Skip to main content
Erschienen in: Comparative Hepatology 1/2005

Open Access 01.12.2005 | Research

Regenerative and fibrotic pathways in canine hepatic portosystemic shunt and portal vein hypoplasia, new models for clinical hepatocyte growth factor treatment

verfasst von: Bart Spee, Louis C Penning, Ted SGAM van den Ingh, Brigitte Arends, Jooske IJzer, Frederik J van Sluijs, Jan Rothuizen

Erschienen in: Comparative Hepatology | Ausgabe 1/2005

Abstract

Background

We analyzed two spontaneous dog diseases characterized by subnormal portal perfusion and reduced liver growth: (i) congenital portosystemic shunts (CPSS) without fibrosis and (ii) primary portal vein hypoplasia (PPVH), a disease associated with fibrosis. These pathologies, that lack inflammation or cholestasis, may represent simplified models to study liver growth and fibrosis. To investigate the possible use of those models for hepatocyte growth factor (HGF) treatment, we studied the functionality of HGF signaling in CPSS and PPVH dogs and compared this to aged-matched healthy controls.

Results

We used quantitative real-time polymerase chain reaction (Q-PCR) to analyze the mRNA expression of HGF, transforming growth factor β1 (TGF-β1), and relevant mediators in liver biopsies from cases with CPSS or PPVH, in comparison with healthy control dogs. CPSS and PPVH were associated with a decrease in mRNA expression of HGF and of MET proto-oncogene (c-MET). Western blot analysis confirmed the Q-PCR results and showed that intracellular signaling components (protein kinase B/Akt, ERK1/2, and STAT3) were functional. The TGF-β1 mRNA levels were unchanged in CPSS whereas there was a 2-fold increase in PPVH indicating an active TGF-β1 pathway, consistent with the observation of fibrosis seen in PPVH. Western blots on TGF-β1 and phosphorylated Smad2 confirmed an activated pro-fibrotic pathway in PPVH. Furthermore, Q-PCR showed an increase in the amount of collagen I present in PPVH compared to CPSS and control, which was confirmed by Western blot analysis.

Conclusion

The pathophysiological differences between CPSS and PPVH can adequately be explained by the Q-PCR measurements and Western blots. Although c-MET levels were reduced, downstream signaling seemed to be functional and provides a rational for HGF-supplementation in controlled studies with CPSS and PPVH. Furthermore both diseases may serve as simplified models for comparison with more complex chronic inflammatory diseases and cirrhosis.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​1476-5926-4-7) contains supplementary material, which is available to authorized users.

Competing interests

The author(s) declare that they have no competing interests.

Authors' contributions

BS performed most Q-PCR measurements and wrote the manuscript. LP participated in the setup of Q-PCR measurements and helped to draft the manuscript. TI histochemically examined samples described in this manuscript. BA helped perform the0 Western blot experiments. JIJ histochemically examined samples described in this manuscript. FS helped collect all samples. JR participated in the study design and helped to draft the initial manuscript. All authors read and approved the final manuscript.

Background

Chronic liver disease is characterized by decreased regeneration of hepatocytes and increased formation of fibrous tissue. These characteristics may be the sequel of various chronic processes such as cholestasis, viral infections, toxin exposure, and metabolic disorders. Dogs have complex liver diseases such as hepatitis and cirrhosis which are highly comparable with the human counterparts. Moreover, coding sequences of dogs proved highly homologous to the human sequences [1], especially compared to the rodent genome. Thus, dogs may fulfill a role as a spontaneous animal model in between toxin-induced or surgical models in rodents, and spontaneous diseases in man. The complex interplay of many factors active in chronic liver disease makes it difficult to unravel the roles of different individual pathogenetic pathways. Dogs display liver diseases, which are potentially valuable models to compare complex with simple pathologic entities.
We have chosen these two congenital dog diseases for comparative analysis of liver growth/regeneration, fibrosis, and hepatic homeostasis: congenital portosystemic shunt (CPSS) and primary portal vein hypoplasia (PPVH). CPSS is characterized by an abnormal single large communication between the portal vein and a major systemic vein (cava or azygos). This results in the virtual absence of portal vein perfusion to the liver from birth onwards. Liver growth remains nearly absent but there is essentially no liver pathology [2, 3]. PPVH is a developmental abnormality in which the terminal vein branches are not or only partially present and, in most cases, in combination with congenital portal fibrosis, but without inflammation [4]. PPVH is associated with portal hypertension and reduced liver growth. Thus, these two congenital diseases represent relatively simple models for reduced liver growth associated with fibrosis (PPVH) or without fibrosis (CPSS). Both diseases have a decrease in liver growth due to differences in portal perfusion which results in a massive reduction of liver size.
Because hepatocyte growth factor (HGF) is one of the most important genes involved in liver growth/regeneration [57], abnormal expression of HGF could play a major role in the decreased liver size in CPSS or PPVH. Therefore, treatment of dogs with HGF could be a possible therapeutic approach. A pre-requisite for treatment is that HGF signaling components are unaffected in those dogs. Consequently, we focused on measuring gene products involved in signaling of HGF and counteracting transforming growth factor β1 (TGF-β1). All biological responses induced by HGF are elicited by binding to its receptor, a transmembrane tyrosine kinase encoded by the MET proto-oncogene (c-MET). The signaling cascade triggered by HGF begins with phosphorylation of the receptor and is mediated by concomitant activation of different cytoplasmic effectors that bind to the same multifunctional binding site. The c-MET mediated response includes two key pathways involved in cell survival and mitogenesis [8]. The first; protein kinase B (PKB/Akt) is activated by phosphoinositide 3-kinase (PI3K) and elicits cell survival [9, 10]. The second; ERK1/2 (also known as p42/44 MAPK), a member of the mitogen-activated protein (MAP) kinase family, is activated by the RAS-RAF-MEK pathway and is responsible for mitogenesis [11]. A third response of HGF is the branching morphogenesis which next to the PKB and ERK pathways requires involvement of the signal transducer and activator of transcription (STAT) 3 pathway [12].
It is well established that an increase of TGF-β1 in liver promotes the formation of extracellular matrix (ECM) components and suppresses hepatocyte proliferation [13, 14]. Prolonged overexpression of TGF-β1 in non-parenchymal cells causes hepatic fibrosis in humans and experimental animals. In several fibrosis models, fibrotic lesions are associated with an increase in collagens and TGF-β1 mRNAs [15]. The intracellular pathway that is activated by TGF-β1 receptors is mediated by Smads. Smad2 is activated via carboxy-terminal phosphorylation by TGF-β1 type I receptor kinases. When bound with co-Smads, they act as TGF-β1-induced transcriptional activators of target genes [16].
Cell homeostasis is the result of balance between cell death, cell proliferation, and growth-arrest. Therefore we investigated expression levels of pro-apoptotic Fas ligand and caspase-3, anti-apoptotic Bcl-2 [17], cell-cycle stimulating TGFα, and cell-cycle inhibitor p27kip. All of these gene-products are regulated directly or indirectly by PKB [9].
The present study was designed to describe the differential gene-expression of the above indicated crucial pathways involved in growth/regeneration, fibrosis, and cellular homeostasis in liver tissues of dogs with CPSS (reduced growth/regeneration without fibrosis) and PPVH (reduced growth/regeneration and fibrosis) in comparison with healthy animals. These simple congenital dog models may be used to unravel the roles of different gene products in those pathways. These well-defined large animal models are intended to serve as the first spontaneous liver diseases to investigate novel regenerative/anti-fibrotic therapies, such as HGF treatment. This study may also serve as a basis for future comparison with more complex diseases like chronic hepatitis and cirrhosis.

Results

Histological grading of fibrosis

No fibrosis was seen in liver biopsies of CPSS dogs. In the PPVH dogs histological examination revealed slight portal fibrosis in one dog, slight to moderate portal fibrosis associated with slight to moderate centrolobular fibrosis in four dogs, and marked portal fibrosis with biliary proliferation in three dogs. The control dogs showed a normal liver without fibrosis. Examples of histological examination of CPSS and PPVH are included as Figures 1A and 1B, respectively.

HGF/c-MET signaling pathway involved in regeneration and growth

One of the main in vivo events during regeneration and growth is the signaling via phosphorylation of the HGF receptor c-MET. Q-PCR analysis revealed that HGF mRNA levels in both CPSS and PPVH were decreased three-fold in comparison with healthy dogs (Figure 2). Moreover, the c-MET levels in CPSS and PPVH were significantly decreased (two- and three-fold, respectively). The levels of the mRNAs for TGFα (proliferation) were decreased six-fold in both CPSS and PPVH. The serine-protease HGF activator mRNA was doubled in dogs with CPSS. In contrast, it was halved in dogs with PPVH. Although not significantly in dogs with CPSS, the cell-cycle inhibitor p27kip mRNA was decreased in both conditions.

TGF-β1 cascade signaling pathway involved in fibrosis

The fibrosis signaling pathway is activated through bindings of the active TGF-β1 dimer to the heteromeric type-I and type-II serine/threonine receptor kinases. As shown in Figure 3, TGF-β1 mRNA levels were increased two-fold in dogs with PPVH, whereas the levels in dogs with CPSS were not changed significantly. The receptor type-I, was induced in both liver diseases but only significantly in PPVH. Receptor type-II was increased in both CPSS and PPVH (4- and 5-fold, respectively), indicating an increased binding capacity. One of the proteolytic enzymes involved in activation of TGF-β1 is urokinase plasminogen activator (uPA). The uPA mRNA level was decreased two-fold in dogs with CPSS and, in contrast, doubled in dogs with PPVH.

Gene-expression of apoptosis-related signaling proteins and hypoxia induced factor

We measured three well-known basic apoptotic components of which two are pro-apoptotic (caspase-3 and Fas ligand) and one is anti-apoptotic (Bcl-2). Figure 4 shows that pro-apoptotic mediator Fas ligand was severely inhibited in both dogs with CPSS and in dogs with PPVH (14- and 8-fold, respectively). Moreover, caspase-3 was halved in both CPSS and PPVH. On the other hand, no induction of the anti-apoptotic Bcl-2 was seen in dogs with CPSS, whereas Bcl-2 in dogs with PPVH was doubled. The mechanisms underlying progressive fibrosis are unknown, but fibrosis and hypoxia could have been a fibrogenic stimulus. Hypoxia coordinately up-regulates matrix production and hypoxia induced factor 1 alpha (HIF1α) [18]. These direct hypoxic effects on the expression of genes involved in fibrogenesis was shown in our dogs with PPVH which indeed had elevated levels of HIF1α.

Gene-expression of extracellular matrix gene products

The analysis of ECM expression was performed on three collagens (I, III and IV) and one glycoprotein (fibronectin). Interstitial collagens types I and III are the most commonly found collagens, collagen type IV is a basal membrane collagen. In Figure 5, collagen I was shown to be significantly increased in PPVH (two-fold), whereas CPSS was unchanged. Collagen III and IV were not significantly changed in both groups. Fibronectin showed to be halved in the CPSS group where PPVH remained normal.

Western blot analysis of HGF, c-MET, PKB, STAT3, ERK, TGF-β1, Smad2, Collagen I, and Caspase-3

PKB plays a pivotal role in liver regeneration and growth upon activation of the c-MET-HGF signaling pathway [10]. Western blot analysis of HGF showed an immunoreactive band at 82 kDa with no apparent quantitative differences (Figure 6A). Non-phosphorylated c-MET was detected in all samples, where it was present as an immunoreactive band of 145 kDa. Results showed a decrease in the amount of c-MET in both diseases. On the other hand, the anti-phosphorylated c-MET antibody showed an immunoreactive band in all samples with no apparent quantitative differences. Non-phosphorylated PKB was detected in all samples, where it was present as a single band of 60 kDa. The anti-phosphorylated PKB antibody showed an immunoreactive band in all samples. Two immunoreactive bands at 42 and 44 kDa representing the MAP kinase ERK1/2 showed to be equally present at the protein level between the diseased groups and healthy controls. Interestingly, this also applied for the phosphorylated form where no apparent quantitative differences were found. The 80 kDa STAT3 protein showed a similar result with no apparent quantitative differences in the non-phosphorylated form; however, the STAT3 protein seemed to be somewhat less phosphorylated at the serine 727 residue in the PPVH group. TGF-β1 exerts its actions through complex intracellular signaling pathways. All downstream signaling routes following binding of an active TGF-β1 to its receptors type-I and II elicit phosphorylation of Smad2. TGF-β1 was seen in all diseases as a single band of 25 kDa under non-denaturing conditions (Figure 6B). Interestingly, the amount of TGF-β1 was induced in PPVH compared to CPSS and controls. Non-phosphorylated Smad2 was detected in all samples, where it was present as a single band of 58 kDa, with no apparent changes in quantity. Also interestingly, the anti-phosphorylated Smad2 antibody showed a slight band in CPSS whereas in PPVH a phosphorylated Smad2 is clearly present. Moreover, anti-collagen I showed an increase in the amount of protein in PPVH compared to CPSS and healthy controls, all together emphasizing the differences in fibrosis between CPSS and PPVH. Although reduced in the CPSS and PPVH group, inactive or uncleaved caspase-3 was detected in all samples (Figure 6C), where it was present as a single band of 34 kDa. Finally, the processed forms of 20 and 13 kDa showed to be increased in CPSS and PPVH towards healthy controls.

Discussion

In order to analyze the possibility of growth factor therapy, two congenital canine liver diseases were molecularly dissected. The expression of a total of 17 gene products involved in liver growth/regeneration, fibrosis, ECM, and cellular homeostasis was measured and normalized to the average amount of two reference genes (Q-PCR). Western blot analysis confirmed the quantitative mRNA results and, furthermore, showed activated pathways. These two independent techniques provided insight into the effects of portal venous hypoperfusion in two canine hepatic diseases; congenital portosystemic shunt (CPSS) without fibrosis and primary portal vein hypoplasia (PPVH) with fibrosis. Taken together, the obtained data provided insights in the feasibility for HGF-treatment.
The normalization performed in this study was obtained by averaging the amount of two different reference genes, glyceraldehyde-3-phosphate dehydrogenase (GAPDH) and hypoxanthine phosphoribosyl transferase (HPRT). No samples were more than 5 percent apart from the individual measured reference genes levels (data not shown). This normalization strategy, using the average amount of two reference genes, is taken as a prerequisite for accurate Q-PCR expression profiling which enables us to measure small expression differences and allows the study of their biological relevance [19].
It is well known that HGF plays an essential role in development [20] and regeneration of the liver, and increases hepatocyte viability. The found decrease in gene-expression of both HGF and its receptor agrees with the reduced liver size in these canine disorders. However, and in contrast to the c-MET levels which correlate nicely with the found protein levels, the amount of HGF mRNA does not seem to reflect protein levels. This can be contributed to HGF which can be a paracrine but also an endocrine factor. Extra-hepatic HGF could have been present in the pancreas or intestinal tract [21].
Although HGF and c-MET mRNA levels were decreased, downstream targets of this tyrosine cascade signaling pathway were still active. Downstream targets, such as Fas ligand and p27kip, were chosen as direct or indirect targets of the HGF-cMET-PI3K-PKB axis. Fas ligand transcription is regulated by FOXO (forkhead box, sub-group "O" transcription factors). Therefore, the decrease in Fas ligand can be explained by an active PKB which directly phosphorylates FOXO [22]. A similar result can be seen in the reduced levels of p27kip mRNA, as this is down-regulated at the gene-transcription level by active PKB [23]. Combined, this indicates that PKB is active in both diseases, which was confirmed by Western blot analysis. It remains to be seen whether other receptor tyrosine kinases (e.g., EGF receptor or insulin receptor) activate this pathway in these dogs [24]. Next to the activated PKB pathway, we have analyzed other c-MET mediated responses in CPSS and PPVH. ERK1/2 showed to be activated in both diseases to a similar level as the healthy controls. The significance of the slightly reduced phosphorylated STAT3 in PPVH, which is phosphorylated by HGF on serine 727 [25], needs to be further investigated. Taken together, the pathways which elicit all major biological functions of c-MET showed to be active in CPSS and PPVH.
Prolonged or overexpression of TGF-β1 acts to suppress cell proliferation, and induces a deposition of ECM proteins, resulting in fibrosis in major organs such as liver [26, 27]. We showed that in PPVH the TGF-β1 pathway through Smad2 is activated, consistent with the fibrosis seen in PPVH. Measurements on fibrosis related gene products revealed no elevated activity of the TGF-β1 pathway in CPSS. Gene expression levels related to the TGF-β1 pathway, including its receptors, and the proteolytic activator of TGF-β1 (uPA) were elevated in PPVH, thus indicating an active Smad pathway that could subsequently lead to fibrosis. Western blot analysis confirmed found TGF-β1 levels. Measurements on collagen gene-expression, especially collagen I, confirm the current paradigm of TGF-β1 signaling in fibrous tissues like PPVH [28]. Contrary, non-fibrotic CPSS did not show any alterations in collagen expression. The observation of phosphorylated Smad2 in healthy liver tissue showed that the phosphorylation of Smad2 is a dynamic process and has already been described in other publications [29, 30].
The expressions of the pro-apoptotic genes Fas ligand and caspase-3 were clearly decreased. Bcl-2 gene-expression was elevated two times in PPVH; but not in CPSS (Figure 4). Western blot analysis showed that the unprocessed form of caspase-3 was present in lesser amount in CPSS and PPVH; however, the amount of processed or active bands compared to healthy control was higher in the diseases compared to healthy controls. This indicates that although the total amount of caspase-3 is lower, there is more cleavage of the caspase-3 to its active forms in the diseases, possibly leading to an increase in apoptosis.
Both HGF and TGF-β1 need extracellular processing to become biologically active. The serine protease HGF activator is responsible for activation of proHGF [31]. Our studies revealed that HGF activator gene-expression was doubled in dogs with CPSS and halved in case of PPVH. This indicated an increased HGF activation in CPSS. Although levels of HGF activator were reduced in PPVH, this does not necessarily indicate a lack of extracellular processing of HGF. Interestingly uPA, the activator of TGF-β1, was expressed at an increased level in dogs with PPVH. This may, via active TGF-β1-receptor interaction, indicate an activation of Smads and thus the formation of collagens.
Differential gene expression measurements on hepatic diseases have been performed in the past; nevertheless, little is known about levels of genes that play an important role in fibrosis. There have been measurements on cirrhosis in man and rat that indicate an up or down-regulated expression of several proteins [32]. Although these results might be significant in severe forms of fibrosis, these data depict an end-point of the disease whereas earlier stages may be more informative.
Regeneration with recombinant HGF has been achieved in rodent models of liver failure [33, 34]. Moreover, besides its regenerative capacity, HGF is known to have an antifibrogenic effect [35, 36] and thus reduces or prevent fibrosis in PPVH. TGF-β1 intervention to halt the progression of liver fibrosis and positively effect regeneration, has been applied successfully [37] even in cirrhosis [38]. The measured gene products involved in fibrosis in PPVH make it a good spontaneous animal model to investigate new therapeutic strategies to influence the HGF and/or TGF-β1 pathways in vivo. Furthermore, most fibrogenic models are induced by toxins such as dimethylnitrosamine (DMN), CCl4, or thioacetamide [39]. The canine PPVH model is not drug-induced; therefore, may be better to compare with human diseases and thus fill the gap between induced rodent models and human diseases.
This study is the first to measure expression profiles of crucial pathways of liver growth/regeneration, fibrosis, and hepatic homeostasis in spontaneous canine liver diseases. The present findings in two diseases with relatively simple pathogenesis may also serve as basis for evaluation of more complex diseases like hepatitis and cirrhosis. Evaluation of such complex diseases in dogs is highly suitable for comparative studies on the roles of different pathways in the pathogenesis of liver diseases in man. Two further conclusions can be deduced from the data presented here. First, the pathophysiological differences between CPSS and PPVH can nicely be explained by the Q-PCR measurements and Western blots. Second, although c-MET levels were reduced, downstream signaling seemed to be functional and provides a rational background to design controlled studies for HGF-supplementation in CPSS and PPVH.

Methods

Animals

All samples are obtained from different dog breeds appearing in the clinic with spontaneous diseases. Samples were randomly chosen and aimed to encompass different dog-breeds and both sexes in each group. The procedures were approved by the Ethical Committee as required under Dutch legislation.

Groups

The congenital portosystemic shunt (n = 11 dogs) and primary portal vein hypoplasia group (n = 8 dogs) were compared with a group of healthy dogs (n = 11 dogs). The inclusion criteria for CPSS were increased fasting plasma ammonia concentration, abnormal ammonia tolerance test (peak ammonia ≥ 150 μmol/l plasma) and ultrasonographic visualization of a small liver and a congenital portosystemic shunt with a diameter as wide as the portal vein trunk. The presence of the shunt was further confirmed with surgery, during which a wedge liver biopsy was taken and immediately put in liquid nitrogen and stored at -70°C, until analysis. In CPSS there is no portal hypertension. The inclusion criteria for PPVH were the visualization of a small liver with ultrasonography, presence of multiple small acquired portosystemic collaterals due to portal hypertension, and an abnormal ammonia tolerance test (peak ammonia ≥ 150 μmol/l plasma). Liver tissue of dogs with PPVH was obtained under local anaesthesia by ultrasound-guided biopsy with a true cut 16G biopsy needle. Two biopsies were immediately immersed in liquid nitrogen, and stored at -70°C, until analysis. The healthy control dogs were age-matched, and had AP, ALT, and fasting bile acids in plasma within the reference range. Ultrasonographically the control dog livers had a normal size, shape, and structure, and there were no histological abnormalities in stained histological sections.

Histological grading of fibrosis

Liver samples were fixed in 10% buffered formalin and routinely embedded in paraffin. Sections (4 μm) were stained with haematoxylin-eosin, the Van Gieson stain, and the reticulin stain according to Gordon and Sweet. Histologically, the presence of fibrosis was evaluated semi-quantitatively (absent, slight, moderate, or marked) as well as with respect to its localization. Fibrosis scoring was performed according to Scheuer, a defined scoring method for fibrosis in hepatitis. The slides were independently examined by one certified veterinary pathologist.

RNA isolation and reverse-transcription polymerase chain reaction

Total cellular RNA was isolated from each frozen canine liver tissue in duplicate, using the RNeasy Mini Kit (Qiagen, Leusden, The Netherlands) according to the manufacturer's instructions. The RNA samples were treated with Dnase-I (Qiagen Rnase-free DNase kit). In total 3 μg of RNA was incubated with poly(dT) primers at 42°C for 45 min, in a 60 μl reaction volume, using the Reverse Transcription System from Promega (Promega Benelux, Leiden, The Netherlands).

Quantitative measurements of the mRNA levels of HGF, TGF-β1, and other related signaling molecules

Q-PCR based on the high affinity double-stranded DNA-binding dye SYBR® green I (BMA, Rockland, ME) was performed in triplicate in a spectrofluorimetric thermal iCycler® (BioRad, Veenendaal, The Netherlands). Data were collected and analyzed with the provided application software. For each Q-PCR, 2 μl (of the 2 times diluted stock) of cDNA was used in a reaction volume of 50 μl containing 1× manufacturer's buffer, 2 mM MgCl2, 0.5 × SYBR® green I, 200 μM dNTP's, 20 pmol of both primers, 1.25 units of AmpliTaq Gold (Applied Biosystems, Nieuwerkerk a/d IJssel, The Netherlands), on 96-well iCycler iQ plates (BioRad). Primer pairs, depicted in Table 1, were designed using PrimerSelect software (DNASTAR Inc., Madison, WI). All PCR protocols included a 5-minute polymerase activation step and continued for 40 cycles at 95°C denaturation for 20 sec, annealing for 30 sec and elongation at 72°C for 30 sec with a final extension for 5 min at 72°C. Annealing temperatures were optimized at various levels ranging from 56°C till 67°C (Table 1). Melt curves (iCycler, BioRad), agarose gel electrophoresis, and standard sequencing procedures were used to examine each sample for purity and specificity (ABI PRISM 3100 Genetic Analyser, Applied Biosystems). Standard curves constructed by plotting the relative starting amount versus threshold cycles were generated using serial 4-fold dilutions of pooled cDNA fractions from both healthy and diseased liver tissues. The amplification efficiency, E (%) = (10(1/-s)-1) * 100 (s = slope), of each standard curve was determined and appeared to be > 95%, and < 105%, over a wide dynamic range. For each experimental sample, the amount of the gene of interest, and of the endogenous references glyceraldehyde-3-phosphate dehydrogenase (GAPDH) and hypoxanthine phosphoribosyl transferase (HPRT) were determined from the appropriate standard curve in autonomous experiments. If relative amounts of GAPDH and HPRT were constant for a sample, data were considered valid and the average amount was included in the study (data not shown). Results were normalized according to the average amount of the endogenous references. The normalized values were divided by the normalized values of the calibrator (healthy group) to generate relative expression levels [40].
Table 1
Nucleotide Sequences of Dog-Specific Primers for Real-Time Q-PCR.
Gene
Primer
Sequence (5'-3')
°C
Product size (bp)
Accession number
GAPDH
Forward
TGT CCC CAC CCC CAA TGT ATC
58
100
AB038240
 
Reversed
CTC CGA TGC CTG CTT CAC TAC CTT
   
HPRT
Forward
AGC TTG CTG GTG AAA AGG AC
56
100
L77488/L77489
 
Reversed
TTA TAG TCA AGG GCA TAT CC
   
HGF
Forward
AAA GGA GAT GAG AAA CGC AAA CAG
58
92
BD105535
 
Reversed
GGC CTA GCA AGC TTC AGT AAT ACC
   
c-MET
Forward
TGT GCT GTG AAA TCC CTG AAT AGA AATC
59
112
AB118945
 
Reversed
CCA AGA GTG AGA GTA CGT TTG GAT GAC
   
TGFα
Forward
CCG CCT TGG TGG TGG TCT CC
63
136
AY458143
 
Reversed
AGG GCG CTG GGC TTC TCG T
   
HGF activator
Forward
ACA CAG ACG TTT GGC ATC GAG AAG TAT
60
128
AY458142
 
Reversed
AAA CTG GAG CGG ATG GCA CAG
   
p27kip
Forward
CGG AGG GAC GCC AAA CAG G
60
90
AY455798
 
Reversed
GTC CCG GGT CAA CTC TTC GTG
   
TGF-β1
Forward
CAA GGA TCT GGG CTG GAA GTG GA
66
113
L34956
 
Reversed
CCA GGA CCT TGC TGT ACT GCG TGT
   
TGF-β1 R I
Forward
CAG TCA CCG AGA CCA CAG ACA AAG T
59
101
AY455799
 
Reversed
TGA AGA TGG TGC ACA AAC AAA TGG
   
TGF-β1 R II
Forward
GAC CTG CTG CCT GTG TGA CTT TG
61
116
AY455800
 
Reversed
GGA CTT CGG GAG CCA TGT ATC TTG
   
UPA
Forward
CTG GGG AGA TGA AGT TTG AGG TGG
64.5
105
AY455801
 
Reversed
TGG AAC GGA TCT TCA GCA AGG C
   
Bcl-2
Forward
TGG AGA GCG TCA ACC GGG AGA TGT
61
87
AB116145
 
Reversed
AGG TGT GCA GAT GCC GGT TCA GGT
   
Fas Ligand
Forward
GGG GTC AGT CCT GCA ACA ACA A
54
94
AY603042
 
Reversed
ATC TTC CCC TCC ATC AGC ATC AG
   
Caspase-3
Forward
ATC ACT GAA GAT GGA TGG GTT GGT
58
140
AB085580
 
Reversed
GAA AGG AGC ATG TTC TGA AGT AGC ACT
   
HIF1α
Forward
TTA CGT TCC TTC GAT CAG TTG TCA
61
106
AY455802
 
Reversed
GAG GAG GTT CTT GCA TTG GAG TC
   
Collagen I
Forward
GTG TGT ACA GAA CGG CCT CA
61
111
AF056303
 
Reversed
TCG CAA ATC ACG TCA TCG
   
Collagen III
Forward
ATA GAG GCT TTG ATG GAC GAA
65
134
AB042266
 
Reversed
CCT CGC TCA CCA GGA GC
   
Collagen IV
Forward
CAC AGC CAG ACA ACA GAT GC
67
151
U07888
 
Reversed
GCA TGG TAC TGA AGC GAC G
   
Fibronectin
Forward
AGG TTG TTA CCA TGG GCA
61
91
U52106
 
Reversed
GCA TAA TGG GAA ACC GTG TAG
   

Statistical analysis

A Kolmogorov-Smirnov test was performed to establish a normal distribution and a Levene's test for the homogeneity of variances. All samples included in this study were normally distributed and homogeneous in variance. The statistical significance of differences between diseased and control animals was determined by using the Student's t-test. A p-value < 0.05 was considered statistically significant. Analysis was performed using SPSS software (SPSS Benelux BV, Gorinchem, The Netherlands).

Western blot analysis

Used antibodies are described in Table 2. For Western blot analysis 30 mg of liver tissue from at least six samples of each group (n = 6 dogs per group, randomly chosen from original group) were pooled and analyzed. Liver tissues were homogenized in RIPA buffer containing 1% Igepal, 0.6 mM phenylmethylsulfonyl-fluoride, 17 μg/ml aprotinine, and 1 mM sodium-orthovanadate (Sigma chemical Co., Zwijndrecht, The Netherlands). Protein concentrations were obtained using a Lowry-based assay (DC Protein Assay, BioRad). Twenty μg of protein of the supernatant was denatured for 3 min at 95°C and electroforesed on 7.5% Tris-HCl polyacrylamide gels (BioRad) and the proteins were transferred onto Hybond-C Extra Nitrocellulose membranes (Amersham Biosciences Europe, Roosendaal, The Netherlands) using a Mini Trans-Blot® Cell blot-apparatus (BioRad). Immunodetection was based on an ECL Western blot analysis system, performed according to the manufacturer's instructions (Amersham Biosciences Europe). The membranes were incubated with 4% ECL blocking solution in TBS for 1 hour under gentle shaking. The incubation of the primary antibody was performed at 4°C over-night for all antibodies (see Table 2) in TBS with 0.1% Tween-20 (Boom B.V., Meppel, The Netherlands). After washing, the membranes were incubated with their respective horseradish peroxidase-conjugated secondary antibody (R&D systems, Europe Ltd., Abingdon, UK) at room temperature for 1 h and exposed to Kodak BioMax Light-1 films (Sigma chemical Co.). Densitometric analysis of immunoreactive bands was performed with a Gel Doc 2000 system coupled to the Quantity One 4.3.0 Software (BioRad).
Table 2
Used antibodies in Western blot experiments.
Antigen
Product Size (kDa)
Dilution
Manufacturer
Secondary antibody
Dilution
HGF
82
1:100
Neomarkers
Anti-mouse HRP
1:20,000
p-c-MET (Tyr 1230/1234/1235)
145
1:750
Abcam
Anti-rabbit HRP
1:20,000
c-MET
145
1:750
Sigma
Anti-goat HRP
1:20,000
p-PKB (Thr 308)
60
1:1,000
Cell-Signaling
Anti-mouse HRP
1:20,000
PKB
60
1:250
BD Biosciences
Anti-mouse HRP
1:20,000
p-STAT3 (Ser 727)
86
1:1,000
Cell Signalling
Anti-rabbit HRP
1:20,000
STAT3
86
1:2,500
BD Biosciences
Anti-rabbit HRP
1:20,000
p-Erk1/2 (Thr 202/Tyr 204)
42/44
1:1,500
Cell Signalling
Anti-rabbit HRP
1:20,000
ERK1/2
42/44
1:1,000
Cell Signalling
Anti-rabbit HRP
1:20,000
TGF-β1
25
1:1,000
Abcam
Anti-rabbit HRP
1:20,000
p-Smad2 (Ser 465/467)
58
1:2,000
Cell-Signaling
Anti-rabbit HRP
1:20,000
Smad2
58
1:500
BD Biosciences
Anti-mouse HRP
1:20,000
Collagen I
95/210
1:500
Calbiochem
Anti-mouse HRP
1:20,000
Caspase-3
34/20/18
1:1,000
Calbiochem
Anti-rabbit HRP
1:20,000
Beta-actin (pan Ab-5)
42
1:2,000
Neomarkers
Anti-mouse HRP
1:20,000

Acknowledgements

The authors are indebted to Dr. Alexandra Pietersen, Dr. Bernard Roelen, and Dr. Peter ten Dijke for their invaluable advice.
Open Access This article is published under license to BioMed Central Ltd. This is an Open Access article is distributed under the terms of the Creative Commons Attribution License ( https://​creativecommons.​org/​licenses/​by/​2.​0 ), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Competing interests

The author(s) declare that they have no competing interests.

Authors' contributions

BS performed most Q-PCR measurements and wrote the manuscript. LP participated in the setup of Q-PCR measurements and helped to draft the manuscript. TI histochemically examined samples described in this manuscript. BA helped perform the0 Western blot experiments. JIJ histochemically examined samples described in this manuscript. FS helped collect all samples. JR participated in the study design and helped to draft the initial manuscript. All authors read and approved the final manuscript.
Literatur
1.
Zurück zum Zitat Guyon R, Lorentzen TD, Hitte C, Kim L, Cadieu E, Parker HG, Quignon P, Lowe JK, Renier C, Gelfenbeyn B, Vignaux F, DeFrance HB, Gloux S, Mahairas GG, Andre C, Galibert F, Ostrander EA: A 1-Mb resolution radiation hybrid map of the canine genome. Proc Natl Acad Sci U S A. 2003, 100: 5296-5301. 10.1073/pnas.0831002100.PubMedPubMedCentralCrossRef Guyon R, Lorentzen TD, Hitte C, Kim L, Cadieu E, Parker HG, Quignon P, Lowe JK, Renier C, Gelfenbeyn B, Vignaux F, DeFrance HB, Gloux S, Mahairas GG, Andre C, Galibert F, Ostrander EA: A 1-Mb resolution radiation hybrid map of the canine genome. Proc Natl Acad Sci U S A. 2003, 100: 5296-5301. 10.1073/pnas.0831002100.PubMedPubMedCentralCrossRef
2.
Zurück zum Zitat Murray CP, Yoo SJ, Babyn PS: Congenital extrahepatic portosystemic shunts. Pediatr Radio. 2003, 33: 614-620. 10.1007/s00247-003-1002-x.CrossRef Murray CP, Yoo SJ, Babyn PS: Congenital extrahepatic portosystemic shunts. Pediatr Radio. 2003, 33: 614-620. 10.1007/s00247-003-1002-x.CrossRef
3.
Zurück zum Zitat van den Ingh TS, Rothuizen J, Meyer HP: Circulatory disorder of the liver in dogs and cats. Vet Q. 1995, 17: 70-76.PubMedCrossRef van den Ingh TS, Rothuizen J, Meyer HP: Circulatory disorder of the liver in dogs and cats. Vet Q. 1995, 17: 70-76.PubMedCrossRef
4.
Zurück zum Zitat van den Ingh TS, Rothuizen J, Meyer HP: Portal Hypertension associated with primary hypoplasia of the hepatic portal vein in dogs. Vet Rec. 1995, 137: 424-427.PubMedCrossRef van den Ingh TS, Rothuizen J, Meyer HP: Portal Hypertension associated with primary hypoplasia of the hepatic portal vein in dogs. Vet Rec. 1995, 137: 424-427.PubMedCrossRef
5.
Zurück zum Zitat Michalopoulos KM, DeFrances MC: Liver regeneration. Science. 1997, 276: 60-66. 10.1126/science.276.5309.60.PubMedCrossRef Michalopoulos KM, DeFrances MC: Liver regeneration. Science. 1997, 276: 60-66. 10.1126/science.276.5309.60.PubMedCrossRef
6.
Zurück zum Zitat Trusolino L, Comoglio PM: Scatter-factor and semaphorin receptors: cell signalling for invasive growth. Nat Rev Cancer. 2002, 2: 289-300. 10.1038/nrc779.PubMedCrossRef Trusolino L, Comoglio PM: Scatter-factor and semaphorin receptors: cell signalling for invasive growth. Nat Rev Cancer. 2002, 2: 289-300. 10.1038/nrc779.PubMedCrossRef
7.
Zurück zum Zitat Fausto N: Liver regeneration. J Hepatol. 2000, 19-31. 10.1016/S0168-8278(00)80412-2. Suppl 1 Fausto N: Liver regeneration. J Hepatol. 2000, 19-31. 10.1016/S0168-8278(00)80412-2. Suppl 1
8.
Zurück zum Zitat Funakoshi H, Nakamura T: Hepatocyte growth factor: from diagnosis to clinical applications. Clin Chim Acta. 2003, 327: 1-23. 10.1016/S0009-8981(02)00302-9.PubMedCrossRef Funakoshi H, Nakamura T: Hepatocyte growth factor: from diagnosis to clinical applications. Clin Chim Acta. 2003, 327: 1-23. 10.1016/S0009-8981(02)00302-9.PubMedCrossRef
9.
Zurück zum Zitat Scheid MP, Woodgett JR: Unravelling the activation mechanisms of protein kinase B/Akt. FEBS Lett. 2003, 546: 108-112. 10.1016/S0014-5793(03)00562-3.PubMedCrossRef Scheid MP, Woodgett JR: Unravelling the activation mechanisms of protein kinase B/Akt. FEBS Lett. 2003, 546: 108-112. 10.1016/S0014-5793(03)00562-3.PubMedCrossRef
10.
Zurück zum Zitat Coffer PJ, Jin J, Woodgett JR: Protein kinase B (c-Akt): a multifunctional mediator of phosphatidylinositol 3-kinase activation. Biochem J. 1998, 335: 1-13.PubMedPubMedCentralCrossRef Coffer PJ, Jin J, Woodgett JR: Protein kinase B (c-Akt): a multifunctional mediator of phosphatidylinositol 3-kinase activation. Biochem J. 1998, 335: 1-13.PubMedPubMedCentralCrossRef
11.
Zurück zum Zitat Paumelle R, Tulasne D, Kherrouche Z, Plaza S, Leroy C, Reveneau S, Vandenbunder B, Fafeur V, Tulashe D, Reveneau S: Hepatocyte growth factor/scatter factor activates the ETS1 transcription factor by a RAS-RAF-MEK-ERK signaling pathway. Oncogene. 2002, 21: 2309-2319. 10.1038/sj.onc.1205297.PubMedCrossRef Paumelle R, Tulasne D, Kherrouche Z, Plaza S, Leroy C, Reveneau S, Vandenbunder B, Fafeur V, Tulashe D, Reveneau S: Hepatocyte growth factor/scatter factor activates the ETS1 transcription factor by a RAS-RAF-MEK-ERK signaling pathway. Oncogene. 2002, 21: 2309-2319. 10.1038/sj.onc.1205297.PubMedCrossRef
12.
Zurück zum Zitat Boccaccio C, Ando M, Tamagnone L, Bardelli A, Michieli P, Battistini C, Comoglio PM: Induction of epithelial tubules by growth factor HGF depends on the STAT pathway. Nature. 1998, 391: 285-288. 10.1038/34657.PubMedCrossRef Boccaccio C, Ando M, Tamagnone L, Bardelli A, Michieli P, Battistini C, Comoglio PM: Induction of epithelial tubules by growth factor HGF depends on the STAT pathway. Nature. 1998, 391: 285-288. 10.1038/34657.PubMedCrossRef
13.
Zurück zum Zitat Derynck R, Akhurst RJ, Balmain A: TGF-β signaling in tumor suppression and cancer progression. Nature Genet. 2001, 29: 117-129. 10.1038/ng1001-117.PubMedCrossRef Derynck R, Akhurst RJ, Balmain A: TGF-β signaling in tumor suppression and cancer progression. Nature Genet. 2001, 29: 117-129. 10.1038/ng1001-117.PubMedCrossRef
14.
Zurück zum Zitat Piek E, Heldin CA, ten Dijke P: Specificity, diversity, and regulation in TGF-β superfamily signalling. FASEB J. 1999, 13: 2105-2124.PubMed Piek E, Heldin CA, ten Dijke P: Specificity, diversity, and regulation in TGF-β superfamily signalling. FASEB J. 1999, 13: 2105-2124.PubMed
15.
Zurück zum Zitat Aihara Y, Kasuya H, Onda H, Hori T, Takeda J: Quantitative analysis of gene expressions related to inflammation in canine spastic artery after subarachnoid hemorrhage. Stroke. 2001, 32: 212-217.PubMedCrossRef Aihara Y, Kasuya H, Onda H, Hori T, Takeda J: Quantitative analysis of gene expressions related to inflammation in canine spastic artery after subarachnoid hemorrhage. Stroke. 2001, 32: 212-217.PubMedCrossRef
16.
Zurück zum Zitat Massague J: How cells read TGF-beta signals. Nat Rev Mol Cell Biol. 2000, 1: 169-178. 10.1038/35043051.PubMedCrossRef Massague J: How cells read TGF-beta signals. Nat Rev Mol Cell Biol. 2000, 1: 169-178. 10.1038/35043051.PubMedCrossRef
17.
18.
Zurück zum Zitat Norman JT, Clark IM, Garcia PL: Hypoxia promotes fibrogenesis in human renal fibroblasts. Kidney Int. 2000, 58: 2351-2366. 10.1046/j.1523-1755.2000.00419.x.PubMedCrossRef Norman JT, Clark IM, Garcia PL: Hypoxia promotes fibrogenesis in human renal fibroblasts. Kidney Int. 2000, 58: 2351-2366. 10.1046/j.1523-1755.2000.00419.x.PubMedCrossRef
19.
Zurück zum Zitat Vandesompele J, De Preter K, Pattyn F, Poppe B, Van Roy N, De Paepe A, Speleman F: Accurate normalization of real-time quantitative RT-PCR data by geometric averaging of multiple internal control genes. Genome Biol. 2002, 3: research0034.1-research0034.11. 10.1186/gb-2002-3-7-research0034.CrossRef Vandesompele J, De Preter K, Pattyn F, Poppe B, Van Roy N, De Paepe A, Speleman F: Accurate normalization of real-time quantitative RT-PCR data by geometric averaging of multiple internal control genes. Genome Biol. 2002, 3: research0034.1-research0034.11. 10.1186/gb-2002-3-7-research0034.CrossRef
20.
Zurück zum Zitat Schmidt C, Bladt F, Goedecke S, Brinkmann V, Zschiesche W, Sharpe M, Gherardi E, Birchmeier C: Scatter factor/hepatocyte growth factor is essential for liver development. Nature. 1995, 373: 699-702. 10.1038/373699a0.PubMedCrossRef Schmidt C, Bladt F, Goedecke S, Brinkmann V, Zschiesche W, Sharpe M, Gherardi E, Birchmeier C: Scatter factor/hepatocyte growth factor is essential for liver development. Nature. 1995, 373: 699-702. 10.1038/373699a0.PubMedCrossRef
21.
Zurück zum Zitat La Rosa S, Uccella S, Capella C, Erba S, Sessa F: Localization of Hepatocyte Growth Factor and Its Receptor met in Endocrine Cells and Related Tumors of the Gut and Pancreas: An Immunohistochemical Study. Endocr Pathol. 2000, 11: 315-329. 10.1385/EP:11:4:315.PubMedCrossRef La Rosa S, Uccella S, Capella C, Erba S, Sessa F: Localization of Hepatocyte Growth Factor and Its Receptor met in Endocrine Cells and Related Tumors of the Gut and Pancreas: An Immunohistochemical Study. Endocr Pathol. 2000, 11: 315-329. 10.1385/EP:11:4:315.PubMedCrossRef
22.
Zurück zum Zitat Barthelemy C, Henderson CE, Pettmann B: Foxo3a induces motoneuron death through the Fas pathway in cooperation with JNK. BMC Neurosci. 2004, 5: 48-10.1186/1471-2202-5-48.PubMedPubMedCentralCrossRef Barthelemy C, Henderson CE, Pettmann B: Foxo3a induces motoneuron death through the Fas pathway in cooperation with JNK. BMC Neurosci. 2004, 5: 48-10.1186/1471-2202-5-48.PubMedPubMedCentralCrossRef
23.
Zurück zum Zitat Chandramohan V, Jeay S, Pianetti S, Sonenshein GE: Reciprocal control of Forkhead box O 3a and c-Myc via the phosphatidylinositol 3-kinase pathway coordinately regulates p27Kip1 levels. J Immunol. 2004, 172: 5522-5527.PubMedCrossRef Chandramohan V, Jeay S, Pianetti S, Sonenshein GE: Reciprocal control of Forkhead box O 3a and c-Myc via the phosphatidylinositol 3-kinase pathway coordinately regulates p27Kip1 levels. J Immunol. 2004, 172: 5522-5527.PubMedCrossRef
24.
Zurück zum Zitat Hanada M, Feng J, Hemmings BA: Structure, regulation and function of PKB/AKT – a major therapeutic target. Biochim Biophys Acta. 2004, 1697: 3-16.PubMedCrossRef Hanada M, Feng J, Hemmings BA: Structure, regulation and function of PKB/AKT – a major therapeutic target. Biochim Biophys Acta. 2004, 1697: 3-16.PubMedCrossRef
25.
Zurück zum Zitat Nakagami H, Morishita R, Yamamoto K, Taniyama Y, Aoki M, Matsumoto K, Nakamura T, Kaneda Y, Horiuchi M, Ogihara T: Mitogenic and antiapoptotic actions of hepatocyte growth factor through ERK, STAT3, and AKT in endothelial cells. Hypertension. 2001, 37: 581-586.PubMedCrossRef Nakagami H, Morishita R, Yamamoto K, Taniyama Y, Aoki M, Matsumoto K, Nakamura T, Kaneda Y, Horiuchi M, Ogihara T: Mitogenic and antiapoptotic actions of hepatocyte growth factor through ERK, STAT3, and AKT in endothelial cells. Hypertension. 2001, 37: 581-586.PubMedCrossRef
26.
Zurück zum Zitat Diez-Marques L, Ortega-Velazquez R, Langa C, Rodriguez-Barbero A, Lopez-Novoa JM, Lamas S, Bernabeu C: Expression of endoglin in human mesangial cells: modulation of extracellular matrix synthesis. Biochim Biophys Acta. 2002, 1587: 36-44.PubMedCrossRef Diez-Marques L, Ortega-Velazquez R, Langa C, Rodriguez-Barbero A, Lopez-Novoa JM, Lamas S, Bernabeu C: Expression of endoglin in human mesangial cells: modulation of extracellular matrix synthesis. Biochim Biophys Acta. 2002, 1587: 36-44.PubMedCrossRef
27.
Zurück zum Zitat Zhu HJ, Burgess AW: Regulation of transforming growth factor-beta signaling. Mol Cell Biol Res Commun. 2001, 4: 321-330. 10.1006/mcbr.2001.0301.PubMedCrossRef Zhu HJ, Burgess AW: Regulation of transforming growth factor-beta signaling. Mol Cell Biol Res Commun. 2001, 4: 321-330. 10.1006/mcbr.2001.0301.PubMedCrossRef
28.
Zurück zum Zitat Tsukada S, Parsons CJ, Rippe RA: Mechanisms of liver fibrosis. Clin Chim Act. Tsukada S, Parsons CJ, Rippe RA: Mechanisms of liver fibrosis. Clin Chim Act.
29.
Zurück zum Zitat Liu C, Gaca MD, Swenson ES, Vellucci VF, Reiss M, Wells RG: Smads 2 and 3 are differentially activated by transforming growth factor-beta (TGF-beta) in quiescent and activated hepatic stellate cells. Constitutive nuclear localization of Smads in activated cells is TGF-beta-independent. J Biol Chem. 2003, 278: 11721-11728. 10.1074/jbc.M207728200.PubMedCrossRef Liu C, Gaca MD, Swenson ES, Vellucci VF, Reiss M, Wells RG: Smads 2 and 3 are differentially activated by transforming growth factor-beta (TGF-beta) in quiescent and activated hepatic stellate cells. Constitutive nuclear localization of Smads in activated cells is TGF-beta-independent. J Biol Chem. 2003, 278: 11721-11728. 10.1074/jbc.M207728200.PubMedCrossRef
30.
Zurück zum Zitat Kondou H, Mushiake S, Etani Y, Miyoshi Y, Michigami T, Ozono K: A blocking peptide for transforming growth factor-beta1 activation prevents hepatic fibrosis in vivo. J Hepatol. 2003, 39: 742-748. 10.1016/S0168-8278(03)00377-5.PubMedCrossRef Kondou H, Mushiake S, Etani Y, Miyoshi Y, Michigami T, Ozono K: A blocking peptide for transforming growth factor-beta1 activation prevents hepatic fibrosis in vivo. J Hepatol. 2003, 39: 742-748. 10.1016/S0168-8278(03)00377-5.PubMedCrossRef
31.
Zurück zum Zitat Kataoka H, Miyata S, Uchinokura S, Itoh H: Roles of hepatocyte growth factor (HGF) activator and HGF activator inhibitor in the pericellular activation of HGF/scatter factor. Cancer Metastasis Rev. 2003, 22: 223-236. 10.1023/A:1023051500010.PubMedCrossRef Kataoka H, Miyata S, Uchinokura S, Itoh H: Roles of hepatocyte growth factor (HGF) activator and HGF activator inhibitor in the pericellular activation of HGF/scatter factor. Cancer Metastasis Rev. 2003, 22: 223-236. 10.1023/A:1023051500010.PubMedCrossRef
32.
Zurück zum Zitat Wang HT, Chen S, Wang J, Ou QJ, Liu C, Zheng SS, Deng MH, Liu XP: Expression of growth hormone receptor and its mRNA in hepatic cirrhosis. World J Gastroenterol. 2003, 9: 765-770.PubMedPubMedCentralCrossRef Wang HT, Chen S, Wang J, Ou QJ, Liu C, Zheng SS, Deng MH, Liu XP: Expression of growth hormone receptor and its mRNA in hepatic cirrhosis. World J Gastroenterol. 2003, 9: 765-770.PubMedPubMedCentralCrossRef
33.
Zurück zum Zitat Kosai K, Matsumoto K, Funakoshi H, Nakamura T: Hepatocyte growth factor prevents endotoxin-induced lethal hepatic failure in mice. Hepatology. 1999, 30: 151-159. 10.1002/hep.510300102.PubMedCrossRef Kosai K, Matsumoto K, Funakoshi H, Nakamura T: Hepatocyte growth factor prevents endotoxin-induced lethal hepatic failure in mice. Hepatology. 1999, 30: 151-159. 10.1002/hep.510300102.PubMedCrossRef
34.
Zurück zum Zitat Yasuda H, Imai E, Shiota A, Fujise N, Morinaga T, Higashio K: Antifibrogenic effect of a deletion variant of hepatocyte growth factor on liver fibrosis in rats. Hepatology. 1996, 24: 636-642.PubMedCrossRef Yasuda H, Imai E, Shiota A, Fujise N, Morinaga T, Higashio K: Antifibrogenic effect of a deletion variant of hepatocyte growth factor on liver fibrosis in rats. Hepatology. 1996, 24: 636-642.PubMedCrossRef
35.
Zurück zum Zitat Ueki T, Kaneda Y, Tsutsui H, Nakanishi K, Sawa Y, Morishita R, Matsumoto K, Nakamura T, Takahashi H, Okamoto E, Fujimoto J: Hepatocyte growth factor gene therapy of liver cirrhosis in rats. Nat Med. 1999, 5: 226-230. 10.1038/5593.PubMedCrossRef Ueki T, Kaneda Y, Tsutsui H, Nakanishi K, Sawa Y, Morishita R, Matsumoto K, Nakamura T, Takahashi H, Okamoto E, Fujimoto J: Hepatocyte growth factor gene therapy of liver cirrhosis in rats. Nat Med. 1999, 5: 226-230. 10.1038/5593.PubMedCrossRef
36.
Zurück zum Zitat Matsuda Y, Matsumoto K, Yamada A, Ichida T, Asakura H, Komoriya Y, Nishiyama E, Nakamura T: Preventive and therapeutic effects in rats of hepatocyte growth factor infusion on liver fibrosis/cirrhosis. Hepatology. 1997, 26: 81-89.PubMedCrossRef Matsuda Y, Matsumoto K, Yamada A, Ichida T, Asakura H, Komoriya Y, Nishiyama E, Nakamura T: Preventive and therapeutic effects in rats of hepatocyte growth factor infusion on liver fibrosis/cirrhosis. Hepatology. 1997, 26: 81-89.PubMedCrossRef
37.
Zurück zum Zitat Ohara K, Kusano M: Anti-transforming growth factor-beta1 antibody improves survival rate following partial hepatectomy in cirrhotic rats. Hepatol Res. 2002, 24: 174-10.1016/S1386-6346(02)00031-1.PubMedCrossRef Ohara K, Kusano M: Anti-transforming growth factor-beta1 antibody improves survival rate following partial hepatectomy in cirrhotic rats. Hepatol Res. 2002, 24: 174-10.1016/S1386-6346(02)00031-1.PubMedCrossRef
38.
Zurück zum Zitat Nakamura T, Sakata R, Ueno T, Sata M, Ueno H: Inhibition of transforming growth factor β prevents progression of liver fibrosis and enhanced hepatocyte regeneration in dimethylnitrosamine-treated rats. Hepatology. 2000, 32: 247-255. 10.1053/jhep.2000.9109.PubMedCrossRef Nakamura T, Sakata R, Ueno T, Sata M, Ueno H: Inhibition of transforming growth factor β prevents progression of liver fibrosis and enhanced hepatocyte regeneration in dimethylnitrosamine-treated rats. Hepatology. 2000, 32: 247-255. 10.1053/jhep.2000.9109.PubMedCrossRef
39.
Zurück zum Zitat Sato M, Kakubari M, Kawamura M, Sugimoto J, Matsumoto K, Ishii T: The decrease in total collagen fibers in the liver by hepatocyte growth factor after formation of cirrhosis induced by thioacetamide. Biochem Pharmacol. 2000, 59: 681-690. 10.1016/S0006-2952(99)00359-7.PubMedCrossRef Sato M, Kakubari M, Kawamura M, Sugimoto J, Matsumoto K, Ishii T: The decrease in total collagen fibers in the liver by hepatocyte growth factor after formation of cirrhosis induced by thioacetamide. Biochem Pharmacol. 2000, 59: 681-690. 10.1016/S0006-2952(99)00359-7.PubMedCrossRef
40.
Zurück zum Zitat Kimura Y, Leung PS, Kenny TP, van de Water J, Nishioka M, Giraud AS, Neuberger J, Benson G, Kaul R, Ansari AA, Coppel RL, Gershwin ME: Differential expression of intestinal trefoil factor in biliary epithelial cells of primary biliary cirrhosis. Hepatology. 2002, 36: 1227-1235. 10.1053/jhep.2002.36157.PubMedCrossRef Kimura Y, Leung PS, Kenny TP, van de Water J, Nishioka M, Giraud AS, Neuberger J, Benson G, Kaul R, Ansari AA, Coppel RL, Gershwin ME: Differential expression of intestinal trefoil factor in biliary epithelial cells of primary biliary cirrhosis. Hepatology. 2002, 36: 1227-1235. 10.1053/jhep.2002.36157.PubMedCrossRef
Metadaten
Titel
Regenerative and fibrotic pathways in canine hepatic portosystemic shunt and portal vein hypoplasia, new models for clinical hepatocyte growth factor treatment
verfasst von
Bart Spee
Louis C Penning
Ted SGAM van den Ingh
Brigitte Arends
Jooske IJzer
Frederik J van Sluijs
Jan Rothuizen
Publikationsdatum
01.12.2005
Verlag
BioMed Central
Erschienen in
Comparative Hepatology / Ausgabe 1/2005
Elektronische ISSN: 1476-5926
DOI
https://doi.org/10.1186/1476-5926-4-7

Weitere Artikel der Ausgabe 1/2005

Comparative Hepatology 1/2005 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Echinokokkose medikamentös behandeln oder operieren?

06.05.2024 DCK 2024 Kongressbericht

Die Therapie von Echinokokkosen sollte immer in spezialisierten Zentren erfolgen. Eine symptomlose Echinokokkose kann – egal ob von Hunde- oder Fuchsbandwurm ausgelöst – konservativ erfolgen. Wenn eine Op. nötig ist, kann es sinnvoll sein, vorher Zysten zu leeren und zu desinfizieren. 

Umsetzung der POMGAT-Leitlinie läuft

03.05.2024 DCK 2024 Kongressbericht

Seit November 2023 gibt es evidenzbasierte Empfehlungen zum perioperativen Management bei gastrointestinalen Tumoren (POMGAT) auf S3-Niveau. Vieles wird schon entsprechend der Empfehlungen durchgeführt. Wo es im Alltag noch hapert, zeigt eine Umfrage in einem Klinikverbund.

Proximale Humerusfraktur: Auch 100-Jährige operieren?

01.05.2024 DCK 2024 Kongressbericht

Mit dem demographischen Wandel versorgt auch die Chirurgie immer mehr betagte Menschen. Von Entwicklungen wie Fast-Track können auch ältere Menschen profitieren und bei proximaler Humerusfraktur können selbst manche 100-Jährige noch sicher operiert werden.

Die „Zehn Gebote“ des Endokarditis-Managements

30.04.2024 Endokarditis Leitlinie kompakt

Worauf kommt es beim Management von Personen mit infektiöser Endokarditis an? Eine Kardiologin und ein Kardiologe fassen die zehn wichtigsten Punkte der neuen ESC-Leitlinie zusammen.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.