Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 474
Filter
Add more filters

Complementary Medicines
Country/Region as subject
Publication year range
1.
Ren Fail ; 43(1): 1496-1505, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34751624

ABSTRACT

In this study, we explored the role and mechanism of repulsive guidance molecule B (RGMb, also known as Dragon) in the protective effects of curcumin against renal fibrosis and verified Dragon's effect on renal tubular epithelial cell apoptosis and cell programmability. Unilateral ureteral obstruction (UUO) was surgically induced in rats to establish a model of renal interstitial fibrosis (RIF). The rats were then treated with curcumin. Curcumin prominently decreased the serum creatinine (SCr) and blood urea nitrogen (BUN) levels, and also improved the tubular injury in the UUO-induced rats. Curcumin significantly downregulated the TGF-ß1, P-Smad2/3, cleaved caspase-3, cleaved caspase-8 and Dragon levels. Dragon knockdown also markedly reduced the TGF-ß1, P-Smad2/3, Smad2/3, cleaved caspase-3, cleaved caspase-8, fibronectin, collagen I, collagen IV, vimentin, and α-SMA expression levels. Conversely, Dragon overexpression caused higher expression levels of these proteins, and curcumin reversed this effect. Furthermore, Dragon knockdown increased the E-cadherin levels, whereas Dragon overexpression decreased these levels. Overexpressing Dragon significantly decreased the cell viability, and curcumin reversed this effect. In conclusion, curcumin acted on Dragon and attenuated RIF in UUO rat models. Curcumin downregulated the TGF-ß1/Smad signaling pathway and inhibited Dragon and fibrogenic molecules in both rats and HK-2 cells.


Subject(s)
Curcumin/pharmacology , Fibrosis/drug therapy , GPI-Linked Proteins/biosynthesis , Kidney/drug effects , Nerve Tissue Proteins/biosynthesis , Receptors, Cell Surface/biosynthesis , Ureteral Obstruction/drug therapy , Animals , Blood Urea Nitrogen , Caspase 3/metabolism , Creatinine/metabolism , Fibrosis/metabolism , Fibrosis/pathology , GPI-Linked Proteins/drug effects , Humans , Kidney/metabolism , Kidney/pathology , Male , Nerve Tissue Proteins/drug effects , Rats , Rats, Sprague-Dawley , Receptors, Cell Surface/drug effects , Signal Transduction/drug effects , Transforming Growth Factor beta1/metabolism , Ureteral Obstruction/metabolism
2.
Sci Rep ; 11(1): 17747, 2021 09 07.
Article in English | MEDLINE | ID: mdl-34493757

ABSTRACT

Deregulation of synaptic function and neurotransmission has been linked with the development of major depression disorder (MDD). Tianeptine (Tian) has been used as antidepressant with anxiolytic properties and recently as a nootropic to improve cognitive performance, but its mechanism of action is unknown. We conducted a proteomic study on the hippocampal synaptosomal fractions of adult male Wistar rats exposed to chronic social isolation (CSIS, 6 weeks), an animal model of depression and after chronic Tian treatment in controls (nootropic effect) and CSIS-exposed rats (lasting 3 weeks of 6-week CSIS) (therapeutic effect). Increased expression of Syn1 and Camk2-related neurotransmission, vesicle transport and energy processes in Tian-treated controls were found. CSIS led to upregulation of proteins associated with actin cytoskeleton, signaling transduction and glucose metabolism. In CSIS rats, Tian up-regulated proteins involved in mitochondrial energy production, mitochondrial transport and dynamics, antioxidative defense and glutamate clearance, while attenuating the CSIS-increased glycolytic pathway and cytoskeleton organization proteins expression and decreased the expression of proteins involved in V-ATPase and vesicle endocytosis. Our overall findings revealed that synaptic vesicle dynamics, specifically exocytosis, and mitochondria-related energy processes might be key biological pathways modulated by the effective nootropic and antidepressant treatment with Tian and be a potential target for therapeutic efficacy of the stress-related mood disorders.


Subject(s)
Antidepressive Agents/pharmacology , Depressive Disorder/drug therapy , Mitochondria/drug effects , Nootropic Agents/pharmacology , Proteome/drug effects , Social Isolation , Synaptic Vesicles/drug effects , Thiazepines/pharmacology , Animals , Anti-Anxiety Agents/pharmacology , Anti-Anxiety Agents/therapeutic use , Antidepressive Agents/therapeutic use , Depressive Disorder/physiopathology , Disease Models, Animal , Drug Evaluation, Preclinical , Gene Expression Regulation/drug effects , Hippocampus/drug effects , Hippocampus/ultrastructure , Male , Mitochondria/physiology , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Nootropic Agents/therapeutic use , Protein Interaction Mapping , Rats , Rats, Wistar , Signal Transduction/drug effects , Thiazepines/therapeutic use
3.
Mol Neurobiol ; 58(11): 5473-5493, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34338970

ABSTRACT

Inflammation, demyelination, glial activation, and oxidative damage are the most pathological hallmarks of multiple sclerosis (MS). Piperine, a main bioactive alkaloid of black pepper, possesses antioxidant, anti-inflammatory, and neuroprotective properties whose therapeutic potential has been less studied in the experimental autoimmune encephalomyelitis (EAE) models. In this study, the efficiency of piperine on progression of EAE model and myelin repair mechanisms was investigated. EAE was induced in female Lewis rats and piperine and its vehicle were daily administrated intraperitoneally from day 8 to 29 post immunization. We found that piperine alleviated neurological deficits and EAE disease progression. Luxol fast blue and H&E staining and immunostaining of lumbar spinal cord cross sections confirmed that piperine significantly reduced the extent of demyelination, inflammation, immune cell infiltration, microglia, and astrocyte activation. Gene expression analysis in lumbar spinal cord showed that piperine treatment decreased the level of pro-inflammatory cytokines (TNF-α, IL-1ß) and iNOS and enhanced IL-10, Nrf2, HO-1, and MBP expressions. Piperine supplementation also enhanced the total antioxidant capacity (FRAP) and reduced the level of oxidative stress marker (MDA) in the CNS of EAE rats. Finally, we found that piperine has anti-apoptotic and neuroprotective effect in EAE through reducing caspase-3 (apoptosis marker) and enhancing BDNF and NeuN expressing cells. This study strongly indicates that piperine has a beneficial effect on the EAE progression and could be considered as a potential therapeutic target for MS treatment. Upcoming clinical trials will provide a deeper understanding of piperine's role for the treatment of the MS.


Subject(s)
Alkaloids/therapeutic use , Anti-Inflammatory Agents/therapeutic use , Antioxidants/therapeutic use , Benzodioxoles/therapeutic use , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Neuroprotective Agents/therapeutic use , Piperidines/therapeutic use , Polyunsaturated Alkamides/therapeutic use , Alkaloids/pharmacology , Animals , Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Apoptosis/drug effects , Astrocytes/metabolism , Benzodioxoles/pharmacology , Caspase 3/biosynthesis , Caspase 3/genetics , Cytokines/biosynthesis , Cytokines/genetics , Disease Progression , Drug Evaluation, Preclinical , Encephalomyelitis, Autoimmune, Experimental/metabolism , Female , Gene Expression Regulation/drug effects , Microglia/metabolism , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Piperidines/pharmacology , Polyunsaturated Alkamides/pharmacology , Random Allocation , Rats , Rats, Inbred Lew
4.
Sci Rep ; 11(1): 10973, 2021 05 26.
Article in English | MEDLINE | ID: mdl-34040073

ABSTRACT

Manual acupuncture (MA) can be used to manage high blood pressure; however, the underlying molecular mechanism remains unknown. To explore the mechanism of acupuncture in the treatment of hypertension, Wistar Kyoto rats (WKYs) and spontaneously hypertensive rats (SHRs) were subjected to either MA stimulation or the corresponding sham procedure as a negative control (Sham-MA) for 1 week. PET-CT scans, transcriptomics and molecular biology were used to evaluate the effect of MA. The results show that MA can regulate blood pressure in SHRs, change the glucose metabolism of the paraventricular hypothalamus (PVH), and affect the mRNA and protein expression levels of differentially expressed genes in the PVH. These genes may lower blood pressure by regulating angiotensin, endothelial function and inflammation. These findings reveal that MA regulates multiple biological processes and genes/proteins of the PVH, and provide a solid theoretical basis for exploring the mechanisms by which MA regulates hypertension.


Subject(s)
Acupuncture Therapy , Essential Hypertension/therapy , Gene Expression Regulation , Molecular Targeted Therapy , Paraventricular Hypothalamic Nucleus/metabolism , Positron Emission Tomography Computed Tomography , Sequence Analysis, RNA , Animals , Blood Pressure/physiology , Blotting, Western , Essential Hypertension/diagnostic imaging , Essential Hypertension/genetics , Gene Ontology , Gene Regulatory Networks , Glucose/metabolism , Male , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Paraventricular Hypothalamic Nucleus/diagnostic imaging , RNA, Messenger/analysis , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Real-Time Polymerase Chain Reaction , Transcriptome
5.
Int J Mol Sci ; 21(18)2020 Sep 16.
Article in English | MEDLINE | ID: mdl-32948011

ABSTRACT

Hypoxic-ischemic encephalopathy (HIE) is still a major cause of neonatal death and disability as therapeutic hypothermia (TH) alone cannot afford sufficient neuroprotection. The present study investigated whether ventilation with molecular hydrogen (2.1% H2) or graded restoration of normocapnia with CO2 for 4 h after asphyxia would augment the neuroprotective effect of TH in a subacute (48 h) HIE piglet model. Piglets were randomized to untreated naïve, control-normothermia, asphyxia-normothermia (20-min 4%O2-20%CO2 ventilation; Tcore = 38.5 °C), asphyxia-hypothermia (A-HT, Tcore = 33.5 °C, 2-36 h post-asphyxia), A-HT + H2, or A-HT + CO2 treatment groups. Asphyxia elicited severe hypoxia (pO2 = 19 ± 5 mmHg) and mixed acidosis (pH = 6.79 ± 0.10). HIE development was confirmed by altered cerebral electrical activity and neuropathology. TH was significantly neuroprotective in the caudate nucleus but demonstrated virtually no such effect in the hippocampus. The mRNA levels of apoptosis-inducing factor and caspase-3 showed a ~10-fold increase in the A-HT group compared to naïve animals in the hippocampus but not in the caudate nucleus coinciding with the region-specific neuroprotective effect of TH. H2 or CO2 did not augment TH-induced neuroprotection in any brain areas; rather, CO2 even abolished the neuroprotective effect of TH in the caudate nucleus. In conclusion, the present findings do not support the use of these medical gases to supplement TH in HIE management.


Subject(s)
Asphyxia Neonatorum/therapy , Brain Damage, Chronic/prevention & control , Carbon Dioxide/therapeutic use , Hydrogen/therapeutic use , Hypothermia, Induced , Hypoxia-Ischemia, Brain/therapy , Neuroprotection/drug effects , Neuroprotective Agents/therapeutic use , Acidosis/blood , Acidosis/etiology , Acidosis/prevention & control , Administration, Inhalation , Animals , Animals, Newborn , Apoptosis Inducing Factor/biosynthesis , Apoptosis Inducing Factor/genetics , Asphyxia Neonatorum/complications , Asphyxia Neonatorum/drug therapy , Brain Damage, Chronic/etiology , Brain-Derived Neurotrophic Factor/biosynthesis , Brain-Derived Neurotrophic Factor/genetics , Carbon Dioxide/administration & dosage , Carbon Dioxide/toxicity , Caspase 3/biosynthesis , Caspase 3/genetics , Caudate Nucleus/pathology , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Disease Models, Animal , Drug Evaluation, Preclinical , Electroencephalography , Evoked Potentials, Visual/drug effects , Gene Expression Regulation/drug effects , Hippocampus/pathology , Hydrogen/administration & dosage , Hydrogen/analysis , Hypoxia-Ischemia, Brain/complications , Hypoxia-Ischemia, Brain/drug therapy , Hypoxia-Ischemia, Brain/pathology , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Neuroprotective Agents/administration & dosage , Organ Specificity , Random Allocation , Swine
6.
Neuromolecular Med ; 22(3): 341-358, 2020 09.
Article in English | MEDLINE | ID: mdl-32048142

ABSTRACT

Alzheimer's disease (AD) is a common neurodegenerative disease that is always accompanied by synaptic loss in the brain. Safflower yellow (SY) is the extract of safflower, a traditional Chinese medicine, which has shown neuroprotective effects in recent studies. However, the mechanism of SY in protecting synapses remains unclear. In this study, we are going to study the mechanism of how SY treats AD in terms of synaptic plasticity. We found, via behavioral experiments, that SY treatment could improve the abilities of learning and memory in APP/PS1 mice. In addition, using Golgi staining and HE staining, we found that SY treatment could reduce the loss of dendritic spines in the pathological condition and could maintain the normal physiological state of the cells in cortex and in hippocampus. In addition, the results of immunofluorescence staining and western blotting showed that SY treatment could significantly increase the expression of synapse-related proteins. Moreover, after being treated with SY, the expression of iNOS (marker of M1 microglia) declined remarkably, and the level of Arginase-1 (marker of M2 microglia) increased significantly. Finally, we found BDNF/TrkB/ERK signaling cascade was activated. These results indicate that SY enhances synaptic plasticity in APP/PS1 mice by regulating microglia activation phenotypes and BDNF/TrkB/ERK signaling pathway.


Subject(s)
Alzheimer Disease/drug therapy , Brain-Derived Neurotrophic Factor/physiology , Chalcone/analogs & derivatives , Drugs, Chinese Herbal/therapeutic use , MAP Kinase Signaling System/drug effects , Membrane Glycoproteins/physiology , Microglia/drug effects , Neuronal Plasticity/drug effects , Phytotherapy , Protein-Tyrosine Kinases/physiology , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , Animals , Arginase/biosynthesis , Arginase/genetics , Cerebral Cortex/chemistry , Cerebral Cortex/drug effects , Cerebral Cortex/pathology , Chalcone/therapeutic use , Dendritic Spines/drug effects , Dendritic Spines/ultrastructure , Disease Models, Animal , Donepezil/pharmacology , Donepezil/therapeutic use , Enzyme Induction/drug effects , Escape Reaction/drug effects , Female , Hippocampus/chemistry , Hippocampus/drug effects , Hippocampus/pathology , Male , Memory, Long-Term/drug effects , Memory, Short-Term/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microglia/physiology , Morris Water Maze Test/drug effects , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Neuronal Plasticity/physiology , Nitric Oxide Synthase Type II/biosynthesis , Nitric Oxide Synthase Type II/genetics , Presenilin-1/genetics , Random Allocation
7.
Sci Rep ; 10(1): 1143, 2020 01 24.
Article in English | MEDLINE | ID: mdl-31980673

ABSTRACT

Neurodegenerative diseases, including Alzheimer's and Parkinson's disease, are characterized by increased protein aggregation in the brain, progressive neuronal loss, increased inflammation, and neurogenesis impairment. We analyzed the effects of a new purine derivative drug, PDD005, in attenuating mechanisms involved in the pathogenesis of neurodegenerative diseases, using both in vivo and in vitro models. We show that PDD005 is distributed to the brain and can rescue cognitive deficits associated with aging in mice. Treatment with PDD005 prevents impairment of neurogenesis by increasing sex-determining region Y-box 2, nestin, and also enhances synaptic function through upregulation of synaptophysin and postsynaptic density protein 95. PDD005 treatment also reduced neuro-inflammation by decreasing interleukin-1ß expression, activation of astrocytes, and microglia. We identified prohibitin as a potential target in mediating the therapeutic effects of PDD005 for the treatment of cognitive deficit in aging mice. Additionally, in the current study, glycogen synthase kinase appears to attenuate tau pathology.


Subject(s)
Cognition Disorders/prevention & control , Hippocampus/drug effects , Molecular Targeted Therapy , Nerve Tissue Proteins/antagonists & inhibitors , Neuroprotective Agents/pharmacology , Repressor Proteins/antagonists & inhibitors , Tauopathies/prevention & control , Aging/psychology , Animals , Blood-Brain Barrier , Brain/metabolism , Cells, Cultured , Cognition Disorders/drug therapy , Donepezil/pharmacology , Drug Evaluation, Preclinical , Endothelial Cells/drug effects , Gene Expression Regulation/drug effects , Glycogen Synthase Kinase 3 beta/biosynthesis , Glycogen Synthase Kinase 3 beta/genetics , Interleukin-1beta/biosynthesis , Interleukin-1beta/genetics , Male , Maze Learning/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mitochondria/drug effects , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Neurogenesis/drug effects , Neuroglia/drug effects , Neuronal Plasticity/drug effects , Neuroprotective Agents/pharmacokinetics , Phosphorylation/drug effects , Prohibitins , Protein Processing, Post-Translational/drug effects , Repressor Proteins/biosynthesis , Repressor Proteins/genetics , Tauopathies/drug therapy , tau Proteins/metabolism
8.
Histochem Cell Biol ; 153(3): 165-175, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31858211

ABSTRACT

In diabetic nephropathy (DN), intercellular communication is disrupted. Connexins (Cx) have a crucial role in that process. Dietary ratios and supplementation with polyunsaturated fatty acids (PUFAs) can alleviate diabetic complications and cause alterations in Cx levels. Although pannexins (Panx) share similarities with members of the Cx family, their function in diabetic nephropathy has still not been fully determined. We studied the influence of PUFA supplementation on the immunoexpression of Px1 and Cx family members in diabetic kidneys of rats. Four groups of rats in experimental DM1 model were supplemented with different dietary n-6/n-3 ratios; ≈7 in control (C) and diabetic groups (STZ), ≈ 60 in the STZ + N6 group and ≈ 1 (containing 16% EPA and 19% DHA) in the STZ + N3 group. Immunoexpression of Cx40, Cx43, Cx45 and Panx1 was evaluated in the renal tissue of diabetic rats using immunohistochemistry. Diabetes significantly decreased the protein expression of Cx40 and Cx43 and increased Panx1 protein expression in the renal cortex (p < 0.05-p < 0.01). There was a significant impact of diet on Cx and Panx1 immunoexpression. Dietary supplementation with a high n-6/n-3 ratio downregulated the protein expression of Cx45 and Panx1 in diabetic rats (p < 0.05-p < 0.01), while Cx43 immunoexpression was increased in diabetic rats fed with high and low n-6/n-3 ratios (p < 0.01-p < 0.001). Hyperglycaemic conditions in DN interfere with cell-to-cell communication and disturb the connection between cells and their immediate environment due to variations in connexin and pannexin immunoexpression. These variations can be regulated by PUFA dietary intake, suggesting their beneficial effect and possible therapeutic option.


Subject(s)
Connexins/antagonists & inhibitors , Diabetes Mellitus, Experimental/drug therapy , Diabetic Nephropathies/drug therapy , Fatty Acids, Unsaturated/pharmacology , Kidney/drug effects , Nerve Tissue Proteins/antagonists & inhibitors , Animals , Connexins/analysis , Connexins/biosynthesis , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/metabolism , Diabetic Nephropathies/chemically induced , Diabetic Nephropathies/metabolism , Dietary Supplements , Fatty Acids, Unsaturated/administration & dosage , Kidney/metabolism , Kidney/pathology , Male , Nerve Tissue Proteins/analysis , Nerve Tissue Proteins/biosynthesis , Rats , Rats, Wistar , Streptozocin
9.
Proc Natl Acad Sci U S A ; 116(47): 23636-23642, 2019 11 19.
Article in English | MEDLINE | ID: mdl-31685615

ABSTRACT

Sonic hedgehog (SHH) signaling plays a pivotal role in 2 different phases during brain development. Early SHH signaling derived from the prechordal plate (PrCP) triggers secondary Shh induction in the forebrain, which overlies the PrCP, and the induced SHH signaling, in turn, directs late neuronal differentiation of the forebrain. Consequently, Shh regulation in the PrCP is crucial for initiation of forebrain development. However, no enhancer that regulates prechordal Shh expression has yet been found. Here, we identified a prechordal enhancer, named SBE7, in the vicinity of a cluster of known forebrain enhancers for Shh This enhancer also directs Shh expression in the ventral midline of the forebrain, which receives the prechordal SHH signal. Thus, the identified enhancer acts not only for the initiation of Shh regulation in the PrCP but also for subsequent Shh induction in the forebrain. Indeed, removal of the enhancer from the mouse genome markedly down-regulated the expression of Shh in the rostral domains of the axial mesoderm and in the ventral midline of the forebrain and hypothalamus in the mouse embryo, and caused a craniofacial abnormality similar to human holoprosencephaly (HPE). These findings demonstrate that SHH signaling mediated by the newly identified enhancer is essential for development and growth of the ventral midline of the forebrain and hypothalamus. Understanding of the Shh regulation governed by this prechordal and brain enhancer provides an insight into the mechanism underlying craniofacial morphogenesis and the etiology of HPE.


Subject(s)
Enhancer Elements, Genetic , Gene Expression Regulation, Developmental , Hedgehog Proteins/physiology , Nerve Tissue Proteins/physiology , Prosencephalon/embryology , Animals , CRISPR-Cas Systems , Eye Proteins/physiology , Gene Knockout Techniques , Genes, Reporter , Hedgehog Proteins/biosynthesis , Hedgehog Proteins/genetics , Holoprosencephaly/genetics , Homeodomain Proteins/physiology , Hypothalamus/abnormalities , Hypothalamus/embryology , Hypothalamus/metabolism , Lac Operon , Mesencephalon/embryology , Mesencephalon/metabolism , Mesoderm/metabolism , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Prosencephalon/abnormalities , Prosencephalon/metabolism , Signal Transduction , Transgenes , Homeobox Protein SIX3
10.
Proc Natl Acad Sci U S A ; 116(47): 23426-23436, 2019 11 19.
Article in English | MEDLINE | ID: mdl-31685616

ABSTRACT

As a central feature of neuroinflammation, microglial dysfunction has been increasingly considered a causative factor of neurodegeneration implicating an intertwined pathology with amyloidogenic proteins. Herein, we report the smallest synthetic molecule (N,N'-diacetyl-p-phenylenediamine [DAPPD]), simply composed of a benzene ring with 2 acetamide groups at the para position, known to date as a chemical reagent that is able to promote the phagocytic aptitude of microglia and subsequently ameliorate cognitive defects. Based on our mechanistic investigations in vitro and in vivo, 1) the capability of DAPPD to restore microglial phagocytosis is responsible for diminishing the accumulation of amyloid-ß (Aß) species and significantly improving cognitive function in the brains of 2 types of Alzheimer's disease (AD) transgenic mice, and 2) the rectification of microglial function by DAPPD is a result of its ability to suppress the expression of NLRP3 inflammasome-associated proteins through its impact on the NF-κB pathway. Overall, our in vitro and in vivo investigations on efficacies and molecular-level mechanisms demonstrate the ability of DAPPD to regulate microglial function, suppress neuroinflammation, foster cerebral Aß clearance, and attenuate cognitive deficits in AD transgenic mouse models. Discovery of such antineuroinflammatory compounds signifies the potential in discovering effective therapeutic molecules against AD-associated neurodegeneration.


Subject(s)
Alzheimer Disease/drug therapy , Anti-Inflammatory Agents/pharmacology , Cognition/drug effects , Microglia/drug effects , Neuroprotective Agents/pharmacology , Phagocytosis/drug effects , Phenylenediamines/pharmacology , Alzheimer Disease/psychology , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Anti-Inflammatory Agents/therapeutic use , Drug Evaluation, Preclinical , Gene Expression Regulation/drug effects , Humans , Inflammasomes/drug effects , Inflammasomes/genetics , Maze Learning , Mice , Mice, Transgenic , Microglia/physiology , Molecular Structure , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Neuroprotective Agents/therapeutic use , Peptide Fragments/genetics , Phenylenediamines/chemistry , Phenylenediamines/therapeutic use , Presenilin-1/genetics , Spatial Memory/drug effects
11.
J Mol Neurosci ; 69(3): 456-469, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31290091

ABSTRACT

Obviously, opiates (e.g., morphine) are associated with the suppression and dysfunction of reproductive axis. It has been reported that substance P (SP) and RF-amid-related peptide-3 (RFRP-3) can exhibit anti-opioid effects in some regions of the nervous system. Moreover, SP and RFRP-3 are deemed as neuropeptides which exert modulatory and regulatory impacts on the function of the reproductive axis. The precise interactions of morphine with SP or RFRP-3 on the parameters of the reproductive activity, however, are not fully known. The present study was aimed to determine the impacts of the interaction of morphine either with SP or RFRP-3 on the hormonal and behavioral parameters of reproductive activity in male rats. In addition, it was aimed at determining whether the effects of these interactions rely on kisspeptin/G protein coupled receptor 54 (GPR54) pathway as the main upstream pulse generator and the mediator of the function of many inputs of gonadotropin-releasing hormone (GnRH)/luteinizing hormone (LH) system or not. Altogether, the resulted data from the sexual behavior tests, radioimmunoassay of LH/testosterone, and real-time quantitative PCR for the assessment of the expression of hypothalamic Kiss1, Gpr54, and Gnrh1 genes following concomitant administration of morphine with SP or RFRP-3 revealed that the suppressing effects of morphine on the parameters of reproductive axis activity can be affected by the administration of either RFRP-3 or SP. It is advocated that SP and RFRP-3, by the modulation of the expression of hypothalamic Kiss1, can possibly antagonize the effects of morphine on GnRH/LH system and sexual behavior.


Subject(s)
Hypothalamus/drug effects , Kisspeptins/physiology , Morphine/pharmacology , Nerve Tissue Proteins/physiology , Neuropeptides/pharmacology , Receptors, Kisspeptin-1/physiology , Sexual Behavior, Animal/drug effects , Substance P/pharmacology , Animals , Drug Interactions , Gene Expression Regulation/drug effects , Gonadotropin-Releasing Hormone/biosynthesis , Gonadotropin-Releasing Hormone/genetics , Gonadotropin-Releasing Hormone/physiology , Hypothalamus/metabolism , Kisspeptins/biosynthesis , Kisspeptins/genetics , Luteinizing Hormone/physiology , Male , Naloxone/pharmacology , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Protein Precursors/biosynthesis , Protein Precursors/genetics , RNA, Messenger/biosynthesis , Rats , Rats, Wistar , Receptors, Kisspeptin-1/biosynthesis , Receptors, Kisspeptin-1/genetics , Signal Transduction/physiology
12.
Neurochem Int ; 129: 104493, 2019 10.
Article in English | MEDLINE | ID: mdl-31220473

ABSTRACT

Glutamatergic excitotoxicity is crucial in the pathogenesis of epileptic seizures. Dexmedetomidine, a potent and highly selective α2 adrenoceptor agonist, inhibits glutamate release from nerve terminals in rat cerebrocortical nerve terminals. However, the ability of dexmedetomidine to affect glutamate-induced brain injury is still unknown. Therefore, the present study evaluated the protective effect of dexmedetomidine against brain damage by using a kainic acid (KA) rat model, a frequently used model for temporal lobe epilepsy. Rats were treated with dexmedetomidine (1 or 5 µg/kg, intraperitoneally) 30 min before the KA (15 mg/kg) intraperitoneal injection. KA-induced seizure score and elevations of glutamate release in rat hippocampi were inhibited by pretreatment with dexmedetomidine. Histopathological and TUNEL staining analyzes showed that dexmedetomidine attenuated KA-induced neuronal death in the hippocampus. Dexmedetomidine ameliorated KA-induced apoptosis, and this neuroprotective effect was accompanied by inhibited the KA-induced caspase-3 expression as well as MAPKs phosphorylation, and reversed Bcl-2 down-expression, coupled with increased Nrf2, BDNF and TrkB expression in KA-treated rats. The results suggest that dexmedetomidine protected rat brains from KA-induced excitotoxic damage by reducing glutamate levels, suppressing caspase-3 activation and MAPKs phosphorylation, and enhancing Bcl-2, Nrf2, BDNF and TrkB expression in the hippocampus. Therefore, dexmedetomidine may be beneficial for preventing or treating brain disorders associated with excitotoxic neuronal damage. In conclusion, these data suggest that dexmedetomidine has the therapeutic potential for treating epilepsy.


Subject(s)
Anticonvulsants/therapeutic use , Brain-Derived Neurotrophic Factor/agonists , Dexmedetomidine/therapeutic use , Glutamic Acid/metabolism , Hippocampus/drug effects , Kainic Acid/antagonists & inhibitors , Neurons/drug effects , Neuroprotective Agents/therapeutic use , Seizures/prevention & control , Animals , Anticonvulsants/pharmacology , Apoptosis/drug effects , Brain-Derived Neurotrophic Factor/biosynthesis , Brain-Derived Neurotrophic Factor/genetics , Dexmedetomidine/pharmacology , Drug Evaluation, Preclinical , Hippocampus/physiopathology , Kainic Acid/toxicity , MAP Kinase Signaling System/drug effects , Male , NF-E2-Related Factor 2/biosynthesis , NF-E2-Related Factor 2/genetics , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Neurons/physiology , Neuroprotective Agents/pharmacology , Rats , Rats, Sprague-Dawley , Receptor, trkB/biosynthesis , Receptor, trkB/genetics , Seizures/chemically induced
13.
Neuromolecular Med ; 21(3): 250-261, 2019 09.
Article in English | MEDLINE | ID: mdl-31037465

ABSTRACT

Depression is one of the most prevalent and crucial public health problem connected to significant mortality and co-morbidity. Recently, numerous studies suggested that dietary flavanones exhibit neuroprotective and antidepressant effects against various psycho-physiological conditions including depression. The present study is focused on the antidepressant and neuroprotective effects of naringenin (NAR) and the involvement of sonic hedgehog (Shh) signaling in the chronic unpredictable mild stress (CUMS)-induced depression. Twenty-four male Wistar rats were randomly assigned into four groups: CON group (saline s.c.), NAR group (NAR 50 mg/kg, p.o.), CUMS group (subjected to CUMS along with saline p.o.), and CUMS + NAR group (NAR 50 mg/kg p.o. along with CUMS) for 28 days including 1-week pre-treatment with NAR. The results showed that NAR was found to inhibit behavioral abnormalities including increased despair in force swim test, and reduced locomotor activity caused by CUMS in open field test. Moreover, Morris water maze revealed that NAR also mitigates CUMS-associated cognitive impairment. In addition to the antidepressant-like effect, NAR mitigates morphological anomalies in the hippocampal CA1 region and cortex. Furthermore, we observed brain-derived neurotrophic factor (BDNF), Shh, GLI1, NKX2.2, and PAX6 were downregulated in the hippocampus of CUMS-exposed rats, which can be upregulated by NAR pre-treatment. GLI1 is main downstream signaling component of Shh signaling cascade, which further regulates the expression of homeodomain transcription factors PAX6 and NKX2.2.


Subject(s)
Antidepressive Agents/therapeutic use , Depression/drug therapy , Flavanones/therapeutic use , Hedgehog Proteins/physiology , Learning Disabilities/drug therapy , Memory Disorders/drug therapy , Neuroprotective Agents/therapeutic use , Signal Transduction/drug effects , Stress, Psychological/drug therapy , Zinc Finger Protein GLI1/physiology , Animals , Antidepressive Agents/pharmacology , Chronic Disease , Depression/etiology , Depression/metabolism , Depression/prevention & control , Disease Models, Animal , Drug Evaluation, Preclinical , Exploratory Behavior/drug effects , Flavanones/pharmacology , Gene Expression Regulation/drug effects , Hippocampus/chemistry , Hippocampus/drug effects , Homeobox Protein Nkx-2.2 , Learning Disabilities/etiology , Learning Disabilities/metabolism , Learning Disabilities/prevention & control , Male , Maze Learning/drug effects , Memory Disorders/etiology , Memory Disorders/metabolism , Memory Disorders/prevention & control , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Neuroprotective Agents/pharmacology , Random Allocation , Rats , Rats, Wistar , Stress, Psychological/physiopathology , Swimming
14.
J Neuroimmune Pharmacol ; 14(3): 462-477, 2019 09.
Article in English | MEDLINE | ID: mdl-30941623

ABSTRACT

Resveratrol (3,5,4'-trihydroxy-trans-stilbene) (RES) is a naturally-derived phytoestrogen found in the skins of red grapes and berries and has potential as a novel and effective therapeutic agent. In the current study, we investigated the role of microRNA (miRNA) in RES-mediated attenuation of experimental autoimmune encephalomyelitis (EAE), a murine model of multiple sclerosis. Administration of RES effectively decreased disease severity, including inflammation and central nervous system immune cell infiltration. miRNA microarray analysis revealed an altered miRNA profile in encephalitogenic CD4+ T cells from EAE mice exposed to RES treatment. Additionally, bioinformatics and in silico pathway analysis suggested the involvement of RES-induced miRNA in pathways and processes that regulated cellular proliferation. Additional studies confirmed that RES affected cell cycle progression and apoptosis in activated T cells, specifically in the brain. RES treatment significantly upregulated miR-124 during EAE, while suppressing associated target gene, sphingosine kinase 1 (SK1), and this too was specific to mononuclear cells in the brains of treated mice, as peripheral immune cells remained unaltered upon RES treatment. Collectively, these studies demonstrate that RES treatment leads to amelioration of EAE development through mechanism(s) potentially involving suppression of neuroinflammation via alteration of the miR-124/SK1 axis, thereby halting cell-cycle progression and promoting apoptosis in activated encephalitogenic T cells. Graphical Abstract Resveratrol alters the miR-124/sphingosine kinase 1 (SK1) axis in encephalitogenic T cells, promotes cell-cycle arrest and apoptosis, and decreases neuroinflammation in experiemental autoimmune encephalomyelitis (EAE).


Subject(s)
Brain/drug effects , CD4-Positive T-Lymphocytes/drug effects , Encephalomyelitis, Autoimmune, Experimental/drug therapy , MicroRNAs/biosynthesis , Nerve Tissue Proteins/biosynthesis , Phosphotransferases (Alcohol Group Acceptor)/biosynthesis , Resveratrol/therapeutic use , Signal Transduction/drug effects , T-Lymphocyte Subsets/drug effects , Animals , Apoptosis/drug effects , Brain/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Cycle/drug effects , Cell Division/drug effects , Drug Evaluation, Preclinical , Encephalomyelitis, Autoimmune, Experimental/metabolism , Enzyme Induction/drug effects , Female , Gene Expression Regulation/drug effects , Gene Ontology , Lymphocyte Activation , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , MicroRNAs/physiology , Microglia/drug effects , Monocytes/drug effects , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/physiology , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/physiology , Resveratrol/pharmacology , T-Lymphocyte Subsets/metabolism
15.
J Neural Transm (Vienna) ; 125(9): 1319-1331, 2018 09.
Article in English | MEDLINE | ID: mdl-29998409

ABSTRACT

Histone acetylation is a key regulatory factor for gene expression in cells. Modulation of histone acetylation by targeting of histone acetyltransferases (HATs) effectively alters many gene expression profiles and synaptic plasticity in the brain. However, the role of HATs on L-DOPA-induced dyskinesia of Parkinson's disease (PD) has not been reported. Our aim was to determine whether HAT inhibitors such as anacardic acid, garcinol, and curcumin from natural plants reduce severity of L-DOPA-induced dyskinesia using a unilaterally 6-hydroxydopamine (6-OHDA)-lesioned PD mouse model. Anacardic acid 2 mg/kg, garcinol 5 mg/kg, or curcumin 100 mg/kg co-treatment with L-DOPA significantly reduced the axial, limb, and orofacial (ALO) score indicating less dyskinesia with administration of HAT inhibitors in 6-OHDA-lesioned mice. Additionally, L-DOPA's efficacy was not altered by the compounds in the early stage of treatment. The expression levels of c-Fos, Fra-2, and Arc were effectively decreased by administration of HAT inhibitors in the ipsilateral striatum. Our findings indicate that HAT inhibitor co-treatment with L-DOPA may have therapeutic potential for management of L-DOPA-induced dyskinesia in patients with PD.


Subject(s)
Anacardic Acids/therapeutic use , Antiparkinson Agents/toxicity , Curcumin/therapeutic use , Dyskinesia, Drug-Induced/drug therapy , Enzyme Inhibitors/therapeutic use , Histone Acetyltransferases/antagonists & inhibitors , Levodopa/toxicity , Parkinsonian Disorders/drug therapy , Terpenes/therapeutic use , Anacardic Acids/pharmacology , Animals , Curcumin/pharmacology , Cytoskeletal Proteins/biosynthesis , Cytoskeletal Proteins/genetics , Drug Evaluation, Preclinical , Dyskinesia, Drug-Induced/etiology , Dyskinesia, Drug-Induced/genetics , Enzyme Inhibitors/pharmacology , Fos-Related Antigen-2/biosynthesis , Fos-Related Antigen-2/genetics , Gene Expression Regulation/drug effects , Histone Code/drug effects , MAP Kinase Signaling System/drug effects , Male , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Oxidopamine/toxicity , Proto-Oncogene Proteins c-fos/biosynthesis , Proto-Oncogene Proteins c-fos/genetics , Specific Pathogen-Free Organisms , Substantia Nigra/drug effects , Substantia Nigra/pathology , Terpenes/pharmacology
16.
Mol Cell Biochem ; 448(1-2): 251-263, 2018 Nov.
Article in English | MEDLINE | ID: mdl-29446045

ABSTRACT

Dietary administration of orotic acid (OA), an intermediate in the pyrimidine biosynthetic pathway, is considered to provide a wide range of beneficial effects, including cardioprotection and exercise adaptation. Its mechanisms of action, when applied extracellularly, however, are barely understood. In this study, we evaluated potential effects of OA on skeletal muscle using an in vitro contraction model of electrically pulse-stimulated (EPS) C2C12 myotubes. By analyzing a subset of genes representing inflammatory, metabolic, and structural adaptation pathways, we could show that OA supplementation diminishes the EPS-provoked expression of inflammatory transcripts (interleukin 6, Il6; chemokine (C-X-C Motif) ligand 5, Cxcl5), and attenuated transcript levels of nuclear receptor subfamily 4 group A member 3 (Nr4A3), early growth response 1 (Egr1), activating transcription factor 3 (Atf3), and fast-oxidative MyHC-IIA isoform (Myh2). By contrast, OA had no suppressive effect on the pathogen-provoked inflammatory gene response in skeletal muscle cells, as demonstrated by stimulation of C2C12 myotubes with bacterial LPS. In addition, we observed a suppressive effect of OA on EPS-induced phosphorylation of AMP-activated protein kinase (AMPK), whereas EPS-triggered phosphorylation/activation of the mammalian target of rapamycin (mTOR) was not affected. Finally, we demonstrate that OA positively influences glycogen levels in EP-stimulated myotubes. Taken together, our results suggest that in skeletal muscle cells, OA modulates both the inflammatory and the metabolic reaction provoked by acute contraction. These results might have important clinical implications, specifically in cardiovascular and exercise medicine.


Subject(s)
Muscle Contraction/drug effects , Myoblasts, Skeletal/metabolism , Orotic Acid/pharmacology , Activating Transcription Factor 3/biosynthesis , Animals , Chemokine CXCL5/biosynthesis , DNA-Binding Proteins/biosynthesis , Early Growth Response Protein 1/biosynthesis , Electric Stimulation , Gene Expression Regulation/drug effects , Interleukin-6/biosynthesis , Mice , Myoblasts, Skeletal/cytology , Nerve Tissue Proteins/biosynthesis , Receptors, Steroid/biosynthesis , Receptors, Thyroid Hormone/biosynthesis , TOR Serine-Threonine Kinases/biosynthesis
17.
Neuromolecular Med ; 20(1): 133-146, 2018 03.
Article in English | MEDLINE | ID: mdl-29468499

ABSTRACT

Autism is a neurodevelopmental disorder characterized by deficits in qualitative impairments in communication, repetitive and social interaction, restricted, and stereotyped patterns of behavior. Resveratrol has been extensively studied pharmacologically and biologically and has anti-inflammatory, antioxidant, and neuroprotective effects on neuronal damage in neurodegenerative disorders. The BTBR T+ Itpr3tf/J (BTBR) autistic mouse model has been explored for treatment of autism, which shows low reciprocal social interactions, impaired juvenile play, and decreased social approach. Here, we explored whether resveratrol treatment decreases neuroimmune dysregulation mediated through toll-like receptor (TLR4) and nuclear factor-κB (NF-κB) signaling pathway in BTBR mice. We investigated the effect of resveratrol treatment on TLR2, TLR3, TLR4, NF-κB, and inducible nitric oxide synthase (iNOS or NOS2) levels in CD4 spleen cells. We also assessed the effect of resveratrol treatment on TLR2, TLR3, TLR4, NF-κB, iNOS, and cyclooxygenase (COX-2) mRNA expression levels in the brain tissue. We further explored TLR2, TLR4, NF-κB, iNOS, and COX-2 protein expression levels in the brain tissue. Resveratrol treatment on BTBR mice significantly decreased CD4+TLR2+, CD4+TLR3+, CD4+TLR4+ CD4+NF-κB+, and CD4+iNOS+ levels in spleen cells. Resveratrol treatment on BTBR mice decreased TLR2, TLR3, TLR4, NF-κB, iNOS, and COX-2 mRNA expression levels in brain tissue. Moreover, resveratrol treatment resulted in decreased protein expression of TLR2, TLR3, TLR4, NF-κB, iNOS, and COX-2 in brain tissue. Taken together, these results indicate that resveratrol treatment improves neuroimmune dysregulation through the inhibition of proinflammatory mediators and TLRs/NF-κB transcription factor signaling, which might be help devise future therapies for neuroimmune disorders.


Subject(s)
Autistic Disorder/drug therapy , Cyclooxygenase 2/physiology , Gene Expression Regulation/drug effects , Neuroprotective Agents/therapeutic use , Nitric Oxide Synthase Type II/physiology , Resveratrol/therapeutic use , Signal Transduction/drug effects , Toll-Like Receptors/physiology , Animals , Autistic Disorder/metabolism , Brain Chemistry/drug effects , Cells, Cultured , Cyclooxygenase 2/biosynthesis , Cyclooxygenase 2/genetics , Drug Evaluation, Preclinical , Inositol 1,4,5-Trisphosphate Receptors , Mice , Mice, Inbred C57BL , Mice, Neurologic Mutants , NF-kappa B/physiology , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Neurons/drug effects , Neuroprotective Agents/pharmacology , Nitric Oxide Synthase Type II/biosynthesis , Nitric Oxide Synthase Type II/genetics , Organ Specificity , Resveratrol/pharmacology , Spleen , Toll-Like Receptors/biosynthesis , Toll-Like Receptors/genetics
18.
Neuromolecular Med ; 20(1): 147-159, 2018 03.
Article in English | MEDLINE | ID: mdl-29476479

ABSTRACT

Acute inflammation can exacerbate brain injury after ischemic stroke. Beyond its well-characterized role in calcium metabolism, it is becoming increasingly appreciated that the active form of vitamin D, 1,25-dihydroxyvitamin D3 (1,25-VitD3), has potent immunomodulatory properties. Here, we aimed to determine whether 1,25-VitD3 supplementation could reduce subsequent brain injury and associated inflammation after ischemic stroke. Male C57Bl6 mice were randomly assigned to be administered either 1,25-VitD3 (100 ng/kg/day) or vehicle i.p. for 5 day prior to stroke. Stroke was induced via middle cerebral artery occlusion for 1 h followed by 23 h reperfusion. At 24 h post-stroke, we assessed infarct volume, functional deficit, expression of inflammatory mediators and numbers of infiltrating immune cells. Supplementation with 1,25-VitD3 reduced infarct volume by 50% compared to vehicle. Expression of pro-inflammatory mediators IL-6, IL-1ß, IL-23a, TGF-ß and NADPH oxidase-2 was reduced in brains of mice that received 1,25-VitD3 versus vehicle. Brain expression of the T regulatory cell marker, Foxp3, was higher in mice supplemented with 1,25-VitD3 versus vehicle, while expression of the transcription factor, ROR-γ, was decreased, suggestive of a reduced Th17/γδ T cell response. Immunohistochemistry indicated that similar numbers of neutrophils and T cells were present in the ischemic hemispheres of 1,25-VitD3- and vehicle-supplemented mice. At this early time point, there were also no differences in the impairment of motor function. These data indicate that prior administration of exogenous vitamin D, even to vitamin D-replete mice, can attenuate infarct development and exert acute anti-inflammatory actions in the ischemic and reperfused brain.


Subject(s)
Brain/drug effects , Cholecalciferol/therapeutic use , Infarction, Middle Cerebral Artery/drug therapy , Neuroprotective Agents/therapeutic use , Animals , Brain/pathology , Cholecalciferol/pharmacology , Cytokines/biosynthesis , Cytokines/genetics , Forkhead Transcription Factors/biosynthesis , Forkhead Transcription Factors/genetics , Gene Expression Regulation/drug effects , Infarction, Middle Cerebral Artery/pathology , Infarction, Middle Cerebral Artery/physiopathology , Inflammation , Inflammation Mediators/metabolism , Macrophages/drug effects , Male , Mice, Inbred C57BL , Microglia/drug effects , Motor Activity/drug effects , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Neuroprotective Agents/pharmacology , Neutrophil Infiltration/drug effects , Nuclear Receptor Subfamily 1, Group F, Member 3/biosynthesis , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/metabolism
19.
Neuromolecular Med ; 20(1): 97-111, 2018 03.
Article in English | MEDLINE | ID: mdl-29411248

ABSTRACT

Our previous studies demonstrated that the multifunctional agent TBN, a derivative of tetramethylpyrazine armed with a nitrone moiety, displayed high therapeutic efficacy in experimental ischemic stroke models. However, its molecular mechanisms of action underlying the neuroprotective effect need further exploration. In the present study, we found that TBN had significant activities scavenging free radicals such as ·OH, O 2·- and ONOO-, inhibiting Ca2+ overload, maintaining mitochondrial function and preventing neuronal damage in primary cortical cultures. Further, TBN was effective in reducing brain infarction and ameliorating impairment of behavioral functions in the permanent middle cerebral artery occlusion (p-MCAo) rat model. TBN down-regulated the expression of pro-apoptotic factors Bax, while up-regulated the expression of anti-apoptotic factor Bcl-2 and increased the expression of pro-survival factors including p-Akt and p-GSK3ß in the peri-infarct cortex of p-MCAo rats. In addition, LY-294002 (a PI3K inhibitor) and MK2206 (an Akt inhibitor) significantly blocked the protective effect of TBN against OGD-induced death of cortical neurons. Taken together, the multifunctional mechanisms including scavenging free radicals, blocking calcium overload, maintaining mitochondrial function and activating the PI3K/Akt/p-GSK3ß cell survival pathway were possibly involved in the neuroprotective effects of TBN, making it a promising clinical candidate for the treatment of ischemic stroke.


Subject(s)
Infarction, Middle Cerebral Artery/drug therapy , Neuroprotective Agents/therapeutic use , Nitrogen Oxides/therapeutic use , Pyrazines/therapeutic use , Animals , Apoptosis Regulatory Proteins/biosynthesis , Apoptosis Regulatory Proteins/genetics , Calcium/metabolism , Cell Hypoxia , Cells, Cultured , Drug Evaluation, Preclinical , Free Radical Scavengers/pharmacology , Free Radical Scavengers/therapeutic use , Glucose/pharmacology , Glutamic Acid/toxicity , Infarction, Middle Cerebral Artery/prevention & control , Lipid Peroxidation/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Motor Activity/drug effects , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Neuroprotective Agents/pharmacology , Nitrogen Oxides/pharmacology , Patch-Clamp Techniques , Protein Kinase Inhibitors/pharmacology , Pyrazines/pharmacology , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects
20.
Biomed Pharmacother ; 93: 1033-1039, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28738518

ABSTRACT

BACKGROUND AND OBJECTIVE: Teucrium polium Lamiaceae. an antidiabetic traditional medicine was observed to stimulate insulin secretion and ß-cell mass restoration in pancreatic of diabetic rats. Bioactive compounds in T. polium such as terpenoids and flavonoids have a wide range of antioxidant properties. Given this background, our research was aimed at probing the effective molecular mechanism of T. polium which in turn protects the pancreatic ß-cells of diabetic rats through attenuation of streptozotocin-induced oxidative stress. MATERIALS AND METHODS: Induction of diabetes by streptozotocin (STZ) injection followed by treatment of the rats with T. polium extract (0.5g/kg) and glibenclamide as a standard drug (600µg/kg) for six consecutive weeks. Besides, biochemical indexes including sera glucose and lipids were investigated once every two weeks. Moreover, oxidative stress markers were also measured in the sixth week of treatment to assess the antioxidant capacity of T. polium. Prior to sacrifice of rats, the oral glucose tolerance test was carried out and thereafter western blot analysis was performed on pancreatic tissues to detect the expression of JNK, FoxO1 and Pdx1 proteins in the experimental groups. RESULTS: Oral administration of T. polium extract not only disclosed the significant anti-hyperglycemic potential of the plant, but also relieved dyslipidemia and oxidative stress related to diabetes. Furthermore, remarkable improvement of glucose tolerance was exhibited among the treated groups, confirming the presence of insulin in the blood. Subsequently, the effective molecular mechanism of T. polium was partially revealed by western blot analyses which detected considerable up-regulation of p-FoxO1 and Pdx1 proteins parallel to reduction of p-JNK expression among the treated diabetic rats. CONCLUSION: This study demonstrates that T. polium extract is able to restore the ß-cell mass and insulin secretion by regulation of pivotal transcription factor of the pancreatic ß-cells Pdx1 in JNK pathway.


Subject(s)
Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Homeodomain Proteins/biosynthesis , Nerve Tissue Proteins/biosynthesis , Plant Extracts/therapeutic use , Teucrium , Trans-Activators/biosynthesis , Animals , Blood Glucose/drug effects , Blood Glucose/metabolism , Diabetes Mellitus, Experimental/blood , Gene Expression , Male , Plant Extracts/isolation & purification , Plant Extracts/pharmacology , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL