Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 74
Filtrar
1.
Metabolism ; 155: 155912, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38609038

RESUMO

Saroglitazar (SARO), a dual peroxisome proliferator activated receptor (PPAR)-α/γ agonist, has been used to treat metabolic diseases such as insulin resistance and diabetic dyslipidemia in patients with non-alcoholic fatty liver disease (NAFLD). SARO, administered at a dose of 4 mg/day, has been consistently studied in clinical trials with different time points ranging from 4 to 24 weeks with NAFLD patients. Due to its PPAR-γ agonistic action, SARO prevents adipose tissue-mediated fatty acid delivery to the liver by increasing insulin sensitivity and regulating adiponectin and leptin levels in adipose tissue. In hepatocytes, SARO induces fatty acid ß-oxidation in mitochondria and transcriptionally activates lipid metabolizing genes in peroxisomes. SARO inhibits insulin resistance, thereby preventing the activation of sterol regulatory element-binding proteins -1c and carbohydrate response element binding protein in hepatocytes through its PPAR-α agonistic action. SARO treatment reduces lipotoxicity-mediated oxidative stress by activating the nuclear factor erythroid 2-related factor 2 and transcriptionally expressing the antioxidants from the antioxidant response element in the nucleus through its PPAR-γ agonistic action. SARO provides a PPAR-α/γ-mediated anti-inflammatory effect by preventing the phosphorylation of mitogen-activated protein kinases (JNK and ERK) and nuclear factor kappa B in hepatocytes. Additionally, SARO interferes with transforming growth factor-ß/Smad downstream signaling, thereby reducing liver fibrosis progression through its PPAR-α/γ agonistic actions. Thus, SARO improves insulin resistance and dyslipidemia in NAFLD, reduces lipid accumulation in the liver, and thereby prevents mitochondrial toxicity, oxidative stress, inflammation, and fibrosis progression. This review summarizes the possible molecular mechanism of SARO in the NAFLD.


Assuntos
Hepatopatia Gordurosa não Alcoólica , PPAR alfa , PPAR gama , Humanos , Hepatopatia Gordurosa não Alcoólica/tratamento farmacológico , Hepatopatia Gordurosa não Alcoólica/metabolismo , PPAR alfa/agonistas , PPAR alfa/metabolismo , PPAR gama/agonistas , PPAR gama/metabolismo , Animais , Fenilpropionatos/uso terapêutico , Fenilpropionatos/farmacologia , Resistência à Insulina , Pirróis
2.
J Biochem Mol Toxicol ; 38(4): e23691, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38500399

RESUMO

Sustained liver injuries predominantly promote oxidative stress and inflammation that lead to the progression of chronic liver disease (CLD), including fibrosis, cirrhosis, and hepatocellular carcinoma. Boldine, an alkaloid isolated from Peumus boldus, has been shown to have antioxidant and anti-inflammatory effects. Currently, there is no definitive treatment option available for CLD. Therefore, we investigated the hepatoprotective effect of boldine against carbon tetrachloride (CCl4 )-induced chronic liver injury in rats. CCl4 (2 mL/kg., b.w., i.p.) was administered twice weekly for 5 weeks to induce chronic liver injury in rats. Separate groups of rats were given boldine (20 mg/kg b.w., and 40 mg/kg b.w.) and silymarin (100 mg/kg b.w.) orally, daily. Serum transaminases, lipid peroxidation, and antioxidant levels were measured, and nuclear factor-κB (NF-κB), tumor necrosis factor-α (TNF-α), cyclooxygenase-2 (cox-2), interleukin-1 ß (IL-1ß), and α-smooth muscle actin (α-SMA) gene and protein expressions were evaluated. CCl4 administration increased liver marker enzymes of hepatotoxicity in serum and oxidative stress markers, inflammatory genes and α-smooth muscle actin expression in liver tissue. Boldine concurrent treatment suppressed CCl4 -induced elevation of transaminase levels in serum, restored enzymic and non-enzymic antioxidants, and downregulated NF-κB, TNF-α, Cox-2 and IL-1ß expressions, thereby suppressing hepatic inflammation. Boldine administration also repressed α-SMA expression. The results of this study demonstrate the antioxidant, anti-inflammatory, and antifibrotic properties of boldine, and it can be a potential therapeutic candidate in the treatment of CLD.


Assuntos
Aporfinas , Doença Hepática Induzida por Substâncias e Drogas , NF-kappa B , Ratos , Animais , NF-kappa B/metabolismo , Antioxidantes/farmacologia , Antioxidantes/metabolismo , Tetracloreto de Carbono/toxicidade , Actinas/metabolismo , Actinas/farmacologia , Fator de Necrose Tumoral alfa/metabolismo , Ciclo-Oxigenase 2/metabolismo , Fígado/metabolismo , Transdução de Sinais , Estresse Oxidativo , Inflamação/metabolismo , Anti-Inflamatórios/farmacologia , Doença Hepática Induzida por Substâncias e Drogas/metabolismo
3.
J Biochem Mol Toxicol ; 38(4): e23694, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38504479

RESUMO

Chronic liver injury due to various etiological factors results in excess secretion and accumulation of extracellular matrix proteins, leading to scarring of liver tissue and ultimately to hepatic fibrosis. If left untreated, fibrosis might progress to cirrhosis and even hepatocellular carcinoma. Thymoquinone (TQ), an active compound of Nigella sativa, has been reported to exhibit antioxidant, anti-inflammatory and anticancer activities. Therefore, the effect of TQ against thioacetamide (TAA)-induced liver fibrosis was assessed in rats. Fibrosis was induced with intraperitoneal administration of TAA (250 mg/kg b.w.) twice a week for 5 weeks. TQ (20 mg/kg b.w.) and silymarin (50 mg/kg b.w.) were orally administered daily for 5 weeks separately in TAA administered groups. Liver dysfunction was reported by elevated liver enzymes, increased oxidative stress, inflammation and fibrosis upon TAA administration. Our study demonstrated that TQ inhibited the elevation of liver marker enzymes in serum. TQ administration significantly increased antioxidant markers, such as superoxide dismutase, catalase, glutathione, glutathione peroxidase and glutathione reductase in the liver tissue of rats. Further, TQ significantly attenuated liver fibrosis, as illustrated by the downregulation of TAA-induced interleukin-ß, tumour necrosis factor-α, inducible nitric oxide synthase and fibrosis markers like transforming growth factor-ß (TGF-ß), α-smooth muscle actin, collagen-1, Smad3 and 7. Therefore, these findings suggest that TQ has a promising hepatoprotective property, as indicated by its potential to effectively suppress TAA-induced liver fibrosis in rats by inhibiting oxidative stress and inflammation via TGF-ß/Smad signaling.


Assuntos
Benzoquinonas , Neoplasias Hepáticas , Fator de Crescimento Transformador beta1 , Ratos , Animais , Fator de Crescimento Transformador beta1/metabolismo , Tioacetamida/toxicidade , Antioxidantes/metabolismo , Fígado/metabolismo , Cirrose Hepática/induzido quimicamente , Cirrose Hepática/tratamento farmacológico , Cirrose Hepática/prevenção & controle , Fator de Crescimento Transformador beta/metabolismo , Inflamação/metabolismo , Estresse Oxidativo , Neoplasias Hepáticas/metabolismo
4.
Cureus ; 16(1): e51609, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38313882

RESUMO

Background Drug-induced liver injury is a common cause of acute liver failure. Isoniazid (INH) is used as a first-line treatment for tuberculosis. Clinical and experimental studies have reported abnormal liver function after INH therapy. Lagerstroemia speciosa Pers., commonly known as banaba, has been traditionally used to treat various ailments including diabetes and obesity due to its antioxidant and anti-inflammatory properties. Aim To investigate the hepatoprotective effect of ethanolic banaba leaf extract (EBLE) against INH-induced hepatotoxicity in rats. Materials and methods A total of 30 male Wistar albino rats (150 - 200 g) were divided into five groups (n = 6). Group I rats were served as a control and were administered dimethyl sulfoxide for the first 30 days and water for the next 30 consecutive days. Group II rats were administered INH (50 mg/kg, p.o.) once in the first 30 consecutive days and sacrificed at Day 30. Group III rats were administered INH for 30 consecutive days and left without treatment for the next 30 days. In Groups IV and V, rats were post-treated orally with EBLE 250 and 500 mg/kg, p.o. (0.3 ml/rat) for 30 days after INH administration. At the end of Day 60, the remaining group of animals were sacrificed. The blood and liver tissues were collected. The marker enzymes of hepatotoxicity, oxidative stress markers, inflammatory markers, and histopathology were analyzed. Results INH administration induced significant elevation of marker enzymes (aspartate transaminase, alanine transaminase, alkaline phosphatase, lactate dehydrogenase, bilirubin, gamma-glutamyl transpeptidase) of hepatotoxicity in the serum. This treatment also increased lipid peroxidation and proinflammatory marker expression (tumor necrosis factor-alpha, transforming growth factor-beta, and nuclear factor kappa B (NF-κB) except inhibitor of NF-κB) and decreased antioxidants such superoxide dismutase, catalase, and glutathione in the liver tissue. All these abnormalities were significantly mitigated after treatment with EBLE. Conclusion The results of this study suggest that EBLE can be used for INH-induced hepatotoxicity.

6.
Cell Biochem Funct ; 42(1): e3916, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38269515

RESUMO

Alcoholic liver disease (ALD) is one of the most common health problems worldwide, especially in developing countries caused by chronic consumption of alcohol on a daily basis. The ALD spectrum is initiated with the early stages of alcoholic fatty liver (steatosis), progressing to alcoholic steatohepatitis, followed by the later stages of fibrosis and in some cases, cirrhosis and hepatocellular carcinoma (HCC). The Wnt/ß-catenin signaling required for healthy liver development, function, and regeneration is found to be aberrated in ALD, attributed to its progression. This review is to elucidate the association of Wnt/ß-catenin signaling with various stages of ALD progression. Alcohol causes downregulation of Wnt/ß-catenin signaling components and thereby suppressing the pathway. Reports have been published that aberrated Wnt/ß-catenin signaling, especially the absence of ß-catenin, results in decreased alcohol metabolism, causing steatosis followed by steatohepatitis via lipid accumulation, lipid peroxidation, liver injury, increased oxidative stress and apoptosis of hepatocytes, contributing to the advancement of ALD. Contrastingly, the progression of later stages of ALD like fibrosis and HCC depends on the increased activation of Wnt/ß-catenin signaling and its components. Existing studies reveal the varied expression of Wnt/ß-catenin signaling in ALD. However, the dual role of the Wnt/ß-catenin pathway in earlier and later stages of ALD is not clear. Therefore, studies on the Wnt/ß-catenin pathway and its components in various manifestations of ALD might provide insight in targeting the Wnt/ß-catenin pathway in ALD treatment.


Assuntos
Carcinoma Hepatocelular , Fígado Gorduroso , Hepatopatias Alcoólicas , Neoplasias Hepáticas , Humanos , beta Catenina , Etanol , Cirrose Hepática
7.
Chem Biol Drug Des ; 103(1): e14369, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-37817304

RESUMO

Acetaminophen (APAP) in high doses causes acute liver injury and acute liver failure. Ethyl gallate (EG) is a natural polyphenol, possessing antioxidant, anti-inflammatory, and anti-microbial properties. Therefore, in this study, we evaluated the protective role of EG against APAP-induced acute liver injury in mice. Acute liver injury was induced by a single dose of APAP (400 mg/kg., i.p.). In separate groups, EG (10 mg/kg), EG (20 mg/kg), and N-acetylcysteine (NAC; 1200 mg/kg., i.p.) were administered concurrently with APAP. The mice were sacrificed after 24 h of treatment. Liver marker enzymes of hepatotoxicity, antioxidant markers, inflammatory markers, and histopathological studies were done. APAP administration caused a significant elevation of marker enzymes of hepatotoxicity and lipid peroxidation. APAP administration also decreased enzymic and nonenzymic antioxidants. Acute APAP intoxication induced nuclear factor κ B, tumor necrosis factor-α, interleukin-1, p65, and p52 and downregulated IκB gene expressions. Our histopathological studies have confirmed the presence of centrilobular necrosis, 24 h after APAP intoxication. All the above abnormalities were significantly inhibited in groups of mice that were concurrently administered with APAP + EG and APAP + NAC. Our in silico analysis further confirms that hydroxyl groups of EG interact with the above inflammatory proteins at the 3,4,5-trihydroxybenzoic acid region. These effects of EG against APAP-induced acute liver injury could be attributed to its antioxidative, free radical scavenging, and anti-inflammatory potentials. Therefore, this study suggests that EG can be an efficient therapeutic approach to protect the liver from APAP intoxication.


Assuntos
Antioxidantes , Doença Hepática Induzida por Substâncias e Drogas , Camundongos , Animais , Antioxidantes/farmacologia , Antioxidantes/uso terapêutico , Antioxidantes/metabolismo , Acetaminofen/toxicidade , Fígado , Ácido Gálico/metabolismo , Ácido Gálico/farmacologia , Anti-Inflamatórios/farmacologia , Doença Hepática Induzida por Substâncias e Drogas/tratamento farmacológico , Doença Hepática Induzida por Substâncias e Drogas/patologia , Estresse Oxidativo
8.
J Appl Toxicol ; 44(2): 245-259, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37661188

RESUMO

Hepatocellular carcinoma (HCC) contributes to more than 80% of all primary cancers globally and ranks fourth in cancer-related deaths, due to the lack of an effective, definite therapeutic drug. Coleus vettiveroides (CV) has been used in Indian traditional medicine to treat diabetes, liver ailments, skin diseases, leukoderma, and leprosy. This study investigates the anticancer effect of CV ethanolic root extract in HepG2 cells. HepG2 cells were treated with CV extract, and its cytotoxicity was analyzed by MTT assay. AO/EB staining, propidium iodide staining, DCFH-DA assay, phalloidine staining, flow cytometry, and qPCR studies were performed for ROS expression, apoptosis and cell cycle analysis. The phytochemical analysis confirmed the presence of quercetin and galangin in CV root extract. The results showed that CV inhibited the proliferation of HepG2 cells, with altered cellular and nuclear morphology. CV was also found to increase intracellular ROS levels and oxidative stress markers in HepG2 cells. CV significantly altered the actin microfilament distribution in HepG2 cells and caused cell cycle arrest at the sub G0 -G1 phase. CV also induced mitochondria-mediated apoptosis, as evidenced by increased expression of p53, Bax, cytochrome C, Apaf-1, PARP, caspase-3 and caspase-9, and downregulated Bcl-2 expression. Therefore, CV exerts its anticancer effect by inducing mitochondrial dysfunction, oxidative stress, cytoskeletal disorganization, cell cycle arrest, and mitochondria-mediated apoptosis, and it could be a potent therapeutic option for HCC.


Assuntos
Carcinoma Hepatocelular , Coleus , Neoplasias Hepáticas , Humanos , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Células Hep G2 , Neoplasias Hepáticas/metabolismo , Coleus/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Apoptose , Etanol
9.
Cureus ; 15(11): e48126, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-38046745

RESUMO

Introduction Cancer continues to be a significant health issue worldwide, with colorectal cancer (CRC) standing out as one of the most prevalent forms of cancer on a global scale. The lifetime risk of developing CRC is about one in 23 (4.3%) for men and one in 25 (4.0%) for women. Moreover, children and adolescents are frequently reported with osteosarcoma with a low five-year survival rate (69% and 67%, respectively). Aim The aim of the study was to analyze the cytotoxic effects of boldine against human CRC (HCT-116) and osteosarcoma cell lines (Saos-2). Materials and methods HCT-116 and Saos-2 cell lines were subjected to different concentrations of boldine treatment (5, 10, 20, 30, 40, and 50 µg/mL) and (10, 20, 40, 60, and 80 µg/mL), respectively, for 24 hours. The cytotoxicity was analyzed by MTT assay, AO/EB staining, DCFH-DA assay, and scratch assay. Results The MTT assay, microscopic analysis, and staining showed that boldine had dose-dependent cytotoxic effects against HCT-116 and Saos-2 cell lines by inhibiting their proliferation, viability, and migration, and inducing ROS-mediated apoptosis. Conclusion The study concluded that boldine had a concentration-dependent cytotoxic effect on human CRC and osteosarcoma cell lines.

10.
Cureus ; 15(11): e49372, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-38149135

RESUMO

INTRODUCTION: Increased colorectal carcinoma (CRC) and osteosarcoma prevalence, low survival rate, poor prognosis, and the limitations of existing anticancer therapies like side effects of drugs, non-specificity, short half-life, etc., pose a need for novel anticancer drugs. Farnesol, an organic sesquiterpene compound, found in the essential oils of various plants has been shown to possess antioxidant, anti-inflammatory, and anticancer properties. However, the anticancer effect of farnesol against CRC and osteosarcoma has not yet been adequately elucidated. AIM: The aim of the study was to analyze the anticancer effects of farnesol against human osteosarcoma and CRC cell lines. MATERIALS AND METHODS: Human osteosarcoma (Saos-2) and colorectal carcinoma (HCT-116) cell lines were procured and cultured at 37oC and 5% CO2. The cells were treated with 10, 20, 40, 60, 80, and 100 µM/ml and 20, 40, 60, 80, 100, and 120 µM/ml of farnesol for 24 hours, respectively. 3-(4,5-dimethylthiazol-2-yl)-2,5 diphenyl tetrazolium bromide assay was performed to assess the cytotoxicity of farnesol on Saos-2 and HCT-116 cells. Acridine orange/ethidium bromide staining was carried out to analyze apoptosis. 4',6-diamidino-2-phenylindole staining was done to observe the nuclear changes. Dichloro-dihydro-fluorescein diacetate staining was performed to assess the farnesol-induced reactive oxygen species (ROS)-mediated cell death. RESULTS: Farnesol reduced the viability and proliferation of Saos-2 and HCT-116 cells in a dose-dependent manner. Farnesol was able to alter the cellular and nuclear morphology of Saos-2 and HCT-116 cells, promoting cell death. Farnesol-induced apoptosis in human osteosarcoma and colorectal carcinoma cell lines. Early apoptosis was observed in farnesol-treated HCT-116 cells. Additionally, ROS-mediated apoptotic cell death was reported in Saos-2 cells. CONCLUSION: Farnesol has the potential to induce cytotoxicity against human osteosarcoma and CRC cell lines.

11.
Cureus ; 15(10): e47609, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-38021731

RESUMO

Background Osteosarcoma is the eighth most common cancer and its prevalence in children makes it a global concern. Existing medications and treatments like high-dose methotrexate possess harmful side effects. Therefore, novel herbal drugs like Nelumbo nucifera are of utmost importance. Aim  To analyze a novel anticancer herbal drug, Nelumbo nucifera leaf extract for its cytotoxic potential against osteosarcoma.  Materials and method Nelumbo nucifera leaf extract was prepared. Saos-2 Cells (human osteosarcoma cell line) were treated with Nelumbo nucifera leaf extract (25, 50, 75, 100, 125, and 150 µg/ml) for 24 hours which were then subjected to MTT assay, morphological analysis and DAPI staining. Results The results suggested that Nelumbo nucifera leaf extract had a concentration-dependent cytotoxic effect on Saos-2 cell line. The extract significantly reduced the number of viable cells, inhibited proliferation and induced morphological changes in Saos-2 cells.  Conclusion Nelumbo nucifera has the potential to induce cytotoxicity against osteosarcoma cell lines and hence, this study provides a novel therapeutic regimen for the treatment of osteosarcoma.

12.
Cell Biochem Funct ; 41(7): 876-888, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37605364

RESUMO

Acute liver injury is caused by various factors, including oxidative stress and inflammation. Coleus vettiveroides, an ayurvedic medicinal plant, is known to possess antioxidant, antibacterial, and antidiabetic properties. In this current study, we investigated the protective effect of C. vettiveroides ethanolic root extract (CVERE) against thioacetamide (TAA)-induced acute liver injury in rats. A single dose of TAA (300 mg/kg, b.w., i.p.) was administered to induce acute liver injury. The treatment groups of rats were concurrently treated with CVERE (125 and 250 mg/kg, b.w., p.o.) and silymarin (100 mg/kg, b.w., p.o.), respectively. After 24 h of the experimental period, TAA-induced liver injury was confirmed by increased activity of serum transaminases and malondialdehyde levels in liver tissue, decreased levels of antioxidants, upregulated expression of the inflammatory marker gene, and altered liver morphology. Whereas CVERE simultaneous treatment inhibited hepatic injury and prevented the elevation of serum aspartate and alanine transaminases, alkaline phosphatase, and lactate dehydrogenase activities. CVERE attenuated TAA-induced oxidative stress by suppressing lipid peroxidation and restoring antioxidants such as superoxide dismutase, catalase, and reduced glutathione. Further, CVERE treatment was found to inhibit nuclear factor κB-mediated inflammatory signaling, as indicated by downregulated pro-inflammatory cytokines including tumor necrosis factor-α and interleukin-1ß. Our findings suggest that CVERE prevents TAA-induced acute liver injury by targeting oxidative stress and inflammation.

13.
J Biochem Mol Toxicol ; 37(11): e23478, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37458150

RESUMO

Acetaminophen (APAP) is known to cause acute liver injury and acute liver failure in Western countries. This study investigates the protective role of farnesol (FAR) (C15 H26 O), a natural sesquiterpene alcohol in essential oils, against APAP-induced acute liver necrosis in mice. Mice were injected with a single dose of APAP (300 mg/kg) via an intraperitoneal route. Different groups of mice were concurrently treated with a single dose of FAR 25 mg/kg, FAR 50 mg/kg, and N-acetylcysteine. APAP administration caused a significant increase in transaminase activities and malondialdehyde (MDA) levels in the serum and liver tissue, respectively, with a concomitant decrease in intracellular antioxidants, including reduced glutathione (GSH) in the liver tissue. APAP intoxication upregulated proinflammatory cytokines such as tumor necrosis factor-α, interleukin-1ß (IL-1ß), IL-6, nuclear factor-κB (NF-κB), and IκB kinase ß in the liver tissue. FAR and N-acetylcysteine (NAC) administrations concurrently with APAP prevented serum transaminase increase in serum and MDA levels in the liver tissue. A high dose of FAR and NAC treatments significantly inhibited GSH and other antioxidant depletion. FAR and NAC treatments also downregulated the expression of proinflammatory markers. FAR treatments protects against APAP-induced acute liver injury and offers antioxidant and anti-inflammatory effects by inhibiting the NF-κB pathway involved in the transcription of genes responsible for inflammatory cytokine synthesis.


Assuntos
Acetaminofen , Doença Hepática Induzida por Substâncias e Drogas , Camundongos , Animais , Acetaminofen/toxicidade , Antioxidantes/metabolismo , Farneseno Álcool/farmacologia , Farneseno Álcool/metabolismo , NF-kappa B/metabolismo , Acetilcisteína/farmacologia , Doença Hepática Induzida por Substâncias e Drogas/tratamento farmacológico , Doença Hepática Induzida por Substâncias e Drogas/prevenção & controle , Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Fígado/metabolismo , Glutationa/metabolismo , Necrose , Transaminases/metabolismo , Transaminases/farmacologia , Alanina Transaminase
14.
Drug Metab Rev ; 55(3): 239-253, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37218081

RESUMO

Tuberculosis (TB) remains a major global health burden. Antitubercular drugs (ATDs) such as isoniazid (INH), rifampicin (RIF), pyrazinamide (PZA), and ethambutol are used as first-line therapy in TB patients. Drug-induced liver injury is one of the common side effects that leads to the discontinuation of ATDs in TB patients. Therefore, this review discusses the molecular pathogenesis of ATDs induced liver injury. The biotransformation of INH, RIF, and PZA in the liver liberates several reactive intermediates, leading to peroxidation of the hepatocellular membrane and oxidative stress. INH + RIF administration decreased the expression of bile acid transporters such as the bile salt export pump and multidrug resistance-associated protein 2 and induced liver injury by sirtuin 1 and farnesoid X receptor pathway. INH inhibits the nuclear translocation of Nrf2 by interfering with its nuclear importer, karyopherin ß1, thereby inducing apoptosis. INF + RIF treatments alter Bcl-2 and Bax homeostasis, mitochondrial membrane potential, and cytochrome c release, thereby triggering apoptosis. RIF administration enhances the expression of genes involved in fatty acid synthesis and hepatocyte fatty acid uptake (CD36). RIF induces the expression of peroxisome proliferator-activated receptor -γ and its downstream proteins and perilipin-2 by activating the pregnane X receptor in the liver to increase fatty infiltration into the liver. ATDs administration induces oxidative stress, inflammation, apoptosis, cholestasis, and lipid accumulation in the liver. However, ATDs toxic potentials are not elaborately studied at the molecular level in clinical samples. Therefore, future studies are warranted to explore ATDs induced liver injuries at the molecular level in clinical samples whenever possible.


Assuntos
Doença Hepática Crônica Induzida por Substâncias e Drogas , Doença Hepática Induzida por Substâncias e Drogas , Humanos , Antituberculosos/efeitos adversos , Doença Hepática Crônica Induzida por Substâncias e Drogas/tratamento farmacológico , Isoniazida , Pirazinamida/uso terapêutico , Rifampina
15.
Environ Toxicol Pharmacol ; 99: 104093, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-36870405

RESUMO

Thioacetamide (TAA) undergoes bioactivation in the liver by the CYP450 2E1 enzyme, resulting in the formation of TAA-S-oxide and TAA-S-dioxide. TAA-S-dioxide induces oxidative stress via lipid peroxidation of the hepatocellular membrane. A single TAA dose (50-300 mg/kg) administration initiates hepatocellular necrosis around the pericentral region after its covalent binding to macromolecules in the liver. Intermittent TAA administration (150-300 mg/kg, weekly thrice, for 11-16 weeks) activates transforming growth factor (TGF)-ß/smad3 downstream signaling in injured hepatocytes, causing hepatic stellate cells (HSCs) to acquire myofibroblast like phenotype. The activated HSCs synthesize a variety of extracellular matrix, leading to liver fibrosis, cirrhosis, and portal hypertension. The TAA induced liver injury varies depending on the animal model, dosage, frequency, and routes of administration. However, TAA induces hepatotoxicity in a reproducible manner, and it is an ideal model to evaluate the antioxidant, cytoprotective, and antifibrotic compounds in experimental animals.


Assuntos
Fígado , Tioacetamida , Animais , Tioacetamida/toxicidade , Tioacetamida/metabolismo , Fígado/metabolismo , Cirrose Hepática/induzido quimicamente , Cirrose Hepática/metabolismo , Hepatócitos/metabolismo , Células Estreladas do Fígado/metabolismo , Células Estreladas do Fígado/patologia , Fator de Crescimento Transformador beta
17.
J Cell Physiol ; 238(4): 673-686, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36745560

RESUMO

Galectin-3 (Gal-3) previously referred to as S-type lectins, is a soluble protein that specifically binds to ß-galactoside carbohydrates with high specificity. Gal-3 plays a pivotal role in a variety of pathophysiological processes such as cell proliferation, inflammation, differentiation, angiogenesis, transformation and apoptosis, pre-mRNA splicing, metabolic syndromes, fibrosis, and host defense. The role of Gal-3 has also been implicated in liver diseases. Gal-3 is activated upon a hepatotoxic insult to the liver and its level has been shown to be upregulated in fatty liver diseases, inflammation, nonalcoholic steatohepatitis, fibrosis, cholangitis, cirrhosis, and hepatocellular carcinoma (HCC). Gal-3 directly interacts with the NOD-like receptor family, pyrin domain containing 3, and activates the inflammasome in macrophages of the liver. In the chronically injured liver, Gal-3 secreted by injured hepatocytes and immune cells, activates hepatic stellate cells (HSCs) in a paracrine fashion to acquire a myofibroblast like collagen-producing phenotype. Activated HSCs in the fibrotic liver secrete Gal-3 which acts via autocrine signaling to exacerbate extracellular matrix synthesis and fibrogenesis. In the stromal microenvironment, Gal-3 activates cancer cell proliferation, migration, invasiveness, and metastasis. Clinically, increased serum levels and Gal-3 expression were observed in the liver tissue of nonalcoholic steatohepatitis, fibrotic/cirrhotic, and HCC patients. The pathological role of Gal-3 has been experimentally and clinically reported in the progression of chronic liver disease. Therefore, this review discusses the pathological role of Gal-3 in the progression of chronic liver diseases.


Assuntos
Galectina 3 , Fígado , Humanos , Fibrose , Galectina 3/genética , Células Estreladas do Fígado/metabolismo , Inflamação/patologia , Cirrose Hepática/patologia , Fígado/lesões , Fígado/patologia
18.
Crit Rev Oncol Hematol ; 182: 103902, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36621514

RESUMO

Activated hepatic stellate cells (HSCs)/myofibroblasts are the important sources of cancer-associated fibroblasts in the liver tumor microenvironment (TME). The crosstalk between activated HSCs and tumor cells mediates HCC progression, metastasis, tumor cell survival, angiogenesis and chemoresistance. In TME, HCC cells secrete various soluble factors responsible for the phenotypic activation of quiescent HSCs. Tumor cells use activated HSC-derived extracellular matrix (ECM) for migration and invasion. Further, in liver TME, activated HSCs and sinusoidal endothelial cells engage in a crosstalk that causes the secretion of angiogenesis and metastasis-related growth factors and cytokines. Activated HSCs and immune cells crosstalk to decrease immune surveillance in the liver TME by increasing the population of T regulatory cells and M2 macrophages or myeloid-derived suppressor cells. Thus, HSCs play a vital role in liver TME cell interactions. Therefore, a deep understanding of HSCs activation and their crosstalk with cancer and immune cells in TME may lead to the development of novel therapeutic strategies to target HCC.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Humanos , Neoplasias Hepáticas/patologia , Carcinoma Hepatocelular/patologia , Células Estreladas do Fígado/metabolismo , Células Estreladas do Fígado/patologia , Microambiente Tumoral , Células Endoteliais
20.
Hepatobiliary Pancreat Dis Int ; 22(4): 333-345, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-36448560

RESUMO

Nonalcoholic fatty liver disease (NAFLD) is a global health concern associated with significant morbidity and mortality. NAFLD is a spectrum of diseases originating from simple steatosis, progressing through nonalcoholic steatohepatitis (NASH), fibrosis, and cirrhosis that may lead to hepatocellular carcinoma (HCC). The pathogenesis of NAFLD is mediated by the triglyceride accumulation followed by proinflammatory cytokines expression leading to inflammation, oxidative stress, and mitochondrial dysfunction denoted as "two-hit hypothesis", advancing with a "third hit" of insufficient hepatocyte proliferation, leading to the increase in hepatic progenitor cells contributing to fibrosis and HCC. Wnt/ß-catenin signaling is responsible for normal liver development, regeneration, hepatic metabolic zonation, ammonia and drug detoxification, hepatobiliary development, etc., maintaining the overall liver homeostasis. The key regulators of canonical Wnt signaling such as LRP6, Wnt1, Wnt3a, ß-catenin, GSK-3ß, and APC are abnormally regulated in NAFLD. Many experimental studies have shown the aberrated Wnt/ß-catenin signaling during the NAFLD progression and NASH to hepatic fibrosis and HCC. Therefore, in this review, we have emphasized the role of Wnt/ß-catenin signaling and its modulators that can potentially aid in the inhibition of NAFLD.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Hepatopatia Gordurosa não Alcoólica , Humanos , Hepatopatia Gordurosa não Alcoólica/patologia , Carcinoma Hepatocelular/patologia , Via de Sinalização Wnt/fisiologia , beta Catenina , Glicogênio Sintase Quinase 3 beta/metabolismo , Neoplasias Hepáticas/patologia , Progressão da Doença , Fígado/patologia , Cirrose Hepática/patologia , Fibrose
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA