Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
2.
Virol J ; 20(1): 241, 2023 10 24.
Article in English | MEDLINE | ID: mdl-37875904

ABSTRACT

BACKGROUND: Coronavirus disease 19 (COVID-19) is the disease caused by SARS-CoV-2, a highly infectious member of the coronavirus family, which emerged in December 2019 in "Wuhan, China". It induces respiratory illness ranging from mild symptoms to severe disease. It was declared a "pandemic" by the World Health Organization (WHO) in March 2020. Since then, a vast number of clinical and experimental studies have been conducted to identify effective approaches for its prevention and treatment. MAIN BODY: The pathophysiology of COVID-19 represents an unprecedented challenge; it triggers a strong immune response, which may be exacerbated by "a cytokine storm syndrome". It also induces thrombogenesis and may trigger multi-organ injury. Therefore, different drug classes have been proposed for its treatment and prevention, such as antivirals, anti-SARS-CoV-2 antibody agents (monoclonal antibodies, convalescent plasma, and immunoglobulins), anti-inflammatory drugs, immunomodulators, and anticoagulant drugs. To the best of our knowledge, this review is the first to present, discuss, and summarize the current knowledge about the different drug classes used for the treatment of COVID-19, with special emphasis on their targets, mechanisms of action, and important adverse effects and drug interactions. Additionally, we spotlight the latest "October 2023" important guidelines (NIH, IDSA, and NICE) and FDA approval or authorization regarding the use of these agents in the management of COVID-19. CONCLUSION: Despite the wide array of therapeutic strategies introduced for the treatment of COVID-19, one of the most prominent therapeutic challenges is SARS-CoV-2 mutations and emerging new variants and subvariants. Currently, the anti-COVID-19 drug pipeline is continuously affording novel treatments to face this growing challenge.


Subject(s)
Antiviral Agents , COVID-19 , Humans , Antiviral Agents/therapeutic use , Antiviral Agents/pharmacology , SARS-CoV-2 , Antibodies, Viral , Antibodies, Monoclonal
3.
Int Immunopharmacol ; 124(Pt A): 110945, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37716161

ABSTRACT

Chemotherapy-induced cognitive impairment (CICI) is a general term describing cognitive dysfunction during/after treatment with chemotherapeutic agents. CICI represents a significant medical problem due to its increasing prevalence with the lack of robust therapeutic approaches. This study aimed at investigating the effects of chronic treatment with amisulpride (5 mg/kg/day) in the management of 5-fluorouracil (5-FU)-induced cognitive deficits in Wistar rats. Rats received 5 intraperitoneal injections of 5-FU (25 mg/kg every 3 days). 5-FU treatment induced impairments in spatial learning (reduction in object location discrimination ratio) and non-spatial learning (reduction in novel object recognition discrimination ratio). Moreover, 5-FU induced a decrease in the activity of the Wnt/GSK-3ß/ß-catenin pathway with a decrease in brain-derived neurotrophic factor (BDNF) level in the hippocampus. These changes were associated with an increase in the expression of the pro-inflammatory cytokines; tumor necrosis factor-α (TNF-α) and interleukin-1ß (IL-1ß), in hippocampal tissue sections accompanied by a decrease in the number of Ki-67 positive cells (indicating a decrease in proliferative capacity), a decrease in the Nissl's granules optical density (denoting neurodegeneration), a decrease in the number of viable intact neurons with an increase in the expression of ß-amyloid and caspase-3. Amisulpride enhanced Wnt/GSK-3ß/ß-catenin signaling, increased BDNF levels, and abrogated 5-FU-induced neuroinflammation, apoptosis, ß-amyloid accumulation, and neurodegenerative changes with an improvement of cognitive performance. This study draws attention to the pro-cognitive effects of amisulpride in 5-FU-exposed rats that could be attributed to enhancing hippocampal Wnt/GSK-3ß/ß-catenin signaling pathway, and this could offer a promising therapeutic option for subjects with CICI.


Subject(s)
Brain-Derived Neurotrophic Factor , Cognitive Dysfunction , Humans , Rats , Animals , Rats, Wistar , Glycogen Synthase Kinase 3 beta/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Amisulpride/pharmacology , Fluorouracil/pharmacology , beta Catenin/metabolism , Wnt Signaling Pathway , Hippocampus , Cognitive Dysfunction/chemically induced , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/metabolism , Cognition
4.
Eur J Pharmacol ; 955: 175916, 2023 Sep 15.
Article in English | MEDLINE | ID: mdl-37460052

ABSTRACT

Several reports indicate a plausible role of calcium (Ca2+) permeable AMPA glutamate receptors (with RNA hypo-editing at the GluA2 Q/R site) and the subsequent excitotoxicity-mediated neuronal death in the pathogenesis of a wide array of neurological disorders including autism spectrum disorder (ASD). This study was designed to examine the effects of chronic risperidone treatment on the expression of adenosine deaminase acting on RNA 2 (Adar2), the status of AMPA glutamate receptor GluA2 editing, and its effects on oxidative/nitrosative stress and excitotoxicity-mediated neuronal death in the prenatal valproic acid (VPA) rat model of ASD. Prenatal VPA exposure was associated with autistic-like behaviors accompanied by an increase in the apoptotic marker "caspase-3" and a decrease in the antiapoptotic marker "BCL2" alongside a reduction in the Adar2 relative gene expression and an increase in GluA2 Q:R ratio in the hippocampus and the prefrontal cortex. Risperidone, at doses of 1 and 3 mg, improved the VPA-induced behavioral deficits and enhanced the Adar2 relative gene expression and the subsequent GluA2 subunit editing. This was reflected on the cellular level where risperidone impeded VPA-induced oxidative/nitrosative stress and neurodegenerative changes. In conclusion, the present study confirms a possible role for Adar2 downregulation and the subsequent hypo-editing of the GluA2 subunit in the pathophysiology of the prenatal VPA rat model of autism and highlights the favorable effect of risperidone on reversing the RNA editing machinery deficits, giving insights into a new possible mechanism of risperidone in autism.


Subject(s)
Autism Spectrum Disorder , Autistic Disorder , Animals , Female , Pregnancy , Rats , Adenosine Deaminase/genetics , Adenosine Deaminase/metabolism , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/metabolism , Autism Spectrum Disorder/chemically induced , Autistic Disorder/chemically induced , Autistic Disorder/drug therapy , Autistic Disorder/genetics , Disease Models, Animal , Glutamic Acid/metabolism , Receptors, AMPA/genetics , Receptors, AMPA/metabolism , Risperidone/pharmacology , RNA/metabolism , RNA Editing , Valproic Acid/adverse effects
5.
Neurotherapeutics ; 20(2): 464-483, 2023 03.
Article in English | MEDLINE | ID: mdl-36918475

ABSTRACT

Significant efforts are increasingly directed towards identifying novel therapeutic targets for autism spectrum disorder (ASD) with a rising role of aberrant glutamatergic transmission in the pathogenesis of ASD-associated cellular and behavioral deficits. This study aimed at investigating the role of chronic memantine (20 mg/kg/day) and aripiprazole (3 mg/kg/day) combination therapy in the management of prenatal sodium valproate (VPA)-induced autistic-like/cognitive deficits in male Wistar rats. Pregnant female rats received a single intraperitoneal injection of VPA (600 mg/kg) to induce autistic-like behaviors in their offspring. Prenatal VPA induced autistic-like symptoms (decreased social interaction and the appearance of stereotyped behavior) with deficits in spatial learning (in Morris water maze) and cognitive flexibility (in the attentional set-shifting task) in addition to decreased hippocampal protein levels of phosphorylated cAMP response element-binding protein (p-CREB), brain-derived neurotrophic factor (BDNF), and gene expression of glutamate transporter-1 (Glt-1) with a decline in GABA/glutamate ratio (both measured by HPLC). These were accompanied by the appearance of numerous neurofibrillary tangles (NFTs) with enhanced apoptosis in hippocampal sections. Memantine/aripiprazole combination increased the protein levels of p-CREB, BDNF, and Glt-1 gene expression with restoration of GABA/glutamate balance, attenuation of VPA-induced neurodegenerative changes and autistic-like symptoms, and improvement of cognitive performance. This study draws attention to the favorable cognitive effects of memantine/aripiprazole combination in autistic subjects which could be mediated via enhancing CREB/BDNF signaling with increased expression of astrocytic Glt-1 and restoration of GABA/glutamate balance, leading to inhibition of hippocampal NFTs formation and neuronal apoptosis.


Subject(s)
Autism Spectrum Disorder , Autistic Disorder , Cognitive Dysfunction , Animals , Female , Male , Pregnancy , Rats , Aripiprazole/adverse effects , Autism Spectrum Disorder/chemically induced , Autism Spectrum Disorder/drug therapy , Autistic Disorder/chemically induced , Autistic Disorder/drug therapy , Brain-Derived Neurotrophic Factor/metabolism , Cognitive Dysfunction/chemically induced , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/complications , Disease Models, Animal , gamma-Aminobutyric Acid/pharmacology , Glutamates/adverse effects , Hippocampus , Homeostasis , Memantine/adverse effects , Rats, Wistar , Valproic Acid
6.
Eur J Pharmacol ; 927: 175046, 2022 Jul 15.
Article in English | MEDLINE | ID: mdl-35623405

ABSTRACT

Oxidative stress induced neurotoxicity is increasingly perceived as an important neuropathologic mechanism underlying the motor and behavioral phenotypes associated with Huntington's disease (HD). Repeated exposure to 3-nitropropionic acid (3-NP) induces neurotoxic changes which closely simulate the neuropathological and behavioral characteristics of HD. This study aimed at evaluating the prophylactic effects of the dual-specificity tyrosine phosphorylation regulated kinase 1A (DYRK1A) inhibitor "harmine" against 3-NP-indued neurotoxicity and HD-like symptoms. The potential prophylactic effect of harmine (10 mg/kg/day; intraperitoneal) was investigated on 3-NP-induced motor and cognitive HD-like deficits, nuclear factor erythroid 2 like 2 (NRF2), AMP kinase (AMPK) and p21 protein levels and the gene expression of haem oxygenase-1 (Ho-1), NAD(P)H: quinone oxidoreductase-1 (Nqo-1) and p62 in addition to redox imbalance and histological neurotoxic changes in the striatum, prefrontal cortex, and hippocampus of male Wistar rats. Harmine successfully increased the protein levels of NRF2, AMPK and p21 and the gene expression of Ho-1, Nqo-1 and p62, restored redox homeostasis, and reduced CASPASE-3 level. This was reflected in attenuation of 3-NP-induced neurodegenerative changes and improvement of rats' motor and cognitive performance. This study draws attention to the protective role of harmine against 3-NP-induced motor and cognitive dysfunction that could be mediated via enhancing NRF2-mediated signaling with subsequent amelioration of oxidative stress injury via NRF2 activators, p21 and AMPK, in the striatum, prefrontal cortex, and hippocampus which could offer a promising therapeutic tool to slow the progression of HD.


Subject(s)
AMP-Activated Protein Kinases , Cyclin-Dependent Kinase Inhibitor p21 , Harmine , Huntington Disease , Neuroprotective Agents , Neurotoxicity Syndromes , AMP-Activated Protein Kinases/metabolism , Adenylate Kinase/metabolism , Animals , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Harmine/pharmacology , Huntington Disease/chemically induced , Huntington Disease/metabolism , Huntington Disease/prevention & control , Male , NF-E2-Related Factor 2/metabolism , Neuroprotective Agents/adverse effects , Neurotoxicity Syndromes/drug therapy , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/prevention & control , Nitro Compounds/antagonists & inhibitors , Nitro Compounds/pharmacology , Oxidative Stress , Propionates/antagonists & inhibitors , Propionates/pharmacology , Rats , Rats, Wistar , Signal Transduction/drug effects
7.
Br J Clin Pharmacol ; 88(9): 4211-4216, 2022 09.
Article in English | MEDLINE | ID: mdl-35244235

ABSTRACT

Tranexamic acid (TXA) is widely utilized to control perioperative bleeding. TXA is considered a safe drug with few serious adverse effects, but many studies report TXA-associated seizures, especially with cardiac surgeries. Usually, TXA-associated seizures persist for a few minutes with no progression into status epilepticus. Here, we report, for the first time, a case of refractory status epilepticus after IV injection of TXA in a paediatric non-cardiac surgery. This case report and literature review aim to increase awareness about TXA-associated seizures and to provide mechanistic-based prevention and treatment recommendations. During adenotonsillectomy for a 4-year-old male child, TXA infusion started after induction of anaesthesia for surgical bleeding prophylaxis. During recovery from anaesthesia, the patient developed tonic-clonic convulsions which did not improve after two IV doses of midazolam but showed an improvement after a dose of propofol. The patient did not regain consciousness and was transferred to the ICU. He had recurrent treatment-resistant attacks of tonic-clonic convulsions. The patient developed acute kidney injury and died after 18 hours. In high-risk patients, using the lowest effective dose with early termination of TXA infusion and prolongation of administration of anaesthetics may prevent seizures. General anaesthetics (propofol and halogenated inhaled anaesthetics) are considered the first line for prevention/treatment of TXA-associated seizures.


Subject(s)
Antifibrinolytic Agents , Propofol , Status Epilepticus , Tranexamic Acid , Antifibrinolytic Agents/therapeutic use , Blood Loss, Surgical/prevention & control , Child , Child, Preschool , Humans , Male , Seizures/chemically induced , Seizures/drug therapy , Seizures/prevention & control , Status Epilepticus/chemically induced , Status Epilepticus/complications , Status Epilepticus/drug therapy , Tranexamic Acid/therapeutic use
8.
Article in English | MEDLINE | ID: mdl-35131336

ABSTRACT

Autism spectrum disorder (ASD) is a neurodevelopmental disorder of heterogenous etiology exhibiting a challenge in understanding its exact neuro-pathophysiology. Recently, peroxisome proliferator activated receptor (PPAR)-α activation was found to play a fundamental role in neuroprotection and improving autistic-like-behaviors in experimental animal models of ASD through alleviating neuroinflammation, oxidative-stress, astrocyte reactivity, tauopathy in addition to its favorable role in metabolic regulation, thus attracting attention as a possible target in treatment of ASD. This study aimed to investigate the role of PPAR-α, astrocytic dysfunction and tauopathy in ASD and detect the possible neuroprotective effects of metformin (MET), through PPAR-α activation, and risperidone (RIS) either monotherapy or in combination in alleviating autistic-like-changes at behavioral and neurobiological levels in male Wistar rats. Pregnant female Wistar rats received valproic-acid (VPA) to induce autistic-like-behavioral and neurobiological alterations in their offspring. Chronic intra-peritoneal MET (100 mg/kg/day) and RIS (1 mg/kg/day) either monotherapy or in combination started from postnatal day (PND) 24 till PND61 (38 days). Prenatal VPA exposure simulated the autistic core behaviors associated with neurochemical and histopathological neurodevelopmental degenerative changes. Both MET and RIS either monotherapy or in combination were able to reverse these changes. The effect of MET was comparable to RIS. Moreover, MET was able to alleviate the RIS induced weight gain and improve cognitive functions highlighting its promising adjunctive role in alleviating ASD pathophysiology. Our study highlighted the favorable effects of MET and RIS both in monotherapy and in combination in alleviating the autistic-like-changes and proposed PPAR-α activation along with restoring astrocytes homeostasis as promising targets in novel therapeutic strategies in ASD.


Subject(s)
Autism Spectrum Disorder , Autistic Disorder , Metformin , Prenatal Exposure Delayed Effects , Tauopathies , Animals , Autism Spectrum Disorder/chemically induced , Autism Spectrum Disorder/drug therapy , Autism Spectrum Disorder/metabolism , Autistic Disorder/chemically induced , Behavior, Animal , Disease Models, Animal , Female , Male , Metformin/adverse effects , Peroxisome Proliferator-Activated Receptors/adverse effects , Pregnancy , Rats , Rats, Wistar , Risperidone/pharmacology , Risperidone/therapeutic use , Valproic Acid/pharmacology
9.
Neurotoxicology ; 83: 77-88, 2021 03.
Article in English | MEDLINE | ID: mdl-33417987

ABSTRACT

Low-dose repeated lipopolysaccharide pre-challenge followed by chronic mild stress (LPS/CMS) protocol has been introduced as a rodent model of depression combining the roles of immune activation and chronic psychological stress. However, the impact of this paradigm on cognitive functioning has not been investigated hitherto. METHODS: This study evaluated LPS/CMS-induced cognitive effects and the role of glycogen synthase kinase-3ß (GSK-3ß) activation with subsequent neuroinflammation and pathological tau deposition in the pathogenesis of these effects using lithium (Li) as a tool for GSK-3 inhibition. RESULTS: LPS pre-challenge reduced CMS-induced neuroinflammation, depressive-like behavior and cognitive inflexibility. It also improved spatial learning but increased GSK-3ß expression and exaggerated hyperphosphorylated tau accumulation in hippocampus and prefrontal cortex. Li ameliorated CMS and LPS/CMS-induced depressive and cognitive deficits, reduced GSK-3ß over-expression and tau hyperphosphorylation, impeded neuroinflammation and enhanced neuronal survival. CONCLUSION: This study draws attention to LPS/CMS-triggered cognitive changes and highlights how prior low-dose immune challenge could develop an adaptive capacity to buffer inflammatory damage and maintain the cognitive abilities necessary to withstand threats. This work also underscores the favorable effect of Li (as a GSK-3ß inhibitor) in impeding exaggerated tauopathy and neuroinflammation, rescuing neuronal survival and preserving cognitive functions. Yet, further in-depth studies utilizing different low-dose LPS challenge schedules are needed to elucidate the complex interactions between immune activation and chronic stress exposure.


Subject(s)
Behavior, Animal/drug effects , Cerebral Cortex/drug effects , Cognition/drug effects , Cognitive Dysfunction/prevention & control , Depression/prevention & control , Encephalitis/prevention & control , Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Hippocampus/drug effects , Lithium Chloride/pharmacology , Protein Kinase Inhibitors/pharmacology , Tauopathies/prevention & control , Animals , Cerebral Cortex/enzymology , Cerebral Cortex/physiopathology , Chronic Disease , Cognitive Dysfunction/enzymology , Cognitive Dysfunction/etiology , Cognitive Dysfunction/physiopathology , Depression/enzymology , Depression/etiology , Depression/physiopathology , Disease Models, Animal , Encephalitis/enzymology , Encephalitis/etiology , Encephalitis/physiopathology , Glycogen Synthase Kinase 3 beta/metabolism , Hippocampus/enzymology , Hippocampus/physiopathology , Inflammation Mediators/metabolism , Lipopolysaccharides , Male , Phosphorylation , Rats, Wistar , Spatial Learning/drug effects , Stress, Psychological/complications , Stress, Psychological/psychology , Tauopathies/enzymology , Tauopathies/etiology , Tauopathies/physiopathology , tau Proteins/metabolism
10.
Article in English | MEDLINE | ID: mdl-33238165

ABSTRACT

Despite the increasing prevalence of autism spectrum disorder (ASD), there is still a deficiency in understanding its exact pathophysiology and treatment, therefore validation of translational ASD animal model is warranted. Although strong evidences support the valproic acid (VPA) model of autism, yet a controversy exists regarding the best timing of exposure whether prenatal or postnatal. Accordingly, this study was designed to compare the time dependent effects of VPA exposure as regard its ability to induce autistic like changes in male Wistar rats. In this study, two different protocols of VPA exposure (prenatal and postnatal) were compared at different levels (behavioral, neurochemical and histopathological). Results of this study revealed that both prenatal and postnatal VPA exposures induced autistic-like behaviors manifested by reduced social interaction, increased repetitive stereotyped behavior and anxiety, cognitive dysfunction, lowered sensitivity to pain, and neurodevelopmental delay. Furthermore, inflammatory cytokines and oxidative/nitrosative stress markers were elevated in prefrontal cortex and hippocampal homogenates. Likewise, histopathological and immunohistochemical assessment confirmed the neurodegenerative and the apoptotic changes in prefrontal cortex, hippocampus and cerebellum exhibited by decreased viable cells number and Nissl's granules optical density, and increased caspase-3 immunoreactivity respectively. Interestingly, ASD core symptoms and histopathological changes were significantly (P < 0.05) altered in prenatal VPA model compared to postnatal VPA model. Additionally, postnatal mortality in prenatal model (4.3%) was much lower compared to the postnatal model (22.7%). In conclusion, our study overweighs the ability of prenatal VPA model over postnatal VPA model to induce behavioral and neuropathological alterations that simulate those observed in autistic individuals with a lower postnatal animal mortality, highlighting the privilege of prenatal over postnatal VPA exposure as a translational model for understanding pathophysiology and developing novel targets for management of ASD.


Subject(s)
Autistic Disorder/chemically induced , Valproic Acid/pharmacology , Animals , Animals, Newborn , Apoptosis/drug effects , Autistic Disorder/pathology , Autistic Disorder/psychology , Brain Chemistry/drug effects , Cytokines/analysis , Disease Models, Animal , Female , Male , Morris Water Maze Test , Open Field Test , Oxidative Stress/drug effects , Pregnancy , Prenatal Exposure Delayed Effects/chemically induced , Rats , Rats, Wistar , Reproducibility of Results , Valproic Acid/administration & dosage
11.
Eur J Pharmacol ; 885: 173411, 2020 Oct 15.
Article in English | MEDLINE | ID: mdl-32800810

ABSTRACT

Accumulating evidence indicates the role of microglial activation and sustained neuroinflammation in the pathogenesis of cognitive dysfunction, a common feature associated with depressive disorders. It also indicates the role of Wnt/ß-catenin pathway in regulation of microglia-mediated neuroinflammation. Amisulpride exhibits antidepressant and pro-cognitive activities in several clinical and experimental studies. Hitherto, the direct effects of amisulpride on Wnt/ß-catenin signaling and microglial activity have not been thoroughly studied. This study aimed at investigating the effects of chronic amisulpride treatment on Wnt/ß-catenin signaling and pro-inflammatory microglial activation and its role in alleviation of depressive-like behavior and cognitive deficits elicited by unpredictable chronic mild stress (UCMS). The effects of amisulpride (3 mg/kg/day) were investigated on behavioral/cognitive deficits, expression of Wnt/ß-catenin pathway and microglial activation in the prefrontal cortex (PFC) of UCMS-exposed male Wistar rats. UCMS induced depressive-like behavior with impairment of performance in novel object recognition test and attentional set-shifting task. These behavioral deficits were associated with decreased total ß-catenin and increased pro-inflammatory microglial activation. Amisulpride improved UCMS-induced behavioral/cognitive deficits, ameliorated Wnt/ß-catenin signaling dysregulation and pro-inflammatory microglial activation. This work highlights the antidepressant and pro-cognitive effects of amisulpride in UCMS-exposed rats that could be mediated by modulation of Wnt/ß-catenin pathway activity and amelioration of pro-inflammatory microglial activation in the prefrontal cortex. This could provide new insights into the putative mechanisms behind the antidepressant and pro-cognitive effects exerted by amisulpride.


Subject(s)
Amisulpride/therapeutic use , Cognition Disorders/drug therapy , Microglia/drug effects , Nootropic Agents/therapeutic use , Prefrontal Cortex/drug effects , Stress, Psychological/psychology , Wnt Signaling Pathway/drug effects , Animals , Attention/drug effects , Behavior, Animal/drug effects , Cognition Disorders/etiology , Depression/prevention & control , Depression/psychology , Encephalitis/drug therapy , Encephalitis/psychology , Macrophage Activation , Male , Motor Activity/drug effects , Rats , Rats, Wistar , Recognition, Psychology/drug effects , Stress, Psychological/drug therapy , beta Catenin/antagonists & inhibitors , beta Catenin/biosynthesis
12.
Toxicol Appl Pharmacol ; 399: 115073, 2020 07 15.
Article in English | MEDLINE | ID: mdl-32454056

ABSTRACT

Microglial in vivo production of pro-inflammatory cytokines is central to the pathogenesis of multiple neurological disorders including depression, with a rising role of Wnt/ß-catenin signaling as potential regulator of microglia-mediated neuro-inflammation. This study aimed at investigating the hippocampal expression of the Wnt/ß-catenin pathway in chronic mild stress (CMS)-exposed rats and the effects of Lithium (Li) on the expression of this pathway as a method to identify a plausible link between exposure to chronic stress, microglial activation, and neuroinflammation. METHODS: The effect of chronic administration of Li was investigated on behavioral changes, hippocampal expression of Wnt-DVL-GSK3ß-ß-catenin signaling pathway, and microglial activation in CMS-exposed male Wistar rats RESULTS: CMS induced a depressive-like behavior associated with increased pro-inflammatory microglial activation and reduced hippocampal expression of the Wnt/ß-catenin signaling pathway. Chronic Li treatment ameliorated stress induced-behavioral changes, reduced microglial activation and enhanced the hippocampal expression of Wnt/ß-catenin signaling pathway. CONCLUSION: This work highlights that Li-induced inhibition of GSK-3ß with subsequent accumulation of ß-catenin could impede pro-inflammatory microglia activation which is a key pathological hallmark associated with depression. Wnt/ß-catenin signaling represents a promising therapeutic target, not only for alleviation of depression, but also for a wide array of neurological disorders.


Subject(s)
Cytokines/metabolism , Inflammation Mediators/metabolism , Lithium/pharmacology , Stress, Physiological/drug effects , Wnt Signaling Pathway/drug effects , beta Catenin/metabolism , Animals , Glycogen Synthase Kinase 3 beta/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Male , Microglia/drug effects , Microglia/metabolism , Rats , Rats, Wistar
13.
Naunyn Schmiedebergs Arch Pharmacol ; 392(6): 669-683, 2019 06.
Article in English | MEDLINE | ID: mdl-30739140

ABSTRACT

Depression and cardiovascular disease (CVD) are two faces of one coin. A pro-inflammatory state was previously suggested in the pathology of both diseases. We investigated the effect of chronic administration (2 weeks) of imipramine (20 mg/kg/day) and pentoxifylline (50 mg/kg/day) on behavioral, aortic histological abnormalities, and level of circulating endothelial progenitor cells (CEPCs) in peripheral blood of male Wistar rats exposed to chronic mild stress (CMS) and high-fat diet. Exposure to CMS and high-fat diet induced a depressive-like behavior alongside aortic immunohistochemical changes associated with an increase in aortic TNF-α level. Markers of CEPCs, VEGFR-2 and CD133, were significantly disturbed in aortic sections, as compared to control animals and those exposed to CMS while fed high-fat diet, although flowcytometric analysis did not show any significant changes in the level of CEPCs in peripheral blood. Chronic pentoxifylline treatment was more effective in ameliorating the histological changes and endothelial damage compared to imipramine. Pro-inflammatory cytokines-induced disturbances in CEPCs could constitute a plausible link between depression and atherosclerotic cardiovascular disease. Current antidepressants reduce symptoms of depression without tackling the underlying link between it and cardiovascular disease. Targeting pro-inflammatory cytokines might open a new therapeutic approach to alleviate depression and reduce the risk of mortality from cardiovascular disease.


Subject(s)
Aorta, Thoracic/drug effects , Diet, High-Fat , Endothelial Progenitor Cells/physiology , Pentoxifylline/therapeutic use , Stress, Psychological/complications , Vascular Diseases/drug therapy , AC133 Antigen/analysis , Animals , Aorta, Thoracic/chemistry , Aorta, Thoracic/pathology , Body Weight/drug effects , Chronic Disease , Flow Cytometry , Male , Pentoxifylline/pharmacology , Rats , Rats, Wistar , Vascular Endothelial Growth Factor Receptor-2/analysis
14.
Behav Pharmacol ; 29(5): 413-425, 2018 08.
Article in English | MEDLINE | ID: mdl-29561292

ABSTRACT

Depression is the disease of the modern era. The lack of response to the available antidepressants, which were developed on the basis of the monoaminergic deficit hypothesis of depression, has encouraged scientists to think about new mechanisms explaining the pathogenesis of depression. In this context, the inflammatory theory has emerged to clarify many aspects of depression that the previous theories have failed to explain. Toll-like receptor-4 (TLR-4) has a regulatory role in the brain's immune response to stress, and its activation is suggested to play a pivotal role in the pathophysiology of depression. In this study, we tested eritoran (ERI), a TLR-4 receptor-4 antagonist, as a potential antidepressant. We investigated the effect of long-term administration of ERI in three different doses on behavioral changes, hippocampal and prefrontal cortex (PFC) neurogenesis, and γ-aminobutyric acid (GABA)/glutamate balance in male Wistar rats exposed to chronic restraint stress (CRS). Long-term administration of ERI ameliorated CRS-induced depressive-like symptoms and hypothalamic-pituitary-adrenal axis hyperactivity alongside reducing levels of hippocampal and PFC inflammatory cytokines, restoring GABA and glutamate balance, and enhancing PFC and hippocampal neurogenesis, by increasing BDNF gene and protein expression in a dose-dependent manner. The results demonstrate an antidepressant-like activity of ERI in Wistar rats exposed to CRS, which may be largely mediated by its ability to reduce neuroinflammation, increase BDNF, and restore GABA/glutamate balance in prefrontal cortex and hippocampus. Nonetheless, further studies are needed to characterize the mechanism of the antidepressant effect of ERI.


Subject(s)
Depression/drug therapy , Disaccharides/pharmacology , Sugar Phosphates/pharmacology , Animals , Antidepressive Agents/pharmacology , Brain-Derived Neurotrophic Factor/metabolism , Depression/etiology , Depressive Disorder/physiopathology , Disaccharides/metabolism , Disease Models, Animal , Glutamic Acid/drug effects , Hippocampus/drug effects , Hypothalamo-Hypophyseal System/drug effects , Male , Neurogenesis/drug effects , Pituitary-Adrenal System/drug effects , Prefrontal Cortex/drug effects , Rats , Rats, Wistar , Stress, Psychological/physiopathology , Sugar Phosphates/metabolism , Toll-Like Receptor 4/antagonists & inhibitors , Toll-Like Receptor 4/drug effects , Toll-Like Receptor 4/metabolism , gamma-Aminobutyric Acid/drug effects
15.
Naunyn Schmiedebergs Arch Pharmacol ; 391(4): 407-422, 2018 04.
Article in English | MEDLINE | ID: mdl-29379991

ABSTRACT

Several hypotheses link high fat diet (HFD) with the pathophysiology of depression and its response to antidepressants. This study aimed to determine the effect of metformin (MET) on the cognitive and antidepressant activity of fluoxetine (FLU) through its effect on c-Jun expression. Behavioral, cognitive function, biochemical, and histopathological studies were performed in non-HFD- and HFD-fed rats exposed to chronic restraint stress (CRS). Stressed group showed cognitive impairment, depressive-like symptoms, disturbed glucose homeostasis and lipid profile, reduced adiponectin level, brain-derived neurotrophic factor (BDNF) expression, and increased corticosterone and c-Jun. All these were aggravated by HFD. MET, FLU and their combination produced significant improvement in lipid profile with significant increase in adiponectin and BDNF expression. Corticosterone, body weight and insulin resistance showed significant decrease in the treated groups. Moreover, there was a significant decrease in hippocampal c Jun expression. There was a significant preferable effect toward the combination. Conclusion, MET may decrease the refractoriness to FLU and improves the cognition in individuals who are fed on HFD.


Subject(s)
Antidepressive Agents/pharmacology , Fluoxetine/pharmacology , Hypoglycemic Agents/pharmacology , Metformin/pharmacology , Stress, Psychological/metabolism , Stress, Psychological/psychology , Animals , Behavior, Animal/drug effects , Brain-Derived Neurotrophic Factor/genetics , Cognition/drug effects , Diet, High-Fat , Drug Synergism , Hippocampus/drug effects , Hippocampus/metabolism , Male , Proto-Oncogene Proteins c-jun/metabolism , Rats, Wistar , Restraint, Physical
16.
Biomed Pharmacother ; 85: 225-238, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27916420

ABSTRACT

BACKGROUND: Depression and non-alcoholic steatohepatitis (NASH) are highly co-morbid, and hepatic JNK pathway may be involved in their relation. AIM: To evaluate the impact of depression on NASH through the involvement of JNK1 and to assess the effect of sitagliptin and metformin on hepatic JNK1 expression in both NASH and NASH associated with depression. METHODS: Eight groups of male Wistar rats were used: naïve rats, non-stressed NASH, non-stressed NASH sitagliptin treated, non-stressed NASH metformin treated, stressed, stressed NASH untreated, stressed NASH sitagliptin treated and stressed NASH metformin treated. Behavioral, biochemical, molecular and histopathological studies were performed. RESULTS: Non-stressed NASH group showed depressive like symptoms, disturbed glucose homeostasis, impairment of liver functions, decrease adiponectin and increase malondialdehyde, which were aggreviated by stress. Sitagliptin produced significant improvement compared to metformin regarding biochemical and histopathological parameters. Furthermore, sitagliptin significantly decreased expression of hepatic JNK1 in both stressed and non-stressed rats. All these changes were accompanied by significant improvement of behavioral changes. CONCLUSIONS: The link between NASH and depression raised the role of JNK activation through increase expression of JNK1. Since sitagliptin was associated with preferable effects than metformin, therefore, it is potentially preferred in the management of either NASH or NASH associated with depression.


Subject(s)
Depression/enzymology , Diet, High-Fat , Liver/drug effects , Metformin/pharmacology , Mitogen-Activated Protein Kinase 8/metabolism , Non-alcoholic Fatty Liver Disease/drug therapy , Sitagliptin Phosphate/pharmacology , Stress, Psychological/enzymology , Animals , Behavior, Animal/drug effects , Biomarkers/blood , Chronic Disease , Depression/blood , Depression/psychology , Disease Models, Animal , Down-Regulation , Gene Expression Regulation, Enzymologic , Insulin Resistance , Liver/enzymology , Liver/ultrastructure , Male , Mitogen-Activated Protein Kinase 8/genetics , Non-alcoholic Fatty Liver Disease/blood , Non-alcoholic Fatty Liver Disease/chemically induced , Non-alcoholic Fatty Liver Disease/enzymology , Rats, Wistar , Signal Transduction/drug effects , Stress, Psychological/blood , Stress, Psychological/psychology , Time Factors
17.
PLoS One ; 10(3): e0120559, 2015.
Article in English | MEDLINE | ID: mdl-25826421

ABSTRACT

UNLABELLED: Several studies reveal that diabetes doubles the odds of comorbid depression with evidence of a pro-inflammatory state underlying its vascular complications. Indeed, little information is available about vascular effects of antidepressant drugs in diabetes. METHOD: We investigated the effect of chronic administration of fluoxetine "FLU" and imipramine "IMIP" on behavioral, metabolic and vascular abnormalities in diabetic and non-diabetic rats exposed to chronic restraint stress (CRS). RESULTS: Both diabetes and CRS induced depressive-like behavior which was more prominent in diabetic/depressed rats; this was reversed by chronic treatment with FLU and IMIP in a comparable manner. Diabetic and non-diabetic rats exposed to CRS exhibited abnormalities in glucose homeostasis, lipid profile and vascular function, manifested by decreased endothelium-dependent relaxation, increased systolic blood pressure and histopathological atherosclerotic changes. Vascular and metabolic dysfunctions were associated with significant increase in aortic expression of TLR-4, and pro-inflammatory cytokines (TNF-α and IL-1ß). FLU ameliorated these metabolic, vascular and inflammatory abnormalities, while IMIP induced either no change or even worsening of some parameters. CONCLUSION: FLU has favorable effect over IMIP on metabolic, vascular and inflammatory aberrations associated with DM and CRS in Wistar rats, clarifying the preference of FLU over IMIP in management of comorbid depression in diabetic subjects.


Subject(s)
Antidepressive Agents/pharmacology , Blood Vessels/drug effects , Diabetes Mellitus, Experimental/metabolism , Fluoxetine/pharmacology , Imipramine/pharmacology , Stress, Physiological , Toll-Like Receptor 4/metabolism , Animals , Behavior, Animal/drug effects , Blood Glucose/metabolism , Blood Pressure/drug effects , Blood Vessels/metabolism , Body Weight/drug effects , Chronic Disease , Insulin/blood , Lipids/blood , Male , Rats , Rats, Wistar
18.
Psychopharmacology (Berl) ; 232(6): 1095-105, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25315361

ABSTRACT

Several studies have pointed to the nicotinic acetylcholine receptor (nAChR) antagonists, such as mecamylamine (MEC), as a potential therapeutic target for the treatment of depression. The present study evaluated the behavioral and neurochemical effects of chronic administration of MEC (1, 2, and 4 mg/kg/day, intraperitoneally (i.p.)) in Wistar rats exposed to chronic restraint stress (CRS, 4 h × 6 W). MEC prevented CRS-induced depressive-like behavior via increasing sucrose preference, body weight, and forced swim test (FST) struggling and swimming while reducing immobility in FST and hypothalamic-pituitary-adrenal (HPA) axis hyperactivity (adrenal gland weight and serum corticosterone). At the same time, MEC amended CRS-induced anxiety as indicated by decreasing central zone duration in open field test and increasing active interaction duration. Additionally, MEC modulated the prefrontal cortex (PFC) level of brain-derived neurotrophic factor (BDNF), 5-hydroxy tryptamine (5-HT), and norepinephrine (NE). In conclusion, the present data suggest that MEC possesses antidepressant and anxiolytic-like activities in rats exposed to CRS. These behavioral effects may be in part mediated by reducing HPA axis hyperactivity and increasing PFC level of BDNF and monoamines. Accordingly, these findings further support the hypothesis that nAChRs blockade might afford a novel promising strategy for pharmacotherapy of depression.


Subject(s)
Behavior, Animal/drug effects , Brain-Derived Neurotrophic Factor/metabolism , Depressive Disorder/metabolism , Mecamylamine/pharmacology , Nicotinic Antagonists/pharmacology , Norepinephrine/metabolism , Prefrontal Cortex/drug effects , Serotonin/metabolism , Animals , Body Weight/drug effects , Corticosterone , Disease Models, Animal , Hypothalamo-Hypophyseal System/drug effects , Hypothalamo-Hypophyseal System/metabolism , Pituitary-Adrenal System/drug effects , Pituitary-Adrenal System/metabolism , Prefrontal Cortex/metabolism , Rats , Rats, Wistar , Restraint, Physical , Stress, Psychological/metabolism , Swimming
19.
Pharmacol Biochem Behav ; 126: 152-62, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25268312

ABSTRACT

OBJECTIVES: The present study examined the effect of combined exposure to repeated challenge using low doses of lipopolysaccharide (LPS) and chronic mild stress (CMS) together. This combined exposure is thought to expose the animals to more realistic challenges, testable on different levels (behavioral, neurochemical, immunohistochemical and gene expression). The role of glial cells was examined, as well. Additionally, the effects of chronic administration of the tricyclic antidepressant imipramine and the anti-TNF-α pentoxyphylline were investigated. METHODS: Wistar rats were exposed to either repeated LPS (50µg/kg i.p.) over 2weeks, CMS protocol for 4weeks or LPS over 2weeks then 4weeks of CMS. Two groups of rats were exposed to LPS/CMS protocol and treated with either imipramine or pentoxifylline. Rats were examined for behavioral, neurochemical and gene expression changes. RESULTS: Animals exposed to LPS/CMS elaborated depressive-like symptoms with significant increase in both serum corticosterone and TNF-α levels compared to those in the saline, LPS or CMS groups. Hippocampal kynurenine/tryptophan ratio and TNF-α gene expression showed significant increase in the LPS/CMS model compared to those in saline, LPS or CMS groups. The immunohistochemical findings scrutinized the topography of the examined effects. Chronic treatment with imipramine or pentoxifylline significantly ameliorated the behavioral, neurochemical, immunohistochemical and TNF-α gene expression changes induced by the LPS/CMS protocol. CONCLUSION: This study gave a clue to the neurobiological processes underlying, at least, the subtypes of depressive disorders. It highlighted the possible interactions between stress and immune-inflammatory pathways in the pathogenesis of depression and suggested a new animal model of depression that addresses these pathways.


Subject(s)
Antidepressive Agents, Tricyclic/therapeutic use , Behavior, Animal/drug effects , Depression/drug therapy , Imipramine/therapeutic use , Lipopolysaccharides/adverse effects , Pentoxifylline/therapeutic use , Stress, Psychological/drug therapy , Animals , Antidepressive Agents, Tricyclic/pharmacology , Corticosterone/blood , Depression/complications , Disease Models, Animal , Gene Expression/drug effects , Hippocampus/metabolism , Imipramine/pharmacology , Kynurenine/metabolism , Male , Pentoxifylline/pharmacology , Rats , Stress, Psychological/complications , Tryptophan/metabolism , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/blood
20.
Can J Physiol Pharmacol ; 92(5): 375-85, 2014 May.
Article in English | MEDLINE | ID: mdl-24754268

ABSTRACT

There is accumulating evidence suggesting that depression is a risk factor for cardiovascular diseases. This study aimed to examine the hypothesis that the proinflammatory cytokine TNF-α would partially explain the link between depression and atherosclerotic endothelial changes. Rats were distributed among 6 groups: (i) control group; (ii) group subjected to chronic mild stress (CMS); (iii) group fed a cholesterol-cholic acid-thiouracil (CCT diet); and (iv) CMS group fed the CCT diet and treated with the vehicle for 8 weeks. The last 2 groups were subjected to CMS-CCT and received thalidomide (THAL) or imipramine (IMIP). Rats were assessed behaviorally (sucrose preference, open field, and forced-swimming tests). TNF-α protein was assessed from the serum, aorta, and liver. Aortic TNF-α gene expression (assessed using RT-PCR), serum lipid profile, and insulin levels were measured. Endothelial function was assessed in isolated aortic rings. The THAL and IMIP groups showed ameliorated CMS-CCT-related behavioral changes. CMS-CCT-induced metabolic and endothelial dysfunctions were improved in the THAL group but were worsened in the IMIP group. RT-PCR showed a significant reduction of aortic TNF-α mRNA expression in the THAL and IMIP treatment groups. These data paralleled the findings for aortic immunohistochemistry. The THAL group, but not the IMIP group, showed improved CMS-CCT-induced changes in the vascular reactivity of the aortic rings. Thus, TNF-α provides a target link between depression, metabolic syndrome, and endothelial dysfunction. This could open a new therapeutic approach to address the comorbidities of depression.


Subject(s)
Diet, Atherogenic/adverse effects , Endothelium, Vascular/drug effects , Immunosuppressive Agents/pharmacology , Stress, Psychological/metabolism , Stress, Psychological/psychology , Thalidomide/pharmacology , Tumor Necrosis Factor-alpha/metabolism , Animals , Antidepressive Agents, Tricyclic/pharmacology , Aorta, Thoracic/metabolism , Aorta, Thoracic/physiopathology , Atherosclerosis/metabolism , Atherosclerosis/pathology , Chronic Disease , Depression/metabolism , Depression/psychology , Endothelium, Vascular/metabolism , Endothelium, Vascular/physiopathology , Imipramine/pharmacology , Insulin Resistance , Male , Muscle Contraction/drug effects , Muscle, Smooth, Vascular/physiopathology , Nitric Oxide/metabolism , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...