Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Sci Rep ; 14(1): 11396, 2024 05 18.
Article in English | MEDLINE | ID: mdl-38762495

ABSTRACT

Acute liver injury, there is a risky neurological condition known as hepatic encephalopathy (HE). Herbacetin is a glycosylated flavonoid with many pharmacological characteristics. The purpose of this study was to assess the ability of herbacetin to protect against the cognitive deficits associated with thioacetamide (TAA) rat model and delineate the underlying behavioral and pharmacological mechanisms. Rats were pretreated with herbacetin (20 and 40 mg/kg) for 30days. On 30th day, the rats were injected with TAA (i.p. 350 mg/kg) in a single dose. In addition to a histpathological studies, ultra-structural architecture of the brain, liver functions, oxidative stress biomarkers, and behavioral tests were evaluated. Compared to the TAA-intoxicated group, herbacetin improved the locomotor and cognitive deficits, serum hepatotoxicity indices and ammonia levels. Herbacetin reduced brain levels of malodialdeyde, glutamine synthetase (GS), tumor necrosis factor- alpha (TNF-α), interleukin 1 B (IL-1ß), annexin v, and increased brain GSH, Sirtuin 1 (SIRT1), and AMP-activated kinase (AMPK) expression levels. Also, herbacetin improve the histopathological changes and ultra- structure of brain tissue via attenuating the number of inflammatory and apoptotic cells. Herbacetin treatment significantly reduced the toxicity caused by TAA. These findings suggest that herbacetin might be taken into account as a possible neuroprotective and cognitive enhancing agent due to its ability to reduce oxidative stress, inflammation and apoptosis associated with TAA.


Subject(s)
AMP-Activated Protein Kinases , Hepatic Encephalopathy , Neuroprotective Agents , Signal Transduction , Sirtuin 1 , Thioacetamide , Animals , Sirtuin 1/metabolism , Hepatic Encephalopathy/drug therapy , Hepatic Encephalopathy/metabolism , Hepatic Encephalopathy/chemically induced , Rats , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Signal Transduction/drug effects , AMP-Activated Protein Kinases/metabolism , Male , Oxidative Stress/drug effects , Up-Regulation/drug effects , Cognition/drug effects , Brain/metabolism , Brain/drug effects , Brain/pathology , Rats, Wistar , Liver/drug effects , Liver/metabolism , Liver/pathology , Disease Models, Animal
2.
Iran J Basic Med Sci ; 27(6): 657-670, 2024.
Article in English | MEDLINE | ID: mdl-38645500

ABSTRACT

Diabetic peripheral neuropathy (DPN) poses a significant threat, affecting half of the global diabetic population and leading to severe complications, including pain, impaired mobility, and potential amputation. The delayed manifestation of diabetic neuropathy (DN) makes early diagnosis challenging, contributing to its debilitating impact on individuals with diabetes mellitus (DM). This review examines the multifaceted nature of DPN, focusing on the intricate interplay between oxidative stress, metabolic pathways, and the resulting neuronal damage. It delves into the challenges of diagnosing DN, emphasizing the critical role played by hyperglycemia in triggering these cascading effects. Furthermore, the study explores the limitations of current neuropathic pain drugs, prompting an investigation into a myriad of pharmaceutical agents tested in both human and animal trials over the past decade. The methodology scrutinizes these agents for their potential to provide symptomatic relief for DPN. The investigation reveals promising results from various pharmaceutical agents tested for DPN relief, showcasing their efficacy in ameliorating symptoms. However, a notable gap persists in addressing the underlying problem of DPN. The results underscore the complexity of DPN and the challenges in developing therapies that go beyond symptomatic relief. Despite advancements in treating DPN symptoms, there remains a scarcity of options addressing the underlying problem. This review consolidates the state-of-the-art drugs designed to combat DPN, highlighting their efficacy in alleviating symptoms. Additionally, it emphasizes the need for a deeper understanding of the diverse processes and pathways involved in DPN pathogenesis.

3.
Toxicol Mech Methods ; 34(4): 454-467, 2024 May.
Article in English | MEDLINE | ID: mdl-38166588

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) has emerged as a major chronic liver illness characterized by increase of lipid content in the liver. This study investigated the role of lauric acid to treat NAFLD in male adult Sprague Dawley rats. In this study, to induce NAFLD in the rats, a high-fat diet (HFD) was administered for eight consecutive weeks. Lauric acid groups received lauric acid (250 and 500 mg/kg; orally), concurrently with HFD for eight consecutive weeks. Lauric acid could ameliorate the serum levels of TG, TC, ALT, AST, blood glucose, and insulin. Moreover, lauric acid significantly elevated the levels of SOD, GSH, catalase, and IL-10. Additionally, it lowered the hepatic levels of MDA, ROS, MPO, 4-HNE, interleukin (IL)-1ß, and tumor necrosis factor (TNF-α). Furthermore, lauric acid significantly up-regulated the hepatic expression of IRS1, AMPK, PI3K, and SIRT1 genes. In parallel, lauric acid could improve the histopathological picture of the liver and reduce the liver apoptosis via decreasing the expression of annexin V (Anx V). Finally, our data proposed that lauric acid could be an effective candidate for the NAFLD treatment.


Subject(s)
Lauric Acids , Non-alcoholic Fatty Liver Disease , Rats , Male , Animals , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/prevention & control , Non-alcoholic Fatty Liver Disease/etiology , Diet, High-Fat/adverse effects , Rats, Sprague-Dawley , Liver , Tumor Necrosis Factor-alpha/metabolism
4.
Article in English | MEDLINE | ID: mdl-37966574

ABSTRACT

Di-(2-ethylhexyl) phthalate (DEHP) is the most abundant phthalate threatening public health-induced neurotoxicity. This neurotoxicity is associated with behavioral and biochemical deficits in male rats. Our study investigated the neuroprotective effect of ferulic acid (FA) on male rats exposed to DEHP. Thirty-two male Wistar rats were assigned to four groups. Group I control rats received corn oil, group II intoxicated rats received 300 mg/kg of DEHP, group III received 300 mg/kg of DEHP + 50 mg/kg of FA, and group IV received 50 mg/kg of FA, all agents administrated daily per os for 30 days. Anxiety-like behavior, spatial working memory, and recognition memory were assessed. Also, brain oxidative stress biomarkers, including brain malondialdehyde (MDA), reduced glutathione (GSH), nitric oxide (NO), superoxide dismutase (SOD), brain-derived neurotrophic factor (BDNF) as well as heme oxygenase-1 (HO-1) were measured. Moreover, brain histopathology examinations associated with immunohistochemistry determination of brain caspase-3 were also evaluated. Furthermore, docking simulation was adapted to understand the inhibitory role of FA on caspase-3 and NO synthase. Compared to DEHP-intoxicated rats, FA-treated rats displayed improved cognitive memory associated with a reduced anxious state. Also, the redox state was maintained with increased BNDF levels. These changes were confirmed by restoring the normal architecture of brain tissue and a decrement in the immunohistochemistry caspase-3. In conclusion, FA has potent antioxidant and antiapoptotic properties that confirm the neuroprotective activity of FA, with a possible prospect for its therapeutic capabilities and nutritional supplement value.

5.
Article in English | MEDLINE | ID: mdl-37950769

ABSTRACT

Links between bronchial asthma and depression have recently become a great subject of interest. The present study was carried out to assess the protective role of hesperidin against ovalbumin (OVA)-induced bronchial asthma that is associated with depression in rats, for this purpose, four groups. Rats were sensitized with intraperitoneal administration of 200 µg OVA/10 mg aluminum hydroxide (Al (OH) 3 for 3 consecutive days then at day 11 followed by intranasal challenge with OVA (1.5 mg/kg) at days 19, 20, and 21. Rats were pretreated with hesperidin (100 & 200 mg/kg) 1h before OVA challenge. At the end of the study, behavioral tests, biochemical indices, and histopathological architectures of lung and brain tissues were evaluated. Our findings showed that hesperidin significantly ameliorated the reduction in motor activity, motor coordination, forced swimming, CD4, CD25 and foxp3, interleukin-10 (IL-10), dopamine, serotonin, and neurotrophin-3 (NT3) as well as alleviated the elevation in transforming growth factor-beta (TGF-ß), tumor necrosis factor-alpha (TNF-α), iL-5, and immunoglobulin E (IgE). In addition, hesperidin reduced cellular infiltration, alveolar sacs damage, the bronchioles wall disruption, and nuclei pyknosis in neuron cells. Finally, hesperidin may provide protection against OVA-induced asthma and depression. This impact could be mediated in part by its anti-inflammatory and immunoregulatory properties.

6.
J Drug Target ; 31(10): 1081-1097, 2023 12.
Article in English | MEDLINE | ID: mdl-37886815

ABSTRACT

One of the most prevalent worldwide problems that affect all ages and genders is skin burn. The goal of our study was to assess the ability of curcumin nanoparticles to cure a rat burn model. Three formulations were selected after several tests were performed including investigation of encapsulation efficiency, particle size and zeta potential measurements. In vitro release was achieved on the three selected formulations. The effectiveness of the chosen formulation for healing was evaluated. The induced burn wound was smeared, starting just after excision, once daily with curcumin nanoparticles for 18 days. Our findings revealed that curcumin nanoparticles improved the burn healing potential by augmenting the skin regeneration indices as evidenced by enhancing the new production of hyaluronic acid and collagen type I. Additionally, curcumin nanoparticles could increase levels of vascular endothelial growth factor and alpha smooth muscle activity while drastically reducing the skin's tumour necrosis factor content, revealing a significant potential for burn healing process that is also reflected in the histopathological and immunohistochemical studies. Finally, our results demonstrated that curcumin nanoparticles revealed a significant potential for burn healing than curcumin alone due to its potent antimicrobial, antioxidant and anti-inflammatory properties.


Subject(s)
Burns , Chitosan , Curcumin , Nanoparticles , Tamarindus , Female , Rats , Male , Animals , Chitosan/chemistry , Curcumin/pharmacology , Curcumin/therapeutic use , Vascular Endothelial Growth Factor A , Nanoparticles/chemistry , Burns/drug therapy
7.
Iran J Basic Med Sci ; 26(8): 941-952, 2023.
Article in English | MEDLINE | ID: mdl-37427328

ABSTRACT

Objectives: Our study was conducted to evaluate the synergistic effect of arginine (ARG) and Lactobacillus plantarum against potassium dichromate (K2Cr2O7) induced-acute hepatic and kidney injury. Materials and Methods: Fifty male Wistar rats were divided into five groups. The control group received distilled water. The potassium dichromate group (PDC) received a single dose of PDC (20 mg/kg; SC). The arginine group (ARG) and Lactobacillus plantarum group received either daily doses of ARG (100 mg/kg, PO) or L. plantarum (109 CFU/ml, PO) for 14 days. The combination group (ARG+L. plantarum) received daily doses of ARG (100 mg/kg) with L. plantarum (109 CFU/ml), orally for 14 days, before induction of acute liver and kidney injury. Forty eight hours after the last dose of PDC, serum biochemical indices, oxidative stress biomarkers, pro-inflammatory cytokines, histopathological and immunohistochemical analysis were evaluated. Results: Combining ARG with L. plantarum restored the levels of serum hepatic & kidney enzymes, hepatic & renal oxidative stress biomarkers, and TLR 4/ NF-κB signaling pathway. Furthermore, they succeeded in decreasing the expression of iNOS and ameliorate the hepatic and renal markers of apoptosis: Caspase-3, Bax, and Bcl2. Conclusion: This study depicts that combining ARG with L. plantarum exerted a new bacteriotherapy against hepatic and renal injury caused by PDC.

8.
Int Immunopharmacol ; 122: 110634, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37451012

ABSTRACT

Wound healing is a series of coordinated events that involve tissue repair and regeneration. Cold atmospheric plasma approach sheds the light on the mechanism that initiates the inflammatory responses throughout the healing cascade. The present study was planned to assess the effect of thymoquinone treated with cold plasma (TQcp) on the rat wound model compared to thymoquinone (TQ). To assess the wound healing potential of TQcp, a full-thickness wound model was used. The induced wound was smeared, starting just after excision, twice daily with TQcp and TQ for 7 days. Our findings revealed that TQcp improved the skin healing potential by augmenting the skin regeneration indices as evidenced by enhancing the new production of hyaluronic acid and collagen type I. TQcp significantly reduced the skin content of tumor necrosis factor- α and inhibited the hypertrophic scarring by up-regulating the skin content of transforming growth factor-beta. Furthermore, TQcp enhanced the levels of interleukin-10, alpha smooth muscle actin and vascular endothelial growth factor, demonstrating a great potential for wound healing that also reflected in the histopathological and ultra-structural picture of the skin. Finally, our results demonstrated that TQcp revealed a significant potential for wound healing than TQ alone.


Subject(s)
Plasma Gases , Vascular Endothelial Growth Factor A , Rats , Animals , Vascular Endothelial Growth Factor A/metabolism , Plasma Gases/metabolism , Plasma Gases/pharmacology , Actins/metabolism , Wound Healing , Skin/pathology , Tumor Necrosis Factor-alpha/metabolism
9.
Environ Sci Pollut Res Int ; 30(35): 84791-84804, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37378730

ABSTRACT

The aim of the present study was to investigate the impact of arginine (ARG), a nitric oxide (NO) precursor, on thioacetamide (TAA)-induced hepatic encephalopathy (HE) in rats by injection of TAA (100 mg/kg, i.p) three times per week for six consecutive weeks. TAA-injected rats were administered ARG (100 mg/kg; p.o.) concurrently with TAA for the six consecutive weeks. Blood samples were withdrawn, and rats were sacrificed; liver and brain tissues were isolated. Results of the present study demonstrated that ARG administration to TAA-injected rats revealed a restoration in the serum and brain ammonia levels as well as serum aspartate transaminase, alanine transaminase, and alkaline phosphatase and total bilirubin levels as well as behavioral alterations evidenced by restoration in locomotor activity, motor skill performance, and memory impairment. ARG showed also improvement in the hepatic and neuro-biochemical values, pro-inflammatory cytokines, and oxidative stress biomarkers. All these results were confirmed by histopathological evaluation as well as ultrastructural imaging of the cerebellum using a transmission electron microscope. Furthermore, treatment with ARG could ameliorate the immunological reactivity of nuclear factor erythroid-2-related factor 2 (Nrf2) and cleaved caspase-3 proteins in the cerebellum and hepatic tissues. From all the previous results, it can be fulfilled that ARG showed a beneficial role in modulating the adverse complications associated with TAA-induced HE in rats via reducing hyperammonemia and downregulating nuclear factor kappa B (NF-κB)-mediated apoptosis.


Subject(s)
Hepatic Encephalopathy , Rats , Animals , Hepatic Encephalopathy/chemically induced , NF-kappa B/metabolism , Thioacetamide/toxicity , Nitric Oxide/metabolism , Down-Regulation , Liver/metabolism , Arginine/adverse effects , Arginine/metabolism , Oxidative Stress
10.
Environ Sci Pollut Res Int ; 30(35): 83356-83375, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37340161

ABSTRACT

Aluminum (Al) is a ubiquitous xenobiotic with known toxicity for both humans and animals. Our study was conducted to investigate the protective role of febuxostat (Feb) against aluminum chloride (AlCl3)-induced hepatorenal injury in rats. Hepatorenal injury was induced by oral administration of AlCl3 (40 mg/kg b.w.), for 2 months. Twenty-four male Sprague-Dawley rats were randomly allocated into four groups (six rats/group). The first group received the vehicle thought the experiment. The second group was considered as a control positive group. The third and fourth groups received oral treatment of Feb (10 mg/kg.b.w.) and (15 mg/kg.b.w.), respectively with AlCl3, concurrently for 2 months. Twenty-four hours, after the last treatment, serum biochemical, molecular, histopathology, and immunohistochemical studies were evaluated. Our findings showed that rats intoxicated with Alcl3 had disturbed biochemical picture. In addition, intoxication with AlCl3 increased oxidative stress and apoptosis, as demonstrated by an increase in malodialdeyde (MDA), carnitine o-acetyltransferase (Crat), and carbonic anhydrase (Car3) with a decrease in glutathione (GSH), MAP kinase-interacting serine/threonine kinase (MNK) and nuclear factor-erythroid 2-related factor 2 (Nrf2) mRNA expression. Furthermore, the levels of tumor necrosis factor-alpha (TNF-α) and the levels of caspase-3 were elevated with sever hepatic and renal pathological changes. Conversely, Feb (15 mg/kg.b.w.) could improve the serum biochemical indices and repressed MDA, Crat, and Car3 levels, whereas it increased GSH, MNK, and Nrf2 levels. Feb inhibited the apoptotic effect of AlCl3 in the liver and kidney by decreasing caspase-3 and TNF-α expression. The protective effect of Feb against AlCl3 toxicity was confirmed by histopathological findings. Moreover, molecular docking studies supported the anti-inflammatory effect of Feb due to its significant binding interactions with cyclooxygenase-1 (COX-1), NF-kappa-B-inducing kinase (NIK), and mitogen-activated protein kinases-p38 (MAPK-p38). The findings suggest that Feb system Feb can avert Alcl3-induced hepatotoxicity and nephrotoxicity by enhancing the antioxidant defense system, and inhibiting the inflammatory cascade and apoptosis.


Subject(s)
Febuxostat , NF-E2-Related Factor 2 , Humans , Rats , Male , Animals , Aluminum Chloride/metabolism , Febuxostat/pharmacology , Febuxostat/metabolism , Caspase 3/metabolism , NF-E2-Related Factor 2/metabolism , Carnitine O-Acetyltransferase/metabolism , Carnitine O-Acetyltransferase/pharmacology , Rats, Sprague-Dawley , Tumor Necrosis Factor-alpha/metabolism , Molecular Docking Simulation , Antioxidants/metabolism , Liver , Oxidative Stress , Aluminum/metabolism , Glutathione/metabolism , Apoptosis
11.
Toxicol Appl Pharmacol ; 468: 116515, 2023 06 01.
Article in English | MEDLINE | ID: mdl-37061009

ABSTRACT

Di -(2-ethylhexyl) phthalate (DEHP) is a widely used phthalate that possesses a public health concern. Different concentrations of DEHP, including 50, 300, and 750 mg/kg were administrated orally for 28 days in male rats. Body weight and vital organs weight were measured as well as anxiety-like behavior, short and long-term memory were investigated. Brain inflammatory cytokines, including IL-1ß, TLR4, NF-κB, TNF-α, and IL1-6 were assessed. Brain caspase-3, neuropeptide-Y (NPY), and brain histopathology were also evaluated. DEHP triggers the release of pro-inflammatory cytokines via inducing the nuclear translocation of the signaling pathway; TLR 4/ NF-κB leads to cognitive impairment and neurodegeneration, which is confirmed by the impaired brain architecture. Also, DEHP upgrades the expression levels of brain caspase-3 and NPY. In conclusion, exposure to high doses of DEHP persuades great toxicity visualized by behavioral, biochemical, and histological impairments when compared to the low doses.


Subject(s)
Diethylhexyl Phthalate , NF-kappa B , Rats , Animals , Male , NF-kappa B/metabolism , Diethylhexyl Phthalate/toxicity , Caspase 3/metabolism , Toll-Like Receptor 4 , Signal Transduction , Cytokines/metabolism
12.
Inflammopharmacology ; 31(3): 1465-1480, 2023 Jun.
Article in English | MEDLINE | ID: mdl-36884189

ABSTRACT

Potassium dichromate (PD) is an environmental xenobiotic commonly recognized as teratogenic, carcinogenic, and mutagenic in animals and humans. The present study was conducted to investigate the role of tangeretin (TNG) as a neuro-protective drug against PD-induced brain injury in rats. Thirty-two male adult Wistar rats were blindly divided into four groups (8 rats/group). The first group received saline intranasally (i.n.). The second group received a single dose of PD (2 mg/kg, i.n.). The third group received TNG (50 mg/kg; orally), for 14 days followed by i.n. of PD on the last day of the experiment. The fourth group received TNG (100 mg/kg; orally) for 14 days followed by i.n. of PD on the last day of the experiment. Behavioral indices were evaluated 18 h after PD administration. Neuro-biochemical indices and histopathological studies were evaluated 24 h after PD administration. Results of the present study revealed that rats intoxicated with PD induced- oxidative stress and inflammation via an increase in malondialdehyde (MDA) and a decrease in nuclear factor erythroid 2-related factor 2 (Nrf2) signaling pathway and glutathione(GSH) levels with an increase in brain contents of tumor necrosis factor-alpha (TNF-α) and interleukin (IL-6). Pre-treatment with TNG (100 mg/kg; orally) ameliorated behavior, cholinergic activities, and oxidative stress and decreased the elevated levels of pro-inflammatory mediators; TNF-α and IL-6 with a decrease in brain content of chromium residues detected by Plasma-Optical Emission Spectrometer. Also, the histopathological picture of the brain was improved significantly in rats that received TNG (100 mg/kg). Additionally, TNG decreased caspase-3 expression in the brain of PD rats. In conclusion, TNG possesses a significant neuroprotective role against PD-induced acute brain injury via modulating the Nrf2 signaling pathway and quenching the release of inflammatory mediators and apoptosis in rats.


Subject(s)
Brain Injuries , Neuroprotective Agents , Humans , Rats , Male , Animals , Rats, Wistar , NF-E2-Related Factor 2/metabolism , Neuroprotective Agents/pharmacology , Inflammation Mediators/metabolism , Tumor Necrosis Factor-alpha/metabolism , Chromium/pharmacology , Interleukin-6/metabolism , Signal Transduction , Oxidative Stress , Glutathione/metabolism , Apoptosis
13.
Can J Physiol Pharmacol ; 99(3): 332-347, 2021 Mar.
Article in English | MEDLINE | ID: mdl-32721224

ABSTRACT

Hepatic encephalopathy depicts the cluster of neurological alterations that occur during acute or chronic hepatic injury. Hyperammonemia, inflammatory injury, and oxidative stress are the main predisposing factors for the direct and indirect changes in cerebral metabolism causing encephalopathy. The aim of this study was to evaluate the possible synergistic effect between aminoguanidine (AG; 100 mg/kg, p.o.) and l-carnosine (CAR; 200 mg/kg, p.o.) on hepatic encephalopathy that was induced by thioacetamide (TAA; 100 mg/kg, i.p.) administered three times weekly for six weeks. Behavioral changes, biochemical parameters, histopathological analysis, and immunohistochemical and ultrastructural studies were conducted 24 h after the last treatment. Combining AG with CAR improved TAA-induced locomotor impairment and motor incoordination evidenced by reduced locomotor activity and decline in motor skill performance, as well as ameliorated cognitive deficits. Moreover, both drugs restored the levels of serum hepatic enzymes and serum and brain levels of ammonia. In addition, the combination significantly modulated hepatic and brain oxidative stress biomarkers, inflammatory cytokines, and cleaved caspase-3 expression. Furthermore, they succeeded in activating nuclear erythroid 2-related factor 2 (Nrf2) expression and heme oxygenase-1 (HO-1) activity and ameliorating markers of hepatic encephalopathy, including hepatic necrosis and brain astrocyte swelling. This study shows that combining AG with CAR exerted a new intervention for hepatic and brain damage in hepatic encephalopathy due to their complementary antioxidant, anti-inflammatory effects and hypoammonemic effects via Nrf2/HO-1 activation and NO inhibition.


Subject(s)
Carnosine/therapeutic use , Guanidines/therapeutic use , Hepatic Encephalopathy/prevention & control , Thioacetamide , Ammonia/metabolism , Animals , Behavior, Animal , Brain/pathology , Brain Chemistry/drug effects , Drug Synergism , Hepatic Encephalopathy/chemically induced , Hepatic Encephalopathy/psychology , Liver/pathology , Liver Function Tests , Male , Motor Activity/drug effects , Motor Skills/drug effects , Rats , Rats, Wistar
14.
Can J Physiol Pharmacol ; 95(4): 427-436, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28157387

ABSTRACT

The present study aimed to evaluate the effect of trigonelline (TRG) on the hepatic complications associated with high-fat high-fructose (HFHF) diet-induced insulin resistance (IR) in rats. IR was induced by giving a saturated fat diet and 10% fructose in drinking water to rats for 8 weeks. Insulin-resistant rats were orally treated with TRG (50 and 100 mg/kg), sitagliptin (SIT; 5 mg/kg), or a combination of TRG (50 mg/kg) and SIT (5 mg/kg) for 14 days. Liver homogenates were used for assessment of hepatic lipids, oxidative stress biomarkers, and inflammatory cytokines. Histopathological and DNA cytometry examinations were carried out for hepatic and pancreatic tissues. Hepatic tissues were examined using Fourier-transform infrared spectroscopy for assessment of any molecular changes. Results of the present study revealed that oral treatment of insulin-resistant rats with TRG or TRG in combination with SIT significantly decreased homeostatic model assessment of IR, hepatic lipids, oxidative stress biomarkers, and the inflammatory cytokines. TRG or TRG in combination with SIT ameliorated the histopathological, DNA cytometry, and molecular alterations induced by a HFHF diet. Finally, it can be concluded that TRG has beneficial effects on the hepatic complications associated with IR due to its hypoglycemic effect and antioxidant potential.


Subject(s)
Alkaloids/therapeutic use , Antioxidants/therapeutic use , Hypoglycemic Agents/therapeutic use , Metabolic Syndrome/complications , Non-alcoholic Fatty Liver Disease/drug therapy , Sitagliptin Phosphate/therapeutic use , Animals , Biomarkers/analysis , Blood Glucose/analysis , Cytokines/analysis , Diet, Carbohydrate Loading/adverse effects , Diet, High-Fat/adverse effects , Dipeptidyl-Peptidase IV Inhibitors/therapeutic use , Disease Models, Animal , Drug Evaluation, Preclinical , Drug Therapy, Combination , Humans , Lipids/analysis , Liver/drug effects , Liver/metabolism , Liver/pathology , Male , Metabolic Syndrome/etiology , Non-alcoholic Fatty Liver Disease/etiology , Oxidative Stress/drug effects , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...