ABSTRACT
PURPOSE: To investigate the effect of clofibrate on inducing liver fatty acid binding protein (FABP1) following a high-fat load in a hepatocyte cell culture model. METHODS: Rat hepatoma cells (CRL-1548) were treated with a fatty acid (FA) mixture consisting of oleate:palmitate (2:1) in the presence of 3% albumin. Cells were treated with 0, 0.5, 1, 2, or 3 mM FA for 24 and 48 hr, or further treated with 500 µM clofibrate (CLO) to induce FABP1 levels. Cytotoxicity was determined using the WST-1 assay. Intracellular lipid droplets were quantitated following staining with Nile Red. Dichlorofluorescein (DCF) was used to assess the extent of intracellular reactive oxygen species (ROS). RESULTS: Cell viability decreased (p < 0.01) with an increase in lipid concentration. Intracellular lipid droplets accumulated significantly (p < 0.001) with an increase in long-chain fatty acid load, which was associated with a statistical increase (p < 0.05) in ROS levels. Early clofibrate treatment showed significant increases in intracellular FABP1 levels with significant decreases in ROS levels (p < 0.05). Silencing FABP1 expression using siRNA revealed that FABP1 was the main contributor for the observed intracellular ROS clearance. CONCLUSIONS: Characteristic cellular damage resulted from released ROS following a high fat load to hepatoma cells. The damage was attenuated through early treatment with clofibrate, which may act as a hepatoprotectant by inducing FABP1 expression and in this manner, suppress intracellular ROS levels. This article is open to POST-PUBLICATION REVIEW. Registered readers (see "For Readers") may comment by clicking on ABSTRACT on the issue's contents page.
Subject(s)
Clofibrate/pharmacology , Lipids/pharmacology , Reactive Oxygen Species/antagonists & inhibitors , Animals , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Fatty Acid-Binding Proteins/antagonists & inhibitors , Fatty Acid-Binding Proteins/biosynthesis , Fatty Acid-Binding Proteins/genetics , Fatty Acids/pharmacology , Lipids/administration & dosage , Lipids/chemistry , Particle Size , RNA, Small Interfering/pharmacology , Rats , Reactive Oxygen Species/metabolism , Structure-Activity Relationship , Tumor Cells, CulturedABSTRACT
Herbal products have been used for centuries among indigenous people to treat symptoms and illnesses. Recently, their use in Western countries has grown significantly, rivaling that of prescription medications. Currently, herbal products are used mainly for weight loss and bodybuilding purposes but also to improve well-being and symptoms of chronic diseases. Many people believe that because they are natural, they must be effective and safe; however, these beliefs are erroneous. Few herbal products have been studied in well-designed controlled trials of patients with liver or other diseases, despite testimony to the contrary. Moreover, current highly effective antiviral drugs make efforts to treat hepatitis C with herbal products redundant. Herbal products are no safer than conventional drugs and have caused liver injury severe enough to require transplantation or cause death. Furthermore, their efficacy, safety, and claims are not assessed by regulatory agencies, and there is uncertainty about their reported and unreported contents. We review the history of commonly used herbal products, as well as their purported efficacies and mechanisms and their adverse effects.
Subject(s)
Chemical and Drug Induced Liver Injury/etiology , Liver Diseases/drug therapy , Plant Preparations/adverse effects , Humans , Liver/drug effects , Plant Preparations/pharmacologyABSTRACT
Over four decades have passed since liver fatty acid binding protein (FABP)1 was first isolated. There are few protein families for which most of the complete tertiary structures, binding properties, and tissue occurrences are described in such detail and yet new functions are being uncovered for this protein. FABP1 is known to be critical for fatty acid uptake and intracellular transport and also has an important role in regulating lipid metabolism and cellular signaling pathways. FABP1 is an important endogenous cytoprotectant, minimizing hepatocyte oxidative damage and interfering with ischemia-reperfusion and other hepatic injuries. The protein may be targeted for metabolic activation through the cross-talk among many transcriptional factors and their activating ligands. Deficiency or malfunction of FABP1 has been reported in several diseases. FABP1 also influences cell proliferation during liver regeneration and may be considered as a prognostic factor for hepatic surgery. FABP1 binds and modulates the action of many molecules such as fatty acids, heme, and other metalloporphyrins. The ability to bind heme is another cytoprotective property and one that deserves closer investigation. The role of FABP1 in substrate availability and in protection from oxidative stress suggests that FABP1 plays a pivotal role during intracellular bacterial/viral infections by reducing inflammation and the adverse effects of starvation (energy deficiency).
Subject(s)
Fatty Acid-Binding Proteins/metabolism , Liver/metabolism , Animals , Fatty Acid-Binding Proteins/genetics , Humans , Lipid Metabolism/genetics , Lipid Metabolism/physiology , Oxidative Stress/genetics , Oxidative Stress/physiologyABSTRACT
BACKGROUND: FABP1 has been reported to possess strong antioxidant properties. Upon successful transfection of the Chang cell line, which has undetectable FABP1 mRNA levels, with human FABP1 cDNA, the Chang cells were shown to express FABP1. Using the transfected and control (normal) Chang cells and subjecting them to oxidative stress, transfected cells were reported to be associated with enhanced cell viability. This study extends those observations by investigating the effect of FABP1 on acetaminophen (AAP)-induced hepatotoxicity. We hypothesized that presence of FABP1 would enhance cell viability compared to control cells (vector transfected cells). METHODS: Following AAP treatment of Chang FABP1 transfected and control cells, cell viability, oxidative stress, and apoptosis were evaluated using lactate dehydrogenase (LDH) release, the fluorescent probe DCF, and Bax expression, respectively. RESULTS: FABP1 cDNA transfected cells showed greater resistance against AAP toxicity than vector transfected cells. Significantly lower LDH levels (p < 0.05) were observed as were lower DCF fluorescence intensity (p < 0.05) in FABP1 cDNA transfected cells compared to vector transfected cells. FABP1 expression also attenuated the expression of Bax following AAP induced toxicity. CONCLUSION: FABP1 attenuated AAP-induced toxicity and may be considered a cytoprotective agent in this in vitro model of drug induced oxidative stress.
Subject(s)
Acetaminophen/toxicity , Analgesics, Non-Narcotic/toxicity , Cytoprotection , Fatty Acid-Binding Proteins/metabolism , Liver/drug effects , Liver/metabolism , Apoptosis , Cell Survival , Cells, Cultured , Fatty Acid-Binding Proteins/genetics , Fluoresceins , Fluorescence , Humans , L-Lactate Dehydrogenase/metabolism , Liver/physiopathology , Oxidative Stress , Transfection , bcl-2-Associated X Protein/metabolismABSTRACT
Quinoid dihydropteridine reductase (QDPR) is an enzyme involved in the metabolic pathway of tetrahydrobiopterin (BH4). BH4 is an essential cofactor of nitric oxide synthase (NOS) and can catalyze arginine to citrulline to release nitric oxide. Point mutations of QDPR have been found in the renal cortex of spontaneous Otsuka Long Evans Tokushima Fatty (OLETF) diabetic rats. However, the role of QDPR in DN is not clear. This study investigates the effects of QDPR overexpression and knockdown on gene expression in the kidney. Rat QDPR cDNA was cloned into pcDNA3.1 vector and transfected in human kidney cells (293T). The expression of NOS, transforming growth factor beta 1 (TGF-ß1), Smad3, and NADPH oxidase were examined by RT-PCR and Western blot analyses. BH4 was assayed by using ELISA. Expression of QDPR was significantly decreased and TGF-ß1 and Smad3 were increased in the renal cortex of diabetic rats. Transfection of QDPR into 293T cells increased the abundance of QDPR in cytoplasm and significantly reduced the expression of TGF-ß1, Smad3, and the NADPH oxidases NOX1 and NOX4. Moreover, abundance of neuronal NOS (nNOS) mRNA and BH4 content were significantly increased. Furthermore, inhibition of QDPR resulted in a significant increase in TGF-ß1 expression. In conclusion, QDPR might be an important factor mediating diabetic nephropathy through its regulation of TGF-ß1/Smad3 signaling and NADPH oxidase.
Subject(s)
Dihydropteridine Reductase/metabolism , Gene Expression , Kidney/metabolism , Transforming Growth Factor beta1/metabolism , Animals , Base Sequence , Cell Line , DNA Primers , Humans , Kidney/cytology , Rats , Rats, WistarABSTRACT
Nitric oxide (NO) has major physiological and cellular effects on muscle growth, repair, and function. In most muscle biopsies from humans with myopathies, sarcolemma-localized neuronal nitric oxide synthase (nNOS) is either reduced or not detected, particularly in dystrophin-deficient Duchenne muscular dystrophy (DMD). Abnormal NO signaling at the sarcolemmal level is integrally involved in the pathogenesis and accounts, at least in part, for the muscle weakness of DMD. Dystrophic muscle fibers exhibit an increased susceptibility to contraction-induced membrane damage. Muscle relaxants function to prevent muscle wasting by decreasing nerve impulses and reducing calcium influx that regulates tensing or tightening of muscle fibers. We have recently developed a new class of nitric esters that combines the pharmacological functions of NO and muscle relaxation. Here, we report the synthesis and properties of the nitric ester (MMPN) of 2-methyl-2-n-propyl-1,3-propanediol (MPP) and its effect in mdx dystrophic mice, a murine model of DMD. MMPN produced significant improvements in biochemical, pathological, and functional phenotypes in the mouse model. The endurance of exercise was extended by 47% in time to exhaustion and 84% in running distance. Serum CK level was decreased by 30%. Additionally, MMPN decreased intracellular free calcium concentration without causing skeletal muscle weakness. No hepatic or renal toxicities were observed during the study. Our investigations unveil a potential new treatment for muscular diseases.
Subject(s)
Muscular Dystrophy, Duchenne/drug therapy , Muscular Dystrophy, Duchenne/metabolism , Neuromuscular Agents/therapeutic use , Animals , Blotting, Western , Cells, Cultured , Male , Mice , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Myostatin/metabolism , Nitric Oxide/metabolismABSTRACT
OBJECTIVE: To determine the hepatoprotective effect of silymarin with Chang cell cultures. Specifically, to investigate the antioxidant properties of silymarin and its protective function in reducing pro-apoptotic markers. METHODS: Intracellular free radical levels were assessed with dichlorofluorescein (DCF) fluorescence after exposing cells to an oxidative stress of 400 µmol/L H2O2 for 20 min. Levels of cellular ATP and bax expression were examined to evaluate the protective effects of silymarin. RESULTS: Silymarin significantly reduced the DCF fluorescence signal. Cell viability, assessed by the MTT assay, showed that silymarin enhanced the cell growth. Drug treatment was also associated with enhanced ATP levels, and reduced Bax and protein mRNA levels. CONCLUSION: Silymarin can function as a hepatoprotectant against free radical damage due to oxidative stress. The protective nature extends to reducing levels of pro-apoptotic Bax protein. Silymarin may be a useful adjuvant for the treatment of specific liver diseases.
Subject(s)
Antioxidants/pharmacology , Apoptosis/drug effects , Hepatocytes/cytology , Silymarin/pharmacology , Adenosine Triphosphate/metabolism , Cell Line , Fluoresceins , Free Radicals/metabolism , Hepatocytes/metabolism , Humans , Hydrogen Peroxide , Protective Agents/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolismABSTRACT
In Duchenne muscular dystrophy (DMD), palliative glucocorticoid therapy can produce myopathy or calcification. Since increased nitric oxide synthase activity in dystrophic mice promotes regeneration, the outcome of two nitric oxide (NO) donor drugs, MyoNovin (M) and isosorbide dinitrate (I), on the effectiveness of the anti-inflammatory drug prednisone (P) in alleviating progression of dystrophy was tested. Dystrophic mdx mice were treated (18 days) as controls or with an NO donor ± P. Fiber permeability and DNA synthesis were labeled by Evans blue dye (EBD) and bromodeoxyuridine uptake, respectively. P decreased body weight gain, M increased quadriceps mass, and I increased heart mass. P increased fiber permeability (%EBD+ fibers) and calcification in diaphragm. Treatment with NO donors + P (M+P, I+P) reduced %EBD+ fibers and calcification vs. P alone. %EBD+ fibers in M+P diaphragm did not differ from control. NO donor treatment reduced proliferation and the population of c-met+ cells and accelerated fiber regeneration. Concurrent with P, NO donor treatment suppressed two important detrimental effects of P in mice, possibly by accelerating regeneration, rebalancing satellite cell quiescence and activation in dystrophy, and/or increasing perfusion. Results suggest that NO donors could improve current therapy for DMD.
Subject(s)
Diaphragm/drug effects , Guaifenesin/analogs & derivatives , Isosorbide Dinitrate/therapeutic use , Muscular Dystrophy, Duchenne/drug therapy , Nitrates/therapeutic use , Nitric Oxide Donors/therapeutic use , Prednisone/adverse effects , Animals , Body Weight/drug effects , Calcinosis/chemically induced , Calcinosis/prevention & control , Diaphragm/physiopathology , Female , Guaifenesin/therapeutic use , Male , Mice , Mice, Inbred mdx , Muscle, Skeletal/pathology , Muscular Diseases/chemically induced , Muscular Diseases/prevention & control , Organ Size , Prednisone/therapeutic use , Regeneration/drug effectsABSTRACT
UNLABELLED: Chronic hepatitis B (CHB) is major global health problem. In China, where about 120,000,000 persons are chronically infected, CHB has been treated for centuries with traditional Chinese medicines (TCMs). This review summarizes and meta-analyzes the results of randomized controlled trials (RCTs) of TCM formulations reported in China in 1998-2008 for treatment of CHB. RCTs comparing either TCM formulations alone or in combination with interferon (IFN) or lamivudine (LAM) versus IFN or LAM were included. Chinese electronic databases were searched. The methodological quality of RCTs was assessed using the Jadad scale. TCMs had a greater beneficial effect (P = 0.0003) than IFN and a slightly better effect (P = 0.01) than LAM on the normalization of serum alanine aminotransferase. TCMs had a similar beneficial effect when compared with IFN or LAM for CHB on antiviral activity as evidenced by the loss of serum hepatitis B e antigen and hepatitis B virus (HBV) DNA. TCMs enhanced IFN and LAM antiviral activities and improvements of liver function. The quality of many studies was poor; reports often lacked information regarding methods of randomization or blinding and adverse events. CONCLUSION: Some TCMs seem effective as alternative remedies for patients with CHB, suggesting that further study of TCMs in the treatment of CHB is warranted, both in preclinical models of HBV infection and in higher quality RCTs worldwide.
Subject(s)
Biomedical Research/statistics & numerical data , Drugs, Chinese Herbal/therapeutic use , Hepatitis B, Chronic/drug therapy , Medicine, Chinese Traditional , Phytotherapy , China , Humans , Randomized Controlled Trials as Topic/statistics & numerical dataABSTRACT
Peroxisome proliferator-activated receptor (PPAR) agonists such as clofibrate are known to affect liver fatty acid binding protein (L-FABP) levels, which in turn influence hepatocellular oxidant status. The mechanism of clofibrate's modulation of L-FABP levels is not clear. In this study we used clofibrate (PPARα agonist), MK886 (PPARα antagonist), and GW9662 (PPARγ antagonist) in determining the regulating mechanism of L-FABP expression and its antioxidant activity in CRL-1548 hepatoma cells. Antioxidant activity was assessed by determining intracellular reactive oxygen species (ROS) using dichlorofluorescein (DCF) fluorescence. The effect of clofibrate on cytosolic activity of the intracellular antioxidant enzymes was also assessed. RT-PCR and mRNA stability assay showed that clofibrate treatment enhanced L-FABP mRNA stability, which resulted in increased L-FABP levels. A nuclear run-off assay and RT-PCR measurements of L-FABP mRNA revealed that clofibrate increased the L-FABP gene transcription rate. The increased L-FABP was associated with reduced cytosolic ROS. Levels of superoxide dismutase, glutathione peroxidase, and catalase were not affected by clofibrate treatment. L-FABP siRNA knockdown studies showed that a reduction in L-FABP expression was associated with increased DCF fluorescence. We conclude that clofibrate enhanced L-FABP gene transcription and mRNA stability, thus affecting L-FABP expression and ultimately cellular antioxidant activity.
Subject(s)
Clofibrate , Fatty Acid-Binding Proteins , PPAR alpha , Anilides/pharmacology , Animals , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/enzymology , Catalase/analysis , Catalase/metabolism , Cell Line, Tumor , Clofibrate/pharmacology , Fatty Acid-Binding Proteins/analysis , Fatty Acid-Binding Proteins/chemistry , Fatty Acid-Binding Proteins/genetics , Fatty Acid-Binding Proteins/metabolism , Fluoresceins/analysis , Gene Expression Regulation, Neoplastic/drug effects , Glutathione Peroxidase/analysis , Glutathione Peroxidase/metabolism , Hepatocytes/drug effects , Hepatocytes/enzymology , Humans , Hydrogen Peroxide/administration & dosage , Indoles/pharmacology , Liver Neoplasms/drug therapy , Liver Neoplasms/enzymology , Oxidative Stress , PPAR alpha/agonists , PPAR alpha/antagonists & inhibitors , PPAR alpha/genetics , PPAR alpha/metabolism , RNA Stability/drug effects , RNA, Messenger/metabolism , Rats , Superoxide Dismutase/analysis , Superoxide Dismutase/metabolismABSTRACT
Hepatocytes expressing liver fatty acid binding protein (L-FABP) are known to be more resistant to oxidative stress than those devoid of this protein. The mechanism for the observed antioxidant activity is not known. We examined the antioxidant mechanism of a recombinant rat L-FABP in the presence of a hydrophilic (AAPH) or lipophilic (AMVN) free radical generator. Recombinant L-FABP amino acid sequence and its amino acid oxidative products following oxidation were identified by MALDI quadrupole time-of-flight MS after being digested by endoproteinase Glu-C. L-FABP was observed to have better antioxidative activity when free radicals were generated by the hydrophilic generator than by the lipophilic generator. Oxidative modification of L-FABP included up to five methionine oxidative peptide products with a total of approximately 80 Da mass shift compared with native L-FABP. Protection against lipid peroxidation of L-FABP after binding with palmitate or alpha-bromo-palmitate by the AAPH or AMVN free radical generators indicated that ligand binding can partially block antioxidant activity. We conclude that the mechanism of L-FABP's antioxidant activity is through inactivation of the free radicals by L-FABP's methionine and cysteine amino acids. Moreover, exposure of the L-FABP binding site further promotes its antioxidant activity. In this manner, L-FABP serves as a hepatocellular antioxidant.
Subject(s)
Antioxidants/metabolism , Fatty Acid-Binding Proteins/metabolism , Amidines/chemistry , Amidines/pharmacology , Animals , Antioxidants/chemistry , Ascorbic Acid/pharmacology , Azo Compounds/chemistry , Azo Compounds/pharmacology , Escherichia coli/chemistry , Escherichia coli/metabolism , Fatty Acid-Binding Proteins/biosynthesis , Fatty Acid-Binding Proteins/chemistry , Fluoresceins/chemistry , Fluorescence , Free Radicals/chemistry , Free Radicals/pharmacology , Glutathione Transferase/biosynthesis , Glutathione Transferase/isolation & purification , Glutathione Transferase/metabolism , Lipid Peroxidation/drug effects , Nitriles/chemistry , Nitriles/pharmacology , Rats , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , alpha-Tocopherol/pharmacologyABSTRACT
Skeletal muscle is a highly-specialized tissue that is capable of contractile function and able to withstand and adapt to daily mechanical and physiological stress. Musculoskeletal disorders, including muscular dystrophies, result in chronic pain and disability, reduced quality of life, burden on family, and increased healthcare costs. Although several mechanisms have been identified for muscle injury and regeneration, mechanisms of these diseases are poorly understood, and targeted and effective pharmacologic treatment(s) are not available. More research is needed in this area in order to develop effective treatment regimens. The purpose of this review is to discuss known mechanisms of muscle injury and repair, and highlight some recent patents and research developments for treatment of skeletal muscle disorders.
Subject(s)
Muscle, Skeletal/drug effects , Muscle, Skeletal/injuries , Muscle, Skeletal/physiopathology , Muscular Diseases/drug therapy , Regeneration/drug effects , Regeneration/physiology , Humans , Patents as TopicABSTRACT
Liver fatty acid binding protein has recently been shown to possess antioxidant properties but its role in liver disease, such as cholestasis, is not known. Since oxidative stress has been recognized as an important contributing factor in liver disease, we investigated the expression and antioxidative function of this protein using the bile-duct ligated model of cholestasis. Rats were divided into 3 groups: sham, bile-duct ligated and bile-duct ligated plus clofibrate. Animals were sacrificed at various time points after bile-duct ligation. RT-PCR and Western blot were used to analyze liver fatty acid binding protein expression. Cellular lipid peroxidation products were assessed by measuring thiobarbituric acid-reactive substances. Liver function was evaluated by measuring serum total bilirubin, alanine aminotransferase and ammonia. Liver fatty acid binding protein mRNA and protein levels were reduced to 51% and 20% of sham, respectively at 2 weeks following bile-duct ligation (p<0.05). The decreased liver fatty acid binding protein was associated with a statistical increase in hepatic lipid peroxidation products (224%) and decrease in hepatic function. Clofibrate treatment restored protein level and improved hepatic function. Clofibrate treatment also reduced hepatic lipid peroxidation products by 68% as compared with the bile-duct ligated group (p<0.05). Liver fatty acid binding protein likely has important antioxidant function during hepatocellular oxidative stress.
Subject(s)
Cholestasis/physiopathology , Clofibrate/pharmacology , Fatty Acid-Binding Proteins/physiology , Hypolipidemic Agents/pharmacology , Oxidative Stress/physiology , Animals , Bile Ducts , Blotting, Western , Disease Models, Animal , Fatty Acid-Binding Proteins/metabolism , Free Radicals , Ligation , Lipid Peroxidation , Liver/pathology , Liver Function Tests , Male , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain ReactionABSTRACT
PURPOSE: We investigated a potential hepatoprotective role of d-cis diltiazem, l-cis diltiazem, thiamine and the combination d-cis diltiazem and thiamine against lipid peroxidation in a piglet liver microsomal model. A modified in vitro dichlorofluorescein assay was developed to assess the extent of peroxidative damage induced by reactive oxygen species in the piglet liver microsomal fraction. METHODS: Microsomal membrane fraction, obtained from 3 week old female piglets, was treated with either the biologically vasoactive d-cis diltiazem or the non-vasoactive stereoisomer l-cis diltiazem (5-1000 microM) for 1 hour at 37 degrees C followed by one hour incubation with the free radical generator AAPH (2,2'-azobis-(2-amidinopropane) dihydrochloride; 1 mM) to initiate lipid peroxidation. In a separate study, piglet liver microsomes were pre-treated with d-cis diltiazem (50 or 500 microM) and thiamine (10-100 microM) to assess the antioxidant activity of the combination. RESULTS: A dose dependant inhibition of membrane lipid peroxidation was observed with d-cis diltiazem (p<0.05) but not with l-cis diltiazem, suggesting that diltiazem is stereospecific in protecting against microsomal lipid peroxidation. Combining diltiazem with thiamine further protected microsomes against lipid peroxidation compared to use of individual drugs. CONCLUSION: We conclude that diltiazem and the combination of diltiazem and thiamine offers a hepatoprotective effect against free radicals.
Subject(s)
Antioxidants/pharmacology , Calcium Channel Blockers/pharmacology , Diltiazem/pharmacology , Lipid Peroxidation/drug effects , Protective Agents/pharmacology , Thiamine/pharmacology , Animals , Antioxidants/administration & dosage , Calcium Channel Blockers/administration & dosage , Diltiazem/administration & dosage , Diltiazem/chemistry , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Synergism , Drug Therapy, Combination , Female , Fluoresceins , Microsomes, Liver , Protective Agents/administration & dosage , Reactive Oxygen Species/metabolism , Stereoisomerism , Swine , Thiamine/administration & dosage , Thiamine/chemistryABSTRACT
The role of bone morphogenetic protein 4 (BMP4), a crucial epithelial-mesenchymal transition (EMT) mediator, in the progression of hepatocellular carcinoma (HCC) patients heretofore has not been elucidated. The present study analyzed BMP4 expression in tumors and paired non-tumorous liver tissue and its correlation with clinicopathological characteristics from two independent cohorts consisting of 420 HCC patients. Functional analysis of BMP4 was performed in Bel-7402 and HCCLM3 HCC cells, and in a murine HCC model. The downstream targets of BMP4 in HCC were screened and confirmed. The results indicated that BMP4 expression was significantly increased in HCC tissue and highly metastatic HCC cells. BMP4 expression was correlated with vein invasion, overall survival and recurrence-free survival of HCC. BMP4 promoted HCC EMT and metastasis in vitro, and consistently in vivo. BMP4 knockdown blocked EMT and tumor metastasis in nude mice. ID2 was up-regulated by recombinant human BMP4, resulting in HCC EMT. Knockdown of ID2 blocked BMP4-induced EMT. In conclusion, BMP4 promotes invasion and metastasis of HCC by an induction of EMT via up-regulating ID2. BMP4 may be a valuable prognostic factor and potential therapeutic target for HCC therapy.
Subject(s)
Bone Morphogenetic Protein 4/metabolism , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/physiopathology , Epithelial-Mesenchymal Transition/genetics , Inhibitor of Differentiation Protein 2/genetics , Liver Neoplasms/physiopathology , Up-Regulation , Female , Humans , Liver Neoplasms/genetics , Male , Middle Aged , Recombinant Proteins/geneticsABSTRACT
BACKGROUND: Bone morphogenetic protein-4 (BMP4) is a key regulator of epithelial-mesenchymal transition (EMT), which is crucial for cancer cells to acquire chemoresistance. The effects of BMP4 on OXA sensitivity in HCC need to be elucidated. METHODS: Functional analysis of BMP4 on EMT-regulated OXA sensitivity was performed in human HCC specimens, in the HCC cell lines HepG2 and HCCLM3, and in a subcutaneous tumor model receiving OXA treatment. The downstream signaling targets of BMP4 in HCC were profiled and confirmed. RESULTS: BMP4 expression was significantly increased in HCC tissue, and was correlated with tumor de-differentiation and unfavorable prognosis. BMP4 promoted HCC EMT and was correlated with OXA resistance. Blocking of BMP4 reversed EMT and increased OXA chemosensitivity in vitro and in vivo. ELK1, a transcription factor involved in EMT, was an important mediator of BMP4-induced OXA resistance in HCC. Blocking of MEK/ERK/ELK1 attenuated BMP4-induced EMT and enhanced OXA sensitivity. CONCLUSIONS: BMP4 induces EMT and OXA chemoresistance via MEK/ERK/ELK1 signaling pathway in HCC. BMP4 may be a valuable therapeutic target for HCC patients receiving OXA-based chemotherapy.
Subject(s)
Bone Morphogenetic Protein 4/genetics , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Organoplatinum Compounds/pharmacology , Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Proliferation/physiology , Drug Resistance, Neoplasm , Epithelial-Mesenchymal Transition , Female , Hep G2 Cells , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , MAP Kinase Kinase 1/genetics , MAP Kinase Kinase 1/metabolism , MAP Kinase Signaling System , Male , Middle Aged , Oxaliplatin , Transfection , ets-Domain Protein Elk-1/metabolismABSTRACT
Contemporary Western medicines approved by the U.S. Food and Drug Administration (FDA) for the treatment of chronic hepatitis B (CHB), although available in China, have high costs, or major side effects and limited effectiveness. Research efforts have focused on looking for natural products as alternative medicines with low cost and good safety for CHB treatment. Chinese medicine (CM) has ancient, time-honored theories about methods of diagnosis and treatment for liver diseases. In recent decades, a large number of clinical trials and pre-clinical studies, which were performed in China and other countries, indicated that CM has potential benefit in several aspects of the treatment of CHB, e.g., anti-inflammatory, anticancer, antioxidant, immunomodulating, antifibrosis, and antiviral. However, there are many concerns regarding the study design and the quality of clinical trials. Further larger, stringently designed, double-blind, placebo control, randomized clinical trials and long-term follow-up are needed to provide conclusive evidence of their efficacy and safety. Components of CM deserve further study in pre-clinical models of HBV infection and in clinical trials world-wide.
Subject(s)
Drugs, Chinese Herbal/therapeutic use , Hepatitis B, Chronic/drug therapy , Medicine, Chinese Traditional , Humans , Randomized Controlled Trials as Topic , World Health OrganizationABSTRACT
Nitric oxide (NO) mediates activation of satellite precursor cells to enter the cell cycle. This provides new precursor cells for skeletal muscle growth and muscle repair from injury or disease. Targeting a new drug that specifically delivers NO to muscle has the potential to promote normal function and treat neuromuscular disease, and would also help to avoid side effects of NO from other treatment modalities. In this research, we examined the effectiveness of the NO donor, iosorbide dinitrate (ISDN), and a muscle relaxant, methocarbamol, in promoting satellite cell activation assayed by muscle cell DNA synthesis in normal adult mice. The work led to the development of guaifenesin dinitrate (GDN) as a new NO donor for delivering nitric oxide to muscle. The results revealed that there was a strong increase in muscle satellite cell activation and proliferation, demonstrated by a significant 38% rise in DNA synthesis after a single transdermal treatment with the new compound for 24 h. Western blot and immunohistochemistry analyses showed that the markers of satellite cell myogenesis, expression of myf5, myogenin, and follistatin, were increased after 24 h oral administration of the compound in adult mice. This research extends our understanding of the outcomes of NO-based treatments aimed at promoting muscle regeneration in normal tissue. The potential use of such treatment for conditions such as muscle atrophy in disuse and aging, and for the promotion of muscle tissue repair as required after injury or in neuromuscular diseases such as muscular dystrophy, is highlighted.
Subject(s)
Guaifenesin/analogs & derivatives , Guaifenesin/chemistry , Muscle Development/drug effects , Neuromuscular Agents/chemistry , Nitrates/chemistry , Nitric Oxide Donors/chemical synthesis , Nitric Oxide Donors/pharmacology , Satellite Cells, Skeletal Muscle/drug effects , Animals , Blotting, Western , Electron Spin Resonance Spectroscopy , Follistatin/metabolism , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Molecular Structure , Myogenic Regulatory Factor 5/metabolism , Myogenin/metabolism , Nitric Oxide/metabolism , Nitric Oxide Donors/chemistry , Satellite Cells, Skeletal Muscle/metabolismABSTRACT
The oxidation of 2',7'-dichlorodihydrofluorescein (2',7'-dichlorofluorescin, DCFH) to a fluorescent product, 2',7'-dichlorofluorescein (DCF), is commonly used to quantitatively measure oxidative stress in cells using a fluorescence microplate reader. However, many cell lines tend to grow non-uniformly in the wells. This non-uniform distribution results in a high degree of variability in the fluorescence signal and decreases the precision of the method. Also, samples treated in large culture plates, dishes or flasks cannot be assayed directly in fluorescence microplate readers. This study reports an improved DCF assay method that lyses cells with DMSO/PBS (90% dimethyl sulphoxide/10% phosphate buffered saline). Oxidative stress was induced with either hydrogen peroxide or an hypoxia-reoxygenation treatment. Cell lysis with DMSO/PBS resulted in highly stable fluorescence signals in comparison to Triton X-100/PBS lysed cells. The precision of DCF fluorescence measurements of DMSO/PBS lysed cells was much better than for attached cells measured directly in 96-well plates. While DCF fluorescence in PBS was strongly quenched by albumin, no quenching occurred in DMSO/PBS. In conclusion this study describes a more convenient and accurate method for measuring cellular oxidative stress that also makes it possible to assay cells treated in large culture plates.
Subject(s)
Dimethyl Sulfoxide/pharmacology , Fluoresceins/pharmacology , Oxidative Stress , Albumins/metabolism , Animals , Cell Line, Tumor , Dimethyl Sulfoxide/metabolism , Humans , Hydrogen Peroxide/metabolism , Hydrogen Peroxide/pharmacology , Hypoxia , K562 Cells , Oxygen/metabolism , Rats , SolutionsABSTRACT
Chlamydia trachomatis is an obligate intracellular bacterium and acquires both building blocks and energy from host cells for growth. The fatty acid-binding protein (FABP) plays an important role in uptake of long-chain fatty acids (LCFA) and energy metabolism by eukaryotic cells. The roles of FABP and LCFA in chlamydial infection were evaluated. Infection of liver cells with chlamydial organisms promoted fatty acid uptake by the infected cells, suggesting that LCFA may benefit chlamydial growth. Introduction of FABP into the liver cells not only enhanced fatty acid uptake, but also increased chlamydial intravacuolar replication and maturation. The FABP-enhanced chlamydial intracellular growth was dependent on the host cell uptake of fatty acids. These results have demonstrated that C. trachomatis can productively infect liver cells and utilize FABP-transported LCFA for its own biosynthesis.