Você está na página 1de 12

Amjad Horani, Nidal Muhanna, Orit Pappo, Alaa Melhem, Carlos E.

Alvarez, Sarit Doron, Wehbi Wehbi, Karussis Dimitrios, Scott L. Friedman and Rifaat Safadi
Am J Physiol Gastrointest Liver Physiol 292:628-638, 2007. First published Oct 12, 2006; doi:10.1152/ajpgi.00137.2006 You might find this additional information useful... This article cites 69 articles, 23 of which you can access free at: http://ajpgi.physiology.org/cgi/content/full/292/2/G628#BIBL This article has been cited by 1 other HighWire hosted article: Amelioration of hepatic fibrosis via beta-glucosylceramide-mediated immune modulation is associated with altered CD8 and NKT lymphocyte distribution R. Safadi, E. Zigmond, O. Pappo, Z. Shalev and Y. Ilan Int. Immunol., August 13, 2007; 0 (2007): dxm069v1-. [Abstract] [Full Text] [PDF] Updated information and services including high-resolution figures, can be found at: http://ajpgi.physiology.org/cgi/content/full/292/2/G628
Downloaded from ajpgi.physiology.org on April 24, 2009

Additional material and information about AJP - Gastrointestinal and Liver Physiology can be found at: http://www.the-aps.org/publications/ajpgi

This information is current as of April 24, 2009 .

AJP - Gastrointestinal and Liver Physiology publishes original articles pertaining to all aspects of research involving normal or abnormal function of the gastrointestinal tract, hepatobiliary system, and pancreas. It is published 12 times a year (monthly) by the American Physiological Society, 9650 Rockville Pike, Bethesda MD 20814-3991. Copyright 2005 by the American Physiological Society. ISSN: 0193-1857, ESSN: 1522-1547. Visit our website at http://www.the-aps.org/.

Am J Physiol Gastrointest Liver Physiol 292: G628 G638, 2007. First published October 12, 2006; doi:10.1152/ajpgi.00137.2006.

Benecial effect of glatiramer acetate (Copaxone) on immune modulation of experimental hepatic brosis
Amjad Horani,1* Nidal Muhanna,1* Orit Pappo,2 Alaa Melhem,1 Carlos E. Alvarez,4 Sarit Doron,1 Wehbi Wehbi,1 Karussis Dimitrios,3 Scott L. Friedman,4 and Rifaat Safadi1
Liver and Gastroenterology Units, Division of Medicine, 2Pathology Department, and 3Division of Neurology, Hadassah University Hospital, Jerusalem, Israel; and 4Division of Liver Diseases, Mount Sinai School of Medicine, New York, New York
Submitted 27 March 2006; accepted in nal form 5 August 2006
1

Horani A, Muhanna N, Pappo O, Melhem A, Alvarez CE, Doron S, Wehbi W, Dimitrios K, Friedman SL, Safadi R. Benecial effect of glatiramer acetate (Copaxone) on immune modulation of experimental hepatic brosis. Am J Physiol Gastrointest Liver Physiol 292: G628 G638, 2007. First published October 12, 2006; doi:10.1152/ajpgi.00137.2006.While CD8 subsets activate hepatic brosis, natural killer (NK) cells exhibit antibrotic activity. Glatiramer acetate (GA) is an immune modulator for multiple sclerosis. We assessed the potential impact of GA on mouse hepatic brogenesis. Hepatic brosis was induced in C57BL/6 mice by intraperitoneal administration of carbon tetrachloride (CCl4) for 6 wk. During the last 2 wk, animals were also treated with either GA (200 /day ip) or medium and compared with naive and brotic mice (8 animals/group). GA markedly attenuated brosis without altering reactive oxygen species production. By morphometric measurement of Sirius redstained tissue sections, the relative brosis area decreased from 5.28 0.32% (mean SE) in the untreated CCl4 group to 2.01 0.28% in CCl4 GA-treated animals, compared with 0.38 0.07% in naive mice. -Smooth muscle actin immunoblotting and mRNA expression revealed a similar pattern. Serum aminotransferase and Ishak-Knodell necroinammatory score were markedly elevated, to the same extent, in both CCl4-treated groups. Fibrosis induction was associated with signicant increase in CD8 subsets and decrease in CD4 T cells. After GA treatment, however, NK content, CD4 CD25 FoxP3 cells, hepatic expression of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), and apoptosis of hepatic stellate cells were all increased. Serum interleukin (IL)-10 levels markedly rose, whereas IL-4 fell. In vitro activation of human hepatic stellate cells cocultured with hepatitis C virus-derived peripheral blood lymphocytes decreased when lymphocytes were preincubated with GA before coculture. In an animal model of hepatic brosis, GA has an antibrotic effect associated with decreased CD8 cells and reduced serum IL-4 levels and increased NK cells, CD4 CD25 FoxP3 cells, TRAIL, and elevated serum IL-10 levels. cirrhosis; stellate cell; immune modulation; lymphocytes; liver injury
HEPATIC FIBROSIS IS THE RESULT

of chronic liver injury, regardless of etiology, during which hepatic stellate cells (HSC) proliferate and differentiate into matrix-producing myobroblastic cells (17, 18). The activity of stellate cells is inuenced by an array of cytokines, some of which are probrotic, e.g., transforming growth factor (TGF) (21), whereas others play an antibrotic role, e.g., interleukin (IL)-10 (62, 68) and interferon (58, 64). The antibrotic effect of IL-10 is further underscored by studies of cultured stellate cells in which

antibodies to IL-10 receptors induce an increase in brosis (68) and is supported by the analysis of the effect of brogenic drugs on IL-10-knockout mice (62) and by attenuation of experimental brosis following electroporative IL-10 gene therapy or transgenic IL-10 (10, 54). Different inammatory cells including natural killer (NK) cells, Kupffer cells, and lymphocytes each play a distinct role in the interplay of pro- and antibrotic stimuli in normal liver and after injury. Lymphocytes express high levels of antibrogenic cytokines such as interferon- and thus have an antibrotic prole (58, 68), whereas lymphocytes that express high levels of brogenic cytokines such as IL-4 have a probrotic role (58, 69). Therefore, subtle alterations in the intrahepatic cytokine ratio can titrate the intrahepatic milieu toward or away from brosis (22, 23). We previously explored (47, 54) the impact of IL-10 on CD8 and CD4 lymphocyte subsets in experimental liver injury. To this end, we generated a transgenic mouse with hepatocyte secretion of rat IL-10, to assess the impact of sustained local expression of the cytokine on hepatic brogenesis. An antibrotic effect of IL-10 was established, which was associated with fewer CD8 lymphocytes. Moreover, adoptive transfer of CD8 lymphocytes from mice with liver injury was found to initiate brosis in naive mice. In contrast to CD8 lymphocytes, NK cells exert an antibrotic effect (40, 46). Collectively, these ndings broaden our understanding of how mediation by various immune factors can affect brosis and point to the manipulation of the CD4, CD8, and NK subsets as a potential means of therapeutically modulating brosis. Glatiramer acetate (GA; also known as Copaxone or copolymer 1) is a synthetic copolymer drug, composed of a random mixture of four amino acids, used in the treatment of multiple sclerosis (MS) (7, 70). GA is a well-tolerated drug that is considered among the safest of the currently approved therapeutic agents for treating MS. Importantly, long-term treatment with GA does not lead to hematologic or liver enzyme abnormalities (27). However, the exact underlying mechanism by which GA exerts its effect in MS still remains elusive. Nevertheless, there is a growing body of evidence that GA reduces the relapse frequency of MS through its effect on dendritic cells (46, 66). In experimental autoimmune encephalitis, the immunologic cross-reaction between the myelin basic protein and GA is thought to be the basis for the suppressive activity of GA, through the induction of antigen-specic suppressor
The costs of publication of this article were defrayed in part by the payment of page charges. The article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact. http://www.ajpgi.org

Downloaded from ajpgi.physiology.org on April 24, 2009

* A. Horani and N. Muhanna contributed equally to this work. Address for reprint requests and other correspondence: R. Safadi, Liver and Gastroenterology Units, Div. of Medicine, Hadassah University Hospital, PO Box 12000, 91120 Jerusalem, Israel (e-mail: safadi@hadassah.org.il). G628

0193-1857/07 $8.00 Copyright 2007 the American Physiological Society

GLATIRAMER ACETATE AMELIORATES HEPATIC FIBROSIS

G629

cells (7, 27, 46, 70). Moreover, it is postulated that in experimental autoimmune encephalomyelitis GA promotes its activity by affecting Th2 CD4 cell development and increasing IL-10 secretion, as well as through in situ bystander suppression of dendritic cells (27, 45, 46). In light of the immunologic activities of GA, we speculated that this drug might have an antibrotic effect on liver injury. Thus we investigated the effect of GA on both the immunologic features of experimental liver injury and the extent of brosis in animals treated for 6 wk with carbon tetrachloride (CCl4), with or without GA, during the nal 2 wk after the toxin had been administered.
MATERIALS AND METHODS

Materials. CCl4 from Sigma (C-5331) and GA (Copaxone) from Teva were used in these studies. Animals. Eight-week-old male C57BL/6 wild-type mice were used. The animals received care according to the National Institutes of Health guidelines. The animal procedures were approved by the local Institutional Animal Care and Use Committee. Hepatic brosis induction. Hepatic brosis was induced by intraperitoneal CCl4 (diluted to 10% with corn oil) administered as a dose of 5 l/g body wt twice a week for 6 wk in 8-wk-old male wild-type C57BL/6 mice (26, 54). During the last 2 wk, the animals were also treated with an intraperitoneal injection of either 200 g of GA or medium per day. Both groups were compared with naive wild-type mice. Eight animals were included in each group. After ketaminexylazine anesthesia, the animals were euthanized 3 days after the nal dose of CCl4, after which serum, liver, and lymphocytes were harvested. Blood samples were obtained and frozen at 20C until being assayed for cytokine levels. Livers were divided for mRNA analysis and stored at 80C for staining, and the rest were used for isolating lymphocytes. Splenocytes and intrahepatic lymphocytes were isolated for uorescence-activated cell sorting (FACS) analysis from all animal groups as detailed below. Biochemistry. Sera from individual mice were obtained. Serum aspartate aminotransferase (AST) and alanine aminotransferase (ALT) levels were measured by an automatic analyzer. Liver histology. The posterior one-third of the liver was xed in 10% formalin for 24 h and then parafn-embedded with an automated tissue processor. Seven-millimeter sections were cut from each liver specimen. Hematoxylin and eosin staining was performed for each animal. The intrahepatic necroinammatory score was evaluated blindly by a histopathologist using the standard histology activity index as described by Knodell et al. (33). Liver sections (15 m) were also stained in 0.1% Sirius red F3B in saturated picric acid (both from Sigma) (54) for computerized Bioquant morphometry system analysis. For the in situ detection of HSC apoptosis, liver sections were double stained with annexin V and -smooth muscle actin (SMA) according to manufacturers instructions. Liver sections were analyzed by confocal microscopy (40). Hepatic brosis quantication. Hepatic brosis was assessed with a computerized Bioquant morphometry system (24, 25, 54, 67). Immunoblotting (using Western blotting) and semiquantitative PCR analysis of -SMA mRNA assessed liver extracts, as previously described (54) and as detailed below. Relative brosis area. Relative brosis area (expressed as % of total liver area) was assessed by analyzing 36 Sirius red-stained liver sections per animal. Each eld was acquired at 10 magnication and then analyzed with a computerized Bioquant morphometry system. To evaluate the relative brosis area, we divided the measured collagen area by the net eld area and then multiplied it by 100. Subtraction of the vascular luminal area from the total eld area yielded the nal calculation of the net brosis area.
AJP-Gastrointest Liver Physiol VOL

Western immunoblotting. Whole liver protein extracts were prepared in liver homogenization buffer [50 mM Tris HCl (pH 7.6), 0.25% Triton X-100, 0.15 M NaCl, 10 mM CaCl2, and a complete mini EDTA-free protease inhibitor cocktail (Roche Diagnostics, Mannheim, Germany)]. Next, proteins (30 g/lane) were resolved on a 10% (wt/vol) SDS-polyacrylamide gel (Novex, Groningen, The Netherlands) under reducing conditions. For immunoblotting, proteins were transferred to a Protran membrane (Schleicher & Schuell, Dassel, Germany). Blots were incubated overnight at 4C in a blocking buffer containing 5% skim milk and then incubated with an anti-SMA mouse monoclonal antibody (Dako, catalog no. M0851), diluted 1/2,000 and maintained for 2 h at room temperature and subsequently diluted 1/10,000 with peroxidase-conjugated goat antimouse IgG (PARIS, Compiegne, France) and maintained for 1 h at ` room temperature. Immunoreactivity was revealed by enhanced chemiluminescence with an ECL kit (Amersham Pharmacia Biotech, Les Ulis, France) (52). For tumor necrosis factor-related apoptosisinducing ligand (TRAIL), incubation with anti-TRAIL-R2 antibodies (dilution 1:1,000; AB1687 or AB16942; Chemicon, Temecula, CA) was used (5). Tissue RNA extraction and -SMA mRNA detection. Total cellular RNA was extracted from frozen liver tissues with TRIzol reagent (GIBCO-BRL, Life Technologies) and DNA digestion. RNA was then used as a template for reverse transcription into single-stranded cDNA with the Reverse Transcription System (Promega, Madison, WI). mRNAs for -SMA and -actin were detected by quantitative real-time PCR with the following primers: -actin (as a housekeeping gene): forward 5 -GAT GAG ATT GGC ATG GCT TT-3 , reverse 5 -AGA GAA GTG GGG TGG CTT TT-3 ; -SMA: forward 5 -TCC TCC CTG GAG AAG AGC TAC-3 , reverse 5 -TAT AGG TGG TTT CGT GGA TGC-3 (54); TRAIL: forward 5 -CCC TGC TTG CAG GTT AAG AG-3 , reverse 5 -GGC CTA AGG TCT TTC CAT CC-3 (46). Real-time PCR analysis of brogenic markers. Synthesis of cDNA was performed with 2 g of total RNA per sample with random primers and reagents contained in the Reverse Transcription System kit, according to the manufacturers protocol (Promega). The reverse transcriptase product was diluted 20 in nuclease-free H2O, and 5 l of each sample was loaded into 96-well plates for real-time PCR in an ABI Prism 7700 Sequence Detection System (Applied Biosystems). -Actin served as internal control, and H2O served as a negative control. Amplication reactions included oligonucleotide primers for each target gene and for -actin, as well as platinum Taq polymerase and SYBR Green DNA-binding dye. Fluorescence signals were analyzed during each of 40 cycles (denaturation 15 s at 95C, annealing 15 s at 56C, and extension 40 s at 72C). Denaturation curves of target genes and -actin, performed at the end of the PCR, and detection of the PCR products by agarose gel electrophoresis conrmed the homogeneity of the DNA products. Relative quantication was calculated with the comparative threshold cycle (CT) method (as described in User Bulletin no. 2, ABI PRISM 7700 Sequence Detection System). CT indicates the fractional cycle number at which the amount of amplied target genes reach a xed threshold within the linear phase of gene amplication and is inversely related to the abundance of mRNA transcripts in the initial sample. Mean CT of duplicate measurements would be used to calculate CT as the difference in CT for target and reference. CT for each sample was compared with the corresponding control CT, and the result was expressed as CT. Relative quantity of product was expressed as fold induction or repression of the target gene compared with the control primers, according to the formula 2 CT . Cytochrome P-450 2E1 activity. Fifty micrograms of liver was homogenized in 5% trichloroacetic acid at a ratio of 1:10 (wt/vol) and centrifuged for 5 min at 8,000 rpm and 4C. Catalytic activity of cytochrome P-450 2E1 (CYP2E1) was determined as the rate of production of p-nitrocatechol from p-nitrophenol (49).
292 FEBRUARY 2007

Downloaded from ajpgi.physiology.org on April 24, 2009

www.ajpgi.org

G630

GLATIRAMER ACETATE AMELIORATES HEPATIC FIBROSIS

HSC isolation. HSC were isolated from all groups by sequential pronase/collagenase digestion followed by Nycodenz density gradient centrifugation as described previously with minor modications in rats (19, 20). After anesthesia and abdominal exploration, liver perfusion via the portal vein with 20 ml MEM (GIBCO BRL) obtained hepatic washout. Perfusion was then followed by 10 ml of DMEMF-12 (GIBCO BRL) containing 0.5 mg Pronase (Roche Diagnostics, catalog no. 1459643) per gram of body weight of the mouse, followed by 10 ml of DMEM-F-12 containing 7 mg of Liberase enzyme 3 (Roche Diagnostics, catalog no. 1814184). The digested liver was mashed ex vivo and was incubated at 37C for 25 min in 50 ml of DMEM-F-12 solution containing 0.05% Pronase and 20 g/ml of DNase I (Roche Diagnostics, catalog no. 1284932). The resulting suspension was ltered through a 150- m steel mesh and centrifuged on an 8.2% Nycodenz (Sigma catalog no. D-2158) cushion at 1400 g for 15 min, which produced a stellate cell-enriched fraction in the upper whitish layer. Samples were stored at 4C until the FACS analysis was performed. Cell isolation, staining, and ow cytometric analysis. Spleens were harvested at the time of euthanasia and fractionated through a 70- m nylon cell strainer. After red blood cell lyses, the splenocytes were washed, suspended in RPMI 1640 medium, and stored at 4C until FACS analysis. Intrahepatic lymphocytes were isolated by perfusion of the liver with digestion buffer. After perfusion, the liver was homogenized and incubated at 37C for 30 min. Next, the digested liver cell suspension was centrifuged to remove hepatocytes and cell clumps. The supernatant was then centrifuged to obtain a pellet of cells depleted of hepatocytes to a nal volume of 1 ml. Lymphocytes were then isolated from this cell suspension by 24% metrizamide gradient separation (39, 54). Human peripheral blood was collected in heparin from patients and healthy control subjects after Institutional Review Board approval. Mononuclear cells were isolated by centrifugation over Ficoll-Hypaque (Pharmacia) according to Boyum (9). After three washes in saline, the cells were resuspended in RPMI 1640 medium supplemented with 50% heat-inactivated fetal calf serum (GIBCO) and 20% dimethyl sulfoxide; cells were stored at 80C. FACS analysis. Briey, lymphocytes were adjusted to 2 107/ml in staining buffer (in saline containing 1% bovine albumin), incubated for 30 min at 4C with antibodies conjugated to uorescein isothiocyanate (FITC), phycoerythrin (PE), or allophycocyanin (APC), washed three times, and then resuspended in xative solution with 2% paraformaldehyde for analysis. Lymphocytes were stained with monoclonal anti-mouse CD4 and CD8 antibodies conjugated by PE and FITC, respectively (BD Biosciences, San Jose, CA). For staining NK cells, we used APC-conjugated rat anti-mouse CD49b/Pan-NK cells possessing monoclonal antibody that identies the majority of the NK cells. NK cells were dened as CD49b CD3 cells, and therefore PE-conjugated rat anti-mouse CD3 staining was used. Staining with peridinin chlorophyll- protein (Per-Cy5)-conjugated rat anti-mouse CD45 (BD Biosciences) was used to identify lymphocytes. T regulatory (Treg) cells were dened as CD4 CD25 FoxP3 with FITC-conjugated rat anti-mouse CD25 and APC-conjugated rat anti-mouse FoxP3. For apoptosis measurements of freshly isolated HSC, propidium iodide (PI) staining of fragmented DNA (42) and phosphatidylserine staining by annexin V conjugated to FITC (65) (R&D Systems, Minneapolis, MN) were used according to the manufacturers instructions. HSC apoptosis was therefore dened as annexin-V but PI . FACS data were acquired with a FACSCalibur ow cytometer (BD Biosciences) set to capture all events. In vitro human study. Peripheral blood lymphocytes from hepatitis C virus (HCV)-cirrhotic patients versus normal controls were cocultured 48 h with human HSC (LX2 cells; Ref. 61). Before coculture with LX2 cells, HCV-derived lymphocytes were preincubated with 1 g/ml GA or medium and washed. Triplets from eight patients were cultured under each condition. After 48 h of incubation, HSC were washed and lymphocyte-free HSC were harvested with a scraper.
AJP-Gastrointest Liver Physiol VOL

Protein lysate of the obtained HSC was assessed by Western blotting of -SMA and -actin. Cytokine measurements. To measure serum IL-4, interferon (IFN)- , and IL-10 levels, OptEIA ELISA kits (Pharmingen, San Diego, CA) were used according to the manufacturers protocol. A standard curve was generated with recombinant cytokines, and concentrations of samples were determined by using a polynomial curve t analysis. Statistical analysis. Lymphocyte subsets, serum aminotransferase levels, and the brosis area of each of the groups were analyzed for signicance with ANOVA and Students t-test. Correlations were analyzed with Pearsons pairwise correlation. Data were analyzed with JMP statistical software version 5.0 (SAS Institute, Cary, NC). The results are presented as means (SD); SE was used in the case of Bioquant analysis because each group includes 360 readings.
RESULTS

GA decreases CCl4-induced hepatic brosis. All animal groups survived 6 wk of CCl4 treatment and were analyzed for the extent of brosis by morphometry. GA treatment in the nal 2 wk of CCl4 mimics a real antibrotic effect since brosis is advanced and well established at the end of the fourth week (54). GA treatment signicantly attenuated hepatic brosis, since brotic septa were more established in CCl4treated mice that did not receive GA compared with those treated with GA based on Bioquant morphometry (Fig. 1A). More specically, hepatic brosis decreased from 5.28 0.32% (mean SE) and 2.85 0.156% in the 6- and 4-wk CCl4-treated groups, respectively, to 2.01 0.28% in the CCl4 plus GA group, compared with 0.38 0.07% in naive mice (Fig. 1B). GA treatment in the 6-wk CCl4-treated animals therefore signicantly ameliorates hepatic brosis compared with GA untreated groups (4 and 6 wk), suggesting a real reverse effect on hepatic brosis. To determine whether reduced brosis in mice treated with GA was associated with decreased stellate cell activation, we evaluated the expression of -SMA on liver protein extracts by immunoblotting. Indeed, the relative extent of brosis correlated closely with -SMA expression in all groups. As shown in Fig. 1C, reduced brosis in the GA-treated mice was associated with diminished expression of -SMA, indicating decreased stellate cell activation in the tissue. Furthermore, -SMA mRNA was also determined in livers with quantitative real-time PCR. -SMA mRNA was increased in the livers of brotic animals not treated with GA to 14.4 (SD 3.8)-fold at 6 wk and to 5.2 (SD 2)-fold at 4 wk (P 0.03). GA treatment, however, signicantly decreased -SMA mRNA expression to 1.5 (SD 1)-fold (Fig. 1D) compared with non-GA-treated groups (P 0.0005 and 0.02, respectively). CYP2E1 activity (Fig. 1E) presented as p-nitrophenol was signicantly lower in the 6-wk CCl4-treated mice [389.7 (SD 214) pmol min 1 mg protein 1] compared with the control mice [744.2 (SD 83) pmol min 1 mg protein 1; P 0.05] because CCl4 is known to lower CYP2E1 levels via radical inactivation and lipid peroxidation (63). GA, however, did not affect CYP2E1 activity [367.5 (SD 3.7) pmol min 1 mg protein 1; P 0.01 vs. naive and P 0.5 vs. non-GA-treated brotic mice], indicating that GA does not alter reactive oxygen species (ROS) production by CCl4. GA does not affect necroinammatory liver injury. Serum AST and ALT levels (Fig. 2A) were signicantly elevated in both CCl4 groups [104.3 (SD 10.8) and 168 (SD 34.2), respectively, and 95 (SD 14.2) and 179 (SD 26), respectively]
292 FEBRUARY 2007

Downloaded from ajpgi.physiology.org on April 24, 2009

www.ajpgi.org

GLATIRAMER ACETATE AMELIORATES HEPATIC FIBROSIS

G631

Downloaded from ajpgi.physiology.org on April 24, 2009


Fig. 1. Tissue sections were stained with Sirius red as described in MATERIALS AND METHODS. A: representative tissue sections. After brosis induction by intraperitoneal carbon tetrachloride (CCl4) injections, brotic septa are less established in glatiramer acetate (GA)-treated animals (magnication 10). B: relative brosis area, expressed as % of total liver area, was assessed by analyzing 36 Sirius red-stained liver sections per animal. Each eld was acquired at 10 magnication and then analyzed with a computerized Bioquant morphometry system. The relative brosis area in the livers of GA-treated animals was lower than that of untreated mice at 4- and 6-wk (W) time points. P values refer to comparisons between indicated groups. C: whole liver protein lysates were extracted, and 30 g of total proteins was loaded per lane and analyzed for -smooth muscle actin (SMA) and -actin expression. -Actin expression was similar in all tested wells. Decreased -SMA expression was found in lysates from GA-treated mice compared with untreated mice receiving CCl4 (results were reproduced in 3 repeated experiments). D: -SMA mRNA expression in the liver by quantitative real time-PCR followed a similar pattern. E: cytochrome P-450 2E1 activity was measured by the rate of oxidation of p-nitrophenol (PNP) to p-nitrocatechol in 50 g of microsomal protein for 20 min at 37C. *P 0.05 for CCl4 vs. naive; **P 0.01 for GA CCl4 vs. naive; P not signicant for CCl4 vs. GA CCl4. The ndings are representative of at least 4 different experiments with the same number of animals (8 10) in each subgroup. AJP-Gastrointest Liver Physiol VOL
292 FEBRUARY 2007

www.ajpgi.org

G632

GLATIRAMER ACETATE AMELIORATES HEPATIC FIBROSIS

hepatic lymphocytes by FACS. As shown in Fig. 3, splenic CD4 T cells signicantly decreased in numbers in both CCl4treated groups [22.20% (SD 0.99) in naive animals, 13.30% (SD 5.17) in CCl4 alone, and 17.1% (SD 3.5) in CCl4 GAtreated brotic animals (Fig. 3A), respectively] (P 0.001). GA tends to increase CD4 content (P 0.08) compared with non-GA-treated brotic mice. Similarly, intrahepatic CD4 declined after induction with CCl4, from 29.76% (SD 2.7) to 14.07% (SD 2.8) and 16% (SD 0.91) (Fig. 3B), respectively (P 0.001). No signicant changes were observed in liver CD4 subsets between the two brotic groups. Both splenic and intrahepatic CD8 cells increased signicantly after brosis induction (P 0.001). Spleen CD8 T cells increased from 8.5% (SD 1.03) in naive animals to 14% (SD 2.7) and 16.2% (SD 2.5) in CCl4 and CCl4 plus GA-treated brotic animals, respectively. Liver CD8 T cells increased from 7.6% (SD 0.9) in naive animals to 21.7% (SD 2.4) and 16.4% (SD 1.7) in CCl4 and CCl4 plus GA-treated brotic animals, respectively. Compared with nontreated brotic mice, GA treatment significantly decreased the number of liver CD8 cells (P 0.001). In addition to characterization of the CD4 and CD8 subsets, NK cells were also analyzed since they have potential antibrotic activity. Spleen NK cells decreased from 7.8% (SD 0.6) in naive animals to 3.9% (SD 1.6) in CCl4-treated mice not

Downloaded from ajpgi.physiology.org on April 24, 2009

Fig. 2. GA did not affect necroinammatory liver injury. A: serum aspartate (AST) and alanine (ALT) aminotransferase levels. B: mean necroinammatory scorings of periportal or periseptal interface hepatitis (A); conuent necrosis (B); and focal lytic necrosis and apoptosis and focal inammation (C). After brosis induction as described in Fig. 1, ALT and AST serum levels and scorings signicantly rose in both groups. A repeated experiment showed the same pattern.

compared with naive mice [45 (SD 25) and 36 (SD 12)] (P 0.001) (CCl4 alone and CCl4 with GA). However, there were no appreciable differences between GA-treated and -untreated CCl4 groups (P 0.26 and P 0.29 for AST and ALT, respectively). The serum biochemistries were correlated with semiquantitative pathological scoring. The extent of periportal or periseptal interface hepatitis; conuent necrosis; and focal lytic necrosis, apoptosis, and focal inammation were 0 (SD 0), 0.2 (SD 0.44), and 0.4 (SD 0.55) in naive animals, respectively (Fig. 2B). Necroinammatory scoring signicantly increased 0.0001): 1 after brosis induction in both CCl4 groups (P (SD 0), 2.67 (SD 0.82), and 1.17 (SD 0.98) in the untreated brotic group and 1.57 (SD 1.97), 3 (SD 0), and 1.57 (SD 0.97) in GA-treated mice, respectively. There was no signicant difference between the two CCl4 groups, with or without GA (P 0.189). Taken together, the lack of any difference in either serum biochemistries or histological scoring in both CCl4-treated groups, with or without GA, suggests that GAs antibrotic effect is not mediated through attenuation of liver injury due to CCl4. Antibrotic effect of GA is associated with lymphocyte and cytokine modulation. To gain insight into the potential mechanism of GAs antibrotic activity, we analyzed splenic and
AJP-Gastrointest Liver Physiol VOL

Fig. 3. A: altered splenocyte subsets. Compared with naive animals, uorescence-activated cell sorting analysis of spleen lymphocytes showed a signicant decrease in CD4 T cells and an increase in CD8 subsets after brosis induction, both in GA-treated and -nontreated mice (ANOVA, P 0.001). A signicant increase in natural killer (NK) cells was seen in the GA-treated group (P 0.009). B: altered intrahepatic subsets. Similar to the spleen ndings, there was a signicant decrease in liver CD4 T cells and an increase in CD8 subsets after brosis induction (ANOVA, P 0.001). The GA-treated group had signicantly fewer CD8 cells and higher NK counts compared with the nontreated group (P 0.001 and P 0.0007, respectively).
292 FEBRUARY 2007

www.ajpgi.org

GLATIRAMER ACETATE AMELIORATES HEPATIC FIBROSIS

G633

Fig. 4. Regulatory cells. In the absence of signicant T regulatory (Treg) cell spleen alterations (A), only increased liver CD4 CD25 FoxP3 T cells correlated well with the GA antibrotic effect (B). Bars represent the same groups detailed in Fig. 3.

receiving GA (P 0.0001). However, they increased to 7.5% (SD 2.8) in the CCl4 plus GA-treated group (P 0.4 vs. naive but P 0.009 vs. nontreated brotic). Liver NK cells decreased from 9.3% (SD 1) in naive mice to 5.8% (SD 0.9) in GA-untreated brotic mice (P 0.0001) and increased to 9.9% (SD 2.1) in the GA-treated brotic group (P 0.3 vs. naive but P 0.0007 vs. nontreated brotic group). Compared with brotic mice, GA treatment signicantly increased the number of spleen and liver NK cells. According to current knowledge, populations of suppressor or Treg cells constitute a pivotal mechanism in regulating immune responses. Treg cells have been implicated in a broad array of immunologic and nonimmunologic diseases (34, 55, 57). Compelling evidence suggests that a delicate equilibrium of controlled self-responsiveness is maintained by a unique population of CD4 CD25 Treg. The result of this process is the survival of autoreactive lymphocytes representing a variety of high-afnity self-reactive T-cell receptors (28). Mounting evidence indicates that Treg cells participate in the regulation of immune responses to microbial infections by contributing to setting the equilibrium between memory and pathogen elimination (8). Treg cells can be isolated from T-cell inltrates in ongoing autoimmune diseases (12), and responses to allergens and tumor antigens appear to be controlled by an interplay between Treg and effector T cells (30, 51, 60). CD4 CD25 FoxP3 Treg cells from tumor-bearing animals impede dendritic cell function by downregulating the activation of the transcription factor NF- B. The suppression mechanism requires TGF- and IL-10 (35). In examining whether the regulatory cell (CD4 CD25 FoxP3 ) phenotype was associated with GA antibrotic effect, we observed no signicant alterations in splenocytes from two brotic groups (Fig. 4A). In the liver, Treg cells signicantly decreased in the CCl4 group (P 0.02) compared with the naive group (Fig. 4B). This decrease of Treg cells may account for the increased content of brogenic CD8 cells. The inability of Treg cells to regulate the inhibition of CD8 T-cell function was reported to contribute to the initiation of autoimmune liver damage (36). However, only liver CD4 CD25 FoxP3 T cells correlated well with an antibrotic effect of GA, because they signicantly increased
AJP-Gastrointest Liver Physiol VOL

(Fig. 4B). Liver CD4 CD25 FoxP3 T cells were 18.5% (SD 2.2) of CD45 cells in the naive group and signicantly decreased to 14.6% (SD 2.5) (P 0.02) in GA-untreated and increased to 23.2% (SD 2.8) (P 0.007) in GA-treated brotic groups, respectively. Compared with GA-nontreated brotic animals, GA treatment signicantly increased Treg cells (P 0.0006). An increase in the suppressant Treg cells after GA treatment may explain the direct effect of the Treg cells regarding the suppression of CD8 cells and subsequently the attenuation of hepatic brosis. Since cytokines can either stimulate or inhibit brosis (21, 38, 48, 62, 68), we rst measured the serum concentrations of the antibrotic cytokine IFN- . As shown in Fig. 5, IFNconcentrations in the naive animals were 6 (SD 4) pg/ml and increased to 35.9 (SD 20.7) pg/ml in CCl4-treated mice not receiving GA (P 0.001) and 34.8 (SD 23.6) pg/ml in CCl4 plus GA-treated brotic animals (P 0.004). There was no effect of GA on IFN- levels in animals treated with CCl4. We next assayed the levels of the probrotic cytokine IL-4. Fibrosis induction led to a signicant elevation of serum IL-4, from 31.3 (SD 26) to 100 (SD 44) pg/ml (P 0.0005). Importantly, GA treatment markedly reduced IL-4 levels after induction of brosis by CCl4 to 55.7 (SD 39.7) pg/ml (P 0.03), suggesting that the antibrotic effect of GA was partly a result of reduced levels of this probrotic cytokine (Fig. 5). The impact of GA was also assessed on serum levels of IL-10, which, in contrast to IL-4, is an antibrotic cytokine (41, 54). Serum IL-10 concentrations were 31 (SD 41.4) pg/ml in naive mice and 34.2 (SD 16.6) pg/ml after the induction of brosis by CCl4 (Fig. 5). GA treatment, however, signicantly elevated the IL-10 level in CCl4-treated mice to 63.1 (SD 25.1) pg/ml (P 0.02). Thus an elevation in the IL-10 level in GA-treated mice may also contribute to its antibrotic activity. HSC apoptosis. HSC apoptosis in freshly isolated cells (Fig. 6A) was signicantly increased from 37.2% (SD 6.4) in naive animals to 49% (SD 1) in brotic animals (P 0.005). GA treatment augmented HSC apoptosis to 53.3% (SD 5.3) (P

Downloaded from ajpgi.physiology.org on April 24, 2009

Fig. 5. Serum cytokine levels reect an antibrotic pattern following GA treatment. Serum interferon (IFN)- , interleukin (IL)-10, and IL-4 levels were determined by ELISA in all animals and are expressed as pg/ml in naive and brotic mice. Compared with naive animals, IFN- concentrations signicantly increased in GA-nontreated and GA-treated brotic animals. No significant changes were seen between brotic groups. Fibrosis induction leads to a signicant increase of IL-4 serum concentrations. GA treatment, however, signicantly reduced their levels. Serum IL-10 concentrations were similar in naive mice and after induction of brosis. GA treatment, however, signicantly increased them. Bars represent the same groups detailed in Fig. 3. Results were regenerated twice, showing the same pattern.
292 FEBRUARY 2007

www.ajpgi.org

G634

GLATIRAMER ACETATE AMELIORATES HEPATIC FIBROSIS

Downloaded from ajpgi.physiology.org on April 24, 2009


Fig. 6. Effect of GA on hepatic stellate cell (HSC) apoptosis. A: fresh HSC isolated from all livers were evaluated for apoptosis with uorescence-activated cell sorting. HSC apoptosis was dened as annexin V but propidium iodide , as described in MATERIALS AND METHODS. HSC apoptosis was signicantly increased in brotic animals compared with naive mice (P 0.005) and further increased after GA treatment (P 0.01 between CCl4 groups). B: double-stained -SMA (red arrow) and annexin V (yellow arrow) immunouorescence staining dened as green (FITC) cells with red periphery revealed increased HSC apoptosis after brosis (left) and failure to stain in naive animals (data not shown). C and D: hepatic tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) expression also increased after GA treatment as detected by real-time PCR and Western blotting.

0.01 vs. GA-untreated group and P 0.0004 vs. naive). Although cleavage of receptors from enzymes used in cell isolation is possible, this effect should have affected all isolated cells equally, yet differences remained between the different groups. In situ apoptosis of tissue HSC by double
AJP-Gastrointest Liver Physiol VOL

staining of -SMA and annexin V conrmed the same ndings. The number of double-stained annexin V -SMA cells (Fig. 6B, left) was markedly increased after brosis induction from 3.8 (SD 2.4) to 8 (SD 3.6) 102 cells/eld (P 0.02), and GA further increased it to 13 (SD 4.3) 102 cells/eld
292 FEBRUARY 2007

www.ajpgi.org

GLATIRAMER ACETATE AMELIORATES HEPATIC FIBROSIS

G635

(P 0.0003 vs. naive and 0.005 vs. non-GA treated brotic animals; Fig. 6B, right). Besides increased apoptosis of HSC, hepatic expression of TRAIL was also increased. TRAIL mRNA expression signicantly increased (P 0.0004) from 1.3 (SD 0.1)-fold in the GA-nontreated brotic group to 2.9 (SD 1.13)-fold in the GA-treated brotic animals (Fig. 6C). TRAIL expression by Western blotting from liver protein extracts showed a similar pattern (Fig. 6D). Effect of GA on activation of human HSC by HCV lymphocytes. To further elucidate the relationship between GA, lymphocytes, and liver brosis and its relevance to human disease, we examined the effect of GA on the capacity of lymphocytes from patients with HCV-associated cirrhosis to activate human HSC. To isolate the GA effect on lymphocytes, we preincubated HCV peripheral blood lymphocytes and HSC with GA or medium for 24 h, after which we washed them before coculture with LX2 cells. HSC activation was then determined by the expression of -SMA. As a control, naive lymphocytes from noninfected healthy controls were also incubated with HSC. As shown in Fig. 7, incubation of lymphocytes from HCV patients increased the activation of HSC as the -SMA expression increased (Fig. 7, center), compared with healthy lymphocytes (Fig. 7, left). Preincubation of HCV lymphocytes with GA, however, resulted in signicant downregulation of HSC activation (Fig. 7, right).
DISCUSSION

GA (Copaxone) is a synthetic copolymer composed of a random mixture of four amino acids that modify the immune response, consequently resulting in central nervous system inammation, demyelination, and axonal loss characteristic of relapsing-remitting multiple sclerosis (RRMS). Different clinical trials have shown GA to have an appreciable effect on the clinical course of RRMS (11, 14, 31, 32, 37, 53, 59). In a prospective, nonrandomized, open-label treatment trial, Khan (31, 32) demonstrated a signicant reduction in the annual relapse rate of GA-treated RRMS at 12- and 18-mo follow-up compared with no treatment. GA was not found to produce an inuenza-like syndrome or neutralizing antibodies similar to those reported in patients treated with IFN- for RRMS. The most commonly reported treatment-related adverse events were localized injection site reactions and transient postinjection systemic reactions, both of which were generally mild and self-limited. On the basis of the available data and current

Fig. 7. Effect of GA on the activation of human HSC by HCV lymphocytes. HCV peripheral blood lymphocytes were preincubated with either GA or medium, and HSC were incubated alone or with GA for 48 h. As a control, naive lymphocytes were used. HSC activation was then determined by the expression of -SMA. Preincubation of lymphocytes from hepatitis C virus (HCV) patients with GA resulted in a marked alleviation of HSC activation (top). -Actin expression was similar under all conditions (bottom). Although the graph shows only 3 patients from each group, all other patients followed the same pattern (data not shown). AJP-Gastrointest Liver Physiol VOL

management guidelines, GA is a valuable rst-line treatment option for patients with RRMS (46, 62). The benecial effect of GA has also been identied in other immune-mediated diseases. For example, GA inhibits type 2 collagen-reactive T-cell clones in animal models of rheumatoid arthritis (16). Furthermore, GA can also inhibit the manifestations of host versus graft rejection (3). In experimental autoimmune uveoretinitis, GA has inhibitory suppressive activity, but in contrast to the models previously described the treated mice failed to produce cytokines such as IL-4, IL-5, and IL-10 (73). The ability of GA to effectively modulate the clinical manifestations and to attenuate the detrimental immune response involved in experimental colitis warrants its further investigation in treating this disease (1). The effect of GA on brosis has not previously been examined. In graft versus host disease, treatment with GA abolished cytotoxic activity toward host targets by induction of the anti-inammatory response and suppression of inammatory cytokine secretion (IL-2 and INF- ) (2). Moreover, GA decreased hepatic brosis even less than that seen at week 4 of CCl4 induction, suggesting a real antibrotic activity. On the other hand, GA did not alter the CYP2E1 activity between GA treated and GA-nontreated brotic mice. GA, therefore, is not affecting the ROS production by the CCl4. Our data reveal that GA exerts an antibrotic effect in the liver, by affecting lymphocyte subsets and also by altering the balance between pro- and antibrotic cytokines. Notably, there was no signicant difference in the inammatory markers (AST, ALT, or Ishak-Knodell score) between the CCl4 and the CCl4 plus GA mice, thus indicating the direct effect of GA on brosis apart from its effect on the degree of liver injury. An additional explanation for GAs lack of an effect on inammation could be that GA blocks lymphocyte-HSC cell-to-cell interaction. In experimental models, brosis induction is associated with an increase in CD8 subsets and with a decrease in CD4 T cells (23, 54). In mouse models, adoptive transfer of CD8 cells from brotic donors to severe combined immunodecient (SCID) mice induces hepatic brosis. In contrast, NK cells are antibrotic (19), as evidenced by their mediating apoptosis of activated stellate cells. We have investigated the role of NK cells on hepatic brogenesis (19). Mouse NK cells express both inhibitory/activating-killing immunoglobulin-related receptors (iKIR/aKIR) specic for class I molecules. Hepatic brosis induced by CCl4 was compared among wild-type male BALBc mice with combined immunodeciency (SCID, lacking B and T cells), SCID-Beige mice (lacking B, T, and NK cells), and naive mice. Hepatic brosis was signicantly increased in all CCl4-treated groups. SCID-Beige mice had more brosis than SCID mice (P 0.0001), as assessed by morphometry of Sirius red-stained tissue sections. After brosis, hepatic NK cells signicantly decreased and the aKIR-to-iKIR ratio signicantly increased, whereas class I receptor expression on HSC decreased (P 0.001). Both freshly isolated and in situ HSC displayed a signicant increase in cellular apoptosis following induction of brosis. In human HSC there was decreased class I receptor expression and increased apoptosis as well, which further increased after blocking of either HSCrelated class I or NK-related killer inhibitory receptors. Apoptosis was inhibited by preincubation of NK cells with the granzyme inhibitor 3,4-dichloroisocoumarin. In conclusion, during liver injury NK cells exhibit antibrotic activity at least
292 FEBRUARY 2007

Downloaded from ajpgi.physiology.org on April 24, 2009

www.ajpgi.org

G636

GLATIRAMER ACETATE AMELIORATES HEPATIC FIBROSIS

in part through stimulation of HSC killing. NK cells kill activated HSC via retinoic acid early inducible 1/NKG2Ddependent and TRAIL-dependent mechanisms, thereby ameliorating liver brosis (46). In this study, GA treatment reduced CD8 in the brotic liver of our animal model, with no signicant effect on the decreased total CD4 subset. The increase of brotic CD8 cells following CCl4 administration may be explained by the marked decrease of the CD4 cells, either total CD4 or CD4 CD25 FoxP3 Treg cells. Hepatic Treg cells, however, signicantly increased after GA treatment, probably resulting in a new reconstitution of the Treg suppressive effect on CD8 cells. CD8 cells therefore decreased in GA-treated livers, which might explain the alleviation of HSC activation and the reduction of brosis. Furthermore, GA increased NK numbers in the liver and spleen after CCl4. In our model, increased NK cells add an additional option for the immunomodulatory effect of GA in brosis reduction. Increased HSC apoptosis and TRAIL support the increased killing of HSC by NK cells after GA treatment (40, 46). The mode of action of the drug in humans with MS is believed to involve the induction of glatiramer-reactive regulatory cells, including CD4 and CD8 T cells. Glatiramer-reactive Th2 cells are believed to enter the brain and, through cross-reactivity with myelin antigens, produce bystander suppression, anti-inammatory effects, and neuroprotection (13, 15). Unlike in our study, GA may suppress autoreactive CD4 effector memory T cells and enhance CD8 regulatory responses in patients with RRMS (6, 29). Therefore, Treg cells may be considered to be of major importance in the pathogenesis of MS (56, 66). Diverse types of Treg cells, with distinct origins and with disparate and multiple functions, have been described. CD4 CD25 cells were reported to be involved in the active suppression exerted by the dual altered peptide ligand on myasthenogenic-associated T-cell responses (43). Another study suggests that levels of circulating CD4 CD25 Treg cells and CD4 CD25(high) Treg cells are not altered in MS patients and are unaffected by substances currently used to modulate the disease, including IFN- and GA (44). In addition to its effects on lymphocyte subsets, GA also affected the balance of pro- and antibrotic cytokines. Specifically, serum IL-10 levels rose, whereas IL-4 levels fell, which were associated with a GA antibrotic effect. There is a growing body of evidence in other models that GA promotes Th2 cell development and increased IL-10 production through modulation of dendritic cells (27, 66). Heat shock protein 60-stimulated Treg cells were reported to suppress target T cells both by cell-to-cell contact and by secretion of TGF- and IL-10 (71). Therefore, CD4 CD25 FoxP3 cells increased by GA might contribute to the enhanced serum IL-10 levels observed in our study. Moreover, GA induces a selective inhibitory effect on the production of dendritic cell-derived inammatory mediators. After exposure to GA, dendritic cells have an impaired capacity to secrete Th1 polarizing factors, but on the other hand, they induce the effectors Th2 cells (50) to secrete IL-4 and to enhance the levels of IL-10 (27). In addition, GA contributes to apoptotic elimination of T helper cells (2, 4). IL-10 is an antibrotic cytokine. It downregulates brogenesis in cultured HSC by decreasing collagen synthesis and increasing the production of collagenase (68). Furthermore,
AJP-Gastrointest Liver Physiol VOL

exogenous IL-10 downregulates the expression of metalloproteinase-2 and metalloproteinase-1, both of which are increased after induction of brosis (72). The present ndings are consistent with our earlier ndings (54) in IL-10 transgenic mice, which have reduced brosis after exposure to CCl4, associated with suppression of CD8 subsets and IL-4 secretion. The direct effect of GA on lymphocytes can explain its antibrotic effect, as shown in the in vitro study (Fig. 6). GA-affected HCV lymphocytes lost their potential to activate HSC. Importantly, those HSC were not directly exposed to the GA itself. In summary, GA exerts antibrotic activity in CCl4-induced brosis through altered numbers of CD8 and NK subsets. These ndings reinforce the growing understanding that subtle alterations in the intrahepatic immunologic milieu can appreciably alter the brotic balance in liver injury. Given its very strong safety prole, GA may also merit further evaluation as an antibrotic agent for patients with chronic brosing liver injury.
Downloaded from ajpgi.physiology.org on April 24, 2009
ACKNOWLEDGMENTS Special thanks to Dr. Dimitrios Losos, department of neurology at Hadassah medical center, for providing GA for this experiment. GRANTS This work was supported by grants from Return to Hadassah, Compensatory, and Horovetz Awards (Grant Nos. 8033601, 8033603, 8033604). REFERENCES 1. Aharoni R, Kayhan B, Arnon R. Therapeutic effect of the immunomodulator glatiramer acetate on trinitrobenzene sulfonic acid-induced experimental colitis. Inamm Bowel Dis 11: 106 115, 2005. 2. Aharoni R, Schlegel PG, Teitelbaum D, Roikhel-Karpov O, Chen Y, Arnon R, Sela M, Chao NJ. Studies on the mechanism and specicity of the effect of the synthetic random copolymer GLAT on graft-versus-host disease. Immunol Lett 58: 79 87, 1997. 3. Aharoni R, Teitelbaum D, Arnon R, Sela M. Copolymer 1 inhibits manifestations of graft rejection. Transplantation 72: 598 605, 2001. 4. Aktas O, Ari N, Rieks M, Hoffmann V, Schimrigk S, Przuntek H, Pohlau D. Multiple sclerosis: modulation of apoptosis susceptibility by glatiramer acetate. Acta Neurol Scand 104: 266 270, 2001. 5. Aktas O, Smorodchenko A, Brocke S, Infante-Duarte C, Topphoff US, Vogt J, Prozorovski T, Meier S, Osmanova V, Pohl E, Bechmann I, Nitsch R, Zipp F. Neuronal damage in autoimmune neuroinammation mediated by the death ligand TRAIL. Neuron 46: 421 432, 2005. 6. Allie R, Hu L, Mullen KM, Dhib-Jalbut S, Calabresi PA. Bystander modulation of chemokine receptor expression on peripheral blood T lymphocytes mediated by glatiramer therapy. Arch Neurol 62: 889 894, 2005. 7. Arnon R, Sela M. Immunomodulation by the copolymer glatiramer acetate. J Mol Recognit 16: 412 421, 2003. 8. Belkaid Y, Rouse BT. Natural regulatory T cells in infectious disease. Nat Immun 6: 353360, 2005. 9. Boyum A. Isolation of mononuclear cells and granulocytes from human blood. Isolation of mononuclear cells by one centrifugation, and of granulocytes by combining centrifugation and sedimentation at 1 g. Scand J Clin Lab Invest Suppl 97: 77 89, 1968. 10. Chou WY, Lu CN, Lee TH, Wu CL, Hung KS, Concejero AM, Jawan B, Wang CH. Electroporative interleukin-10 gene transfer ameliorates carbon tetrachloride-induced murine liver brosis by MMP and TIMP modulation. Acta Pharmacol Sin 27: 469 476, 2006. 11. Comi G, Filippi M, Wolinsky JS. European/Canadian multicenter, double-blind, randomized, placebo-controlled study of the effects of glatiramer acetate on magnetic resonance imagingmeasured disease activity and burden in patients with relapsing multiple sclerosis. European/Canadian Glatiramer Acetate Study Group. Ann Neurol 49: 290 297, 2001. 12. De Kleer IM, Wedderburn LR, Taams LS, Patel A, Varsani H, Klein M, de Jager W, Pugayung G, Giannoni F, Rijkers G, Albani S, Kuis
292 FEBRUARY 2007

www.ajpgi.org

GLATIRAMER ACETATE AMELIORATES HEPATIC FIBROSIS W, Prakken B. CD4 CD25bright regulatory T cells actively regulate inammation in the joints of patients with the remitting form of juvenile idiopathic arthritis. J Immunol 172: 6435 6443, 2004. Dhib-Jalbut S. Glatiramer acetate (Copaxone) therapy for multiple sclerosis. Pharmacol Ther 98: 245255, 2003. Filippi M, Rovaris M, Rocca MA, Sormani MP, Wolinsky JS, Comi G. Glatiramer acetate reduces the proportion of new MS lesions evolving into black holes. Neurology 57: 731733, 2001. Franciotta D, Zardini E, Bergamaschi R, Andreoni L, Cosi V. Interferon gamma and interleukin 4 producing T cells in peripheral blood of multiple sclerosis patients undergoing immunomodulatory treatment. J Neurol Neurosurg Psychiatry 74: 123126, 2003. Fridkis-Hareli M, Neveu JM, Robinson RA, Lane WS, Gauthier L, Wucherpfennig KW, Sela M, Strominger JL. Binding motifs of copolymer 1 to multiple sclerosis- and rheumatoid arthritis-associated HLA-DR molecules. J Immunol 162: 4697 4704, 1999. Friedman SL. Liver brosisfrom bench to bedside. J Hepatol 38, Suppl 1: S38 S53, 2003. Friedman SL. Molecular regulation of hepatic brosis, an integrated cellular response to tissue injury. J Biol Chem 275: 22472250, 2000. Friedman SL, Rockey DC, McGuire RF, Maher JJ, Boyles JK, Yamasaki G. Isolated hepatic lipocytes and Kupffer cells from normal human liver: morphological and functional characteristics in primary culture. Hepatology 15: 234 243, 1992. Geerts A, Eliasson C, Niki T, Wielant A, Vaeyens F, Pekny M. Formation of normal desmin intermediate laments in mouse hepatic stellate cells requires vimentin. Hepatology 33: 177188, 2001. Gressner AM, Weiskirchen R, Breitkopf K, Dooley S. Roles of TGFbeta in hepatic brosis. Front Biosci 7: d793 d807, 2002. Guler ML, Gorham JD, Hsieh CS, Mackey AJ, Steen RG, Dietrich WF, Murphy KM. Genetic susceptibility to Leishmania: IL-12 responsiveness in TH1 cell development. Science 271: 984 987, 1996. Heinzel FP, Sadick MD, Holaday BJ, Coffman RL, Locksley RM. Reciprocal expression of interferon gamma or interleukin 4 during the resolution or progression of murine leishmaniasis. Evidence for expansion of distinct helper T cell subsets. J Exp Med 169: 59 72, 1989. Hui AY, Liew CT, Go MY, Chim AM, Chan HL, Leung NW, Sung JJ. Quantitative assessment of brosis in liver biopsies from patients with chronic hepatitis B. Liver Int 24: 611 618, 2004. Hung DY, Chang P, Cheung K, McWhinney B, Masci PP, Weiss M, Roberts MS. Cationic drug pharmacokinetics in diseased livers determined by brosis index, hepatic protein content, microsomal activity, and nature of drug. J Pharmacol Exp Ther 301: 1079 1087, 2002. Jiang Y, Kang YJ. Metallothionein gene therapy for chemical-induced liver brosis in mice. Mol Ther 10: 1130 1139, 2004. Johnson KP, Brooks BR, Cohen JA, Ford CC, Goldstein J, Lisak RP, Myers LW, Panitch HS, Rose JW, Schiffer RB, Vollmer T, Weiner LP, Wolinsky JS. Extended use of glatiramer acetate (Copaxone) is well tolerated and maintains its clinical effect on multiple sclerosis relapse rate and degree of disability. Neurology 57: S46 S53, 2001. Jordan MS, Boesteanu A, Reed AJ, Petrone AL, Holenbeck AE, Lerman MA, Naji A, Caton AJ. Thymic selection of CD4 CD25 regulatory T cells induced by an agonist self-peptide. Nat Immun 2: 301306, 2001. Karandikar NJ, Crawford MP, Yan X, Ratts RB, Brenchley JM, Ambrozak DR, Lovett-Racke AE, Frohman EM, Stastny P, Douek DC, Koup RA, Racke MK. Glatiramer acetate (Copaxone) therapy induces CD8 T cell responses in patients with multiple sclerosis. J Clin Invest 109: 641 649, 2002. Karlsson MR, Rugtveit J, Brandtzaeg P. Allergen-responsive CD4 CD25 regulatory T cells in children who have outgrown cows milk allergy. J Exp Med 199: 1679 1688, 2004. Khan OA, Tselis AC, Kamholz JA, Garbern JY, Lewis RA, Lisak RP. A prospective, open-label treatment trial to compare the effect of IFNbeta-1a (Avonex), IFNbeta-1b (Betaseron), and glatiramer acetate (Copaxone) on the relapse rate in relapsing-remitting multiple sclerosis. Eur J Neurol 8: 141148, 2001. Khan OA, Tselis AC, Kamholz JA, Garbern JY, Lewis RA, Lisak RP. A prospective, open-label treatment trial to compare the effect of IFNbeta-1a (Avonex), IFNbeta-1b (Betaseron), and glatiramer acetate (Copaxone) on the relapse rate in relapsing-remitting multiple sclerosis: results after 18 months of therapy. Mult Scler 7: 349 353, 2001. Knodell RG, Ishak KG, Black WC, Chen TS, Craig R, Kaplowitz N, Kiernan TW, Wollman J. Formulation and application of a numerical AJP-Gastrointest Liver Physiol VOL

G637

34. 35.

13. 14. 15.

36.

16.

37.

17. 18. 19.

38.

39.

20. 21. 22. 23.

40.

41.

42.

24. 25.

43.

44.

26. 27.

45.

46.

28.

47.

29.

48. 49.

30. 31.

50.

51. 52.

32.

53.

33.

54.

scoring system for assessing histological activity in asymptomatic chronic active hepatitis. Hepatology 1: 431 435, 1981. Kronenberg M, Rudensky A. Regulation of immunity by self-reactive T cells. Nature 435: 598 604, 2005. Larmonier N, Marron M, Zeng Y, Cantrell J, Romanoski A, Sepassi M, Thompson S, Chen X, Andreansky S, Katsanis E. Tumor-derived CD4 CD25 regulatory T cell suppression of dendritic cell function involves TGF-beta and IL-10. Cancer Immunol Immunother 2006. Longhi MS, Ma Y, Mitry RR, Bogdanos DP, Heneghan M, Cheeseman P, Mieli-Vergani G, Vergani D. Effect of CD4 CD25 regulatory T-cells on CD8 T-cell function in patients with autoimmune hepatitis. J Autoimmun 25: 6371, 2005. Mancardi GL, Sardanelli F, Parodi RC, Melani E, Capello E, Inglese M, Ferrari A, Sormani MP, Ottonello C, Levrero F, Uccelli A, Bruzzi P. Effect of copolymer-1 on serial gadolinium-enhanced MRI in relapsing remitting multiple sclerosis. Neurology 50: 11271133, 1998. Mathurin P, Xiong S, Kharbanda KK, Veal N, Miyahara T, Motomura K, Rippe RA, Bachem MG, Tsukamoto H. IL-10 receptor and coreceptor expression in quiescent and activated hepatic stellate cells. Am J Physiol Gastrointest Liver Physiol 282: G981G990, 2002. Mehal WZ, Azzaroli F, Crispe IN. Antigen presentation by liver cells controls intrahepatic T cell trapping, whereas bone marrow-derived cells preferentially promote intrahepatic T cell apoptosis. J Immunol 167: 667 673, 2001. Melhem A, Muhanna N, Bishara A, Alvarez CE, Ilan Y, Bishara T, Horani A, Nassar M, Friedman SL, Safadi R. Anti-brotic activity of NK cells in experimental liver injury through killing of activated HSC. J Hepatol 45: 60 71, 2006. Nelson DR, Lauwers GY, Lau JY, Davis GL. Interleukin 10 treatment reduces brosis in patients with chronic hepatitis C: a pilot trial of interferon nonresponders. Gastroenterology 118: 655 660, 2000. Nicoletti I, Migliorati G, Pagliacci MC, Grignani F, Riccardi C. A rapid and simple method for measuring thymocyte apoptosis by propidium iodide staining and ow cytometry. J Immunol Methods 139: 271279, 1991. Paas-Rozner M, Sela M, Mozes E. A dual altered peptide ligand down-regulates myasthenogenic T cell responses by up-regulating CD25and CTLA-4-expressing CD4 T cells. Proc Natl Acad Sci USA 100: 6676 6681, 2003. Putheti P, Pettersson A, Soderstrom M, Link H, Huang YM. Circulating CD4 CD25 T regulatory cells are not altered in multiple sclerosis and unaffected by disease-modulating drugs. J Clin Immunol 24: 155161, 2004. Putheti P, Soderstrom M, Link H, Huang YM. Effect of glatiramer acetate (Copaxone) on CD4 CD25high T regulatory cells and their IL-10 production in multiple sclerosis. J Neuroimmunol 144: 125131, 2003. Radaeva S, Sun R, Jaruga B, Nguyen VT, Tian Z, Gao B. Natural killer cells ameliorate liver brosis by killing activated stellate cells in NKG2Ddependent and tumor necrosis factor-related apoptosis-inducing liganddependent manners. Gastroenterology 130: 435 452, 2006. Ramadori G, Saile B. Inammation, damage repair, immune cells, and liver brosis: specic or nonspecic, this is the question. Gastroenterology 127: 9971000, 2004. Reeves HL, Friedman SL. Activation of hepatic stellate cellsa key issue in liver brosis. Front Biosci 7: d808 d826, 2002. Reinke LA, Moyer MJ. p-Nitrophenol hydroxylation. A microsomal oxidation which is highly inducible by ethanol. Drug Metab Dispos 13: 548 552, 1985. Rieks M, Hoffmann V, Aktas O, Juschka M, Spitzer I, Brune N, Schimrigk S, Przuntek H, Pohlau D. Induction of apoptosis of CD4 T cells by immunomodulatory therapy of multiple sclerosis with glatiramer acetate. Eur Neurol 50: 200 206, 2003. Robinson DS. The role of regulatory T lymphocytes in asthma pathogenesis. Curr Allergy Asthma Rep 5: 136 141, 2005. Rockey DC, Boyles JK, Gabbiani G, Friedman SL. Rat hepatic lipocytes express smooth muscle actin upon activation in vivo and in culture. J Submicrosc Cytol Pathol 24: 193203, 1992. Rovaris M, Codella M, Moiola L, Ghezzi A, Zaffaroni M, Mancardi G, Capello E, Sardanelli F, Comi G, Filippi M. Effect of glatiramer acetate on MS lesions enhancing at different gadolinium doses. Neurology 59: 1429 1432, 2002. Safadi R, Ohta M, Alvarez CE, Fiel MI, Bansal M, Mehal WZ, Friedman SL. Immune stimulation of hepatic brogenesis by CD8 cells www.ajpgi.org

Downloaded from ajpgi.physiology.org on April 24, 2009

292 FEBRUARY 2007

G638

GLATIRAMER ACETATE AMELIORATES HEPATIC FIBROSIS response: new insight into the role of tissue inhibitors of matrix metalloproteinases. J Immunol 167: 70177026, 2001. Vermes I, Haanen C, Steffens-Nakken H, Reutelingsperger C. A novel assay for apoptosis. Flow cytometric detection of phosphatidylserine expression on early apoptotic cells using uorescein labelled Annexin V. J Immunol Methods 184: 39 51, 1995. Vieira PL, Heystek HC, Wormmeester J, Wierenga EA, Kapsenberg ML. Glatiramer acetate (copolymer-1, copaxone) promotes Th2 cell development and increased IL-10 production through modulation of dendritic cells. J Immunol 170: 4483 4488, 2003. Vogten JM, Drixler TA, te Velde EA, Schipper ME, van Vroonhoven TJ, Voest EE, Borel Rinkes IH. Angiostatin inhibits experimental liver brosis in mice. Int J Colorectal Dis 19: 387394, 2004. Wang SC, Ohata M, Schrum L, Rippe RA, Tsukamoto H. Expression of interleukin-10 by in vitro and in vivo activated hepatic stellate cells. J Biol Chem 273: 302308, 1998. Wang ZE, Reiner SL, Zheng S, Dalton DK, Locksley RM. CD4 effector cells default to the Th2 pathway in interferon gamma-decient mice infected with Leishmania major. J Exp Med 179: 13671371, 1994. Wolinsky JS. Glatiramer acetate for the treatment of multiple sclerosis. Expert Opin Pharmacother 5: 875 891, 2004. Zanin-Zhorov A, Cahalon L, Tal G, Margalit R, Lider O, Cohen IR. Heat shock protein 60 enhances CD4 CD25 regulatory T cell function via innate TLR2 signaling. J Clin Invest 116: 20222032, 2006. Zhang LJ, Chen YX, Chen ZX, Huang YH, Yu JP, Wang XZ. Effect of interleukin-10 and platelet-derived growth factor on expressions of matrix metalloproteinases-2 and tissue inhibitor of metalloproteinases-1 in rat brotic liver and cultured hepatic stellate cells. World J Gastroenterol 10: 2574 2579, 2004. Zhang M, Chan CC, Vistica B, Hung V, Wiggert B, Gery I. Copolymer 1 inhibits experimental autoimmune uveoretinitis. J Neuroimmunol 103: 189 194, 2000.

55. 56.

57. 58. 59. 60.

61. 62. 63. 64.

and attenuation by transgenic interleukin-10 from hepatocytes. Gastroenterology 127: 870 882, 2004. Sakaguchi S. Naturally arising Foxp3-expressing CD25 CD4 regulatory T cells in immunological tolerance to self and non-self. Nat Immun 6: 345352, 2005. Schmied M, Duda PW, Krieger JI, Trollmo C, Haer DA. In vitro evidence that subcutaneous administration of glatiramer acetate induces hyporesponsive T cells in patients with multiple sclerosis. Clin Immunol 106: 163174, 2003. Shevach EM. CD4 CD25 suppressor T cells: more questions than answers. Nat Rev Immunol 2: 389 400, 2002. Shi Z, Wakil AE, Rockey DC. Strain-specic differences in mouse hepatic wound healing are mediated by divergent T helper cytokine responses. Proc Natl Acad Sci USA 94: 1066310668, 1997. Simpson D, Noble S, Perry C. Glatiramer acetate: a review of its use in relapsing-remitting multiple sclerosis. CNS Drugs 16: 825 850, 2002. Sutmuller RP, van Duivenvoorde LM, van Elsas A, Schumacher TN, Wildenberg ME, Allison JP, Toes RE, Offringa R, Melief CJ. Synergism of cytotoxic T lymphocyte-associated antigen 4 blockade and depletion of CD25 regulatory T cells in antitumor therapy reveals alternative pathways for suppression of autoreactive cytotoxic T lymphocyte responses. J Exp Med 194: 823 832, 2001. Taimr P, Higuchi H, Kocova E, Rippe RA, Friedman S, Gores GJ. Activated stellate cells express the TRAIL receptor-2/death receptor-5 and undergo TRAIL-mediated apoptosis. Hepatology 37: 8795, 2003. Thompson K, Maltby J, Falloweld J, McAulay M, Millward-Sadler H, Sheron N. Interleukin-10 expression and function in experimental murine liver inammation and brosis. Hepatology 28: 15971606, 1998. Tierney DJ, Haas AL, Koop DR. Degradation of cytochrome P450 2E1: selective loss after labilization of the enzyme. Arch Biochem Biophys 293: 9 16, 1992. Vaillant B, Chiaramonte MG, Cheever AW, Soloway PD, Wynn TA. Regulation of hepatic brosis and extracellular matrix genes by the Th

65.

66.

67.

68.

69.

70. 71.

Downloaded from ajpgi.physiology.org on April 24, 2009

72.

73.

AJP-Gastrointest Liver Physiol VOL

292 FEBRUARY 2007

www.ajpgi.org

Você também pode gostar