Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
Pharmacology ; 105(1-2): 39-46, 2020.
Article in English | MEDLINE | ID: mdl-31554003

ABSTRACT

BACKGROUND: The potential protective effects of resveratrol (RES) on the modulation of hepatic biomarkers of apoptosis and survival, p53-Bax axis, and B-cell lymphoma 2 (Bcl-2) in an animal model of paracetamol-induced acute liver injury have not been investigated before. METHODS: The model group of rats received a single dose of paracetamol (2 g/kg, orally), whereas the protective group of rats were pretreated for 7 days with RES (30 mg/kg, i.p.) before they were given a single dose of paracetamol. All rats were then sacrificed 24-h post paracetamol ingestion. RESULTS: Histology images showed that paracetamol overdose induced acute liver injury, which was substantially protected by RES. Paracetamol significantly (p < 0.05) modulated p53, apoptosis regulator Bax, Bcl-2, tumor necrosis factor-alpha, interleukin-6, inducible nitric oxide synthase, malondialdehyde, superoxide dismutase, glutathione peroxidase, alanine aminotransferase, and aspartate aminotransferase, which were significantly protected by RES. We further demonstrated a significant (p< 0.01) correlation between either p53 or Bcl-2 scoring and the levels of inflammatory, nitrosative stress, and liver injury biomarkers. CONCLUSION: We demonstrate a substantial protection by RES pretreatment against paracetamol-induced modulation of p53-Bax axis, Bcl-2, and other acute liver injury biomarkers in rats.


Subject(s)
Acetaminophen/toxicity , Analgesics, Non-Narcotic/toxicity , Antioxidants/pharmacology , Chemical and Drug Induced Liver Injury , Proto-Oncogene Proteins c-bcl-2/metabolism , Resveratrol/pharmacology , Tumor Suppressor Protein p53/genetics , Alanine Transaminase/metabolism , Animals , Apoptosis/drug effects , Aspartate Aminotransferases/metabolism , Biomarkers/metabolism , Chemical and Drug Induced Liver Injury/genetics , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Disease Models, Animal , Drug Overdose , Liver/drug effects , Liver/pathology , Male , Oxidative Stress/drug effects , Rats, Wistar
2.
J Cell Physiol ; 234(5): 5722-5727, 2019 05.
Article in English | MEDLINE | ID: mdl-30417468

ABSTRACT

Psychiatric disorders such as anxiety, phobias, and post-traumatic stress disorder are considered of high global prevalence. Currently, a therapeutic approach to treat these disorders using beta-blockers, which antagonize the beta-adrenergic receptors (B1, B2, and B3) is being studied. This approach claims that beta-blockers, such as propranolol, inhibit fear memory reconsolidation. However, there are several studies refuting such claims by discrediting their experimental design and pointing out both the drugs pharmacokinetic properties and confounding factors. In this review, we explore the different effects of central beta-adrenergic agonists and antagonists on the fear memory consolidation providing mixed-evidence, limitations, and future directions.


Subject(s)
Adrenergic beta-Antagonists/therapeutic use , Brain/drug effects , Fear/drug effects , Memory Consolidation/drug effects , Mental Disorders/drug therapy , Propranolol/therapeutic use , Stress Disorders, Post-Traumatic/drug therapy , Adrenergic beta-Antagonists/adverse effects , Adrenergic beta-Antagonists/pharmacokinetics , Animals , Brain/metabolism , Brain/physiopathology , Humans , Mental Disorders/metabolism , Mental Disorders/physiopathology , Mental Disorders/psychology , Propranolol/adverse effects , Propranolol/pharmacokinetics , Stress Disorders, Post-Traumatic/metabolism , Stress Disorders, Post-Traumatic/physiopathology , Stress Disorders, Post-Traumatic/psychology
3.
J Cell Physiol ; 234(6): 9328-9337, 2019 06.
Article in English | MEDLINE | ID: mdl-30334569

ABSTRACT

The potential inhibitory effect of the antidiabetic and anti-inflammatory drug, metformin on thioacetamide (TAA)-induced hepatotoxicity associated with the inhibition of mammalian target of rapamycin (mTOR)-hypoxia-inducible factor-1α (HIF-1α) axis has not been investigated before. Therefore, we tested whether metformin can protect against liver injuries including fibrosis induced by TAA possibly via the downregulation of mTOR-HIF-1α axis and profibrogenic and inflammatory biomarkers. Rats either injected with TAA (200 mg/kg; twice a week for 8 weeks) before being killed after 10 weeks (model group) or were pretreated with metformin (200 mg/kg) daily for 2 weeks before TAA injections and continued receiving both agents until the end of the experiment, at Week 10 (protective group). Using light and electron microscopy examinations, we observed in the model group substantial damage to the hepatocytes and liver tissue such as collagen deposition, infiltration of inflammatory cells, and degenerative cellular changes with ballooned mitochondria that were substantially ameliorated by metformin. Metformin also significantly ( p < 0.05) inhibited TAA-induced HIF-1α, mTOR, the profibrogenic biomarker α-smooth muscle actin, tissue inhibitor of metalloproteinases-1, tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), alanine aminotransferase (ALT) and aspartate aminotransferase in harvested liver homogenates and blood samples. In addition, a significant ( p < 0.01) positive correlation between hypoxia scoring (HIF-1α) and the serum levels of TNF-α ( r = 0.797), IL-6 ( r = 0.859), and ALT ( r = 0.760) was observed. We conclude that metformin protects against TAA-induced hepatic injuries in rats, which is associated with the inhibition of mTOR-HIF-1α axis and profibrogenic and inflammatory biomarkers; thus, may offer therapeutic potential in humans.


Subject(s)
Biomarkers/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Inflammation/pathology , Liver Cirrhosis/pathology , Liver/pathology , Metformin/pharmacology , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Animals , Chronic Disease , Hepatocytes/drug effects , Hepatocytes/pathology , Hepatocytes/ultrastructure , Liver/drug effects , Liver/injuries , Liver/metabolism , Liver Cirrhosis/metabolism , Male , Protective Agents/pharmacology , Rats , Signal Transduction/drug effects , Thioacetamide
4.
Ultrastruct Pathol ; 43(4-5): 190-198, 2019.
Article in English | MEDLINE | ID: mdl-31522593

ABSTRACT

Cardiovascular disease secondary to diabetes represents a significant challenge to the health community. The advanced glycation end products (AGEs) play an important role in diabetes-mediated vascular injury. We tested whether metformin can suppress aortic AGEs production and protect against aortic injuries (aortopathy) and hypertension in streptozotocin-induced type 2 diabetes mellitus (T2DM) animal model. T2DM was induced in rats two weeks after being fed on a high carbohydrate and fat diet (HCFD), and continued on a HCFD until being sacrificed at week 12 (model group). The protective group was put on metformin two weeks before diabetic induction and continued on metformin and HCFD until the end of the experiment, at week 12. Using electron microscopy examinations, we observed in the model group substantial damage to the ultrastructure of aortic endothelial and vascular smooth muscle layers as demonstrated by markedly distorted vacuolated endothelial and vascular smooth muscle cells with pyknotic nuclei detached from the underlying basement membrane, which were protected by metformin. Also, metformin significantly (p < .05) decreased both systolic and diastolic blood pressure, aortic levels of AGEs, and blood levels of oxidative stress and inflammatory biomarkers. We conclude that metformin protects against T2DM-induced aortopathy and hypertension, possibly via the inhibition of AGEs, inflammation, and oxidative stress.


Subject(s)
Antioxidants/pharmacology , Aorta/drug effects , Diabetes Mellitus, Type 2 , Glycation End Products, Advanced/metabolism , Metformin/pharmacology , Animals , Aorta/pathology , Aorta/ultrastructure , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Diet, High-Fat/adverse effects , Endothelial Cells/drug effects , Endothelial Cells/ultrastructure , Glycation End Products, Advanced/drug effects , Hypertension/etiology , Hypoglycemic Agents/pharmacology , Male , Microscopy, Electron, Transmission , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/ultrastructure , Rats
5.
J Cell Physiol ; 233(8): 5768-5779, 2018 08.
Article in English | MEDLINE | ID: mdl-29205344

ABSTRACT

Diabetes Mellitus is a chronic generalized disorder due to insulin insufficiency or resistance. Skeletal muscles represent one of the most important target organs that is affected by insulin signaling. The aim of the current work was to investigate the effect of metformin versus vitamin D (and also simultaneous administration) therapy in type 2 diabetic (T2D) rats on the state of the muscle and insulin sensitivity. Thirty six male rats constituted the animal model and have been divided into five groups: control, Diabetic, Diabetic + Metformin, Diabetic + Vitamin D, Diabetic + Metformin + Vitamin D. Blood samples were taken for biochemical measurements of serum calcium, interleukin-6 (IL-6), Triglycerides (TG), glucose, insulin, and calculation of HOMA-IR, and then rats were sacrificed, dissected for removal of gastrocnemius muscle that is used for both biochemical, histopathological and electron microscopy examination. Oral administration of vitamin D alone or in combination with metformin improved insulin sensitivity in skeletal muscles, and sustained the metabolic complications along with muscle atrophy and inflammation in T2D rats. We demonstrated super-beneficial action on insulin resistance of additional vitamin D therapy in T2DM rats that were insufficiently controlled by metformin alone.


Subject(s)
Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 2/drug therapy , Hypoglycemic Agents/therapeutic use , Insulin Resistance/physiology , Metformin/therapeutic use , Muscle, Skeletal/metabolism , Vitamin D/therapeutic use , Animals , Blood Glucose/drug effects , Drug Synergism , Male , Muscular Atrophy/drug therapy , Muscular Atrophy/prevention & control , Rats
6.
PeerJ ; 11: e14724, 2023.
Article in English | MEDLINE | ID: mdl-36815993

ABSTRACT

Acute kidney injury (AKI) is a prevalent medical condition accompanied by mutual affection of other organs, including the liver resulting in complicated multiorgan malfunction. Macrophages play a vital role during tissue injury and healing; they are categorized into "classically activated macrophages" (M1) and "alternatively activated macrophages" (M2). The present study investigated and compared the conventional fluid therapy vs Dipeptidyl peptidase 4 inhibitor (DPP-4i) vildagliptin on the liver injury induced by AKI and evaluated the possible molecular mechanisms. Thirty rats comprised five groups (n = 6 rats/group): control, AKI, AKI+saline (received 1.5 mL of normal saline subcutaneous injection), AKI+vildagliptin (treated with oral vildagliptin 10 mg/kg), AKI+saline+vildagliptin. AKI was induced by intramuscular (i.m) injection of 50% glycerol (5 ml/kg). At the end of the work, we collected serum and liver samples for measurements of serum creatinine, blood urea nitrogen (BUN), alanine aminotransferase (ALT), aspartate aminotransferase (AST), tumor necrotic factor-α (TNF-α), and interleukin-10 (IL-10). Liver samples were processed for assessment of inducible nitric oxide synthase (iNOS) as a marker for M1, arginase 1 (Arg-1) as an M2 marker, c-fos, c-Jun, mitogen-activated protein kinase (MAPK), activator protein 1 (AP-1), and high-mobility-group-box1 (HMGB1) protein. The difference was insignificant regarding the relative expression of AP-1, c-Jun, c-fos, MAPK, and HMGB between the AKI+saline group and the AKI+Vildagliptin group. The difference between the same two groups concerning the hepatic content of the M1 marker (iNOS) and the M2 marker Arg-1 was insignificant. However, combined therapy produced more pronounced changes in these markers, as the difference in their relative expression between the AKI+saline+Vildagliptin group and both the AKI+saline group and the AKI+Vildagliptin group was significant. Accordingly, we suggest that the combined saline and vildagliptin hepatoprotective effect involves the downregulation of the MAPK/AP-1 signaling pathway.


Subject(s)
Acute Kidney Injury , Transcription Factor AP-1 , Rats , Animals , Vildagliptin , Saline Solution , Acute Kidney Injury/etiology , Liver/metabolism , Macrophages/metabolism
7.
Brain Sci ; 10(4)2020 Mar 25.
Article in English | MEDLINE | ID: mdl-32218213

ABSTRACT

Acute restraint stress (ARS) is an unavoidable stress situation and may be encountered in different clinical situations. The aim of the current study was to investigate the effects of ARS on the hippocampus and cerebellum, assess the impact of these effects on the behavior and cognitive function, and determine whether pretreatment with ceftriaxone would attenuate the damages produced by ARS on the hippocampus and cerebellum. Four groups of male mice were included in this study: The control group, ARS group, ceftriaxone group, and ARS + ceftriaxone group. Tail suspension test, Y-maze task, and open field tests were used to assess depression, working spatial memory, and anxiety. The biochemical analyses included measurements of serum cortisol, tumor necrotic factor (TNF), interleukin-6, hippocampal expression of bone morphogenetic protein 9 (BMP9), lysosomal-associated membrane protein 1 (LAMP1), glutamate transporter 1 (GLT1), heat shock protein 90, cerebellar expression of S100 protein, glutamic acid decarboxylase (GAD), and carbon anhydrase. Histopathological examination of the brain sections was conducted on the hippocampus and cerebellum by hematoxylin and eosin stains in addition to ultrastructure evaluation using electron microscopy. Our results suggested that ceftriaxone had neuroprotective properties by attenuating the effects of ARS on the hippocampus and cerebellum in mice. This effect was demonstrated by the improvement in the cognitive and behavioral tests as well as by the preservation of the hippocampal and cerebellar architecture.

8.
F1000Res ; 2: 151, 2013.
Article in English | MEDLINE | ID: mdl-24555069

ABSTRACT

BACKGROUND: The hippocampus is a key brain area for many forms of learning and memory and is particularly sensitive to changes in glucose homeostasis. AIM OF THE WORK: To investigate in experimentally induced type 1 and 2 diabetes mellitus in rat model the effect of  diabetes mellitus on cognitive functions and related markers of hippocampal synaptic plasticity, and the possible impact of blocking N-methyl-D-aspartic acid (NMDA) receptors by memantine. MATERIALS AND METHODS: Seven rat groups were included: non-diabetic control and non-diabetic receiving memantine; type-1 diabetic groups - untreated, treated with insulin alone and treated with insulin and memantine; and type 2 diabetic groups - untreated and memantine treated. Cognitive functions were assessed by the Morris Water Maze and passive avoidance test. Biochemical analysis was done for serum glucose, serum insulin and insulin resistance. Routine histological examination was done, together with immunohistochemistry for detection of the hippocampal learning and memory plasticity marker, namely activity regulated cytoskeletal-associated protein (Arc), and the astrocytes reactivity marker, namely glial fibrillary acidic protein (GFAP).  RESULTS: Both type 1 and 2 untreated diabetic groups showed significantly impaired cognitive performance compared to the non-diabetic group. Treating the type 1 diabetic group with insulin alone significantly improved cognitive performance, but significantly decreased GFAP and Arc compared to the untreated type 1 group. In addition, the type 2 diabetic groups showed a significant decrease in hippocampus GFAP and Arc compared to the non-diabetic groups. Blocking NMDA receptors by memantine significantly increased cognitive performance, GFAP and Arc in the type 1 insulin-memantine group compared to the type 1-insulin group and significantly increased Arc in the type 2-memantine group compared to the untreated type 2 diabetic group. The non-diabetic group receiving memantine was, however, significantly adversely affected. CONCLUSION: Cognitive functions are impaired in both types of diabetes mellitus and can be improved by blockage of NMDA receptors which may spark a future therapeutic role for these receptors in diabetes-associated cognitive dysfunction.

9.
Int. j. morphol ; 36(3): 969-974, Sept. 2018. graf
Article in English | LILACS | ID: biblio-954216

ABSTRACT

Kidney injury secondary to diabetes is the most common cause of kidney failure. We sought to determine whether pretreatment with the insulin-sensitizing drug metformin prior to the induction of diabetes can protect the kidney against the development of diabetic nephropathy (DN) induced by a combination of a high-fat diet and streptozotocin. Rats were either injected with vehicle (control group) or with a single injection of streptozotocin (STZ) (50 mg/kg) two weeks after being fed on a high-fat diet (HFD) (model group) and continued on HFD until being sacrificed 10 weeks post diabetic induction. The protective group that also fed on a HFD for 12 weeks was put on metformin (200 mg/kg/day) two weeks before STZ injection and continued on metformin until the sacrifice day. Harvested kidney tissues were examined by light microscopy after staining with hematoxylin and eosin (H&E) and periodic acid Schiff (PAS). Blood samples were assayed for sugar, urea, creatinine, and biomarkers of inflammation. Compared to a normal tissue histology in the control group, there was a profound damage to the kidney in the model group as demonstrated by markedly dilated capsular space, increased mesangial matrix expansion, congested blood vessels, and many tubular epithelial cells showing small pyknotic nuclei and vacuolated cytoplasm, which were significantly but not completely protected by metformin. Our findings also show that metformin significantly inhibited the inflammatory biomarkers, tumor necrosis factor-alpha (TNF-α) and C-reactive protein (CRP) induced by diabetes and HFD as well as significantly inhibiting blood sugar, urea, and creatinine. However, the levels of TNF-α, CRP, glucose, and creatinine in the metformin-treated group was still significant to the control group. Thus, we demonstrated an efficient but not complete protection by metformin pretreatment against DN induced by a combination of HFD and streptozotocin in rats.


La lesión renal secundaria a la diabetes es la causa más común de insuficiencia renal. Intentamos determinar si el pre tratamiento con metformina, un fármaco sensibilizante a la insulina antes de la inducción de diabetes, puede proteger al riñón del desarrollo de la nefropatía diabética (DN) inducida por una combinación de una dieta alta en grasas y estreptozotocina. Las ratas fueron inyectadas con el medio (grupo de control) o con una inyección única de estreptozotocina (STZ) (50 mg / kg) dos semanas después de ser alimentadas con una dieta alta en grasas (HFD) (grupo modelo) y continuaron en HFD hasta ser sacrificadas 10 semanas después de la inducción diabética. El grupo protector que también se alimentó con un HFD durante 12 semanas recibió metformina (200 mg / kg / día) dos semanas antes de la inyección de STZ y continuó con metformina hasta el día en que fueron sacrificadas. Las muestras de riñón se examinaron mediante microscopía óptica después de la tinción con Hematoxilina y Eosina y ácido peryódico de Schiff (PAS). Las muestras de sangre se analizaron para determinar niveles de azúcar, urea, creatinina y biomarcadores de inflamación. Comparado con una histología tisular normal en el grupo control, hubo un daño profundo al riñón en el grupo modelo como lo demuestra el espacio capsular marcadamente dilatado, el aumento de la expansión de la matriz mesangial, los vasos sanguíneos congestionados y muchas células epiteliales tubulares que muestran pequeños núcleos picnóticos y citoplasma vacuolado, que fueron significativamente pero no completamente protegidos por la metformina. Nuestros hallazgos también muestran que la metformina inhibe significativamente los biomarcadores inflamatorios, el factor de necrosis tumoral alfa (TNF-a) y la proteína C reactiva (PCR) inducida por diabetes y DFH, e inhibe significativamente el azúcar en sangre, la urea y la creatinina. Sin embargo, los niveles de TNF-a, CRP, glucosa y creatinina en el grupo tratado con metformina todavía eran significativos para el grupo de control. Por lo tanto, demostramos una protección eficiente pero no completa mediante pretratamiento con metformina contra DN inducida por una combinación de HFD y estreptozotocina en ratas.


Subject(s)
Animals , Male , Rats , Diabetic Nephropathies/drug therapy , Hypoglycemic Agents/administration & dosage , Metformin/administration & dosage , Dietary Fats , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/drug therapy , Disease Models, Animal
SELECTION OF CITATIONS
SEARCH DETAIL