metricas
covid
Buscar en
Annals of Hepatology
Toda la web
Inicio Annals of Hepatology Genistein decreases liver fibrosis and cholestasis induced by prolonged biliary ...
Journal Information
Vol. 6. Issue 1.
Pages 41-47 (January - March 2007)
Share
Share
Download PDF
More article options
Visits
1391
Vol. 6. Issue 1.
Pages 41-47 (January - March 2007)
Open Access
Genistein decreases liver fibrosis and cholestasis induced by prolonged biliary obstruction in the rat
Visits
1391
Alfonso Leija Salas1, Griselda Ocampo1, German Garrido Fariña2, Jorge Reyes-Esparza1, Lourdes Rodríguez-Fragoso1,
Corresponding author
mlrodrigl@yahoo.com.mx

Address for correspondence:
1 Facultad de Farmacia, Universidad Autónoma del Estado de Morelos. Cuernavaca, Morelos. México
2 FES-Cuautitlán, Universidad Nacional Autónoma de México. Cuautitlán, Izcalli Estado de México
This item has received

Under a Creative Commons license
Article information
Abstract
Full Text
Bibliography
Download PDF
Statistics
Figures (5)
Show moreShow less
Tables (1)
Table I. Effect of genistein on Total Bilirubins (TB), Serum Alkaline phosphatase (AP), Serum Alanine amino trasferase (ALT) and γ-Glutamil transpeptidase (γ-GTP)a.
Abstract

Fibrosis accompanies most chronic liver disorders and is a major factor contributing to hepatic failure. Therefore, the need for an effective treatment with the aim of modifying the clinical course of this disease is evident. The aim of this work is to determine whether genistein, which has been shown to modulate the physiology and pathophysiology of liver, is able to decrease experimental liver fibrosis and cholestasis. In male Wistar rats, the common bile duct was ligated. Administration of genistein (5 μg rat-1, day-1, p.o.) began four weeks after biliary obstruction and continued for a further four weeks. The liver was used for histological and ultrastructural analysis and for collagen quantification (hydroxyproline content). The degradation of Matrigel® and collagen type I was determined in homogenized liver. Bilirubins and enzyme activities were measured in serum. Genistein was able to improve normal liver histology, ultrastructure, collagen content, and biochemical markers of liver damage. It also increased Matrigel® and collagen type I degradation. In summary, the present report shows that genistein inhibits the fibrosis and cholestasis induced by prolonged biliary obstruction in the rat. Genistein has therapeutic potential against liver fibrosis.

Key words:
Genistein
fibrosis
cirrhosis
liver
Full Text

Abbreviations:

ECM: extracellular matriz, NASH: non-alcoholic steatohepatitis, HSC: hepatic stellate cells, SMA: smooth muscle α-actin, ROS: reactive oxygen species, TPK: protein tyrosine kinase, TGF-β 1: transforming growth factor,

Introduction

Liver fibrosis results from chronic damage to the liver in conjunction with the accumulation of extracellular matrix proteins, which is a characteristic of most types of chronic liver diseases.1-4 The main causes of liver fibrosis in industrialized countries include chronic HCV infection, alcohol abuse, and nonalcoholic steatohepatitis (NASH). The accumulation of ECM proteins distorts the hepatic architecture by forming a fibrous scar, and the subsequent development of nodules of regenerating hepatocytes defines cirrhosis.5,6

Hepatic fibrosis is considered a model of wound-healing response to chronic liver injury, and it is characterized by the activation of hepatic stellate cells (HSCs). The activation of HSCs involves the transdifferentiation from a quiescent state into myofibroblast-like cells with the appearance of smooth muscle a-actin (SMA) and loss of cellular vitamin A storage.7 The activated HSCs are distinguished by accelerated proliferation and enhanced production of ECM components. Cross-talks between parenchymal and nonparenchymal cells constitute the major interactions in the development of hepatic injury and fibrosis. Soluble factors, such as cytokines, chemokines, or reactive oxygen species are the mediators in these cross-talks, and are possible targets for therapeutic consideration8].

Genistein (4,5,7-trihydroxyisoflavone), a soy-derived isoflavone, has recently attracted much attention in the medical scientific community. Genistein is a potent protein tyrosine kinase inhibitor that attenuates growth factor-and cytokine-stimulated proliferation of both normal and cancer cells.10 Extensive epidemiological, in vitro, and animal studies have been performed, and most studies indicated that genistein has beneficial effects on a multitude of human disorders, including cancers, cardiovascular diseases, osteoporosis, and postmenopausal symptoms.11,13 The role of genistein in the physiology and pathophysiology of liver has been studied in the last decade.14-20 More than a dozen reports regarding the effect of genistein on HSCs have appeared. Antifibrotic effects of genistein in vitro have been shown.21,23 However, at present there are no reports regarding the effect of genistein on fibrogenesis in vivo. Therefore, the aim of this study was to evaluate the effect of genistein on the fibrosis and cholestasis induced by prolonged biliary obstruction in the rat.

Materials and methodsAnimal treatment and biliary obstruction

Male Wistar rats weighing 200 g were used. Animals had free access to food (Standard Purina Chow; St Louis, MO) and water. Obstructive jaundice was induced by double ligation and sectioning of the common bile duct. Control rats were sham operated. Genistein (Sigma Chemical Co., St Louis, MO) was dissolved in water and administered at a dose of 5μg per rat through an intragastric tube. This chosen dose was based on previous studies by our group.24 Administration of genistein began four weeks after biliary obstruction and was continued for a further four weeks. Another group of animals was bile duct ligated, but received only water, instead of the drug (fibrosis group). Each group consisted of six rats. The animals were sacrificed eight weeks after surgery under light ether anesthesia; blood was collected by heart puncture, and the liver was rapidly removed. Small liver sections fixed in Bouin’s medium were used for trichromic staining for histological examination under light microscopy. This investigation followed the Guiding Principles in the Care and Use of Animals from the National Institutes of Health 25.

Collagen quantification

Collagen concentration was determined by measuring hydroxyproline content in fresh liver samples after digestion with acid.26 The procedure was as follows. Fresh liver samples (100 mg) were placed in ampoules, 2 ml of 6 N HCl was added, and the ampoules were sealed and hydrolyzed at 100 °C for 48 h. The samples were then evaporated at 50 °C for 24 h and resuspended in 3 ml of sodium acetate/citric acid buffer (pH 6.0); 0.5 g of activated charcoal was added, and the mixture was stirred vigorously then centrifuged at 5000 xg for 10 min. The mixture was kept for 20 min at room temperature and the reaction was stopped by the addition of 2 M sodium thiosulfate and 1 N sodium hydroxide. The aqueous layer was transferred to test tubes. The oxidation product from hydroxyproline was converted to pyrrole by boiling the samples. The pyrrole-containing samples were incubated with Ehrlich’s reagent for 30 min and their absorbance was read at 560 nm. Recovery of known amounts of standards was carried out on similar liver samples to provide calibration samples.

Tissue extraction of proteases

Liver samples were homogenized with 10 volumes of buffer (0.075 M) potassium acetate, 0.3 M NaCl, 0.01 M EDTA, 0.1 M L-arginine and 0.25% Triton X-100; pH 4.2) on ice. After being kept in ice water for 3 h, the homogenized samples were centrifuged at 12 000 xg and 4 °C for 10 min. Supernatants were stored at -70 °C until assay.

Proteolytic activity assays

Assay plates were prepared by diluting ice-cold collagen I in 0.2% acetic acid with an equal volume of neutralizing buffer (100 mM Tris-HCl, 200 mM NaCl, 0.04% NaN3; pH 7.8) to give a final collagen concentration of 700 μg/mL. Aliquots (50 ¡uL) of the diluted collagen were added quickly to microwell modules and incubated at 30 °C for 40 h (16 h in a humidified atmosphere, then 24 h in a dried atmosphere) to gel and dry the collagen. The same procedure was performed using Matrigel® basement membrane extract. Aliquots of 100 μg were used per well and dried overnight after polymerization at room temperature.

The enzymatic essay was performed as described by Nethery et al.27 Enzyme samples were brought to assay temperature (35 °C) and replicate aliquots (100 μL) were added to the protein-coated wells. Microwells containing samples were incubated for 2 h in a humidified container equilibrated at assay temperature. The samples were decanted from the wells, which were washed twice in quick succession with assay buffer (50 mM Tris-HCl, 100 mM NaCl, 10mM CaCl2, 0.02% NaN3; pH 7.5). Stain solution (100 uL of 0.25 g Coomassie Blue R250, 10% acetic acid, 50% methanol) was incubated in the wells for 25 min at 25 °C. After the stain was drained off, the wells were washed three times with distilled water and left to dry at room temperature. The absorbance at 590 nm was measured using a Titertek Multiscan automatic spectro-photometer (Flow Laboratories, Titertek, Huntsville, AL) with the instrument blank set on unused microwells. Collagenase and urokinase were used as standard samples.

Serum enzyme activities and bilirubins

Serum was obtained for the following determinations: the activities of alkaline phosphatase, alanine amino transferase, μ-glutamyl transpeptidase, and for bilirubin content (Kit Merck, México)

Electron microscopic analysis

For ultrastructural analysis, liver blocks of ca 0.5-0.7 mm3 were fixed with 2.5% glutaraldehyde in 0.1 M phosphate buffer (pH 7.2) for 1 h and 1% OsO4 was added to continue fixing (30 min at 27 °C and 30 mi4n at 4 °C). After dehydration with increasing concentrations of ethyl alcohol, the blocks were embedded in epoxy resin. Ultrathin sections (80 nm) were cut and then examined with a Jeol-200 EX electron microscope at an accelerating voltage of 80 kV.

Statistical analysis

Data are reported as means ± standard deviation of three independent experiments conducted in quadruplicate. Statistical analysis was performed using a non parametric ANOVA. Individual differences between treatments was analyzed by a Tukey’s test. Significant differences were established at p < 0.001.

Results

Figure 1 shows the histological analysis of liver sections. Prolonged biliary obstruction was accompanied by an increase in collagen deposition around the portal triad (Figure 1C). In addition, the normal architecture was lost and a marked ductular proliferation was observed. Genistein treatment for four weeks in bile duct-ligated rats restored the normal architecture of the liver. A significant decrease in the collagen content was observed in the bile duct-ligated rats treated with genistein (Figure ID).

Figure 1.

Histological study of liver sections from rats treated with genistein. Liver sections from: (A) Control (Sham operated) rats; (B) Genistein; (C) bile duct-ligated rats; (D) bile duct-ligated rats and treated with genistein. Liver tissue were stained with Masson trichrome, collagen can be recognized by blue staining (200 X).

(0.34MB).

Serum markers of cholestasis and liver injury are shown in Table I. Total bilirubins increased more than 50-fold compared with controls. Serum activities of alkaline phosphatase, alanine aminotransferase, and γ-glutamiltranspeptidase increased approximately three-, six-, and sixfold, respectively, compared to the control group (p < 0.001). Genistein administration to bile duct-ligated rats significantly reduced the markers of cholestasis and liver injury.

Table I.

Effect of genistein on Total Bilirubins (TB), Serum Alkaline phosphatase (AP), Serum Alanine amino trasferase (ALT) and γ-Glutamil transpeptidase (γ-GTP)a.

Treatment  Total bilirubins (μmol/L)  AP μmol/L/min)  ALT (μmol/Lmin)  γ-GTP (μmol/L/min) 
Sham  2.5 ± 0.8  45 ± 9  32 ± 6  11.8 ± 8 
Genistein  1.9 ± 1.0  58 ± 15  26 ± 10  9.5 ± 10 
Fibrosis  145 ± 36b  167 ± 43b  196 ± 25b  76 ± 16b 
Fibrosis + Genistein  4 5 ± 12bc  67 ± 17c  95 ± 18bc  20 ± 9c 
a

Results are expressed as the mean value of experiments performed in duplicated assays with samples from six animals ± SEM.

b

p < 0.05 vs Sham group

c

p < 0.05 vs Fibrosis group

Liver collagen content was estimated in liver samples by measuring hydroxyproline. Biliary obstruction induced a sixfold increase in liver collagen content. Treatment of bile duct-ligated animals with genistein significantly decreased the collagen content; p < 0.001 (Figure 2).

Figure 2.

Liver collagen content determined in sham-operated rats (sham), genistein treated rats, bile duct-ligated rats (fibrosis) and bile duct-ligated rats and treated with genistein (fibrosis +genistein). Each bar represents the mean ± SEM in experiments performed in duplicate. All groups consisted of six animals. * Means different from the Sham group (p<0.001). # means different from the fibrosis group (p < 0.001).

(0.03MB).

Degradation of collagen type I is shown in Figure 3. Bile duct ligation increased twofold the degradation of collagen type I; genistein administration to bile duct-ligated rats increased this activity significantly (3.6-fold; p < 0.001). Matrigel® degradation by liver homogenates was used as an indicator of the connective tissue degradation capacity of the liver. Figure 4 show that genistein doubled Matrigel® degradation when administered to bile duct-ligated rats only.

Figure 3.

Type I collagen degradation by liver homogenates determined in sham-operated rats (sham), genistein treated rats, bile duct-ligated rats (fibrosis) and bile duct-ligated rats and treated with genistein (fibrosis +genistein). Each bar represents the mean ± SEM in experiments performed in duplicate. All groups consisted of six animals. * Means different from the Sham group (p < 0.001). # means different from the fibrosis group (p < 0.001).

(0.03MB).
Figure 4.

Quantitative Matrigel® degradation by liver homogenates deterHned in sham-operated rats (sham), genistein treated rats, bile duct-ligated rats (fibrosis) and bile duct-ligated rats and treated with genistein (fibrosis +genistein). Each bar represents the mean ± SEM in experiments performed in duplicate. All groups consisted of six animals. * Means different from the Sham group (p < 0.001). # means different from the fibrosis group (p < 0.001).

(0.02MB).

Ultrastructural analysis of the livers is shown in Figure 5. Panel A shows an electron micrograph of a liver section from a control rats (sham) in which hepatocyte are well organized. However, bile duct-ligated rats a high disorganization of hepatocytes with diverse grade of degeneration. We observed the presence of some HSCs activated associated with some collagen fibers (panel C). On the other hand, bile duct-ligated rats treated with genistein restored the ultra structure of hepatocytes which correlated with the improvement of liver function (panel D).

Figure 5.

Electron micrograph of livers from: (A) Control (Sham operated) rats; (B) Genistein; (C) bile duct-ligated rats; (D) bile duct-ligated rats and treated with genistein. HSCs = Hepatic stellate cells; H: hepatocytes, n: nucleus, cf: collagen fibers, (M1500).

(0.2MB).
Discussion

Hepatic fibrosis is a dynamic process resulting from chronic damage up to cirrhosis, characterized by accumulation of ECM components in the liver, caused by both markedly increased and unbalanced degradation of connective tissue components.28-30 Previous reports have shown that bile duct ligation in the rat for four weeks produces cirrhosis and a sixfold increase in liver collagen content.31-33 Our results show that genistein is able to ameliorate the cholestasis, fibrosis, liver architecture, and biochemical markers of liver damage when administered to rats that had been bile duct ligated for four weeks. The mechanism of the action of this compound is probably associated with its ability to reduce the proliferation of HSCs and then the production of hepatic collagen, as well as by increasing matrix degradation; however, the possibility of other actions cannot be disregarded.

The major obstacle to antifibrotic drug development is the slow evolution of fibrosis, which takes years or even decades in humans. Consequently, there is an evident need for an effective treatment with the aim of modifying the clinical course of this disease. A recent insight into the molecular pathogenesis of hepatic fibrosis and the role of activated HSCs provides hope for future development of successful therapy. Genistein is a new drug with hepatic protective properties that may be beneficial in liver fibrosis.

Fibrosis is a well-known phenomenon that leads to loss of normal architecture and function. Thus, restoration of liver homeostasis (i.e., regulation of serum bilirubins and enzymes) by genistein could be explained, at least in part, by the antifibrotic effect of this compound. However, the amelioration of serum bilirubins and enzyme activities cannot be fully explained with the present data. One possibility is that genistein affected the serum bilirubin and enzyme concentration by mechanisms other than by preventing fibrosis. These mechanisms could include decreased production of bilirubins and enzymes or their increased elimination via an anti-oxidative pathway.

Natural flavonoids possess reactive phenolic groups and show antioxidative properties in vitro. Aneja et al.34 showed that the pretreatment of animals with genistein markedly increases the intracellular reduced glutathione (GSH) levels in animals treated with CCl4 and restores them to normal levels. They suggest that the induction of GSH levels may be due to the enhancement of GSH synthesizing enzymes such as c-glutamyl cysteine synthetase and GSH synthetase, which are key enzymes in its biosynthesis.35-36 They also speculate that genistein may cooperate with physiological defense molecules such as reduced glutathione in such a way as to protect animals against oxidative stress. Recently, Lee et al.14 reported that genistein at higher levels decreased hepatic fat accumulation possibly by increasing fatty acid oxidation and uncoupling protein; in low doses, genistein increased mitochondrial enzyme activities in mice with fatty liver and obesity induced by high-fat diets. Taking these data all together could explain why genistein ameliorates liver function in bile duct-ligated rats.

There is a wealth of evidence that HSCs orchestrate most of the important events in liver fibrogenesis. After liver injury, HSCs become activated to a profibrogenic myofibroblast phenotype and can regulate net deposition of collagens and other matrix proteins in the liver.8932 it has been shown that genistein is able to inhibit PDGF-driven proliferative activity of rat HSCs 2, and also inhibits the TGF-31-stimulated collagen synthesis.23 Genistein also influences proliferation of HSCs, suppresses the expression of a-SMA in HSCs, and inhibits the intensity of c-fos, c-jun, and cyclin D1 expression of HSCs.37 In this work we have shown that genistein reduced the total amount of liver collagen and ameliorated the liver architecture; therefore, it is possible that genistein also influences the HSCs in vivo.

The effects of genistein on other hepatic cells have also been studied. The inhibition of cell proliferation and the induction of apoptosis via activation of caspase-3 have been observed in genistein-treated liver cancer-bearing animals.38 It has been shown that genistein modulates gene expression in hepatocytes.19 On the other hand, it has been observed that 100, amol/L genistein increased the synthesis of nitric oxide by sinusoidal endot-helial cells from the early stage (stage I) of fibrosis.39 Those data suggest that genistein may play an important role in regulating the function of all cells residing in the liver, not only in physiological conditions, but also in liver disease.

Extracellular matrix deposition is a constant feature in liver cirrhosis. Proteolytic enzymes are thought to play a primary role in the degradation of the connective tissue generated in processes such as fibrosis.40 Different enzymatic systems may contribute to the overall process of ECM degradation, including plasminogen activators, matrix metalloproteinases (i.e., collagenases), and cathe-psin D, and it is likely that these enzymatic systems can act either independently or in a concerted manner.41 The involvement of these powerful proteolytic systems in pathologies such as hepatic fibrosis reinforces the idea that specific mechanisms must exist for its regulation and could be a target for therapeutic strategies in that process. Thus, the increase in the proteolytic activity mediated by genistein administration may be one of the antifibrotic mechanisms of this compound. It has been reported that genistein can modulate the secretion of urokinase type plasminogen activator and other metalloproteinases in human cancer cell lines.42-44 Nevertheless, further investigations concerningthemodulationof the proteolytic activity in the liver disease by genistein are needed.

Insummary, the present report showed that genistein inhibits the fibrosis and cholestasis induced by prolonged biliaryobstruction in the rat. Genistein has therapeutic potential against liver fibrosis.

References
[1.]
De Franchis R., Hadengue A., Lau G., Lavanchy D., Lok A., McIntyre N., Mele A., Paumgartner G., Pietrangelo A., Rodes J., Rosenberg W., Valla D..
EASL International Consensus Conference on Hepatitis B. 13-14 September, 2002 Geneva, Switzerland. Consensus statement (long version).
J Hepatol, 39 (2003), pp. S3-25
[2.]
Alberti A., Chemello L., Benvegnu L..
Natural history of hepatitis C.
J Hepatol, 31 (1999), pp. 17-24
[3.]
Mendez-Sanchez N., Chavez-Tapia N.C., Uribe M..
Hepatocyte transplantation for acute and chronic liver diseases.
Ann Hepatol, 4 (2005), pp. 212-215
[4.]
Neuschwander-Tetri B.A., Caldwell S.H..
Nonalcoholic steatohepatitis: summary of an AASLD Single Topic Conference.
Hepatology, 37 (2003), pp. 1202-1219
[5.]
Knodell R.G., Ishak K.G., Black W.C., Chen T.S., Craig R., Kaplowitz N., Kiernan T.W., Wollman J..
Formulation and application of a numerical scoring system for assessing histological activity in asymptomatic chronic active hepatitis.
Hepatology, 1 (1981), pp. 431-435
[6.]
Ishak K.G..
Chronic hepatitis: morphology and nomenclature.
Mod Pathol, 7 (1994), pp. 690-713
[7.]
Tilg H., Kaser A., Moschen A.R..
How to modulate inflammatory cytokines in liver diseases.
Liver International, 26 (2006), pp. 1029-1039
[8.]
Friedman S.L..
Molecular regulation of hepatic fibrosis, an integrated cellular response to tissue injury.
J Biol Chem, 275 (2000), pp. 2247-2250
[9.]
Wu J., Zern M.A..
Hepatic stellate cells: a target for the treatment of liver fibrosis.
J Gastroenterol, 35 (2000), pp. 665-672
[10.]
Polkowski K., Mazurek A.P..
Biological properties of genistein. A review of in vitro and in vivo data.
Acta Pol Pharm, 57 (2000), pp. 135-155
[11.]
Goldwyn S., Lazinsky A., Wei H..
Promotion of health by soy isoflavones: efficacy, benefit and safety concerns.
Drug Metabol & Drug Interact, 17 (2000), pp. 261-289
[12.]
Caldarelli A., Diel P., Vollmer G..
Effect of phytoestrogens on gene expression of carbonic anhydrase II in rat uterus and liver.
J Steroid Biochem Mol Biol, 97 (2005), pp. 251-256
[13.]
Wang H.K..
The therapeutic potential of flavonoids.
Expert Opin Investig Drugs, 9 (2000), pp. 2103-2119
[14.]
Lee Y.M., Choi J.S., Kim M.H., Jung M.H., Lee Y.S., Song J..
Effects of dietary genistein on hepatic lipid metabolism and mitochondrial function in mice fed high-fat diets.
Nutrition, 22 (2006), pp. 956-964
[15.]
Atherton K.M., Mutch E., Ford D..
Metabolism of the soyabean isoflavone daidzein by CYP1A2 and the extra-hepatic CYPs 1A1 and 1B1 affects biological activity.
Biochem Pharmacol, 72 (2006), pp. 624-631
[16.]
Ae Park S., Choi M.S., Cho S.Y., Seo J.S., Jung U.J., Kim M.J., Sung M.K., Park Y.B., Lee M.K..
Genistein and daidzein modulate hepatic glucose and lipid regulating enzyme activities in C57BL/KsJ-db/ db mice.
Life Sci, 79 (2006), pp. 1207-1213
[17.]
Zhao J.H., Arao Y., Sun S.J., Kikuchi A., Kayama F..
Oral administration of soy-derived genistin suppresses lipopolysaccharide-induced acute liver inflammation but does not induce thymic atrophy in the rat.
Life Sci, 78 (2006), pp. 812-819
[18.]
Tikkanen M.J., Wahala K., Ohala S., Vihma V., Adlercreutz H..
Effect of soybean phytoestrogen intake on low-density lipoprotein oxidation resistance.
Proc Natl Acad Sci USA, 95 (1998), pp. 3106-3110
[19.]
Geis R.B., Diel P., Degen G.H., Vollmer G..
Effects of genistein on the expression of hepatic genes in two rat strains (Sprague-Dawley and Wistar).
Toxicol Lett, 157 (2005), pp. 21-29
[20.]
Fang Y.C., Chen B.H., Huang R.F., Lu Y.F..
Effect of genistein supplementation on tissue genistein and lipid peroxidation of serum, liver and low-density lipoprotein in hamsters.
J Nutr Biochem, 15 (2004), pp. 142-148
[21.]
Kang L.P., Qi L.H., Zhang J.P., Shi N., Zhang M., Wu T.M., Chen J..
Effect of genistein and quercetin on proliferation, collagen synthesis, and type I procollagen mRNA levels of rat hepatic stellate cells.
Acta Pharmacol Sin, 22 (2001), pp. 793-796
[22.]
Badria F.A., Dawidar A.A., Houssen W.E., Shier W.T..
In vitro study of flavonoids, fatty acids, and steroids on proliferation of rat hepatic stellate cells.
Z Naturforsch [C], 60 (2005), pp. 139-142
[23.]
Qi L.H., Kang L.P., Zhang J.P., Shi N., Zhang M., Wu T.M..
Antifibrotic effects of genistein and quercetin in vitro.
Yao Xue Xue Bao, 36 (2001), pp. 648-651
[24.]
Rodríguez F.L., Campos G.F., Ocampo M.G., Leija A., Garrido F.G., Reyes E.J..
Genistein decreases liver fibrosis and cholestasis from bile duct ligated rats.
The FASEB Journal, 19 (2004), pp. A437
[25.]
Committee on Care and Use of Laboratory Animals of the Institute for Laboratory Animal Resources, Commission on Life Sciences, National Research Council: Guide for the Care and Use of Laboratory Animals. Publication 86-23, 18 and the Animal Welfare Act of 1966, as amended.
[26.]
Rojkind M., Gonzalez P..
An improved method for determining specific radioactivities of proline-14C in collagen and non collagenous protein.
Anal Biochem, 57 (1974), pp. 1-7
[27.]
Nethery A., Lyons J.G., O’grady R.L..
A spectrophotometric collagenase assasy.
Anal Biochem, 159 (1986), pp. 390-395
[28.]
Bataller R., Brenner D.A..
Liver fibrosis.
J Clin Invest, 115 (2005), pp. 209-218
[29.]
Schuppan D., Ruehl M., Somasundaram R., Hahn E.G..
Matrix as a modulator of hepatic fibrogenesis.
Sem Liver Dis, 21 (2001), pp. 351-372
[30.]
Friedman S.L..
Liver fibrosis - from bench to bedside.
J Hepatol, 38 (2003), pp. s38-53
[31.]
Kinnman N., Housset C..
Peribiliary myofibroblasts in biliary type liver fibrosis.
Front Biosci, 7 (2002), pp. d496-503
[32.]
Issa R., Williams E., Trim N., Kendall T., Arthur M.J., Reichen J., Benyon R.C., Iredale J.P..
Apoptosis of hepatic stellate cells: involvement in resolution of biliary fibrosis and regulation by soluble growth factors.
Gut, 48 (2001), pp. 548-557
[33.]
Bueno M.R., Daneri A., Armendariz-Borunda J..
Cholestasis-induced fibrosis is reduced by interferon alpha-2a and is associated with elevated liver metalloprotease activity.
J Hepatol, 33 (2000), pp. 915-925
[34.]
Aneja R., Upadhyaya G., Prakash S., Dass S.K., Chandra R..
Ameliorating effect of phytoestrogens on CCl4-induced oxidative stress in the livers of male Wistar rats.
Artif Cells Blood Substit Immobil Biotechnol, 33 (2005), pp. 201-213
[35.]
Griffin D.W..
Mechanism of action, metabolism and toxicity of buthionine and its higher homologies, potent inhibitors of glutathione synthesis.
J Biol Chem, 257 (1982), pp. 13704-13712
[36.]
Seelig G.F., Meister A..
Glutathione biosynthesis gamma glutamylcysteine synthetase from rat kidney.
Methods Enzymol, 113 (1985), pp. 379-390
[37.]
Liu X.J., Yang L., Mao Y.Q., Wang Q., Huang M.H., Wang Y.P., Wu H.B..
Effects of the tyrosine protein kinase inhibitor genistein on the proliferation, activation of cultured rat hepatic stellate cells.
World J Gastroenterol, 8 (2002), pp. 739-745
[38.]
Chodon D., Banu S.M., Padmavathi R., Sakthisekaran D..
Inhibition of cell proliferation and induction of apoptosis by genistein in experimental hepatocellular carcinoma.
Mol Cell Biochem, (2006), pp. 28
[39.]
Liu X., Huang M., Cheng N., Xiao W., Wang Y..
Effects of genistein on the fenestrate, proliferation and nitric oxide synthesis of liver sinusoidal endothelial cells from carbon tetrachloride-induced experimental hepatic fibrosis rats.
Zhonghua Gan Zang Bing Za Zhi, 10 (2002), pp. 200-203
[40.]
Senties-Gomez M.D., Galvez-Gastelum F.J., Meza-Garcia E., Armendariz-Borunda J..
Hepatic fibrosis: role of matrix metalloproteases and TGF-beta.
Gac Med Mex, 141 (2005), pp. 315-322
[41.]
Bedossa P., Paradis V..
Regression of hepatic fibrosis physio-pathological aspects and clinical reality.
Press Med, 32 (2003), pp. 704-710
[42.]
Fajardo I., Diez E., Rodriguez-Nieto S., Rodriguez-Caso C., Quesada A.R., Sanchez-Jimenez F., Medina M.A..
Effects of genistein and 2-methoxyestradiol on matrix metalloproteinases and their inhibitors secreted by Ehrlich ascites tumor cells.
Anticancer Res, 20 (2000), pp. 1691-1694
[43.]
Garcia de Veas R., Schweigerer L., Medina M.A..
Modulation of the proteolytic balance plasminogen activator/plasminogen activator inhibitor by enhanced N-myc oncogene expression or application of genistein.
Eur J Cancer, 34 (1998), pp. 1736-1740
[44.]
Santibanez J.F., Navarro A., Martinez J..
Genistein inhibits proliferation and in vitro invasive potential of human prostatic cancer cell lines.
Anticancer Res, 17 (1997), pp. 1199-1204
Copyright © 2007. Fundación Clínica Médica Sur, A.C.
Article options
es en pt

¿Es usted profesional sanitario apto para prescribir o dispensar medicamentos?

Are you a health professional able to prescribe or dispense drugs?

Você é um profissional de saúde habilitado a prescrever ou dispensar medicamentos