosis mice. One hundred adult female Institute of Cancer Research (ICR) mice were randomly divided into 4 groups: control group (group C, distilled water by gavage); exercise group (group E, distilled water by gavage and treadmill exercise); fluoride group [group F, 24 mg/kg sodium fluoride (NaF) by gavage]; fluoride plus exercise group (group F + E, 24 mg/kg NaF by gavage and treadmill exercise). After 8 weeks, hippocampal morphological structure, microglial activation and RNA transcriptome of mice in each group were evaluated by hematoxylin and eosin (HE) staining, Nissl staining, immunohistochemistry (IHC), quantitative real time PCR (QRT-PCR) and transcriptome sequencing. We discovered that the number of M1-type microglia in fluorosis-mice hippocampus was significantly increased when compared to group C; group F + E showed a decrease in the number of M1-type microglia with the comparison to group F. In addition, the hippocampal transcriptome analysis showed that 576 differential expression genes (DEG) were confirmed in group F, compared to group C, and 670 DEG were differently expressed in group F + E when compared to group F. Gene Ontology (GO) analysis showed that changed genes were implicated in regulation of transcription, DNA-templated, integral component of membrane and adenosine triphosphate (ATP) binding. Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis of 670 DEG was helpful to find neuroactive ligand-receptor interaction pathway. In conclusion, these results indicate that treadmill running inhibits the excessive activation of microglia in hippocampus of the fluoride-toxic mice, accompanied with the alteration of neuroactive ligand-receptor interaction pathway.
Collapse
Affiliation(s)
- Jixiang Wang
- Shanxi Key Laboratory of Ecological Animal Science and Environmental Veterinary Medicine, College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, China
| | - Baijuan Yue
- Shanxi Key Laboratory of Ecological Animal Science and Environmental Veterinary Medicine, College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, China
| | - Xuhua Zhang
- Shanxi Key Laboratory of Ecological Animal Science and Environmental Veterinary Medicine, College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, China
| | - Xin Guo
- Shanxi Key Laboratory of Ecological Animal Science and Environmental Veterinary Medicine, College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, China
| | - Zilong Sun
- Shanxi Key Laboratory of Ecological Animal Science and Environmental Veterinary Medicine, College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, China
| | - Ruiyan Niu
- Shanxi Key Laboratory of Ecological Animal Science and Environmental Veterinary Medicine, College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, China.
| |
Collapse
|
5
|
Aliter KF, Al-Horani RA. Potential Therapeutic Benefits of Dipyridamole in COVID-19 Patients. Curr Pharm Des 2021; 27:866-875. [PMID: 33001004 PMCID: PMC7990686 DOI: 10.2174/1381612826666201001125604] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 08/31/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND COVID-19 pandemic is caused by coronavirus also known as severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). The viral infection continues to impact the globe with no vaccine to prevent the infection or highly effective therapeutics to treat the millions of infected people around the world. The disease starts as a respiratory infection, yet it may also be associated with a hypercoagulable state, severe inflammation owing to excessive cytokines production, and a potentially significant oxidative stress. The disease may progress to multiorgan failure and eventually death. OBJECTIVE In this article, we summarize the potential of dipyridamole as an adjunct therapy for COVID-19. METHODS We reviewed the literature describing the biological activities of dipyridamole in various settings of testing. Data were retrieved from PubMed, SciFinder-CAS, and Web of Science. The review concisely covered relevant studies starting from 1977. RESULTS Dipyridamole is an approved antiplatelet drug, that has been used to prevent stroke, among other indications. Besides its antithrombotic activity, the literature indicates that dipyridamole also promotes a host of other biological activities including antiviral, anti-inflammatory, and antioxidant ones. CONCLUSION Dipyridamole may substantially help improve the clinical outcomes of COVID-19 treatment. The pharmacokinetics profile of the drug is well established which makes it easier to design an appropriate therapeutic course. The drug is also generally safe, affordable, and available worldwide. Initial clinical trials have shown a substantial promise for dipyridamole in treating critically ill COVID-19 patients, yet larger randomized and controlled trials are needed to confirm this promise.
Collapse
Affiliation(s)
- Kholoud F. Aliter
- Department of Chemistry, School of STEM, Dillard University, New Orleans LA70122, USA
| | - Rami A. Al-Horani
- Division of Basic Pharmaceutical Sciences, College of Pharmacy, Xavier University of Louisiana, New Orleans LA70125, USA
| |
Collapse
|
6
|
Lana D, Ugolini F, Giovannini MG. An Overview on the Differential Interplay Among Neurons-Astrocytes-Microglia in CA1 and CA3 Hippocampus in Hypoxia/Ischemia. Front Cell Neurosci 2020; 14:585833. [PMID: 33262692 PMCID: PMC7686560 DOI: 10.3389/fncel.2020.585833] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 10/09/2020] [Indexed: 12/13/2022] Open
Abstract
Neurons have been long regarded as the basic functional cells of the brain, whereas astrocytes and microglia have been regarded only as elements of support. However, proper intercommunication among neurons-astrocytes-microglia is of fundamental importance for the functional organization of the brain. Perturbation in the regulation of brain energy metabolism not only in neurons but also in astrocytes and microglia may be one of the pathophysiological mechanisms of neurodegeneration, especially in hypoxia/ischemia. Glial activation has long been considered detrimental for survival of neurons, but recently it appears that glial responses to an insult are not equal but vary in different brain areas. In this review, we first take into consideration the modifications of the vascular unit of the glymphatic system and glial metabolism in hypoxic conditions. Using the method of triple-labeling fluorescent immunohistochemistry coupled with confocal microscopy (TIC), we recently studied the interplay among neurons, astrocytes, and microglia in chronic brain hypoperfusion. We evaluated the quantitative and morpho-functional alterations of the neuron-astrocyte-microglia triads comparing the hippocampal CA1 area, more vulnerable to ischemia, to the CA3 area, less vulnerable. In these contiguous and interconnected areas, in the same experimental hypoxic conditions, astrocytes and microglia show differential, finely regulated, region-specific reactivities. In both areas, astrocytes and microglia form triad clusters with apoptotic, degenerating neurons. In the neuron-astrocyte-microglia triads, the cell body of a damaged neuron is infiltrated and bisected by branches of astrocyte that create a microscar around it while a microglial cell phagocytoses the damaged neuron. These coordinated actions are consistent with the scavenging and protective activities of microglia. In hypoxia, the neuron-astrocyte-microglia triads are more numerous in CA3 than in CA1, further indicating their protective effects. These data, taken from contiguous and interconnected hippocampal areas, demonstrate that glial response to the same hypoxic insult is not equal but varies significantly. Understanding the differences of glial reactivity is of great interest to explain the differential susceptibility of hippocampal areas to hypoxia/ischemia. Further studies may evidence the differential reactivity of glia in different brain areas, explaining the higher or lower sensitivity of these areas to different insults and whether glia may represent a target for future therapeutic interventions.
Collapse
Affiliation(s)
- Daniele Lana
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| | - Filippo Ugolini
- Department of Health Sciences, Section of Anatomopathology, University of Florence, Florence, Italy
| | - Maria G Giovannini
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| |
Collapse
|
7
|
Remote ischemic postconditioning attenuates damage in rats with chronic cerebral ischemia by upregulating the autophagolysosome pathway via the activation of TFEB. Exp Mol Pathol 2020; 115:104475. [PMID: 32473154 DOI: 10.1016/j.yexmp.2020.104475] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 05/07/2020] [Accepted: 05/26/2020] [Indexed: 11/23/2022]
Abstract
The transcription factor EB (TFEB) is known for its role in lysosomal biogenesis, and it coordinates this process by driving autophagy and lysosomal gene expression during ischemia. In the present study, we aimed to explore the role of the TFEB-regulated autophagolysosome pathway (ALP) in rats with chronic cerebral ischemia (CCI) that were treated with remote ischemic postconditioning (RIPC). A modified 2-vessel occlusion (2-VO) method was utilized to establish the CCI rat model, and the CCI rats were identified by the Morris water maze test and histological staining. After the CCI rats were treated with RIPC, the damage to the rat cortex and hippocampal tissues and the status of the ALP were determined. Western blot analysis and immunofluorescence assays were performed to observe the nuclear translocation of TFEB. The rats were injected with TFEB siRNA via the lateral ventricle to investigate the effect of TFEB siRNA on the RIPC-treated CCI rats. The results suggested that RIPC of the CCI rats alleviated nerve injury, induced TFEB translocation into the nucleus, upregulated autophagy-related protein expression, and activated ALP machinery. Furthermore, TFEB siRNA decreased the levels of TFEB and impaired the neuroprotective effects of RIPC on the CCI rats. Collectively, we highlighted that RIPC attenuates damage in CCI rats via the activation of the TFEB-mediated ALP.
Collapse
|
8
|
Abstract
Ischemic necrosis has been most dreaded complication of flap reconstruction. Therefore, researchers have tried to improvise modalities to prevent or treat it since the onset of flap surgery. So far these researches have failed to identify a pharmacological therapy equally effective as surgical delay in augmenting skin flap viability. In the path of search for this substance, dipyridamole attracted our attention as an antiaggregant agent. Put together with pathophysiological mechanisms underlying ischemic flap necrosis, we concluded dipyridamole might have beneficial effect on survival of skin flaps. In this research random pattern dorsal rat skin flap model of McFarlane is used. Subjects are separated in a randomized fashion between two groups. Experiment group is given dipyridamole with a dose of 20 mg/kg twice daily. Control group is given same amount of saline. At seventh day viability of skin flaps is assessed and compared between groups. Also on 7th day, pathologic specimens are obtained and evaluated histopathologically in terms of neutrophil and lymphocyte infiltration, edema and fibrosis. Necrosis percentage in experiment group is found to be significantly lower than that of control group (p < 0.01*). Neutrophil infiltration and edema found to be significantly lower in dipyridamole group (p < 0.05*). No significant difference is observed in lymphocyte infiltration and fibrosis. Dipyridamole is shown in this research to be effective in augmenting viability of random pattern skin flaps in rats. Nevertheless, more extensive researches are needed to fully determine its precise mechanism, side effects and appropriate doses.
Collapse
Affiliation(s)
- Alper Burak Uslu
- Department of Plastic, Reconstructive and Aesthetic Surgery, Ankara Numune Training and Research Hospital, Ankara, Turkey
| |
Collapse
|
9
|
Mustapha M, Nassir CMNCM, Aminuddin N, Safri AA, Ghazali MM. Cerebral Small Vessel Disease (CSVD) - Lessons From the Animal Models. Front Physiol 2019; 10:1317. [PMID: 31708793 PMCID: PMC6822570 DOI: 10.3389/fphys.2019.01317] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Accepted: 09/30/2019] [Indexed: 12/28/2022] Open
Abstract
Cerebral small vessel disease (CSVD) refers to a spectrum of clinical and imaging findings resulting from pathological processes of various etiologies affecting cerebral arterioles, perforating arteries, capillaries, and venules. Unlike large vessels, it is a challenge to visualize small vessels in vivo, hence the difficulty to directly monitor the natural progression of the disease. CSVD might progress for many years during the early stage of the disease as it remains asymptomatic. Prevalent among elderly individuals, CSVD has been alarmingly reported as an important precursor of full-blown stroke and vascular dementia. Growing evidence has also shown a significant association between CSVD's radiological manifestation with dementia and Alzheimer's disease (AD) pathology. Although it remains contentious as to whether CSVD is a cause or sequelae of AD, it is not far-fetched to posit that effective therapeutic measures of CSVD would mitigate the overall burden of dementia. Nevertheless, the unifying theory on the pathomechanism of the disease remains elusive, hence the lack of effective therapeutic approaches. Thus, this chapter consolidates the contemporary insights from numerous experimental animal models of CSVD, to date: from the available experimental animal models of CSVD and its translational research value; the pathomechanical aspects of the disease; relevant aspects on systems biology; opportunities for early disease biomarkers; and finally, converging approaches for future therapeutic directions of CSVD.
Collapse
Affiliation(s)
- Muzaimi Mustapha
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| | | | - Niferiti Aminuddin
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
- Department of Basic Medical Sciences, Kulliyyah of Pharmacy, International Islamic University Malaysia, Kuantan, Malaysia
| | - Amanina Ahmad Safri
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| | - Mazira Mohamad Ghazali
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| |
Collapse
|
10
|
Afrin S, Rahman Y, Alhaji Isa M, Ahmed S, Tabish M. Biophysical insights into the binding characteristics of bovine serum albumin with dipyridamole and the influence of molecular interaction with β cyclodextrin. J Biomol Struct Dyn 2019; 38:3046-3058. [DOI: 10.1080/07391102.2019.1651220] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Shumaila Afrin
- Department of Biochemistry, Faculty of Life Sciences, A.M. University, Aligarh, U.P., India
| | - Yusra Rahman
- Department of Biochemistry, Faculty of Life Sciences, A.M. University, Aligarh, U.P., India
| | - Mustafa Alhaji Isa
- Department of Microbiology, Faculty of Sciences, University of Maiduguri, Maiduguri, P.M.B., Nigeria
| | - Shahbaz Ahmed
- Department of Biochemistry, Faculty of Life Sciences, A.M. University, Aligarh, U.P., India
| | - Mohammad Tabish
- Department of Biochemistry, Faculty of Life Sciences, A.M. University, Aligarh, U.P., India
| |
Collapse
|
11
|
Amelina IP, Solovieva EY. [Oxidative stress and inflammation as links in a chain in patients with chronic cerebrovascular diseases]. Zh Nevrol Psikhiatr Im S S Korsakova 2019; 119:106-114. [PMID: 31156231 DOI: 10.17116/jnevro2019119041106] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Cerebrovascular diseases (CVD) are the main cause of death and permanent disability. The urgency of the problem of chronic CVD is associated with an increase of the absolute number of elderly and senile age in the population, a trend towards slowly increasing, sluggish pathological processes. It is obvious that any somatic disease in such patients is comorbid to cerebrovascular diseases that suggests a unified mechanism of the pathogenesis for both the main and concomitant diseases. The article notes that microangiopathy is the most common cause of CVD. The main etiopathogenetic factor affecting cerebral vessels of small caliber is endothelial dysfunction, systemic inflammation and oxidative stress. Understanding the molecular components that underlie functional abnormalities and damage of small blood vessels gives the key to the modern strategies in therapy, forming the foundation for an adequate pathogenetically justified therapy. This impact should be gradual, complex and aimed at correcting pathochemical disorders in general and neurotransmitter imbalance in particular. The drug dipyridamole, which has pleiotropic effects, can be considered as one of the pathogenetically justified means in complex drug therapy in patients with CVD.
Collapse
Affiliation(s)
- I P Amelina
- N.I. Pirogov Russian National Research Medical University of the Ministry of Health of the Russian Federation, Russian Federation, Moscow
| | - E Yu Solovieva
- N.I. Pirogov Russian National Research Medical University of the Ministry of Health of the Russian Federation, Russian Federation, Moscow
| |
Collapse
|
12
|
Dipyridamole plus Triflusal versus Triflusal Alone in Infarct Reduction after Middle Cerebral Artery Occlusion. J Stroke Cerebrovasc Dis 2018; 27:1283-1287. [DOI: 10.1016/j.jstrokecerebrovasdis.2017.12.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 11/16/2017] [Accepted: 12/11/2017] [Indexed: 01/26/2023] Open
|
13
|
Chen C, Zheng Y, Wu T, Wu C, Cheng X. Oral administration of grape seed polyphenol extract restores memory deficits in chronic cerebral hypoperfusion rats. Behav Pharmacol 2018; 28:207-213. [PMID: 27984208 DOI: 10.1097/fbp.0000000000000276] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Chronic cerebral hypoperfusion (CCH) has been recognized as an important cause of both vascular dementia and Alzheimer's disease (AD), the two most prominent neurodegenerative diseases causing memory impairment in the elderly. However, an effective therapy for CCH-induced memory impairment has not yet been established. Grape seed polyphenol extract (GSPE) has powerful antioxidant properties and protects neurons and glia during ischemic injury, but its potential use in the prevention of CCH-induced memory impairment has not yet been investigated. Here, CCH-related memory impairment was modeled in rats using permanent bilateral occlusion of the common carotid artery. A Morris water maze task was used to evaluate memory, the levels of acetylcholinesterase, choline acetyltransferase, acetylcholine were used to evaluate cholinergic function, and oxidative stress was assessed by measuring the enzyme activity of superoxide dismutase, glutathione peroxidase, malonic dialdehyde, and catalase. We found that oral administration of GSPE for 1 month can rescue memory deficits. We also found that GSPE restores cholinergic neuronal function and represses oxidative damage in the hippocampus of CCH rats. We propose that GSPE protects memory in CCH rats by reducing ischemia-induced oxidative stress and cholinergic dysfunction. These findings provide a novel application of GSPE in CCH-related memory impairments.
Collapse
Affiliation(s)
- Chen Chen
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou City, Henan Province, China
| | | | | | | | | |
Collapse
|
14
|
Wei N, Zheng K, Xue R, Ma SL, Ren HY, Huang HF, Wang WW, Xu JJ, Chen KS. Suppression of microRNA-9-5p rescues learning and memory in chronic cerebral hypoperfusion rats model. Oncotarget 2017; 8:107920-107931. [PMID: 29296213 PMCID: PMC5746115 DOI: 10.18632/oncotarget.22415] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 08/17/2017] [Indexed: 12/11/2022] Open
Abstract
Chronic cerebral hypoperfusion has been associated with cognitive impairment in dementias, such as Alzheimer's disease (AD) and vascular disease (VaD), the two most common neurodegenerative diseases in aged people. However, the effective therapeutic approaches for both AD and VaD are still missing. MicroRNAs (miRNAs) are small non-coding RNAs that play important roles in the epigenetic regulation in many neurological disorders; the critical roles of miRNAderegulation had been implicated in both AD and VaD. In the current study, we reported that miR-9-5p is elevated in the serum and cerebrospinalfluid of patientswith VaD. The miR-9-5p wasalso increased in both the hippocampus and cortex of rats with 2-vessel occlusionsurgery. Furthermore, application ofmiR-9-5p antagomirs attenuated the memory impairments in rats with 2-vessel occlusion surgery both in the Morris water maze and inhibitory avoidance step-down tasks. Furthermore, miR-9-5p antagomirs reducedthe inhibition oflong-term potentiation and loss of dendritic spines in chronic cerebral hypoperfusionrats. Additionally, the cholinergic neuronal function was rescued by miR-9-5p antagomirs, as well as the neuronal loss and the oxidative stress. We concluded that miR-9-5p inhibition may be a potential therapeutic target for the memory impairments caused by chronic cerebral hypoperfusion.
Collapse
Affiliation(s)
- Na Wei
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450002, People's Republic of China.,Henan Key Laboratory of Tumor Pathology, Zhengzhou 450002, People's Republic of China.,Department of Pathology, School of Basic Medicine, Zhengzhou University, Zhengzhou 450002, People's Republic of China
| | - Kai Zheng
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Rui Xue
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450002, People's Republic of China
| | - Sheng-Li Ma
- Department of Emergency, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450002, People's Republic of China
| | - Hua-Yan Ren
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450002, People's Republic of China.,Henan Key Laboratory of Tumor Pathology, Zhengzhou 450002, People's Republic of China.,Department of Pathology, School of Basic Medicine, Zhengzhou University, Zhengzhou 450002, People's Republic of China
| | - Hui-Fen Huang
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450002, People's Republic of China.,Henan Key Laboratory of Tumor Pathology, Zhengzhou 450002, People's Republic of China.,Department of Pathology, School of Basic Medicine, Zhengzhou University, Zhengzhou 450002, People's Republic of China
| | - Wei-Wei Wang
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450002, People's Republic of China.,Henan Key Laboratory of Tumor Pathology, Zhengzhou 450002, People's Republic of China.,Department of Pathology, School of Basic Medicine, Zhengzhou University, Zhengzhou 450002, People's Republic of China
| | - Jing-Jing Xu
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450002, People's Republic of China.,Henan Key Laboratory of Tumor Pathology, Zhengzhou 450002, People's Republic of China.,Department of Pathology, School of Basic Medicine, Zhengzhou University, Zhengzhou 450002, People's Republic of China
| | - Kui-Sheng Chen
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450002, People's Republic of China.,Henan Key Laboratory of Tumor Pathology, Zhengzhou 450002, People's Republic of China.,Department of Pathology, School of Basic Medicine, Zhengzhou University, Zhengzhou 450002, People's Republic of China
| |
Collapse
|
15
|
Tian Y, Yang C, Shang S, Cai Y, Deng X, Zhang J, Shao F, Zhu D, Liu Y, Chen G, Liang J, Sun Q, Qiu Z, Zhang C. Loss of FMRP Impaired Hippocampal Long-Term Plasticity and Spatial Learning in Rats. Front Mol Neurosci 2017; 10:269. [PMID: 28894415 PMCID: PMC5581399 DOI: 10.3389/fnmol.2017.00269] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2017] [Accepted: 08/09/2017] [Indexed: 11/13/2022] Open
Abstract
Fragile X syndrome (FXS) is a neurodevelopmental disorder caused by mutations in the FMR1 gene that inactivate expression of the gene product, the fragile X mental retardation 1 protein (FMRP). In this study, we used clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9) technology to generate Fmr1 knockout (KO) rats by disruption of the fourth exon of the Fmr1 gene. Western blotting analysis confirmed that the FMRP was absent from the brains of the Fmr1 KO rats (Fmr1exon4-KO ). Electrophysiological analysis revealed that the theta-burst stimulation (TBS)-induced long-term potentiation (LTP) and the low-frequency stimulus (LFS)-induced long-term depression (LTD) were decreased in the hippocampal Schaffer collateral pathway of the Fmr1exon4-KO rats. Short-term plasticity, measured as the paired-pulse ratio, remained normal in the KO rats. The synaptic strength mediated by the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) was also impaired. Consistent with previous reports, the Fmr1exon4-KO rats demonstrated an enhanced 3,5-dihydroxyphenylglycine (DHPG)-induced LTD in the present study, and this enhancement is insensitive to protein translation. In addition, the Fmr1exon4-KO rats showed deficits in the probe trial in the Morris water maze test. These results demonstrate that deletion of the Fmr1 gene in rats specifically impairs long-term synaptic plasticity and hippocampus-dependent learning in a manner resembling the key symptoms of FXS. Furthermore, the Fmr1exon4-KO rats displayed impaired social interaction and macroorchidism, the results consistent with those observed in patients with FXS. Thus, Fmr1exon4-KO rats constitute a novel rat model of FXS that complements existing mouse models.
Collapse
Affiliation(s)
- Yonglu Tian
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University-IDG/McGovern Institute for Brain Research, Peking UniversityBeijing, China.,Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking UniversityBeijing, China
| | - Chaojuan Yang
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University-IDG/McGovern Institute for Brain Research, Peking UniversityBeijing, China
| | - Shujiang Shang
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University-IDG/McGovern Institute for Brain Research, Peking UniversityBeijing, China
| | - Yijun Cai
- CAS Key Laboratory of Primate Neurobiology, Institute of Neuroscience, Chinese Academy of SciencesShanghai, China
| | - Xiaofei Deng
- Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of SciencesBeijing, China
| | - Jian Zhang
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University-IDG/McGovern Institute for Brain Research, Peking UniversityBeijing, China
| | - Feng Shao
- Department of Psychology, Peking UniversityBeijing, China
| | - Desheng Zhu
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University-IDG/McGovern Institute for Brain Research, Peking UniversityBeijing, China
| | - Yunbo Liu
- Institute of Laboratory Animal Science, Peking Union Medical College/Chinese Academy of Medical SciencesBeijing, China
| | - Guiquan Chen
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing UniversityNanjing, China
| | - Jing Liang
- Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of SciencesBeijing, China
| | - Qiang Sun
- CAS Key Laboratory of Primate Neurobiology, Institute of Neuroscience, Chinese Academy of SciencesShanghai, China
| | - Zilong Qiu
- CAS Key Laboratory of Primate Neurobiology, Institute of Neuroscience, Chinese Academy of SciencesShanghai, China
| | - Chen Zhang
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University-IDG/McGovern Institute for Brain Research, Peking UniversityBeijing, China.,Key Laboratory for Neuroscience, Ministry of Education/National Health and Family Planning Commission, Peking UniversityBeijing, China
| |
Collapse
|
16
|
Lana D, Ugolini F, Melani A, Nosi D, Pedata F, Giovannini MG. The neuron-astrocyte-microglia triad in CA3 after chronic cerebral hypoperfusion in the rat: Protective effect of dipyridamole. Exp Gerontol 2017; 96:46-62. [PMID: 28606482 DOI: 10.1016/j.exger.2017.06.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 06/07/2017] [Accepted: 06/08/2017] [Indexed: 12/01/2022]
Abstract
We investigated the quantitative and morphofunctional alterations of neuron-astrocyte-microglia triads in CA3 hippocampus, in comparison to CA1, after 2 Vessel Occlusion (2VO) and the protective effect of dipyridamole. We evaluated 3 experimental groups: sham-operated rats (sham, n=15), 2VO-operated rats treated with vehicle (2VO-vehicle, n=15), and 2VO-operated rats treated with dipyridamole from day 0 to day 7 (2VO-dipyridamole, n=15), 90days after 2VO. We analyzed Stratum Pyramidalis (SP), Stratum Lucidum (SL) and Stratum Radiatum (SR) of CA3. 1) ectopic neurons increased in SL and SR of 2VO-vehicle, and 2VO-dipyridamole rats; 2) apoptotic neurons increased in SP of 2VO-vehicle rats and dipyridamole reverted this effect; 3) astrocytes increased in SP, SL and SR of 2VO-vehicle and 2VO-dipyridamole rats; 4) TNF-α expression increased in astrocytes, blocked by dipyridamole, and in dendrites in SR of 2VO-vehicle rats; 5) total microglia increased in SL and SR of 2VO-vehicle and 2VO-dipyridamole rats; 6) triads increased in SR of 2VO-vehicle rats and dipyridamole reverted this effect. Microglia cooperated with astrocytes to phagocytosis of apoptotic neurons and debris, and engulfed ectopic non-fragmented neurons in SL of 2VO-vehicle and 2VO-dipyridamole rats, through a new mechanism called phagoptosis. CA3 showed a better adaptive capacity than CA1 to the ischemic insult, possibly due to the different behaviour of astrocytes and microglial cells. Dipyridamole had neuroprotective effects.
Collapse
Affiliation(s)
- Daniele Lana
- Department of Health Sciences, Section of Pharmacology and Clinical Oncology, University of Florence, Viale Pieraccini 6, 50139 Firenze, Italy.
| | - Filippo Ugolini
- Department of Health Sciences, Section of Pharmacology and Clinical Oncology, University of Florence, Viale Pieraccini 6, 50139 Firenze, Italy.
| | - Alessia Melani
- Department of NEUROFARBA, Section of Pharmacology and Toxicology, University of Florence, Viale Pieraccini 6, 50139 Firenze, Italy.
| | - Daniele Nosi
- Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla, 50139 Firenze, Italy.
| | - Felicita Pedata
- Department of NEUROFARBA, Section of Pharmacology and Toxicology, University of Florence, Viale Pieraccini 6, 50139 Firenze, Italy.
| | - Maria Grazia Giovannini
- Department of Health Sciences, Section of Pharmacology and Clinical Oncology, University of Florence, Viale Pieraccini 6, 50139 Firenze, Italy.
| |
Collapse
|
17
|
Puri N, Mohey V, Singh M, Kaur T, Pathak D, Buttar HS, Singh AP. Dipyridamole attenuates ischemia reperfusion induced acute kidney injury through adenosinergic A1 and A2A receptor agonism in rats. Naunyn Schmiedebergs Arch Pharmacol 2016; 389:361-8. [PMID: 26728617 DOI: 10.1007/s00210-015-1206-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 12/21/2015] [Indexed: 01/16/2023]
Abstract
Dipyridamole (DYP) is an anti-platelet agent with marked vasodilator, anti-oxidant, and anti-inflammatory activity. The present study investigated the role of adenosine receptors in DYP-mediated protection against ischemia reperfusion-induced acute kidney injury (AKI) in rats. The rats were subjected to bilateral renal ischemia for 40 min followed by reperfusion for 24 h. The renal damage induced by ischemia reperfusion injury (IRI) was assessed by measuring creatinine clearance, blood urea nitrogen, uric acid, plasma potassium, fractional excretion of sodium, and microproteinuria in rats. The oxidative stress in renal tissues was assessed by quantification of thiobarbituric acid-reactive substances, superoxide anion generation, and reduced glutathione level. The hematoxylin-eosin staining was carried out to observe histopathological changes in renal tissues. DYP (10 and 30 mg/kg, intraperitoneal, i.p.) was administered 30 min before subjecting the rats to renal IRI. In separate groups, caffeine (50 mg/kg, i.p.), an adenosinergic A1 and A2A receptor antagonist was administered with and without DYP treatment before subjecting the rats to renal IRI. The ischemia reperfusion-induced AKI was demonstrated by significant changes in serum as well as urinary parameters, enhanced oxidative stress, and histopathological changes in renal tissues. The administration of DYP demonstrated protection against AKI. The prior treatment with caffeine abolished DYP-mediated reno-protection suggesting role of A1 and A2A adenosine receptors in DYP-mediated reno-protection in rats. It is concluded that adenosine receptors find their definite involvement in DYP-mediated anti-oxidative and reno-protective effect against ischemia reperfusion-induced AKI.
Collapse
Affiliation(s)
- Nikkita Puri
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, 143005, India
| | - Vinita Mohey
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, 143005, India
| | - Manjinder Singh
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, 143005, India
| | - Tajpreet Kaur
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, 143005, India.,Department of Pharmacology, Khalsa College of Pharmacy, Amritsar, India
| | - Devendra Pathak
- Department of Veterinary Anatomy, Guru Angad Dev Veterinary and Animal Sciences University, Ludhiana, India
| | - Harpal Singh Buttar
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, 143005, India.,Department of Pathology and Laboratory Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Amrit Pal Singh
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, 143005, India.
| |
Collapse
|
18
|
Rogosnitzky M, Isakov I, Wlassoff W, Ingram A, Barishak YR. Ocular Applications of Dipyridamole: A Review of Indications and Routes of Administration. J Ocul Pharmacol Ther 2015; 32:83-9. [PMID: 26696547 PMCID: PMC4779994 DOI: 10.1089/jop.2015.0128] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Dipyridamole was introduced decades ago as a treatment for angina, subsequently found to inhibit platelet aggregation. It is most commonly used, and approved for use in thromboembolism prevention, following surgery. Some of its recognized effects such as adenosine uptake inhibition, elevation of cAMP and cGMP levels, vasodilation, and tissue perfusion are important in various ocular disorders. For this reason, dipyridamole represents an interesting candidate as a therapeutic target for the treatment of eye disorders affecting different ocular structures. The aim of this article is to review the evidence and current understanding of the mechanisms by which dipyridamole exerts its effects on different ocular tissues, discuss the role of dipyridamole in clinical practice, and highlight areas of use and routes of administration.
Collapse
Affiliation(s)
- Moshe Rogosnitzky
- 1 Ocular Research Department, Center for Drug Repurposing, Ariel University , Ariel, Israel .,2 Eye Research Program, MedInsight Research Institute , Hampshire, United Kingdom
| | - Itzhak Isakov
- 1 Ocular Research Department, Center for Drug Repurposing, Ariel University , Ariel, Israel
| | | | - April Ingram
- 2 Eye Research Program, MedInsight Research Institute , Hampshire, United Kingdom
| | - Y Robert Barishak
- 1 Ocular Research Department, Center for Drug Repurposing, Ariel University , Ariel, Israel
| |
Collapse
|
19
|
Ghatak S, Banerjee A, Sikdar SK. Ischaemic concentrations of lactate increase TREK1 channel activity by interacting with a single histidine residue in the carboxy terminal domain. J Physiol 2015; 594:59-81. [PMID: 26445100 DOI: 10.1113/jp270706] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Accepted: 09/21/2015] [Indexed: 02/01/2023] Open
Abstract
KEY POINTS The physiological metabolite, lactate and the two-pore domain leak potassium channel, TREK1 are known neuroprotectants against cerebral ischaemia. However, it is not known whether lactate interacts with TREK1 channel to provide neuroprotection. In this study we show that lactate increases TREK1 channel activity and hyperpolarizes CA1 stratum radiatum astrocytes in hippocampal slices. Lactate increases open probability and decreases longer close time of the human (h)TREK1 channel in a concentration dependent manner. Lactate interacts with histidine 328 (H328) in the carboxy terminal domain of hTREK1 channel to decrease its dwell time in the longer closed state. This interaction was dependent on the charge on H328. Lactate-insensitive mutant H328A hTREK1 showed pH sensitivity similar to wild-type hTREK1, indicating that the effect of lactate on hTREK1 is independent of pH change. A rise in lactate concentration and the leak potassium channel TREK1 have been independently associated with cerebral ischaemia. Recent literature suggests lactate to be neuroprotective and TREK1 knockout mice show an increased sensitivity to brain and spinal cord ischaemia; however, the connecting link between the two is missing. Therefore we hypothesized that lactate might interact with TREK1 channels. In the present study, we show that lactate at ischaemic concentrations (15-30 mm) at pH 7.4 increases TREK1 current in CA1 stratum radiatum astrocytes and causes membrane hyperpolarization. We confirm the intracellular action of lactate on TREK1 in hippocampal slices using monocarboxylate transporter blockers and at single channel level in cell-free inside-out membrane patches. The intracellular effect of lactate on TREK1 is specific since other monocarboxylates such as pyruvate and acetate at pH 7.4 failed to increase TREK1 current. Deletion and point mutation experiments suggest that lactate decreases the longer close dwell time incrementally with increase in lactate concentration by interacting with the histidine residue at position 328 (H328) in the carboxy terminal domain of the TREK1 channel. The interaction of lactate with H328 is dependent on the charge on the histidine residue since isosteric mutation of H328 to glutamine did not show an increase in TREK1 channel activity with lactate. This is the first demonstration of a direct effect of lactate on ion channel activity. The action of lactate on the TREK1 channel signifies a separate neuroprotective mechanism in ischaemia since it was found to be independent of the effect of acidic pH on channel activity.
Collapse
Affiliation(s)
- Swagata Ghatak
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore, Karnataka, 560012, India
| | - Aditi Banerjee
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore, Karnataka, 560012, India
| | - Sujit Kumar Sikdar
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore, Karnataka, 560012, India
| |
Collapse
|
20
|
Cheng Y, Zhang Y, Song H, Feng J. Intercellular adhesion molecule-1 expression in the hippocampal CA1 region of hyperlipidemic rats with chronic cerebral ischemia. Neural Regen Res 2015; 7:1312-7. [PMID: 25657661 PMCID: PMC4308801 DOI: 10.3969/j.issn.1673-5374.2012.17.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2012] [Accepted: 05/21/2012] [Indexed: 11/18/2022] Open
Abstract
Chronic cerebral ischemia is a pathological process in many cerebrovascular diseases and it is induced by long-term hyperlipidemia, hypertension and diabetes mellitus. After being fed a high-fat diet for 4 weeks, rats were subjected to permanent occlusion of bilateral common carotid arteries to establish rat models of chronic cerebral ischemia with hyperlipidemia. Intercellular adhesion molecule-1 expression in rat hippocampal CA1 region was determined to better understand the mechanism underlying the effects of hyperlipidemia on chronic cerebral ischemia. Water maze test results showed that the cognitive function of rats with hyperlipidemia or chronic cerebral ischemia, particularly in rats with hyperlipidemia combined with chronic cerebral ischemia, gradually decreased between 1 and 4 months after occlusion of the bilateral common carotid arteries. This correlated with pathological changes in the hippocampal CA1 region as detected by hematoxylin-eosin staining. Immunohistochemical staining showed that intercellular adhesion molecule-1 expression in the hippocampal CA1 region was noticeably increased in rats with hyperlipidemia or chronic cerebral ischemia, in particular in rats with hyperlipidemia combined with chronic cerebral ischemia. These findings suggest that hyperlipidemia aggravates chronic cerebral ischemia-induced neurological damage and cognitive impairment in the rat hippocampal CA1 region, which may be mediated, at least in part, by up-regulated expression of intercellular adhesion molecule-1.
Collapse
Affiliation(s)
- Yingying Cheng
- Department of Neurology, The First Bethune Hospital of Jilin University, Changchun 130021, Jilin Province, China
| | - Ying Zhang
- Department of Neurology, The First Bethune Hospital of Jilin University, Changchun 130021, Jilin Province, China
| | - Hongmei Song
- Department of Neurology, The First Bethune Hospital of Jilin University, Changchun 130021, Jilin Province, China
| | - Jiachun Feng
- Department of Neurology, The First Bethune Hospital of Jilin University, Changchun 130021, Jilin Province, China
| |
Collapse
|
21
|
Bortolotto JW, Melo GMD, Cognato GDP, Vianna MRM, Bonan CD. Modulation of adenosine signaling prevents scopolamine-induced cognitive impairment in zebrafish. Neurobiol Learn Mem 2014; 118:113-9. [PMID: 25490060 DOI: 10.1016/j.nlm.2014.11.016] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Revised: 11/21/2014] [Accepted: 11/27/2014] [Indexed: 12/29/2022]
Abstract
Adenosine, a purine ribonucleoside, exhibits neuromodulatory and neuroprotective effects in the brain and is involved in memory formation and cognitive function. Adenosine signaling is mediated by adenosine receptors (A1, A2A, A2B, and A3); in turn, nucleotide and nucleoside-metabolizing enzymes and adenosine transporters regulate its levels. Scopolamine, a muscarinic cholinergic receptor antagonist, has profound amnesic effects in a variety of learning paradigms and has been used to induce cognitive deficits in animal models. This study investigated the effects of acute exposure to caffeine (a non-selective antagonist of adenosine receptors A1 and A2A), ZM 241385 (adenosine receptor A2A antagonist), DPCPX (adenosine receptor A1 antagonist), dipyridamole (inhibitor of nucleoside transporters) and EHNA (inhibitor of adenosine deaminase) in a model of pharmacological cognitive impairment induced by scopolamine in adult zebrafish. Caffeine, ZM 241385, DPCPX, dipyridamole, and EHNA were acutely administered independently via i.p. in zebrafish, followed by exposure to scopolamine dissolved in tank water (200μM). These compounds prevented the scopolamine-induced amnesia without impacting locomotor activity or social interaction. Together, these data support the hypothesis that adenosine signaling may modulate memory processing, suggesting that these compounds present a potential preventive strategy against cognitive impairment.
Collapse
Affiliation(s)
- Josiane Woutheres Bortolotto
- Laboratório de Neuroquímica e Psicofarmacologia, Departamento de Biologia Celular e Molecular, Programa de Pós-Graduação em Biologia Celular e Molecular, Faculdade de Biociências, Pontifícia Universidade Católica do Rio Grande do Sul, Avenida Ipiranga, 6681, 90619-900 Porto Alegre, RS, Brazil; ZebLab - Grupo de Pesquisa em modelos experimentais em zebrafish, Faculdade de Biociências, Pontifícia Universidade Católica do Rio Grande do Sul, Avenida Ipiranga, 6681, 90619-900 Porto Alegre, RS, Brazil
| | - Gabriela Madalena de Melo
- Laboratório de Neuroquímica e Psicofarmacologia, Departamento de Biologia Celular e Molecular, Programa de Pós-Graduação em Biologia Celular e Molecular, Faculdade de Biociências, Pontifícia Universidade Católica do Rio Grande do Sul, Avenida Ipiranga, 6681, 90619-900 Porto Alegre, RS, Brazil; ZebLab - Grupo de Pesquisa em modelos experimentais em zebrafish, Faculdade de Biociências, Pontifícia Universidade Católica do Rio Grande do Sul, Avenida Ipiranga, 6681, 90619-900 Porto Alegre, RS, Brazil
| | - Giana de Paula Cognato
- Laboratório de Neuroquímica e Psicofarmacologia, Departamento de Biologia Celular e Molecular, Programa de Pós-Graduação em Biologia Celular e Molecular, Faculdade de Biociências, Pontifícia Universidade Católica do Rio Grande do Sul, Avenida Ipiranga, 6681, 90619-900 Porto Alegre, RS, Brazil; Programa de Pós-Graduação em Bioquímica e Bioprospecção, Universidade Federal de Pelotas, Campus Universitário Capão do Leão, s/n°, CEP 96010-900, Pelotas, RS, Brazil
| | - Mônica Ryff Moreira Vianna
- Laboratório de Biologia e Desenvolvimento do Sistema Nervoso, Departamento de Ciências Morfofisiológicas, Programa de Pós-Graduação em Biologia Celular e Molecular, Faculdade de Biociências, Pontifícia Universidade Católica do Rio Grande do Sul, Avenida Ipiranga, 6681, 90619-900 Porto Alegre, RS, Brazil; ZebLab - Grupo de Pesquisa em modelos experimentais em zebrafish, Faculdade de Biociências, Pontifícia Universidade Católica do Rio Grande do Sul, Avenida Ipiranga, 6681, 90619-900 Porto Alegre, RS, Brazil
| | - Carla Denise Bonan
- Laboratório de Neuroquímica e Psicofarmacologia, Departamento de Biologia Celular e Molecular, Programa de Pós-Graduação em Biologia Celular e Molecular, Faculdade de Biociências, Pontifícia Universidade Católica do Rio Grande do Sul, Avenida Ipiranga, 6681, 90619-900 Porto Alegre, RS, Brazil; ZebLab - Grupo de Pesquisa em modelos experimentais em zebrafish, Faculdade de Biociências, Pontifícia Universidade Católica do Rio Grande do Sul, Avenida Ipiranga, 6681, 90619-900 Porto Alegre, RS, Brazil.
| |
Collapse
|
22
|
Lana D, Melani A, Pugliese AM, Cipriani S, Nosi D, Pedata F, Giovannini MG. The neuron-astrocyte-microglia triad in a rat model of chronic cerebral hypoperfusion: protective effect of dipyridamole. Front Aging Neurosci 2014; 6:322. [PMID: 25505884 PMCID: PMC4245920 DOI: 10.3389/fnagi.2014.00322] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Accepted: 11/04/2014] [Indexed: 11/13/2022] Open
Abstract
Chronic cerebral hypoperfusion during aging may cause progressive neurodegeneration as ischemic conditions persist. Proper functioning of the interplay between neurons and glia is fundamental for the functional organization of the brain. The aim of our research was to study the pathophysiological mechanisms, and particularly the derangement of the interplay between neurons and astrocytes-microglia with the formation of "triads," in a model of chronic cerebral hypoperfusion induced by the two-vessel occlusion (2VO) in adult Wistar rats (n = 15). The protective effect of dipyridamole given during the early phases after 2VO (4 mg/kg/day i.v., the first 7 days after 2VO) was verified (n = 15). Sham-operated rats (n = 15) were used as controls. Immunofluorescent triple staining of neurons (NeuN), astrocytes (GFAP), and microglia (IBA1) was performed 90 days after 2VO. We found significantly higher amount of "ectopic" neurons, neuronal debris and apoptotic neurons in CA1 Str. Radiatum and Str. Pyramidale of 2VO rats. In CA1 Str. Radiatum of 2VO rats the amount of astrocytes (cells/mm(2)) did not increase. In some instances several astrocytes surrounded ectopic neurons and formed a "micro scar" around them. Astrocyte branches could infiltrate the cell body of ectopic neurons, and, together with activated microglia cells formed the "triads." In the triad, significantly more numerous in CA1 Str. Radiatum of 2VO than in sham rats, astrocytes and microglia cooperated in the phagocytosis of ectopic neurons. These events might be common mechanisms underlying many neurodegenerative processes. The frequency to which they appear might depend upon, or might be the cause of, the burden and severity of neurodegeneration. Dypiridamole significantly reverted all the above described events. The protective effect of chronic administration of dipyridamole might be a consequence of its vasodilatory, antioxidant and anti-inflammatory role during the early phases after 2VO.
Collapse
Affiliation(s)
- Daniele Lana
- Section of Pharmacology and Clinical Oncology, Department of Health Sciences, University of Florence Florence, Italy
| | - Alessia Melani
- Section of Pharmacology and Toxicology, Department of NEUROFARBA, University of Florence Florence, Italy
| | - Anna Maria Pugliese
- Section of Pharmacology and Toxicology, Department of NEUROFARBA, University of Florence Florence, Italy
| | | | - Daniele Nosi
- Department of Experimental and Clinical Medicine, University of Florence Florence, Italy
| | - Felicita Pedata
- Section of Pharmacology and Toxicology, Department of NEUROFARBA, University of Florence Florence, Italy
| | - Maria Grazia Giovannini
- Section of Pharmacology and Clinical Oncology, Department of Health Sciences, University of Florence Florence, Italy
| |
Collapse
|
23
|
Balakumar P, Nyo YH, Renushia R, Raaginey D, Oh AN, Varatharajan R, Dhanaraj SA. Classical and pleiotropic actions of dipyridamole: Not enough light to illuminate the dark tunnel? Pharmacol Res 2014; 87:144-50. [PMID: 24861566 DOI: 10.1016/j.phrs.2014.05.008] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Revised: 05/01/2014] [Accepted: 05/13/2014] [Indexed: 12/01/2022]
Abstract
Dipyridamole is a platelet inhibitor indicated for the secondary prevention of transient ischemic attack. It inhibits the enzyme phosphodiesterase, elevates cAMP and cGMP levels and prevents platelet aggregation. Dipyridamole inhibits the cellular uptake of adenosine into red blood cells, platelets and endothelial cells that results in increased extracellular availability of adenosine, leading to modulation of cardiovascular function. The antiplatelet action of dipyridamole might offer therapeutic benefits in secondary stroke prevention in combination with aspirin. Inflammation and oxidative stress play an important role in atherosclerosis and thrombosis development, leading to stroke progression. Studies demonstrated anti-inflammatory, anti-oxidant and anti-proliferative actions of dipyridamole. These pleiotropic potentials of dipyridamole might contribute to improved therapeutic outcomes when used with aspirin in preventing secondary stroke. Dipyridamole was documented as a coronary vasodilator 5 decades ago. The therapeutic failure of dipyridamole as a coronary vasodilator is linked with induction of 'coronary steal' phenomenon in which by dilating resistance vessels in non-ischemic zone, dipyridamole diverts the already reduced blood flow away from the area of ischemic myocardium. Dipyridamole at high-dose could cause a marked 'coronary steal' effect. Dipyridamole, however, at low-dose could have a minimal hemodynamic effect. Low-dose dipyridamole treatment has a therapeutic potential in partially preventing diabetes mellitus-induced experimental vascular endothelial and renal abnormalities by enhancing endothelial nitric oxide signals and inducing renovascular reduction of oxidative stress. In spite of plenteous research on dipyridamole's use in clinics, its precise clinical application is still obscure. This review sheds lights on pleiotropic pharmacological actions and therapeutic potentials of dipyridamole.
Collapse
Affiliation(s)
- Pitchai Balakumar
- Pharmacology Unit, Faculty of Pharmacy, Asian Institute of Medicine, Science and Technology (AIMST) University, Semeling, 08100 Bedong, Kedah Darul Aman, Malaysia.
| | - Ying Hui Nyo
- Pharmacology Unit, Faculty of Pharmacy, Asian Institute of Medicine, Science and Technology (AIMST) University, Semeling, 08100 Bedong, Kedah Darul Aman, Malaysia
| | - Raja Renushia
- Pharmacology Unit, Faculty of Pharmacy, Asian Institute of Medicine, Science and Technology (AIMST) University, Semeling, 08100 Bedong, Kedah Darul Aman, Malaysia
| | - Devarajan Raaginey
- Pharmacology Unit, Faculty of Pharmacy, Asian Institute of Medicine, Science and Technology (AIMST) University, Semeling, 08100 Bedong, Kedah Darul Aman, Malaysia
| | - Ann Nah Oh
- Pharmacology Unit, Faculty of Pharmacy, Asian Institute of Medicine, Science and Technology (AIMST) University, Semeling, 08100 Bedong, Kedah Darul Aman, Malaysia
| | - Rajavel Varatharajan
- Pharmacology Unit, Faculty of Pharmacy, Asian Institute of Medicine, Science and Technology (AIMST) University, Semeling, 08100 Bedong, Kedah Darul Aman, Malaysia
| | - Sokkalingam A Dhanaraj
- Pharmaceutical Technology Unit, Faculty of Pharmacy, AIMST University, Semeling, 08100 Bedong, Kedah Darul Aman, Malaysia
| |
Collapse
|
24
|
|