Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Chem Biol Interact ; 398: 111065, 2024 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-38795875

RESUMEN

Multiple cycles of cisplatin result in a permanent loss of kidney function with severe and life-limited chronic kidney disease (CKD) after successful cisplatin therapy. Recently, studies have showed that the activation of G-protein coupled estrogen receptor (GPER) could protect against kidney disease. This study aimed to test the potential of the G1 compound, a GPER selective agonist, to prevent CKD development after cisplatin therapy. Male C57BL/6 mice were exposed to 2 cycles of 2.5 mg/kg cisplatin in a regimen miming clinical exposure (1 injection daily for 5 days, followed by a 16-day recovery period between cycles). G1 (50 or 100 µg/kg) was administered daily for 6 weeks. G1 dose-dependently improved kidney function biomarkers (serum creatinine, creatinine clearance, and protein excretion) and histopathological changes compared to the cisplatin-treated group. Collagen 3 expression was dose-dependently decreased in G1-treated groups that was parallel to the reduction of fibrosis in Masson's trichrome-stained sections. G1 administration also increased total antioxidant capacity (TAC) and nuclear factor erythroid 2-related factor 2 (Nrf2) and reduced the level of malondialdehyde and the proinflammatory cytokine, tumor necrosis factor-α. In addition, G1 downregulated the expression of inflammasome NLRP3 and nuclear factor kappa B p65 (NF-κB p65) in a dose-dependent manner. In conclusion, these data suggest that G1 could be a new therapeutic tool for CKD prevention post cisplatin therapy. These effects might be mediated through the activation of Nrf2 and the inhibition of NF-κB/NLRP3 signaling.

2.
J Biochem Mol Toxicol ; 37(3): e23266, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36468814

RESUMEN

The current study was designed to investigate the potential anti-inflammatory and antioxidant effects of fingolimod against Ovalbumin (Ova)-induced allergic airway inflammation compared to dexamethasone. Fingolimod was given (0.5 mg/kg/day, p.o.) for sensitized mice 1 h before Ova challenge from Days 19 to 24. Fingolimod significantly inhibited Ova-induced elevation of inflammatory cells and eosinophils numbers in bronchoalveolar lavage fluid (BALF) and reduced concentrations of immunoglobulin E in serum and of sphingosine-1-phosphate, interleukin (IL)-4, and IL-13 in BALF. Fingolimod inhibited microvascular leakage and edema as reflected by the decreased lung/body weight index. These findings were supported by histopathological examination results showing that fingolimod substantially decreased perivascular edema and inflammatory cell infiltration. Fingolimod also attenuated Ova-induced oxidative stress as evidenced by decreased malondialdehyde concentration along with increasing concentrations of reduced glutathione and superoxide dismutase in lung tissues. Fingolimod also significantly decreased monocyte chemoattractant protein-1 (MCP-1), p-ERK, and p-P38 in lung tissues of Ova-challenged mice. In conclusion, the current study demonstrated the anti-inflammatory and antioxidant effects of fingolimod in allergic airway inflammation that might be associated with the downregulation of mitogen activated kinases signaling to decrease T helper 2 cytokine secretion (IL-4 and IL-13) and MCP-1 expression, along with the inhibition of oxidative stress.


Asunto(s)
Asma , Animales , Ratones , Antiinflamatorios/farmacología , Antioxidantes/metabolismo , Líquido del Lavado Bronquioalveolar , Citocinas/metabolismo , Modelos Animales de Enfermedad , Clorhidrato de Fingolimod , Inflamación/metabolismo , Interleucina-13 , Pulmón/metabolismo , Ratones Endogámicos BALB C , Ovalbúmina , Sistema de Señalización de MAP Quinasas
3.
Basic Clin Pharmacol Toxicol ; 130(5): 569-580, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-35274449

RESUMEN

BACKGROUND: Because of the approved immunomodulatory activities of fingolimod, the current study aimed at studying the curative potential of fingolimod against experimentally induced ulcerative colitis (UC) via modulating pro/anti-inflammatory cytokines release and AKT/mTOR signalling. METHODS: UC was induced in rats by intracolonic instillation of acetic acid. Fingo (0.5 mg/kg/day, p.o.) was given for 8 consecutive days that started 48 h after UC induction. RESULTS: Fingolimod increased body weight growth rate and colon body/weight and colon weight/length indices compared to the UC group. Fingolimod significantly decreased clinical evaluation score and macroscopic score compared to the UC group. The curative potential of fingolimod was further confirmed by histopathological examination revealing marked attenuation of mucosal injury and inflammatory cells infiltration. Fingolimod significantly decreased colon malondialdehyde content and increased colon glutathione contents compared to the UC group. Fingolimod also significantly decreased the expressions of pro-inflammatory cytokines interleukin-9 and T-helper 17 along with increasing the expression of anti-inflammatory interleukin-10 and transforming growth factor-ß compared to the UC group. In addition, fingolimod decreased the expressions of AKT and mTOR compared to the UC group. CONCLUSION: Fingolimod attenuated acetic acid-induced UC through its immunomodulatory effect by shifting the balance to favour anti-inflammatory cytokine production rather than pro-inflammatory cytokines and modulating the AKT/mTOR signalling.


Asunto(s)
Colitis Ulcerosa , Ácido Acético/metabolismo , Ácido Acético/toxicidad , Animales , Antiinflamatorios/efectos adversos , Colitis Ulcerosa/inducido químicamente , Colitis Ulcerosa/tratamiento farmacológico , Colitis Ulcerosa/metabolismo , Colon/metabolismo , Citocinas/metabolismo , Clorhidrato de Fingolimod/efectos adversos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Serina-Treonina Quinasas TOR/metabolismo
4.
Environ Sci Pollut Res Int ; 29(4): 5812-5826, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34431048

RESUMEN

There is increasing interest in the use of natural products to treat many diseases, considering the minimal toxicity, availability, and low cost. Propolis, a natural resinous product produced by honeybees, has been proven for its antioxidant and anti-inflammatory properties. Therefore, this study was designed to investigate the protective potential of propolis extract against nicotine-induced pulmonary and hepatic damage in rats. Sprague Dawley rats were divided into six groups: control, propolis (200 and 300 mg/kg, p.o.), nicotine (10 mg/kg, i.p), and nicotine plus propolis-treated groups. Nicotine and propolis were given every day for 8 weeks. Then, blood and bronchoalveolar lavage fluid (BALF) were collected for assessing liver and lung functions. Liver and lung tissues were also harvested to assess oxidative stress and inflammatory biomarkers in addition to histopathological and immunohistochemical analysis. Both doses of propolis significantly decreased AST, ALT, ALP, and total and differential cell counts in a dose-dependent manner. Propolis extract significantly attenuated oxidative stress in both lung and liver tissues. The restoration of antioxidant status (GSH level, SOD activities) and reduction of nitric oxide and MDA content was more so in propolis 300-treated than propolis 200-treated group. This was parallel to the improvement seen in histopathological examination. Propolis 200 and 300 significantly decreased Nrf2 expression and increased HO-1 expression in a dose-dependent manner. Moreover, immunohistochemical examination revealed that propolis 200 and 300 decreased the expression of iNOS in lung and liver tissues while decreased TNF-α expression in lung tissues only. Propolis extract could have a protective potential against nicotine-induced pulmonary and hepatic damage via activating Nrf2/HO-1 signaling.


Asunto(s)
Própolis , Animales , Antioxidantes , Pulmón , Nicotina , Estrés Oxidativo , Extractos Vegetales , Própolis/farmacología , Ratas , Ratas Sprague-Dawley
5.
Hum Exp Toxicol ; 40(12_suppl): S573-S582, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34802289

RESUMEN

Inflammation and oxidative stress are the major pathways involved in ischemia-reperfusion (I/R)-induced renal injury. This study was designed to evaluate the potential effect of pomegranate against I/R-induced renal injury. I/R injury was induced in nephrectomized rats by unilateral occlusion of the left renal pedicle for 45 min followed by 24 h of perfusion. Pomegranate succeeded to decrease serum levels of creatinine, potassium, and urea nitrogen, along with increasing creatinine clearance. Pomegranate also decreased I/R-induced changes in histopathological examination. Pomegranate attenuated the renal inflammatory response reflected by the suppression of nuclear factor κB p65 DNA binding activity, the upregulation of inhibitory protein kappa B-alpha mRNA expression, the downregulation of mRNA and protein expression of tumor necrosis factor α, in addition to the reduced myeloperoxidase activity and mRNA expression. Additionally, pomegranate attenuated oxidative stress likely through the modulation of lipid peroxidation and antioxidant levels reflected by the decreased MDA content and the increased glutathione level and superoxide dismutase activity. Results confirm the potential protective effect of pomegranate against I/R-induced renal injury through its anti-inflammatory and anti-oxidant effects mediated through the upregulation of inhibitory protein kappa B-alpha, the inhibition of NF-κB activity, and the associated TNF-α release, neutrophil infiltration, and oxidative stress.


Asunto(s)
Riñón/irrigación sanguínea , FN-kappa B/metabolismo , Extractos Vegetales/farmacología , Granada (Fruta)/química , Daño por Reperfusión/prevención & control , Animales , Ratas
6.
J Biochem Mol Toxicol ; 35(8): e22817, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34047436

RESUMEN

Cisplatin is a chemotherapeutic agent whose therapeutic use is greatly limited by the associated organs' toxicity and particularly, testicular toxicity. Cisplatin-induced testicular damage reported being mediated through mitochondria-mediated apoptosis, inflammation, and oxidative stress. Evidence showed that tranilast (TRN) has the ability to restore the oxidative status and modulate TRAIL/caspase-8 signaling. This led us to hypothesize that TRN could abrogate cisplatin-induced testicular and epididymal injuries via inhibiting oxidative stress and modulating proliferation and TRAIL/caspase-8/cJNK signaling. Cisplatin injection induced oligospermia and abnormalities in testicular and epididymal structure along with impaired oxidative status. TRN administration (100 or 300 mg/kg) for 7 days post-cisplatin injection preserved spermatogenesis and restored testicular and epididymal architecture, but restoration was more so in TRN300 than TRN100. This was in line with the restoration of balanced oxidative status as indicated by the increased total antioxidant capacity, glutathione and superoxide dismutase activity, and the decreased malondialdehyde content in testes (p < 0.05 vs. cisplatin). TRN increased the cell proliferation revealed by the increased expression of proliferating cell nuclear antigen in a dose-dependent manner (p < 0.05 vs. cisplatin) whereas only TRN300 decreased testicular cJNK, TRAIL, and caspase-8 expression (p < 0.05 vs. cisplatin). Moreover, TRN dose-dependently inhibited the pro-inflammatory transcription factor NF-kB and the cytokine TNF-α expressions in testes. In conclusion, TRN300 was more effective than TRN100 in alleviating cisplatin-induced testicular and epididymal injuries and in enhancing spermatogenesis. This curative effect of TRN might be mediated through its antioxidant and anti-inflammatory impacts along with its modulatory impact on cJNK/TRAIL/caspase-8 signaling favoring proliferation rather than apoptosis.


Asunto(s)
Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Cisplatino/efectos adversos , Oligospermia , ortoaminobenzoatos/farmacología , Animales , Cisplatino/farmacología , Epidídimo/lesiones , Epidídimo/metabolismo , Masculino , Oligospermia/inducido químicamente , Oligospermia/metabolismo , Oligospermia/prevención & control , Ratas , Ratas Sprague-Dawley , Testículo/lesiones , Testículo/metabolismo
7.
Fundam Clin Pharmacol ; 35(6): 940-954, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33829539

RESUMEN

Colon rectal cancer (CRC) is the second commonest malignancy in developed countries and a significant cause of mortality. Tenofovir reportedly reduces the risk of hepatocellular carcinoma and interferes with cell cycle and cell proliferation. The current study investigated the potential antitumor effect of tenofovir against experimentally induced CRC. CRC was induced by 1,2-dimethylhydrazine (DMH, 20 mg/kg, once a week) and high-fat diet (HFD) in Wistar rats. Rats received tenofovir at a dose of 25 or 50 mg/kg (i.p.) for 24 weeks. Tenofovir-25 failed to significantly decrease the total number of dysplasia, adenoma and adenocarcinoma and to improve histopathological changes; however, tenofovir-50 resulted in no tumors seen in the colon lumen and a significant decrease in the total number of dysplasia and no adenoma or adenocarcinoma observed compared to DMH/HFD group. Tenofovir-25 failed to attenuate DMH/HFD-induced cell proliferation, whereas tenofovir-50 significantly decreased cell proliferation revealed by the decreased PCNA expression. Tenofovir-25 also failed to attenuate DMH/HFD-induced oxidative stress, whereas tenofovir-50 significantly attenuated oxidative stress as indicated by the decreased MDA concentration and SOD activity along with the increased GSH concentrations. Moreover, tenofovir-50 decreased Bcl-2 and cyclin D1 expressions in colon tissues compared with DMH/HFD group. Tenofovir-50 also significantly decreased INF-ɤ concentration in colon tissues. These findings suggest that the high dose of tenofovir (50 mg/kg) has antitumor potential against DMH/HFD-induced CRC, which might be mediated through the inhibition of cell proliferation, oxidative stress, and inflammation.


Asunto(s)
Neoplasias del Colon , Inhibidores de la Transcriptasa Inversa , Animales , Ratas , 1,2-Dimetilhidrazina , Colon , Neoplasias del Colon/inducido químicamente , Neoplasias del Colon/tratamiento farmacológico , Dieta Alta en Grasa , Ratas Wistar , Inhibidores de la Transcriptasa Inversa/uso terapéutico , Tenofovir
8.
J Biochem Mol Toxicol ; 35(5): e22745, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33749060

RESUMEN

Liver fibrosis is a public health burden that is highly associated with morbidity and mortality. Therefore, this study aims to explore the anti-fibrotic effects of low dose of paclitaxel (PTX) against thioacetamide (TAA)-induced liver fibrosis in rats and the possible mechanisms involved. TAA was administered at a dose of 200 mg/kg twice weekly for 6 weeks in rats to induce liver fibrosis similar to that in humans. Liver dysfunction was shown by increased alanine aminotransferase, aspartate aminotransferase, alkaline phosphatase, and γ-glutamyl transferase, along with histopathological changes. Liver fibrosis was confirmed by Masson's Trichome staining, increased collagen content, and elevated α-smooth muscle actin (α-SMA) protein expression. In addition, TAA induced liver apoptosis as indicated by the increased terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL)-positive cells in liver tissues. This study demonstrated that the administration of PTX (0.3 mg/kg/i.p.) three times a week for 6 weeks significantly alleviated functional and biochemical changes induced by TAA in addition to improving the liver architecture. PTX attenuated liver fibrosis as reflected by the decreased collagen content and α-SMA protein expression. Additionally, PTX attenuated liver apoptosis as indicated by the decreased TUNEL-positive cells. Moreover, PTX prevented TAA-induced elevation of transforming growth factor-ß1 (TGF-ß1), platelet-derived growth factor-BB (PDGF-BB), and tissue inhibitor of metalloproteinase 1 (TIMP-1) levels in liver tissues. These findings suggest that the low dose of PTX prevented TAA-induced liver fibrosis in rats, possibly by inhibiting the expression of TGF-ß1 and PDGF-BB and subsequently suppressing the apoptosis and the expression of TIMP-1.


Asunto(s)
Cirrosis Hepática , Paclitaxel/farmacología , Tioacetamida/toxicidad , Animales , Modelos Animales de Enfermedad , Cirrosis Hepática/inducido químicamente , Cirrosis Hepática/metabolismo , Cirrosis Hepática/prevención & control , Masculino , Ratas , Ratas Sprague-Dawley
9.
Life Sci ; 271: 119154, 2021 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-33539910

RESUMEN

Ulcerative colitis (UC) is a chronic inflammatory bowel disease that increases the risk of colorectal cancer. UC is highly associated with the disturbance of the immune system leading to oxidative stress and chronic inflammation of intestine. Therefore, the current study was conducted to investigate the potential anti-oxidant and anti-inflammatory effects of MMF against acetic acid-induced UC that might be associated with the regulation of Nrf-2 and NLRP3 inflammasome signaling. UC was induced in Sprague-Dawley rats by intracolonic instillation of acetic acid. Forty-eight hours post UC induction, MMF (50 mg/kg/day, orally) was given for 8 consecutive days. Then, colon tissues and blood samples were collected. Results showed that MMF significantly attenuated the acetic acid-induced functional, biochemical, and inflammatory injuries in colon. MMF significantly decreased oxidative stress as indicated by the decreased malondialdehyde concentration and the increased total antioxidant capacity, glutathione, catalase, and superoxide dismutase concentrations in colon tissues. MMF also significantly increased Nrf-2 and decreased NLRP3 inflammasome expressions. Moreover, MMF decreased expression of interferon-gamma and increased expression of interferon-alpha. MMF also significantly decreased expression of pro-inflammatory cytokines, interleukin (IL)-1ß and IL-18. These results suggest that MMF has antioxidant and anti-inflammatory effects against acetic acid-induced UC through the upregulation of Nrf-2, and INF-α expression in addition to the suppression of NLRP3 inflammasome and subsequent release of IL1ß, IL-18 and INF-γ.


Asunto(s)
Colitis Ulcerosa/tratamiento farmacológico , Colitis Ulcerosa/metabolismo , Sistemas de Liberación de Medicamentos/métodos , Ácido Micofenólico/administración & dosificación , Factor 2 Relacionado con NF-E2/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Ácido Acético/toxicidad , Animales , Colitis Ulcerosa/inducido químicamente , Inhibidores Enzimáticos/administración & dosificación , Inflamasomas/antagonistas & inhibidores , Inflamasomas/metabolismo , Masculino , Proteína con Dominio Pirina 3 de la Familia NLR/antagonistas & inhibidores , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley
10.
Life Sci ; 260: 118426, 2020 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-32937159

RESUMEN

AIMS: Tobacco smoking is a major health problem associated with lung and liver damage. Lung and liver damage secondary to tobacco smoking is mediated through nicotine-induced oxidative stress. Therefore, we hypothesized that antioxidant treatment with tiron may improve nicotine-induced lung and liver damage. MATERIALS AND METHODS: Rats were divided into six groups, a control, nicotine (10 mg/kg/day, i.p.; for 8 weeks) and tiron (100 or 200 mg/kg/day, i.p.; for 8 weeks) with or without nicotine administration. KEY FINDINGS: Tiron improved survival rate and attenuated lung and liver damage as reflected by decreased total and differential cell counts, lactate dehydrogenase (LDH) activity in bronchoalveolar lavage fluid (BALF) and decreased alanine aminotransferase (ALT), aspartate aminotransferase (AST) and alkaline phosphatase (ALP) in serum; also histopathological examination confirmed the protective effect of tiron in lung and liver tissues of nicotine treated rats. Tiron attenuated dyslipidemia, which is associated with nicotine. These ameliorative effects of tiron may be mainly due to its antioxidant effect as proved by a significant decrease in malondialdehyde (MDA) content, reactive oxygen species (ROS) and total nitrite/nitrate (NOx) levels, and increase in reduced glutathione (GSH) level, catalase (CAT) and superoxide dismutase (SOD) activities. This is likely related to suppression of protein levels of NADPH oxidase enzyme (NOX1), inducible nitric oxide synthase (iNOS), nuclear factor kappa B (NF-κB) and tumor necrosis factor alpha (TNF-α); and up-regulation of protein levels of nuclear factor erythroid-2 (Nrf2). SIGNIFICANCE: This makes tiron (synthetic analogue of vitamin E) good candidate for future use to minimize nicotine's hazards among smokers.


Asunto(s)
Sal Disódica del Ácido 1,2-Dihidroxibenceno-3,5-Disulfónico/farmacología , Antioxidantes/farmacología , Enfermedad Hepática Inducida por Sustancias y Drogas/prevención & control , Lesión Pulmonar/prevención & control , Nicotina/toxicidad , Animales , Antiinflamatorios no Esteroideos/farmacología , Peso Corporal/efectos de los fármacos , Líquido del Lavado Bronquioalveolar/química , Recuento de Células , Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Enfermedad Hepática Inducida por Sustancias y Drogas/mortalidad , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Enzimas/sangre , L-Lactato Deshidrogenasa/metabolismo , Lípidos/sangre , Lesión Pulmonar/inducido químicamente , Lesión Pulmonar/mortalidad , Lesión Pulmonar/patología , Masculino , NADPH Oxidasa 1/sangre , NADPH Oxidasa 1/metabolismo , FN-kappa B/sangre , FN-kappa B/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Sustancias Protectoras/farmacología , Ratas Sprague-Dawley
11.
Life Sci ; 253: 117729, 2020 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-32348836

RESUMEN

Obstructive nephropathy is a common clinical case that causes chronic kidney disease and ultimately progresses to end-stage renal disease. The activation of peroxisome proliferator-activated receptor-α (PPAR-α) reduces tubulointerstitial fibrosis and inflammation associated with obstructive nephropathy. AIMS: This study was carried out to investigate the potential effect of saroglitazar, dual PPAR-α/γ agonist, in alleviating renal fibrosis induced by unilateralureteral obstruction (UUO). MAIN METHODS: Twenty-four male Sprague Dawley rats were haphazardly divided into four groups of six rats each, including sham operated group, vehicle- or saroglitazar-treated UUO and saroglitazar groups. Rats received oral gavage of saroglitazar (3 mg/kg/day) for 13 days. On day 14, all rats were sacrificed; blood and renal tissues were collected. KEY FINDINGS: Saroglitazar inhibited UUO-induced oxidative stress; it decreased the elevated levels of MDA and nitric oxide and increased levels of GSH and SOD in renal tissue. Moreover, saroglitazar repressed UUO-induced inflammation; it decreased the renal levels of nuclear factor kappa B (NF-κB) and interleukin-6 (IL-6). Furthermore, saroglitazar inhibited the accumulation of extracellular matrix via decreasing collagen, hydroxylproline and matrix metalloproteinase-9 (MMP-9) levels. Saroglitazar also decreased the expression of both the alpha smooth muscle actin (α-SMA) and tumor growth factor-beta (TGF-ß). These effects were in parallel with reduction in mothers against decapentaplegic homolog 3 (smad3) expression and plasminogen activator inhibitor-1 (PAI-1) levels. SIGNIFICANCE: Collectively, the protective impact of saroglitazar might be attributed to its antioxidant, anti-inflammatory and anti-fibrotic effects against UUO-induced tubulointerstitial fibrosis through its regulatory effect on TGF-ß1/Smad3 signaling pathway.


Asunto(s)
Inflamación/prevención & control , Enfermedades Renales/prevención & control , Estrés Oxidativo/efectos de los fármacos , Fenilpropionatos/farmacología , Pirroles/farmacología , Obstrucción Ureteral/tratamiento farmacológico , Animales , Antiinflamatorios/farmacología , Antioxidantes/farmacología , Fibrosis , Inflamación/patología , Enfermedades Renales/fisiopatología , Masculino , PPAR alfa/agonistas , PPAR gamma/agonistas , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Obstrucción Ureteral/complicaciones
12.
Naunyn Schmiedebergs Arch Pharmacol ; 392(12): 1569-1576, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31367862

RESUMEN

Liver fibrosis is a challenging global health problem resulting from chronic liver injury with no treatment currently available. It has been shown that activators for different peroxisome proliferator-activated receptor (PPAR) isoforms (α, γ, and δ) can affect different pathways in liver fibrosis. To evaluate the effects of the dual PPAR-α/γ agonist saroglitazar (SGZ) against thioacetamide (TAA)-induced fibrosis in rats, SGZ was administered for 6 weeks together with TAA injection. Administration of SGZ ameliorated TAA-induced elevation in hepatic biomarkers. SGZ was able to inhibit periportal and intralobular fibrous connective tissue proliferation, to decrease hydroxyproline content, and to lower alpha smooth muscle actin (α-SMA) protein expression. To unearth the antifibrotic mechanism of SGZ, the role of several fibrotic markers was studied. SGZ possesses inhibitory effect on protein levels of leptin, transforming growth factor-beta 1 (TGF-ß1) and platelet-derived growth factor-BB (PDGF-BB). Furthermore, SGZ rectified matrix degradation through decreasing tissue inhibitor of metalloproteinases-1 (TIMP-1). This study suggests that SGZ could have a possible antifibrotic effect via suppression of leptin that can repress TGF-ß1 and PDFG-BB, with subsequent inhibition of TIMP-1.


Asunto(s)
Cirrosis Hepática/tratamiento farmacológico , PPAR alfa/agonistas , PPAR gamma/agonistas , Fenilpropionatos/uso terapéutico , Pirroles/uso terapéutico , Actinas/metabolismo , Animales , Becaplermina/metabolismo , Leptina/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Cirrosis Hepática/inducido químicamente , Cirrosis Hepática/metabolismo , Cirrosis Hepática/patología , Masculino , Fenilpropionatos/farmacología , Pirroles/farmacología , Ratas Sprague-Dawley , Tioacetamida , Inhibidor Tisular de Metaloproteinasa-1/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo
13.
Environ Toxicol Pharmacol ; 43: 182-92, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27011232

RESUMEN

Acute liver injury secondary to sepsis is a major challenge in intensive care unit. This study was designed to investigate potential protective effects of pomegranate against sepsis-induced acute liver injury in rats and possible underlying mechanisms. Pomegranate was orally given (800mg/kg/day) for two weeks before sepsis induction by cecal ligation and puncture (CLP). Pomegranate improved survival and attenuated liver inflammatory response, likely related to downregulation of mRNA expression of toll like recptor-4, reduced nuclear translocation and DNA binding activity of proinflammatory transcription factor NF-κB subunit p65, decreased mRNA and protein expression of tumor necrosis factor-alpha and reduction in myeloperoxidase activity and mRNA expression. Pomegranate also decreased CLP-induced oxidative stress as reflected by decreased malondialdehyde content, and increased reduced glutathione level and superoxide dismutase activity. These results confirm the antiinflammatory and antioxidant effects of pomegranate in CLP-induced acute liver injury mediated through inhibiting TLR4/NF-κB pathway, lipid peroxidation and neutrophil infiltration.


Asunto(s)
Lythraceae , FN-kappa B/metabolismo , Estrés Oxidativo/efectos de los fármacos , Extractos Vegetales/farmacología , Sustancias Protectoras/farmacología , Animales , Antiinflamatorios/farmacología , Antioxidantes/farmacología , Ciego/cirugía , Inflamación/metabolismo , Ligadura , Hígado/metabolismo , Hígado/fisiología , FN-kappa B/antagonistas & inhibidores , Punciones , Ratas
14.
Inflammopharmacology ; 22(3): 187-94, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23989863

RESUMEN

Fulminant hepatic failure (FHF) is a life-threatening syndrome characterized by massive hepatic necrosis and high mortality. There is no effective therapy for the disease other than liver transplantation. This study aimed to investigate the effect of agmatine, inducible nitric oxide synthase (iNOS) inhibitor, on D-galactosamine and lipopolysaccharide (GalN/LPS)-induced FHF in mice and explore its possible mechanism(s). Male Swiss albino mice were injected with a single dose agmatine (14 mg/kg, IP) 8 h prior to challenge with a single intraperitoneal injection of both GalN (800 mg/kg) and LPS (50 µg/kg). Agmatine significantly attenuated all GalN/LPS-induced biochemical and pathological changes in liver. It prevented the increase of serum transaminases and alkaline phosphatase (ALP). In addition, agmatine markedly attenuated GalN/LPS-induced necrosis and inflammation. Agmatine significantly reduced oxidative stress and enhanced antioxidant enzymes. Importantly, agmatine decreased total nitric oxide (NO) and pro-inflammatory cytokine, tumor necrosis factor-alpha (TNF-α). These findings reveal that agmatine has hepatoprotective effects against GalN/LPS-induced FHF in mice that may be related to its ability to suppress oxidative stress, NO synthesis and TNF-α production. Therefore, agmatine may serve as a novel therapeutic strategy for hepatic inflammatory diseases.


Asunto(s)
Agmatina/uso terapéutico , Galactosamina/toxicidad , Lipopolisacáridos/toxicidad , Fallo Hepático Agudo/inducido químicamente , Fallo Hepático Agudo/prevención & control , Animales , Fallo Hepático Agudo/patología , Masculino , Ratones , Sustancias Protectoras/uso terapéutico
15.
J Physiol Biochem ; 70(1): 141-9, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24037794

RESUMEN

The present study was undertaken to investigate the effect of the new formyl peptide receptor 2/lipoxin A4 receptor agonist BML-111 on acetaminophen (APAP)-induced liver injury in mice and explore its possible mechanism(s). Male Swiss albino mice were intraperitoneally injected with BML-111 (1 mg/kg) twice daily for five consecutive days prior to a single intraperitoneal injection of APAP (500 mg/kg). Results have shown that APAP injection caused liver damage as indicated by significant increase in serum alanine aminotransferase (ALT), aspartate aminotransferase (AST), and alkaline phosphatase (ALP). Liver histopathological examination revealed marked necrosis and inflammation. Additionally, APAP decreased activities of hepatic glutathione (GSH) and superoxide dismutase (SOD) with significant increase in the hepatic malondialdehyde (MDA) content. Furthermore, APAP increased serum nitrite/nitrate (NO(2) (-)/NO(3) (-)) level and hepatic tumor necrosis factor alpha (TNF-α). Pretreatment with BML-111 significantly reversed all APAP-induced pathological changes. BML-111 prevented the increase of AST, ALT, and ALP. Also, BML-111 markedly attenuated APAP-induced necrosis and inflammation. It decreased MDA with increase in SOD and GSH. Importantly, BML-111 decreased NO(2) (-)/NO(3) (-) level and TNF-α. These findings suggest that BML-111 has hepatoprotective effects against APAP-induced liver injury in mice. Its protective effect may be attributed to its ability to counteract the inflammatory ROS generation and regulate cytokine effects.


Asunto(s)
Enfermedad Hepática Inducida por Sustancias y Drogas/prevención & control , Ácidos Heptanoicos/farmacología , Acetaminofén , Alanina Transaminasa/sangre , Fosfatasa Alcalina/sangre , Animales , Aspartato Aminotransferasas/sangre , Enfermedad Hepática Inducida por Sustancias y Drogas/sangre , Evaluación Preclínica de Medicamentos , Glutatión/metabolismo , Ácidos Heptanoicos/uso terapéutico , Hígado/efectos de los fármacos , Hígado/enzimología , Hígado/patología , Masculino , Malondialdehído/metabolismo , Ratones , Óxido Nítrico/sangre , Estrés Oxidativo , Superóxido Dismutasa/metabolismo , Factor de Necrosis Tumoral alfa/sangre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA