The insulin-like growth factor (IGF) signaling axis and hepatitis C virus-associated carcinogenesis (Review)

  • Authors:
    • Aldona Kasprzak
    • Agnieszka Adamek
  • View Affiliations

  • Published online on: October 16, 2012     https://doi.org/10.3892/ijo.2012.1666
  • Pages: 1919-1931
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Insulin-like growth factor (IGF) signaling plays an important autocrine, paracrine and endocrine role in growth promotion involving various tissues and organs. Synthesis of both IGFs (IGF-1 and IGF-2) in normal conditions takes place mainly in the liver even if the proteins can be produced in every cell of the human body. The alterations in the IGF signaling axis in human hepatocarcinogenesis are described, but mechanisms of the interactions between expression of oncogenic hepatitis C virus (HCV) proteins and components of the IGF system in progression of chronic hepatitis C to primary hepatocellular carcinoma (HCC) have been poorly recognised. In advanced stages of liver diseases, lowered serum levels of IGF-1 and IGF-2 have been documented. This was supposed to reflect significant damage to liver parenchyma, a decreased number of growth hormone receptors and a decreased genomic expression of IGF binding proteins (IGF BPs). In HCC, a decreased tissue expression of IGF-1, and an increased expression of IGF-1 receptor (IGF-1R) were noted, compared to the control. Potential mechanisms of augmented IGF-2 expression in HCC were also described and dysregulation of IGF signaling in HCC was concluded to occur predominantly at the level of IGF-2 bioavailability. The review aimed at presentation of involvement of IGF-1, IGF-1R and IGF BPs (mostly IGF BP-3 and IGF BP-6) in HCV-related hepatocarcinogenesis. Manifestation of various mRNA transcripts and IGF-1 proteins and their potential involvement in carcinogenesis are also discussed.

Contents

Introduction

Components of insulin-like growth factor (IGF) signaling axis

IGF system and hepatocellular carcinoma

Tissue expression of IGF axis components in HCC

Role of IGF axis in HCV-related hepatocellular carcinoma

IGF axis components and HCV genome

Conclusions

Introduction

In HCV-related hepatocarcinogenesis, participation of viral proteins: core (C), non-structural 3 (NS3) and NS5 proteins themselves used to be accentuated (13). Several investigations point to a relationship between subcellular localisation, concentration, specific molecular form of the proteins and presence of specific domains and oncogenesis (47). HCV proteins were found to be involved in control of cell cycle, through their interaction with such cell cycle control proteins as p53, p21, cyclins, proliferating cell nuclear antigen (PCNA), transcription factors (mainly nuclear factor κB, NF-κB), proto-oncogenes (c-fos, c-jun), growth factors/cytokines, e.g. tumour necrosis factor (TNF)-α, transforming growth factor (TGF)-β and their receptors and proteins of apoptosis (813). A possible interactions between oncogenic HCV proteins and components of IGF axis in human HCC continue to be discussed (14).

Components of insulin-like growth factor (IGF) signaling axis

Two key regulatory proteins of this axis are known: IGF-1 and IGF-2, which manifest ∼50% sequence identical to that of insulin (15). They are included in the insulin-related family together with relaxin while genes of the family members were located on distinct genomic fragments (chromosome 2 and 11p-q13) (16). To date, 6 types of the IGF BPs have been well characterised (IGF BP1–6), plus two subsequent ones, less well recognised (IGF BP-7 and 8) (1719). The basic functions of IGF BPs include modulation of IGF-1 and IGF-2 bioactivity, mainly through interactions with receptors of the factors and with insulin receptor (IR). Moreover, IGF BPs extend half-life of IGFs in blood, store them in selected tissue compartments and inhibit activity of IGFs by lowering accessibility of their receptors. They may act independently of the receptors, inducing mitogenesis and cell migration (19). They participate in the interactions with other growth factors (e.g. TGF-β) (18). The most commonly manifested circulating form of IGF BP, is IGF BP-3, which binds over 95% of IGFs. The protein as a dimer forms a complex with the acid-labile protein (ALS) subunit (18). IGF BP-2, -3 and -5 contain a nuclear localisation signal and may influence on activity of transcription. The IGF BP-3 itself may act also as an inhibitor of cell growth (20,21). Free IGF-1 has a half-life of ∼8 min in serum. This can be increased to ∼30 min if bound to IGF BP-3 and to ∼15 h in the ternary complex with IGF BP-3 and ALS (22).

IGFs affect a cell specifically binding three various surface receptors: IGF-1R, IGF-2R and IR. In most of activities of IGF-1 and IGF-2, IGF-1R plays the main role of a mediator. Apart from mediating in mitogenic and anti-apoptotic activities of IGFs, the receptor is engaged in cell transformation (23) and, therefore, this review accentuates first of all the role of IGF-1R.

IGF-1

Gene of IGF-1 manifests a single copy in human genome located at the long arm of chromosome 12 (12q22–24.1), and contains 6 exons and 5 introns (2426) (Fig. 1). The six exons are alternatively spliced into multiple transcripts, encoding specific circulating and tissue-specific isoforms of the IGF-1 peptide (27,28). At the 5′ end of the gene, different promoters in combination with alternative transcription start sites and differential splicing generate the mutually exclusive Class I (1) and Class II (2) IGF-1 isoforms (29,30) (Fig. 1). At the 3′ end of the gene, alternative splicing gives rise to at least three subsets of RNA transcripts, each encoding three distinct carboxy-terminal portions of the unique E-domain extension-peptide (E-peptide) as well as the 3′-untranslated region (3′UTR) (17,25,3136). Exon 3 encodes parts of the signal peptide and the mature peptide common to all isoforms, while exon 4 encodes the rest of the mature peptide and the proximal part of the E domain. Even if according to some authors at least nine IGF-1 isoforms can be transcribed (35,37), the six main IGF-1 transcripts isoform in mice and men with apppropriate nomenclature are present (Fig. 1). Expression and composition of nucleotides in exons 5 and 6 determine formation of isoform variants: A (Ea), B (Eb) and C (Ec) within classes I and II (27). The predominant transcript IGF-1Ea has an exon 4 spliced directly to exon 6. Inclusion of exon 5 results in two transcript variants in humans: (1) IGF-1Ec (IGF-Ib in rodents) (mechano growth factor, MGF), and (2) IGF-1Eb (26,27,3740). The variant designated as IGF-1Eb was described only in humans and it contains mRNA with exon 5 spliced to exon 4 (41).

IGF-1 mRNA isoforms yield, as a result, also various isoforms of IGF-1 protein (27,28). A post-translational modification may further influence the biological activity of IGF-1 (42). The mature molecule of IGF-1 is coded only by exons 3 and 4. In circulation, the prevailing IGF-1 peptide variant involves IGF-1Ea (35). IGF-1 transcript and protein isoforms play various functions, depending on tissue involved, e.g. in skeletal muscle many isoforms of IGF-1 play critical role in development, growth and repair of the tissue (28,43). IGF-1Eb mRNA correlates with markers of muscle satellite cells and myoblast proliferation, whereas upregulation of IGF-1Ea mRNA is correlated with differentiation to mature myofibers (28). The exon 1-encoded signal peptide seemed to have stronger secretory properties than the exon 2-encoded peptide (44).

In mammals, transcription of IGF-1 gene remains under control of two promoters (P1 and P2) (26,45). The variety of IGF-1 isoforms stems from the use of alternative promoters combined with an elaborate pattern of alternative splicing of the primary transcript (35). In mammals, promoter P1 initiates over 90% transcripts (17). It is thought that P2 promoter encoding the endocrine IGF-1 form, remains under control of growth hormone (GH) (46). Both P1 and P2 promoters regulate transcription over scattered start sites, and it is suggested that heterogeneous transcription initiation compensates for the lack of typical proximal promoter elements, such as TATA- or CCAAT-boxes (47).

The nucleotide sequences of cDNAs predicting two different IGF-1 protein precursors and defining the size of these peptides (153 and 195 amino acids) were described for the first time by Rotwein (34). In liver cells IGF-1 isoform C (comprises 10% of IGF-1A isoform transcript) was also detected. Its presence was confirmed also in cultured HepG2 cells, in which its expression was increased following supplementation of GH. The transcript codes for the prepro-IGF-1 of 158 amino acids (aa), with an E-peptide sequence of 24 aa (33).

Within controlling sequences of both IGF-I gene promoters and their 5′UTR sequences several sites were noted for a potential specific binding of transcription factors, which may control its expression, including CCAAT/enhancer binding protein (C/EBP) α and β and hepatocyte nuclear factor-α (HNF-α) (48,49).

A distinct role is suggested for various IGF-1 splicing profiles as prognostic biomarkers in human cancer development (5052). Mature IGF-1 and IGF-2 peptides, similarly to proinsulin, consist of domains B, C, and A (amino-terminal end) and an additional domain D at the carboxy-terminal end, absent from proinsulin (15).

IGF-1 is a secretory protein of 7649 Da molecular mass, consisting of a single polypeptide chain with 70 aa, exerting a variable effect on cells and tissues (17). In postnatal period liver remains to be the main source of circulating IGF-1 and the protein is produced mainly under effect of GH. Secretion of GH is affected also by age, gender, diet and nutrition, insulin and sex hormones (17). Studies on RNA level demonstrated that in adipose tissue amounts of the IGF-1 transcript are equal to those in liver (17). IGF-1 produced in liver exerts mainly endocrine activity while IGF-1 synthetised by other tissues acts in a para- and/or autocrine way. Interestingly, even if normal liver represents the organ with the highest expression of IGF-1, it contains almost undetectable levels of IGF-1R mRNA. In contrast to hepatocytes, presence of IGF-1R was proven on Kupffer cells, myofibroblasts and hepatic stellate cells (20).

Serum concentration of IGF-1 changes with patient’s age and is dependent on gender. In childhood it grows systematically, most rapidly before and during pubescence, when it reaches the highest levels (53). The gender-related difference in IGF-1 concentration (of ∼20 μg/l) appears already in the first three years of age and it is most pronounced (in girls higher by ∼70 μg/l) at the age of ∼11–13 years. Following 20th year of age a reverse change takes place and higher IGF-1 concentrations are noted in men and the mean inter-gender difference is 6–26 μg/l (54). Other investigators detected a gradually decreasing with age concentrations of IGF-1, independent of gender (5557). Following a marked decrease in IGF-1 concentration following 25th year of life, a systematic slow reduction in the level to 80th year of age is noted (53). Free IGF-1 accounted for ∼1% of the total IGF-1 and its variation with age was similar to total IGF-1 (58).

Independently of GH level, serum concentration of IGF-1 is affected by nutritional status. In childhood and during pubescence no correlation was noted between IGF-1 concentration and body mass index (BMI) (53). In persons with BMI <21 and BMI >29 decreased levels of IGF-1 are noted (59). In cases of obesity it achieves decreasingly lower values. Ther highest standard deviation score (SDS) values for IGF-1 were noted in women with BMI of 27.5 to 30, and in men with BMI ranging between 22.5 and 25 (60). In men of 40–75 years of age also race-dependent differences were noted in serum concentration of IGF-1. Caucasians had the highest median IGF-1 level (224 ng/ml), followed by Asian (208 ng/ml) and African Americans (205 ng/ml) (61).

In women an inverse correlation was documented between IGF-1 and insulin levels (62). Low concentrations of IGF-1 were proven to be strictly linked to growing risk of glucose intolerance and development of type 2 diabetes mellitus (DM) (63). In DM IGF-1 levels decreases (64) and manifests an inverse correlation with concentration of glycosylated haemoglobin (HbA1c) (65). Insulin therapy of DM decreases portal concentrations of insulin. The decreased concentrations of insulin may lead to an insufficient production of IGF-1 in liver. In patients with DM concentration of IGF BP-1 increases also (65,66). In rats with induced diabetes, reductions in circulating IGF-1 levels and hepatic IGF-1 mRNA were noted as well as a significant increase (>400%) in liver IGF BP-1 mRNA (67). IGF-1 stimulates glucose uptake by peripheral tissues. This insulin-like effect may develop with mediation of IGF-1R or IR (68).

Concentrations of IGF-1 and IGF BP-3 are stable during a day even if the main factor which stimulates production and secretion of the two proteins is GH. In children and youth concentrations of IGF-1 and IGF BP-3 well correlate with 24 h output of GH and reflect spontaneous secretion of GH in healthy individuals. Levels of IGF-1 are more sensitive to GH control than levels of IGF BP-3 (69).

IGF-2

Human gene of IGF-2 is located on chromosome 11 (11p15.5) and it consists of 9 exons. Control of the expression takes place from four promoters (P1-P4). Activity of IGF-2 gene is controlled by genomic imprinting (70). IGF-2 protein consists of 67 aa and manifests 65% of homology with IGF-1. Similarly to IGF-1 it exerts a mitogenic effect on various cells. A markedly higher expression of the protein is detected in embryonal life (71), in which it significantly modulates growth and differentiation of muscles (70). It exerts its effect on cells due to binding with, first of all, IGF-1R. It binds also to the cation-independent IGF-2 mannose-6-phosphate receptor (IGF-2R/MPR) (72,73). IGF-2 binds with high affinity also to isoform A of IR (IR-A), amplifying mainly the mitogenic effect. More numerous IR-A were demonstrated in fetal tissues and certain cancers which indirectly provides evidence for a significant involvement of IGF-2 in embryogenesis and carcinogenesis (73). Normal serum concentration of IGF-2 in adult individuals amounts to ∼700 ng/ml. In contrast to IGF-1, concentrations of IGF-2 manifest no significant alterations during maturation. After birth they are low, increase in parallel to age until pubescence and, then, remain unaltered till the end of life. Gender-related differences are small (2%) (58).

IGF-1 receptor, type I IGF receptor (IGF-1R)

Gene of IGF-1R consists of 21 exons and is located on chromosome 15q25–26, synthetised as a single-chain preproprotein, composed of 1367 aa, from which a signal peptide of 30 aa is excised. The remaining part of the molecule undergoes glycosylation and fragments to two subunits, α and β, joined by disulphide bridges. The receptor manifests the most pronounced affinity to IGF-1 and a two-fold lower one to IGF-2 (17,68,74). Product of IGF-1R gene undergoes alternative splicing with formation of two different mRNAs, differing in only three nucleotides (CAG). Using the model of transfected hamsters the receptor devoid of the three nucleotides (CAG) was found to stimulate about two-fold more effectively IGF-1 in its mitogenic activity. Higher expression of either isoform of the transcript was documented in various tumours. Downregulation of the mature receptor resulted in massive apoptosis of neoplastic cells (75). Removal of the IGF-1R coding gene or inhibition of its expression eliminated cell transformation in vitro (23).

IGF-1 binds with IGF-1R to stimulate cellular proliferation or inhibit apoptosis through different pathways, increasing the risk of carcinogenesis (7678) (Fig. 2). IGF-1R/IGF axis may positively control cell cycle progression in many phases, but the major direct effect is probably exerted at the G1-S interface, and this is mediated through the phosphatidylinositol 3-kinase/serine/threonine-specific protein kinase (PI-3K/Akt) and/or extracellular signal-regulated kinase (ERK) pathways (79). IGF-1 exerts a mitogenic effect influencing stimulation of cyclin D1 and of some proto-oncogenes (c-FOS, c-JUN) expression. Moreover, it acts anti-apoptotically and modulates body immune response by control of cytokine production (e.g. IL-3 and IL-14) (68). It was also demonstrated that short peptides of IGF-1 precursors may promote growth of normal and malignant cells in bronchial epithelium (17). IGF-1 controls expression of over 50 genes linked to mitogenesis and cell differentiation. However, it exerts mitogenic activity mainly through stimulation of DNA synthesis and of cyclin D1 expression (68).

IGF-2 receptor, type II IGF receptor (IGF-2R)

The human IGF-2R-coding gene is situated on chromosome 6q25–q27 (80). Extracellular domain consists of 15 segments, each containing from 134 to 191 aa and a small region homologous to the collagen-binding domain of fibronectin (81). Mouse gene of 93 kb in length, includes 48 exons and codes for a protein with 2482 aa (72). The receptor itself is identical with cation-independent IGF-2/MPR (72,81). IGF-2R binds IGF-2 ∼500 higher than IGF-1 (82). IGF-2R is a multifunctional protein and plays a principal role in sorting and transport of lysosomal enzymes from sites of their synthesis to appropriate cellular compartments and in control of proteins containing mannose-6-phosphate. The receptor is of potential significance for maturation and clearance of growth factors and it mediates IGF-2-activated signal transduction through a G-protein-coupled mechanism (72,81). In studies on mice IGF-2R was demonstrated to be paternally imprinted, and to be crucial for regulating normal fetal growth, circulating IGF-2, and heart development (82).

IGF system and hepatocellular carcinoma

Hepatocellluar carcinoma is the third leading cause of cancer-related death worldwide (14,84). The suggested roles of both IGFs and IGF-1R in development of HCC reflect, first of all, observations indicating strong mitogenic effects of the factors in in vitro conditions (21,85,86). Different types of cultured cells produce IGF-1 mRNA (87) and insulin receptor substrate 1 (IRS-1) (86). IRS-1 undergoes overexpression in human HCC (88,89). The dominating pathways activated by IGF-1 in hepatocytes and hepatoma cell lines involve the PI3K/Akt (90,91) and signal transducer and activator family protein (STAT) signaling pathways (92). IGF-1 has been implicated in NF-κB-mediated transcriptional regulation of inflammatory cytokines and endothelial cell adhesions receptors such as intracellular adhesion molecule-1 (ICAM-1) (93). Studies on HepG2 and HuH-7 cells demonstrated that the cells synthetised and secreted also IGF-2 and inhibition of the protein production resulted in a reduced cellular proliferation (87). Recent studies on HCV-related HCC demonstrated overexpression of IGF-2 gene (resulting from reactivation of fetal promoters P3 and P4), IGF BP3 down-regulation, decreased IGF-1 expression and allelic losses of IGF-2R. Administration of IGF-1R-selective inhibitors (A12) reduced IGF-1-induced effects and was associated with a significant reduction of liver tumour growth (84) (Fig. 3).

Figure 3

Schematic representation of the GH-IGF axis in normal physiology (A) and the proposed model for how it is affected by HCV-associated liver injury (B). (A) In postnatal period liver remains to be the main source of circulating IGF-1 and the protein is produced mainly under effect of growth hormone (GH) and its receptor (GHR) on liver cells (17). IGF-1 produced in liver exerts mainly endocrine activity while IGF-1 synthesised by other tissues acts in a para- and/or autocrine way (20). IGFs affect a cell specifically binding three various surface receptors: IGF-1R, IGF-2R and IR. In most of activities of IGF-1 and IGF-2, IGF-1R plays the main role of a mediator (23). (B) In HCV-infected patients, GH insufficiency (GH*) and persistent GH resistance (GHR*) of hepatocytes and low IGF-1 levels were documented (139). Other studies demonstrated an increased IGF-1R mRNA and a decreased IGF-1 protein synthesis in patients with chronic hepatitis C (144). HCV replication was associated with overexpression of IGF-2 in the cirrhotic livers (145). Studies on HCV-related HCCs documented a decreased IGF-1 expression, an increased expression of IGF-2 in 20–50% of HCCs (84,85,146), IGF BP3 downregulation and allelic losses of IGF 2R (84). The increased synthesis of IGF-2 (mRNA and protein) was associated with an increased cellular proliferation in HCCs (146). Compared with a non-cirrhotic liver, all cirrhotic specimens showed reduced expression of IGF-2R/M6PR protein. It was suggested that downregulation of hepatocellular IGF-2R/M6PR and upregulation of IGF-2 might be early events in hepatocarcinogenesis (131). HCV core protein was shown to increase endogenous expression of IGF-2 in HepG2 cell line (149).

IGF-1

A decreased serum levels of both IGFs were found to parallel progression of liver diseases, independently of their etiology (94,95). Besides, patients with HCC and hypoglycaemia were found to carry higher IGF-2 concentrations than patients without hypoglycaemia. The higher IGF-2 than those of IGF-1 levels were accompanied by more advanced histological lesions in the liver (94). Kratzsch et al demonstrated a continuous decline in the concentrations of IGF-1, IGF BP-3 and serum GH-binding activity during progression of cirrhosis (95). Other investigations documented decreased levels of both IGFs in chronic liver diseases but the positive correlation with Child score was confirmed only for IGF-2 (96). It should be added that only in a small number of studied patients (17/55) viral etiology of chronic lesions in liver was confirmed (96). In chronic hepatitis B and C were also demonstrated elevated levels of IGF-1 and lowered levels of IGF BP-3 (97). Studies on serum IGF-1 concentrations in HCC and in metastatic liver cancers demonstrated markedly lowered concentrations of IGF-1 in either type of hepatic tumour, as compared to the control (98). The decrease in IGF-1 was more pronounced in cases with viral-associated than in virus-negative HCCs (98). In liver cirrhosis lower IGF-1 concentrations were observed in comparison with control (99,100), as well as its increased concentration following anti-viral therapy (100). In HCV-infected children serum IGF-1 level was significantly lower compared with the control group (101). In adult patients with chronic hepatitis C the reduction of serum IGF-1 level was found to precede by ∼9 months development of HCC (102).

IGF-2

Similarly to IGF-1, concentration of IGF-2 also markedly decreases in patients with liver cirrhosis, as compared to healthy individuals. Advancement of liver cirrhosis, is paralleled by gradual decreases in IGF-2 levels. The lowest concentrations of the factor are encountered in patients with incurable ascites and extended activated partial tromboplastin time (APTT). During half-a-year observation of the patients, those with very low concentrations of IGF-2 (<200 ng/ml) died manifesting hepatic insufficiency (103). A successful liver transplantation was followed by a significant increase in IGF-2 concentration in almost all of the patients (104). In patients with HCC levels of IGF-2 decrease (but the difference was not statistically significant) and thus the protein cannot serve as a diagnostic marker of the tumour (105). The amount of IGF-2 mRNA was also lowered in patients with liver cirrhosis as compared to the control (106). The studies on rats with experimentally induced liver cancer, demonstrated a decreased expression of IGF-1 mRNA, and an increased expression of IGF-2 mRNA (107). Start of IGF-2 (fetal type of IGF expression) production in HCC is supposed to represent a late phenomenon in carcinogenesis of rats (107). Regarding human HCC activation of IGF axis (e.g. IGF-2 overexpression, IGF-1R activation) was observed in 21% of early liver tumours (83).

Recently, among 102 patients with HCC and 306 control patients, Weng et al detected a significant difference in manifestation of IGF-2 +3580 gene polymorphism (85). Moreover, they noted that the combination of IGF-2 +3580 AA and IGF-2R GG genotype may present a significantly lower risk for HCC. The authors concluded that the most significant IGF system-linked factors in development of HCC involved polymorphisms of IGF-2 and IGF-2R and their combinations (85). Hypomethylation of IGF-2 gene in exon 8–9 was present in 90% HCV-infected patients with liver cirrhosis, who would develop HCC within 10 years. The test could be used as a screening tool in diagnosis of HCC, since frequency of HCC in patients with hypomethylation of the gene is markedly higher than in patients with hypermethylation in the region (30.8% vs. 5.9%) (108).

IGF-1R

Role of IGFs receptors in liver carcinogenesis remains to be fully recognised. On one hand there exist papers pointing to an increased expression of IGF-1R in preneoplastic focal lesions in liver, leading to HCC, in HCC itself and in human hepatoma cells (20), on the other attempts to stimulate mitogenesis with IGF-1 in cultured rat cell lines of HCC proved to be unsuccessful (109). Hepatitis B virus (HBV) and HCV are known to be capable of activating promoters of IGF-2 and IGF-2R genes, thereby increasing the production of this ligand and receptor by liver cancer cells (110112). An increase in IGF-1R mRNA in liver cirrhosis has been demonstrated, but no change in the amount of IGF-2R mRNA was detected, as compared to the control (106). Activation of IGF-1R was significantly associated with activation of IGF-2 mRNA levels in ∼20% of HCCs (84). Other authors observed decreased levels of IGF-1R in 39% patients with cirrhosis, as compared to the control (21).

IGF BPs

Studies on IGF BPs in hepatocarcinogenesis were conducted on animal models (113), in normal liver (114), in patients with chronic hepatitis, cirrhosis and HCC (21,115118). In the studies, viral background of HCC could not be repeatedly demonstrated.

IGF-1, IGF BP-3 and ALS showed a similar pattern of change associated with age (58). Normal levels of the molecules were low upon birth and increased with age through puberty, with a slow age-dependent decrease thereafter. Slightly higher levels of the three compounds were present in women. Levels of IGF BP-6 increased with age and reached higher levels in men as compared to women (58). Healthy liver produces mainly IGF BP-1 and BP-3. IGF BP-1 (mRNA and protein) were demonstrated in hepatocytes while IGF BP-3 was detected mainly in Kupffer cells (114). IGF BP-3 mRNA was demonstrated in portal venous and sinusoidal endothelium (113).

Decreased levels of IGF BP-3 were shown in patients with cirrhotic liver, as compared to the control (115). The lowered serum concentration of IGF BP-3 was accompanied by absence of protease activity in cirrhotic patients (115,119). It should be noted that concentration of IGF BP-3 is also dependent on age (120,121). Serum concentrations increase with progressing age to reach maximum levels at pubescence. Comparison of IGF BP-3 and IGF-1 concentrations revealed that they did not exibit the same developmental pattern at puberty. IGF-1 levels increased to relatively higher levels than IGF BP-3 (120). Median IGF BP-3 concentration was similar between Caucasian and Asian middle-aged men but was more than 13% lower in African Americans. Median molar IGF-1:IGF BP-3 ratio was greatest in Caucasian and lowest in Asians (61). IGF BP-3 levels manifested no differences before and after menopause (122). Also, concentration of IGF BP-3 failed to correlate with BMI (117). A relationship was detected between lower levels of serum IGF-1, decreased IGF-1/IGF BP-3 ratio, higher serum concentrations of IGF BP-3 on one hand and liver steatosis on the other (116).

IGF BP-6 level significantly increases with age beginning at birth up to pubescence. In most age groups adult men manifested higher concentrations of IGF BP-6 than those noted in women (58).

In liver cirrhosis significantly lower serum level of IGF BP-3 is noted, as compared to healthy individuals (103). Also, it becomes reduced with an advancement in cirrhosis. Therefore, concentration of this protein provides a real potential index of progression manifested by hepatic lesions (103,117). A positive correlation was detected between concentration of IGF BP-3 and of albumin as well as a negative correlation with bilirubin concentration, size of spleen and activity of aspartate aminotransferase (AST) (117,121). Concentration of IGF BP-3 return to normal level following a successful liver transplantation (104). In patients with HCC concentrations of IGF BP-3 are decreased in cases with a disturbed nutrition, markedly deteriorated liver function and reduced secretion of GH (105). An increase in IGF-1/IGF BP-3 ratio in patients with HCC was also demonstrated as compared to patients with cirrhotic liver and a similar extent of liver insufficiency (118). In >70% HCC patients, IGF BP-3 expression was lowered as compared to normal liver. A less pronounced tissue expression of the protein was observed in tumour cells as compared to control (21). Decreased IGF BP-3 mRNA levels correlated with smaller tumour size, less vascular invasion, and a lower incidence of early recurrence (84).

Analysis of gene expression using cDNA microarrays performed in 20 primary liver tumours demonstrated 170 genes with a decreased regulation in HCC, including IGF BP-3 and ALS (123). In turn, application of 75 antibodies to evaluate markers for early detection of viral-associated HCC, identified 7 proteins which significantly differentiated patients with HCC and those with chronic hepatitis, including IGF BP-6 (83).

A lowered serum concentration of IGF BP-3 was noted also in patients with variably advanced chronic hepatitis (including 12 HCV-positive patients). No correlation could be disclosed between grading/staging on one hand and activity of transaminases on the other (97). Other investigators failed to detect significant differences between concentration of IGF BP-3 in patients with liver cirrhosis which would be related to its aetiology (alcohol vs. HBV vs. HCV (117). Other studies demonstrated a significantly decreased concentration of IGF BP-3 in patients with chronic hepatitis C as compared to healthy individuals (121).

Tissue expression of IGF axis components in HCC

In all studied tissues of HCC (n=28) a decreased expression of IGF-1 and IGF BP-3 and in 32% of them an increased expression of IGF-2 was detected (21). Decreased amounts of IGF-1 mRNA (84) or both IGF’s mRNA (124126) in HCCs were demonstrated as compared to the control. In case of IGF-1 transcript, the decrease was more pronounced than that in IGF-2 (124126). In hepatocarcinogenesis in rats appearance of IGF-2 mRNA was demonstrated upon a decrease in IGF-1 mRNA (107). An increased expression of IGF-2 (protein and mRNA) was demonstrated in HBV-related HCC (127). IGF-1, IGF-2 and IR mRNAs were detected at various stages of HCC development and in cultured cells. The studies showed that isolated hepatocytes synthetise IGF-2 mRNA with a switch between fetal and adult mRNA profiles occurring 21 days after birth (128).

Other studies demonstrated presence of IGF-2 in 60% HCC arising in HBV-associated cirrhosis, as compared to only 26% tumours in HBV-negative patients (129). Studies employing DNA microarrays showed that imbalances in levels of IGF-2 and H19 transcripts were correlated with advanced tumour stage and poor outcome in HCC patients (130). High focal levels of IGF-2 mRNA were found in some hepatocytes of all HBV and HCV-related cirrhotic livers (131). In subgroups of HCV-related HCC an increased tissue expression of IGF-2 (protein, mRNA) was noted (20,83,111). Compared with noncirrhotic liver, all cirrhotic specimens showed reduced expression of IGF-2R/M6PR protein. It was suggested that downregulation of hepatocellular IGF-2R/M6PR and upregulation of IGF-2 might be early events in hepatocarcinogenesis (131). Tovar et al observed losses in the IGF2R locus in ∼25% of HCCs (84).

The increased expression of IGF-1R was detected in cirrhosis, in HCC and in human hepatoma cells, as compared to normal liver (84,86,87,132,133). More pronounced membraneous location of IGF-1R was documented in HepG2 cells, as compared to Chang liver cell lines (134). Overexpression of IGF-1R in the cell lines was supposed to indicate a malignant phenotype of the cells. Administration of anti-IGF-1R monoclonal antibody (αIR3) inhibited cell proliferation and induced apoptosis in HepG2 cells (134). It has been proven that mutations resulting in upregulation of IGF-1R gene in certain HCC include p53mt249 (135). The same mutation enhances transcription from fetal IGF-2 promoter P4 (136). Recent studies have drawn attention to the role of polymorphism in IGF-1 gene promoter in carcinogenesis. One of IGF-1 gene polymorphisms recognised in greatest detail seems to involve the sequence consisting of several CA repeats, present in 5′UTR of IGF-1 gene (41). The mutations of IGF-2R/M6PR gene give rise to truncated receptor protein and significant aa substitutions, and provide evidence that this gene functions as a tumour suppressor in human liver carcinogenesis (137).

Role of IGF axis in HCV-related hepatocellular carcinoma

Serum concentrations of IGF-1 were significantly lower in HCV-associated HCC than in healthy subjects. Moreover, the lower levels of IGF-1 were detected in all patients below 55th year of age and with homeostasis model assessment of insulin resistance (HOMA-IR) <2.53 as compared to the control (138). Moreover in >80% of HCV-infected patients, severe GH insufficiency was documented and about half of these patients had low IGF-1 level. Basal and stimulated GH concentration increased significantly during therapy, but IGF-1 remained low (139).

IGF-1 is known to regulate, coordinatively with insulin, glucose homeostasis (106). Within nondiabetic HCV patients, IGF-1 serum levels correlated negatively with β-cell function (HOMA-β), and positively with insulin-sensitivity (HOMA-S) (140). In parallel, a negative correlation has been detected between IGF-1 serum levels and both alanine aminotransferase (ALT) and AST (140,141). In HCV patients with DM a positive correlation was observed between fasting insulin levels and HOMA-β (140). Such results suggest (but with caution) that low IGF-1 level might play a role in the development of insulin resistance in HCV viremic patients (140). This would be consistent with observations that IGF-1 may be an early marker of functional reserve or hepatocellular capacity (117,142). Hung et al detected DM more frequently in patients with HCV infection than those with HBV infection. Also in HCV-related HCC, blood glucose, insulin level and HOMA-IR were higher than in patients with chronic hepatitis and advanced fibrosis. The authors concluded that insulin resistance (regardless of the presence of diabetes) is significantly associated with HCC development in patients with chronic HCV infection (143).

Only few investigations pertained hepatic expression of IGFs and their receptors at different stages of chronic hepatitis C in humans (144). The studies demonstrated an increased IGF-1R mRNA, but not IGF-1 protein in patients with CHC as compared to the control (144). HCV replication was associated with the overexpression of IGF-2 in the cirrhotic livers (145). Studies on HCV-related HCCs documented increased expression of IGF-2 in 50% of HCCs. The increased synthesis of IGF-2 (mRNA and protein) was associated with an increased cellular proliferation in HCCs (146). The results observed in tissue samples suggest that IGF-2 might be responsible for IGF-1R activation (83).

Data from examination of serum and tissue levels involving IGF axis components in the patients with advanced liver diseases (including HCV-associated HCC) are summarized in Tables I and II.

Table I

Expression levels of IGF axis components in human hepatocellular carcinoma (including HCV-related HCC).

Table I

Expression levels of IGF axis components in human hepatocellular carcinoma (including HCV-related HCC).

Type of tumourEtiology/patients (n)Method of detectionSummary of the findingsReferences
HCC38Northern blotIGF-2 mRNA was increased 40–100 fold in 9 of 40 liver cancer surgical specimensCariani et al, 1988 (126)
HCC25RIALower levels of IGF-1 and more advanced liver cancer in HCC patients; relatively higher IGF-2 levels in comparison with those who did not have hypoglycaemiaWu et al, 1988 (94)
HepatomaNorthern blotLow IGF-1 mRNA expression in 7/7 specimens, low IGF-2 expression in 4/7 specimensSu et al, 1989 (124)
HCC HBV+ICC and ISHIGF-2 mRNA was increased in HCC specimensLamas et al, 1991 (127)
HCCHBV+, HBV, 54ICCDetection of IGF-2 expression in 9/15 HBV-positive and in 10/39 HBV-negative patientsD’Erico et al, 1994 (129)
HCCHCV+, 7RT-PCRUndetectable expression of transcripts derived from adult IGF-2 P1 promoter; increased expression of fetal promoter-derived transcripts, but lower than in cirrhosisNardone et al, 1996 (111)
HCCHCV+, 35ICCPositive immunoreaction of IGF-2 in 69% of HCC; IGF-2 was positive in 80% of HCC with fatty change but only in 60% of those without steatosisSohda et al, 1997 (112)
HCCHCV+, 7; HBV+, 3ICC, ISHIncreased expression of IGF-2 mRNA and protein in 5/10 HCCsSohda et al, 1997 (146)
HCCHBV+, 25; HBV, 5Northern blotNormal adult promoter of IGF-2 was repressed in all but two of HCC; re-expression of two fetal transcripts in 12 tumours; significant association of IGF-2 with direct tumour invasionNg et al, 1998 (125)
HCCHCV+, 10; HBV+, 10cDNA microarrays (23 040 genes)Upregulation of mitosis-promoting genes (e.g. cyclin G1, MAPK1, MAPK3), tumor-associated genes (CD34, glypican 3, recoverin, matrix metalloproteinase 11), increased expression of genes encoding for 4 enzymes (CYP2E, AKR1C4, EPHX1 and FMo3) exclusively in HCV-positive HCCOkabe et al, 2001 (123)
HCC28ICC, Western blotDecreased expression of IGF-1 in 100% HCCs; low levels of IGF BP3 and type I receptor (IGF-1R) in 39% of HCCs; overexpression of IGF-2 in 32%Huynh et al, 2002 (21)
HCCDNA microarrayIncreased IGF-2 expression was correlated with advanced tumor stage and poor prognosisIizuka et al, 2004 (130)
HCCHCV+, 10; HBV+, 26; B/C +, 1; None/unknown, 11Antibody array platform and immunoassayIdentification of 7 proteins that significantly differentiate HCC patients from hepatitis patients (e.g. IGF BP-6); among the 75 unique probes, three: AFP, β-catenin, CSF1 were upregulated in HCC; L-selectin, IGF BP-6, IL-6R and VCAM-1 were very slightly downregulated in HCC vs. hepatitisSun et al, 2008 (83)
HCCHCV+, 104RT PCR; SNP array, oligonucleotide microarray; cell viability, proliferation and death assays and miRNA transfection; Western blot; ICCOverexpression of IGF-2 (due to reactivation of fetal promoters P3 and P4) in 21% of early HCCs, IGFBP3 downregulation and allelic losses of IGF2R (25% of cases); IGF-1 mRNA expression levels were significantly decreased among HCC samplesTovar et al, 2010 (84)
HCC65RIADecreased serum IGF-1 level in HCCs (especially HCV-infected patients) as compared with healthy subjectsSu et al, 2010 (138)
HCC63ICCDecreased IGF-1 tissue expression in comparison with control livers; no correlations between hepatic expression of IGF-1 in HCC and histologic malignancyKasprzak et al, 2011 (150)

[i] RIA, radioimmunoassay; ICC, immunocytochemistry; ISH, in situ hybridization; RT-PCR, reverse transcription polymerase chain reaction; MAPK, p38 mitogen-activated protein kinase; AFP, α-fetoprotein; CSF, colony stimulating factor; IL, interleukin; VCAM, vascular cell adhesion molecule.

Table II

Expression levels of IGF axis components in various stages of chronic liver diseases (including chronic hepatitis C).

Table II

Expression levels of IGF axis components in various stages of chronic liver diseases (including chronic hepatitis C).

Liver pathologyEtiology/patients (n)Method of detectionSummary of the findingsReferences
Chronic hepatitis Cirrhosis15
15
RIABoth IGF-1 and IGF-2 levels decrease as liver disease progressedWu et al, 1988 (94)
CirrhosisImmunoassayA continuous decline in the concentrations of IGF-1, IGF BP-3 and GH-binding activityKratzsch et al, 1995 (95)
Active chronic hepatitis CirrhosisHCV+, 9; HCV+, 9RT-PCRIncreased expression of transcripts derived from adult IGF-2 P1 promoter; abundantly expressed transcripts from fetal P3 promoter in most CAH patients and all cirrhotic patients; transcripts from fetal P4 promoter detected at high levels in 3/9 CAH patients and in the majority of cirrhotic patientsNardone et al, 1996 (111)
CirrhosisHCV+, 31PCRHCV replication was significantly associated with the overexpression of TGF-α and IGF-2Tanaka et al, 1996 (145)
CirrhosisHCV+, 11; HBV+, 6; others, 21RIAIGF-1 and IGF-2 were lower in patients with cirrhosis than in control; only IGF-2 level was correlated with Child scoreNikolić et al, 2000 (96)
Chronic hepatitisHCV+, 12 HBV+, 5RIAIGF-1 levels were significantly higher than that of the control group; IGF BP-3 levels in chronic hepatitis C were significantly lower than in control groupOkan et al, 2000 (97)
CirrhosisHCV+, 114Immunoradiometric assaySignificant reduction in IGF-1 level preceded the diagnosis of HCC by 9.3±3.1 months independent of the grade of impairment of liver functionMazziotti et al, 2002 (102)
CirrhosisHCV+, 3 HBV+, 6 B/D coinfection, 1ICC, ISHHigh focal levels of IGF-2 RNA were found in some hepatocytes of all, livers HBV- or HCV-induced cirrhosis; all cirrhotic specimens showed reduced hepatocellular expression of M6P/IGF-2R proteinSedlaczek et al, 2003 (131)
Cirrhosisof viral origin, 22Immunoradiometric assaySerum IGF-1 levels were found very significantly lower than in healthy individualsVyzantiadis et al, 2003 (99)
CirrhosisHBV+, 44Immunoradiometric assaySerum levels of IGF-1, IGF-2 and IGF BP-3 were significantly lower than in controls and were associated with the severity of liver dysfunction; IGF-1<30 ng/ml; IGF-2<200 ng/ml and IGF BP-3<6 ng/ml implied a negative prognosis for patients with liver cirrhosisWu et al, 2004 (103)
Chronic hepatitis Cirrhosis
14
ICC, ISHPatients with chronic liver disease have a significant reduction in their hepatic production of IGF-1, whereas IGF-2 tends to be elevated (mRNA, protein)Morali et al, 2005 (106)
Chronic hepatitis before and after therapyHCV+, 34; after therapy, 10RT-PCR, ICCAn increase of IGF-IR mRNA content in all CHC patients in comparison with normal liver; no relevant modification in IGF-1 mRNA content; a decrease IGF-IR mRNA in patients who achieved sustained virological responseStefano et al, 2006 (144)
Chronic hepatitis before and after therapyHCV+, 40ImmunoassayMean IGF-1 values were significantly lower in patients with advanced fibrosis than in the others; serum levels of IGF-1 increased during combined therapyLorenzo-Zúñiga et al, 2007 (100)
Chronic hepatitisHCV+, 29; HBV+, 12; others, 9ELISASignificantly reduced serum levels of IGF-1 in all children with liver diseasesMahdy et al, 2007 (101)
Chronic hepatitisHBC+ or HBV+, 19ImmunoassayAmong top scoring 7 proteins that significantly differentiate HCC from chronic hepatitis was IGF BP-6 (higher levels in CHC than in HCC)Sun et al, 2008 (83)
Chronic hepatitisHCV+, 30ICCSignificantly lower expression of IGF-1 tissue expression in patients with CHC compared with control livers, but with no differences in comparison with HCC; positive correlations between IGF-1 and C and NS3 HCV proteins; negative correlations between IGF-1 and ALT and AST activitiesKasprzak et al, 2011 (150)

[i] RIA, radioimmunoassay; RT-PCR, reverse transcription polymerase chain reaction; CAH, chronic active hepatitis; CHC, chronic hepatitis C; ICC, immunocytochemistry; ISH, in situ hybridization.

IGF axis components and HCV genome

IGF-1 may play a role both in persistence of chronic hepatic inflammation through control of signaling pathways linked to proinflammatory cytokines and receptors for endothelial adhesion molecules (e.g. ICAM-1) (93), and in induction of acute inflammatory reaction, triggered by tumour cells during early stages of liver metastasis (79,147,148). In recent years increased attention is devoted also to complex interactions between HCV proteins and IGF axis. HCV core protein was shown to increase endogenous expression of IGF-2 in HepG2 cell line, regulating positively its transcription, and it may promote cell divisions (149).

Our own studies in chronic hepatitis C patients demonstrated positive correlation between tissue expression of IGF-1 and two HCV proteins: core and NS3 while studies on tissue material originating from HCC detected no significant correlations between tissue expression of IGF-1 and the histological malignancy of the tumour (150).

Conclusions

Several components of the IGF signaling axis, such as IGF-1, IGF-2 and IGF-1R, are dysregulated during HCV-related human HCC. Only few investigations pertained hepatic expression of IGFs and their receptors at different stages of chronic hepatitis C. The studies demonstrated an increased IGF-1R synthesis, the abberant IGF-2 expression (decreased/increased), and decreased synthesis of IGF-1 as an events in human hepatocarcinogenesis. Recognition of the role played by HCV in different splicing profiles of IGF-1 gene in progression of chronic hepatitis C will require further studies. Better understanding of the HCV protein and IGF axis component interactions will facilitate development of novel approaches to prognose and to treat the virus-related HCC.

Acknowledgements

The work was supported by a grant (no. NN401009437) from the Minister of Education and Science, Warsaw, Poland. The authors thank Dr Marek Konkol for assistance with the color figures.

References

1. 

Anzola M: Hepatocellular carcinoma: role of hepatitis B and hepatitis C viruses proteins in hepatocarcinogenesis. J Viral Hepat. 11:383–393. 2004. View Article : Google Scholar : PubMed/NCBI

2. 

Kasprzak A and Adamek A: Role of hepatitis C virus proteins (C, NS3, NS5A) in hepatic oncogenesis. Hepatol Res. 38:1–26. 2008. View Article : Google Scholar : PubMed/NCBI

3. 

Wang F, Yoshida I, Takamatsu M, Fujita T, Oka K and Hotta H: Complex formation between hepatitis C virus core protein and p21Waf1/Cip1/Sdi1. Biochem Biophys Res Commun. 273:479–484. 2000. View Article : Google Scholar : PubMed/NCBI

4. 

Chang SC, Yen JH, Kang HY, Jang MH and Chang MF: Nuclear localization signals in the core protein of hepatitis C virus. Biochem Biophys Res Commun. 205:1284–1290. 1994. View Article : Google Scholar : PubMed/NCBI

5. 

Suzuki R, Sakamoto S, Tsutsumi T, et al: Molecular determinants for subcellular localization of hepatitis C virus core protein. J Virol. 79:1271–1281. 2005. View Article : Google Scholar : PubMed/NCBI

6. 

Yamanaka T, Uchida M and Doi T: Innate form of HCV core protein plays an important role in the localization and the function of HCV core protein. Biochem Biophys Res Commun. 294:521–527. 2002. View Article : Google Scholar : PubMed/NCBI

7. 

Nielsen SU, Bassendine MF, Burt AD, Bevitt DJ and Toms GL: Characterization of the genome and structural proteins of hepatitis C virus resolved from infected human liver. J Gen Virol. 85:1497–1507. 2004. View Article : Google Scholar : PubMed/NCBI

8. 

Cho JW, Baek WK, Suh SI, Yang SH, Chang J, Sung YC and Suh MH: Hepatitis C virus core protein promotes cell proliferation through the upregulation of cyclin E expression levels. Liver. 21:137–142. 2001. View Article : Google Scholar : PubMed/NCBI

9. 

Jung EY, Lee MN, Yang HY, Yu D and Jang KL: The repressive activity of hepatitis C virus core protein on the transcription of p21(waf1) is regulated by protein kinase A-mediated phosphorylation. Virus Res. 79:109–115. 2001. View Article : Google Scholar : PubMed/NCBI

10. 

Kwun HJ, Jung EY, Ahn JY, Lee MN and Jang KL: p53-dependent transcriptional repression of p21 (waf1) by hepatitis C virus NS3. J Gen Virol. 82:2235–2241. 2001.PubMed/NCBI

11. 

Taniguchi H, Kato N, Otsuka M, Goto T, Yoshida H, Shiratori Y and Omata M: Hepatitis C virus core protein upregulates transforming growth factor-beta 1 transcription. J Med Virol. 72:52–59. 2004. View Article : Google Scholar : PubMed/NCBI

12. 

Sato Y, Kato J, Takimoto R, et al: Hepatitis C virus core protein promotes proliferation of human hepatoma cells through enhancement of transforming growth factor alpha expression via activation of nuclear factor-kappaB. Gut. 55:1801–1808. 2006. View Article : Google Scholar

13. 

Kasprzak A, Adamek A, Biczysko W, et al: Intracellular expression of the proliferative marker Ki-67 and viral proteins (NS3, NS5A and C) in chronic, long lasting hepatitis C virus (HCV) infection. Folia Histochem Cytobiol. 45:357–366. 2007.PubMed/NCBI

14. 

Breuhahn K, Longerich T and Schirmacher P: Dysregulation of growth factor signalling in human hepatocellular carcinoma. Oncogene. 25:3787–3800. 2006. View Article : Google Scholar : PubMed/NCBI

15. 

Daughday WH and Rotweien P: Insulin-like growth factors I and II. Peptide, messenger ribonucleic acid and gene structures, serum and tissue concentrations. Endocr Rev. 10:68–91. 1989. View Article : Google Scholar : PubMed/NCBI

16. 

Rotwein P, Naylor SL and Chirgwin JM: Human insulin-related DNA sequences map to chromosomes 2 and 11. Somat Cell Mol Genet. 12:625–631. 1986. View Article : Google Scholar : PubMed/NCBI

17. 

Zarilli R, Bruni CB and Riccio A: Multiple levels of control of insulin-like growth factor gene expression. Mol Cell Endocrinol. 101:R1–R14. 1994. View Article : Google Scholar : PubMed/NCBI

18. 

Murphy LJ: Insulin-like growth factor-binding proteins: functional diversity or redundancy? J Mol Endocrinol. 21:97–107. 1998. View Article : Google Scholar : PubMed/NCBI

19. 

Kostecka Z and Blahovec J: Insulin-like growth factor binding proteins and their functions (minireview). Endocr Regul. 33:90–94. 1999.PubMed/NCBI

20. 

Scharf JG, Dombrowski F and Ramadori G: The IGF axis and hepatocarcinogenesis. Mol Pathol. 54:138–144. 2001. View Article : Google Scholar : PubMed/NCBI

21. 

Huynh H, Chow PKH, Ooi LLP and Soo KC: A possible role for insulin-like growth factor-binding protein-3 autocrine/paracrine loops in controlling hepatocellular carcinoma cell proliferation. Cell Growth Differ. 13:115–122. 2002.

22. 

Rechler MM and Clemmonds DR: Regulatory actions of insulin-like growth factor-binding proteins. Trends Endocrinol Metab. 9:176–183. 1998. View Article : Google Scholar : PubMed/NCBI

23. 

Baserga R: The insulin-like growth factor I receptor: a key to tumor growth? Cancer Res. 55:249–255. 1995.PubMed/NCBI

24. 

Höppener JW, de Pagter-Holthuizen P, Geurts van Kessel AHM, et al: The human gene encoding insulin- like growth factor I is located on chromosome 12. Hum Genet. 69:157–160. 1985.PubMed/NCBI

25. 

Bell GI, Stempien MM, Fong NM and Rall LB: Sequences of liver cDNAs encoding two different mouse insulin-like growth factor 1 precursors. Nucleic Acids Res. 14:7873–7882. 1986. View Article : Google Scholar : PubMed/NCBI

26. 

Adamo ML: Regulation of insulin-like growth factor I gene expression. Implications for normal and pathological growth. Diabetes Rev. 3:2–27. 1995.

27. 

Temmerman L, Slonimsky E and Rosenthal N: Class 2 IGF-1 isoforms are dispensabile for viability, growth and maintenance of IGF-1 serum levels. Growth Horm IGF Res. 20:255–263. 2010. View Article : Google Scholar : PubMed/NCBI

28. 

Matheny RW Jr, Nindl BC and Adamo ML: Minireview: Mechano-Growth Factor: a putative product of IGF-I gene expression involved in tissue repair and regeneration. Endocrinology. 151:865–875. 2010. View Article : Google Scholar : PubMed/NCBI

29. 

Adamo ML, Ben-Hur H, LeRoith D and Roberts CT Jr: Transcription initiation in the two leader exons of the rat IGF-1 gene occurs from disperse versus localized sites. Biochem Biophys Res Commun. 176:887–893. 1991. View Article : Google Scholar : PubMed/NCBI

30. 

Simmons JG, Van Wyk JJ, Hoyt EC and Lund PK: Multiple transcription start sites in the rat insulin-like growth factor-I gene give rise to IGF-I mRNAs that encode different IGF-I precursors and are processed differently in vitro. Growth Factors. 9:205–221. 1993. View Article : Google Scholar : PubMed/NCBI

31. 

Jansen M, van Schaik FM, Ricker AT, et al: Sequence of cDNA encoding human insulin-like growth factor I precursor. Nature. 306:609–611. 1983. View Article : Google Scholar : PubMed/NCBI

32. 

Roberts CT Jr, Lasky SR, Lowe WI Jr, Seaman WT and LeRoith D: Molecular cloning of rat insulin-like growth factor I complementary deoxyribonucleic acids: differential messenger ribonucleic acid processing and regulation by growth hormone in extrahepatic tissues. Mol Endocrinol. 1:243–248. 1987. View Article : Google Scholar

33. 

Chew SL, Lavender P, Clark AJ and Ross RJ: An alternatively spliced human insulin-like growth factor-1 transcript with hepatic tissue expression that diverts away from mitogenic IBE1 peptide. Endocrinology. 136:1939–1944. 1995.PubMed/NCBI

34. 

Rotwein P: Two insulin-like growth factor I messenger RNAs are expressed in human liver. Proc Natl Acad Sci USA. 83:77–81. 1986. View Article : Google Scholar : PubMed/NCBI

35. 

Shavlakadze T, Winn N, Rosenthal N and Grounds MD: Reconciling data from transgenic mice that overexpress IGF-I specifically in skeletal muscle. Growth Horm IGF Res. 15:4–18. 2005. View Article : Google Scholar : PubMed/NCBI

36. 

Winn N, Paul A, Musaro A and Rosenthal N: Insulin-like growth factor isoforms in skeletal muscle aging, regeneration, and disease. Cold Spring Harb Symp Quant Biol. 67:507–518. 2002. View Article : Google Scholar : PubMed/NCBI

37. 

Rotwein P: Molecular biology of IGF-1 and IGF-2. The IGF System. Rosenfeld RG and Roberts CT Jr: Humana Press; Totowa, NJ: pp. 19–35. 1999, View Article : Google Scholar

38. 

Siegfried JM, Kasprzyk PG, Treston AM, Mulshine JL, Quinn KA and Cuttitta F: A mitogenic peptide amide encoded within the E peptide domain of the insulin-like growth factor IB prohormone. Proc Natl Acad Sci USA. 89:8107–8111. 1992. View Article : Google Scholar : PubMed/NCBI

39. 

Barton ER: The ABCs of IGF-I isoforms: impact on muscle hypertrophy and implications for repair. Appl Physiol Nutr Metab. 31:791–797. 2006. View Article : Google Scholar : PubMed/NCBI

40. 

Barton ER: Viral expression of insulin-like growth factor-1 isoforms promotes different responses in skeletal muscle. J Appl Physiol. 100:1778–1784. 2006. View Article : Google Scholar : PubMed/NCBI

41. 

Rotwein P, Pollock KM, Didier DK and Krivi GG: Organization and sequence of the human insulin-like growth factor I gene. Alternative RNA processing produces two insulin-like growth factor I precursor peptides. J Biol Chem. 261:4828–4832. 1986.PubMed/NCBI

42. 

Bach MA, Roberts CT Jr, Smith EP and LeRoith D: Alternative splicing produces messenger RNAs encoding insulin-like growth factor-I prohormones that are differentially glycosylated in vitro. Mol Endocrinol. 4:899–904. 1990. View Article : Google Scholar : PubMed/NCBI

43. 

Hameed M, Lange KH, Andersen JL, Schjerling P, Kjaer M, Harridge SD and Goldspink G: The effect of recombinant human growth hormone and resistance training of IGF-I mRNA expression in the muscles of elderly men. J Physiol. 555:231–240. 2004. View Article : Google Scholar : PubMed/NCBI

44. 

Tan DS, Cook A and Chew SL: Nucleolar localization of an isoform of the IGF-I precursor. BMC Cell Biol. 3:172003. View Article : Google Scholar

45. 

Adamo ML, Neuenschwander S, LeRoith D and Roberts CT Jr: Structure, expression, and regulation of the IGF-I gene. Adv Exp Med Biol. 343:1–11. 1993. View Article : Google Scholar : PubMed/NCBI

46. 

Wang X, Yang Y and Adamo ML: Characterization of the rat insulin-like growth factor I gene promoters and identification of a minimal exon 2 promoter. Endocrionology. 138:1528–1536. 1997.PubMed/NCBI

47. 

Mittanck DW, Kim SW and Rotweien P: Essential promoter elements are located within the 5′ untranslated region of human insulin-like growth factor-I exon I. Mol Cell Endocrinol. 126:153–163. 1997.

48. 

Nolten LA, van Schaik FM, Steenbergh PH and Sussenbach JS: Expression of the insulin-like growth factor I gene is stimulated by the liver-enriched transcription factors C/EBP alpha and LAP. Mol Endocrinol. 8:1636–1645. 1994.PubMed/NCBI

49. 

Nolten LA, Steenbergh PH and Sussenbach JS: Hepatocyte nuclear factor 1 alpha activates promoter 1 of the human insulin-like growth factor I gene via two distinct binding sites. Mol Endocrinol. 9:1488–1499. 1995.PubMed/NCBI

50. 

Armakolas A, Philippou A, Panteleakou Z, Nezos A, Sourla A, Petraki C and Koutsilieris M: Preferential expression of IGF-IEc (MGF) transcript in cancerous tissues of human prostate: evidence for a novel and autonomous growth factor activity of MGF E peptide in human prostate cancer cells. Prostate. 70:1233–1242. 2010. View Article : Google Scholar

51. 

Koczorowska MM, Kwasniewska A and Gozdzicka-Jozefiak A: IGF1 mRNA isoform expression in the cervix of HPV-positive woman with pre-cancerous and cancer lesions. Exp Ther Med. 2:149–156. 2011.PubMed/NCBI

52. 

Górecki DC, Beresewicz M and Zabłocka B: Neuroprotective effects of short peptides derived from the Insulin-like growth factor 1. Neurochem Int. 51:451–458. 2007.PubMed/NCBI

53. 

Juul A, Bang P, Hertel NT, et al: Serum insulin-like growth factor-I in 1030 healthy children, adolescents, and adults: relation to age, stage of puberty, testicular size, and body mass index. J Clin Endocrinol Metabol. 78:744–752. 1994.PubMed/NCBI

54. 

Brabant G, von zur Muhlen A, Wurster Ch, et al: Serum Insulin-like Growth factor I reference values for an automated chemiluminescence immunoassay system: results from a multi-center study. Horm Res. 60:53–60. 2003. View Article : Google Scholar

55. 

Aimaretti G, Boschetti M, Corneli G, et al: Normal age-dependent values of serum insulin growth factor-I: results from a healthy Italian population. J Endocrinol Invest. 31:445–449. 2008. View Article : Google Scholar : PubMed/NCBI

56. 

Rosario P: Normal values of serum IGF-1 in adults: results from a Brazilian population. Arq Bras Endocrinol Metab. 54:477–481. 2010. View Article : Google Scholar : PubMed/NCBI

57. 

Andreassen M, Nielsen K, Raymond I, Kristensen LØ and Faber J: Characteristics and reference ranges of Insulin-Like Growth factor-I measured with a commercially avaible immunoassay in 724 healthy adult Caucasians. Scand J Clin Lab Invest. 69:880–885. 2009. View Article : Google Scholar : PubMed/NCBI

58. 

Yu H, Mistry J, Nicar MJ, Khosravi MJ, Diamandis A, van Doorn J and Juul A: Insulin-like growth factors (IGF-I, free IGF-I, and IGF-II) and insulin-like growth factor binding proteins (IGFBP-2, IGFBP-3, IGFBP-6, and ALS) in blood circulation. J Clin Lab Anal. 13:166–172. 1999. View Article : Google Scholar : PubMed/NCBI

59. 

Holmes MD, Pollak MN and Hankinson SE: Lifestyle correlates of plasma insulin-like growth factor I and insulin-like growth factor binding protein 3 concentrations. Cancer Epidemiol Biomarkers Prev. 11:862–867. 2002.

60. 

Schneider HJ, Saller B, Klotsche J, März W, Erwa W, Wittchen HU and Stalla GK: Oposite associations of age-dependent insulin-like growth factor-I standard deviation scores with nutritional state in normal weight and obese subject. Eur J Endocrinol. 154:699–706. 2006. View Article : Google Scholar : PubMed/NCBI

61. 

Platz EA, Pollak MN, Rimm EB, Majeed N, Tao Y, Willett WC and Giovannucci E: Racial variation in insulin-like growth factor-1 and binding protein-3 concentrations in middlle-aged men. Cancer Epidemiol Biomarkers Prevent. 8:1107–1110. 1999.PubMed/NCBI

62. 

Lecomte P, Lecureuil N, Lecureuil M, Lemonnier Y, Mariotte N, Valat C and Garrigue MA: Sex differences in the control of sex-hormone-binding globulin in the elderly: role of insulin-like growth factor-I and insulin. Eur J Endocrinol. 139:178–183. 1998. View Article : Google Scholar : PubMed/NCBI

63. 

Sandhu MS, Heald AH, Gibson JM, Cruickshank JK, Dunger DB and Wareham NJ: Circulating concentrations of insulin like growth factor-I and development of glucose intolerance: a prospective observational study. Lancet. 359:1740–1745. 2002. View Article : Google Scholar : PubMed/NCBI

64. 

Sesti G, Sciacqua A, Cardellini M, et al: Plasma concentration of IGF-I is independently associated with insulin sensitivity in subjects with different degrees of glucose tolerance. Diabetes Care. 28:120–125. 2005. View Article : Google Scholar : PubMed/NCBI

65. 

Livingstone C and Ferns G: Insulin-like growth factor-related proteins and diabetic complications. Br J Diabetes Vasc Dis. 3:326–331. 2003. View Article : Google Scholar

66. 

Froesch E, Hussain M, Schmid Ch and Zapf J: Insulin-like growth factor I: physiology, metabolic effects and clinical uses. Diabetes Metab Rev. 12:195–215. 1996. View Article : Google Scholar : PubMed/NCBI

67. 

Pao CI, Farmer PK, Begovic S, Goldstein S, Wu GJ and Philips LS: Expression of hepatic insulin-like growth factor-I and insulin-like growth factor-binding protein-1 genes is transcriptionally regulated in streptozotocin-diabetic rats. Mol Endocrinol. 6:969–977. 1992.PubMed/NCBI

68. 

LeRoith D, Bondy C, Yakar S, Liu JL and Butler A: The somatomedin hypothesis: 2001. Endocr Rev. 22:53–74. 2001. View Article : Google Scholar

69. 

Blum W, Albertsson-Wikland K, Rosberg S and Ranke M: Serum levels of insulin-like growth factor (IGF-I) and IGF binding protein 3 reflect spontaneous growth hormone secretion. J Clin Endocrinol Metabol. 76:1610–1616. 1993.PubMed/NCBI

70. 

Khandwala HM, McCutcheon IE, Flyvbjerg A and Friend KE: The effects of insulin-like growth factors on tumorigenesis and neoplastic growth. Endocr Rev. 21:215–244. 2000. View Article : Google Scholar : PubMed/NCBI

71. 

O’Dell SD and Day IN: Insulin-like growth factor II (IGF-II). Int J Biochem Cell Biol. 30:767–771. 1998.

72. 

Szebenyi G and Rotweien P: The mouse insulin-like growth factor II/cation-independent mannose 6-phosphate (IGF-II/MPR) receptor gene: molecular cloning and genomic organization. Genomics. 19:120–129. 1994. View Article : Google Scholar : PubMed/NCBI

73. 

Frasca F, Pandini G, Scalia P, et al: Insulin receptor isoform A, a newly recognized, high-affinity insulin-like growth factor II receptor in fetal and cancer cells. Mol Cell Biol. 19:3278–3288. 1999.PubMed/NCBI

74. 

Stewart CEH and Rotwein P: Growth, differentiation, and survival: multiple physiological functions for insulin-like growth factors. Physiol Rev. 76:1005–1026. 1996.PubMed/NCBI

75. 

Baserga R: The IGF-1 receptor in cancer biology. Int J Cancer. 107:873–877. 2003. View Article : Google Scholar : PubMed/NCBI

76. 

Grimberg A: Mechanisms by which IGF-I may promote cancer. Cancer Biol Ther. 2:630–635. 2003. View Article : Google Scholar : PubMed/NCBI

77. 

Delafontaine P, Song YH and Li Y: Expression, regulation, and function of IGF-1, IGF-1R, and IGF-1 binding proteins in blood vessels. Arterioscler Thromb Vasc Biol. 24:435–444. 2004. View Article : Google Scholar : PubMed/NCBI

78. 

Yu H and Rohan T: Role of the insulin-like growth factor family in cancer development and progression. J Natl Cancer Inst. 92:1472–1489. 2000. View Article : Google Scholar : PubMed/NCBI

79. 

Samani AA, Yakar S, LeRoith D and Brodt P: The role of the IGF system in cancer growth and metastasis: overview and recent insights. Endocr Rev. 28:20–47. 2007. View Article : Google Scholar : PubMed/NCBI

80. 

Laureys G, Barton DE, Ullrich A and Francke U: Chromosomal mapping of the gene for the type II insulin-like growth factor receptor/cation-independent mannose 6-phosphate receptor in man and mouse. Genomics. 3:224–229. 1988. View Article : Google Scholar : PubMed/NCBI

81. 

Morgan DO, Edman JC, Standring DN, Fried VA, Smith MC, Roth RA and Rutter WJ: Insulin-like growth factor II receptor as a multifunctional binding protein. Nature. 329:301–307. 1987. View Article : Google Scholar : PubMed/NCBI

82. 

Lau MM, Stewart CE, Liu Z, Bhatt H, Rotwein P and Stewart CL: Loss of the imprinted IGF2/cation-independent mannose 6-phosphate receptor results in fetal overgrowth and perinatal lethality. Genes Dev. 8:2953–2963. 1994. View Article : Google Scholar

83. 

Sun H, Chua M-S, Yang D, Tsalenko A, Peter B and So S: Antibody arrays identify potential diagnostic markers of hepatocellular carcinoma. Biomark Insights. 3:1–18. 2008.PubMed/NCBI

84. 

Tovar V, Alsinet C, Villanueva A, et al: IGF activation in a molecular subclass of hepatocellular carcinoma and pre-clinical efficacy of IGF-1R blockage. J Hepatol. 52:550–559. 2010. View Article : Google Scholar : PubMed/NCBI

85. 

Weng CJ, Hsieh YH, Tsai CM, et al: Relationship of insulin-like growth factors system gene polymorphisms with the susceptibility and pathological development of hepatocellular carcinoma. Ann Surg Oncol. 17:1808–1815. 2010. View Article : Google Scholar : PubMed/NCBI

86. 

Scharf JG, Schmidt-Sandte W, Pahernik SA, Ramadori G, Braulke T and Hartmann H: Characterization of the insulin-like growth factor axis in a human hepatoma cell line (PLC). Carcinogenesis. 19:2121–2128. 1998. View Article : Google Scholar : PubMed/NCBI

87. 

Tsai TF, Yauk YK, Chou CK, et al: Evidence of autocrine regulation in human hepatoma cell lines. Biochem Biophys Res Commun. 153:39–45. 1988. View Article : Google Scholar : PubMed/NCBI

88. 

Tanaka S, Mohr L, Schmidt EV, Sugimachi K and Wands JR: Biological effects of human insulin receptor substrate-1 over-expression in hepatocytes. Hepatology. 26:598–604. 1997. View Article : Google Scholar : PubMed/NCBI

89. 

Nishiyama M and Wands JR: Cloning and increased expression of an insulin receptor substrate-1-like gene in human hepatocellular carcinoma. Biochem Biophys Res Commun. 183:280–285. 1992. View Article : Google Scholar : PubMed/NCBI

90. 

Coutant A, Rescan C, Gilot D, Loyer P, Guguen-Guillouzo C and Baffet G: PI3K- FRAP/mTOR pathway is critical for hepatocyte proliferation whereas MEK/ERK supports both proliferation and survival. Hepatology. 36:1079–1088. 2002. View Article : Google Scholar : PubMed/NCBI

91. 

Alexia C, Fallot G, Lasfer M, Schweizer-Groyer G and Groyer A: An evaluation of the role of insulin-like growth factors (IGF) and of type-I IGF receptor signaling in hepatocarcinogenesis and in the resistance of hepatocellular cells against drug-induced apoptosis. Biochem Pharmacol. 86:1003–1015. 2004. View Article : Google Scholar : PubMed/NCBI

92. 

LeRoith D: Insulin-like growth factor I receptor signaling-overlapping or redundant pathways. Endocrinology. 141:1287–1288. 2000.PubMed/NCBI

93. 

Knittel T, Dinter C, Kobold D, Neubauer K, Mehde M, Eichhorst S and Ramadori G: Expression and regulation of cell adhesion molecules by hepatic stellate cells (HSC) of rat liver: involvement of HSC in recruitment of inflammatory cells during hepatic tissue repair. Am J Pathol. 154:153–167. 1999. View Article : Google Scholar

94. 

Wu JC, Daughaday WH, Lee SD, et al: Radioimmunoassay of serum IGF-I and IGF-II in patients with chronic liver diseases and hepatocellular carcinoma with or without hypoglycemia. J Lab Clin Med. 112:589–594. 1988.PubMed/NCBI

95. 

Kratzsch J, Blum WF, Schenker E and Keller E: Regulation of growth hormone (GH), insulin-like growth factor (IGF) I, IGF binding proteins -1, -2, -3 and GH binding protein during progression of liver cirrhosis. Exp Clin Endocrinol Diabetes. 103:285–291. 1995. View Article : Google Scholar : PubMed/NCBI

96. 

Nikolić JA, Todorović V, Bozić M, et al: Serum insulin-like growth factor (IGF)-II is more closely associated with liver disfunction than is IGF-I in patients with cirrhosis. Clin Chim Acta. 294:169–177. 2000.PubMed/NCBI

97. 

Okan A, Comlekci A, Akpinar H, Okan I, Yesil S, Tankurt E and Simsek I: Serum concentration of insulin-like growth factor-I and insulin-like growth factor binding protein-3 in patients with chronic hepatitis. Scand J Gastroenterol. 35:1212–1215. 2000. View Article : Google Scholar : PubMed/NCBI

98. 

Stuver SO, Kuper H, Tzonou A, Lagiou P, Spanos E and Hsieh CC: Insulin-like growth factor 1 in hepatocellular carcinoma and metastatic liver cancer in men. Int J Cancer. 87:118–121. 2000. View Article : Google Scholar : PubMed/NCBI

99. 

Vyzantiadis T, Theodoridou S, Giouleme O, Harsoulis P, Evgenidis N and Vyzantiadis A: Serum concentrations of insulin-like growth factor-I (IGF-I) in patients with liver cirrhosis. Hepatogastroenterology. 50:814–816. 2003.PubMed/NCBI

100. 

Lorenzo-Zúñiga V, Bartoli R, Masnou H, Montoliu S, Morillas RM and Planas R: Serum concentration of insulin-like growth factor-I (IGF-I) as a marker of liver fibrosis in patients with chronic hepatitis C. Dig Dis Sci. 52:3245–3250. 2007.PubMed/NCBI

101. 

Mahdy KA, Ahmed HH, Mannaa F and Abdel-Shaheed A: Clinical benefits of biochemical markers of bone turnover in Egyptian children with chronic liver diseases. World J Gastroenterol. 13:785–790. 2007. View Article : Google Scholar : PubMed/NCBI

102. 

Mazziotti G, Sorvillo F, Morisco F, et al: Serum insulin-growth factor I evaluation as a useful tool for predicting the risk of developing hepatocellular carcinoma in patients with hepatitis C virus-related cirrhosis: a prospective study. Cancer. 95:2539–2545. 2002. View Article : Google Scholar

103. 

Wu YL, Ye J, Zhang S, Zhong J and Xi RP: Clinical significance of serum IGF-I, IGF-II and IGFBP-3 in liver cirrhosis. World J Gastroenterol. 10:2740–2743. 2004.PubMed/NCBI

104. 

Weber MM, Auernhammer CJ, Lee PD, Engelhardt D and Zachoval R: Insulin-like growth factors and insulin-like growth factor binding proteins in adult patients with severe liver disease before and after orthotropic liver transplantation. Horm Res. 57:105–112. 2002. View Article : Google Scholar

105. 

Ranke MB, Maier KP, Schweizer R, Stadler B, Schleicher S, Elmlinger MW and Flehmig B: Pilot study of elevated levels of insulin-like growth factor-binding protein-2 as indicators of hepatocellular carcinoma. Horm Res. 60:174–180. 2003. View Article : Google Scholar : PubMed/NCBI

106. 

Morali G, Shitrit AB, Eran M, Freier S, Reinus C and Braverman D: Hepatic production of insulin-like growth factors in normal and diseased liver. Hepatogastroenterology. 52:1511–1515. 2005.PubMed/NCBI

107. 

Nordstedt G, Levinovitz A, Möller C, Eriksson LC and Anderson G: Expression of insulin-like growth factor I (IGF-I) and IGF-II mRNA during hepatic development, proliferation and carcinogenesis in the rat. Carcinogenesis. 9:209–213. 1988. View Article : Google Scholar : PubMed/NCBI

108. 

Couvert P, Carrie A, Paries J, et al: Liver insulin-like growth factor 2 methylation in hepatitis C virus cirrhosis and further occurrence of hepatocellular carcinoma. World J Gastroenterol. 14:5419–5427. 2008. View Article : Google Scholar : PubMed/NCBI

109. 

Price JA, Kovach SJ, Johnson T, Koniaris LG, Cahill PA, Sitzmann JV and McKillop IH: Insulin-like growth factor I is a comitogen for hepatocytes growth factor in a rat model of hepatocellular carcinoma. Hepatology. 36:1089–1097. 2002. View Article : Google Scholar : PubMed/NCBI

110. 

Kim SO, Park JG and Lee YI: Increased expression of the insulin-like growth factor I (IGF-I) receptor gene in hepatocellular carcinoma cell lines: implications of the IGF-I receptor gene activation by hepatitis B virus X gene product. Cancer Res. 56:3831–3836. 1996.PubMed/NCBI

111. 

Nardone G, Romano M, Calabro A, et al: Activation of fetal promoters of insulin-like growth factors II gene in hepatitis C virus-related chronic hepatitis, cirrhosis, and hepatocellular carcinoma. Hepatology. 23:1304–1312. 1996. View Article : Google Scholar : PubMed/NCBI

112. 

Sohda T, Kamimura S, Iwata K, Shijo H and Okumura M: Immunohistochemical evidence of insulin-like growth factor II in human small hepatocellular carcinoma with hepatitis C virus infection: relationship to fatty change in carcinoma cells. J Gastroenterol Hepatol. 12:224–228. 1997. View Article : Google Scholar

113. 

Chin E, Zhou J, Dai J, Baxter RC and Bondy CA: Cellular localization and regulation of gene expression for components of the insulin-like growth factor ternary binding protein complex. Endocrinology. 134:2498–2504. 1994.PubMed/NCBI

114. 

Arany E, Afford S, Strain AJ, Winwood PJ, Arthur MJ and Hill DJ: Differential cellular synthesis of insulin-like growth factor binding protein-1 (IGFBP-1) and IGFBP-3 within human liver. J Clin Endocrinol Metabol. 79:1871–1876. 1994.PubMed/NCBI

115. 

Ross RJM, Chew SL, D’Souza Li L, et al: Expression of IGF-I and IGF-binding protein genes in cirrhotic liver. J Endocrinol. 149:209–216. 1996. View Article : Google Scholar : PubMed/NCBI

116. 

Völzke H, Nauck M, Rettig R, Dörr M, Higham C, Brabant G and Wallaschofski H: Association between hepatic steatosis and serum IGF1 and IGFBP-3 levels in a population-based sample. Eur J Endocrinol. 161:705–713. 2009.PubMed/NCBI

117. 

Colakoğlu O, Taşkiran B, Colakoğlu G, Kizildağ S, Ari Ozkan F and Unsal B: Serum insulin like growth factor-1 (IGF-1) and insulin growth factor binding protein-3 (IGFBP-3) levels in liver cirrhosis. Turk J Gastroenterol. 18:245–249. 2007.

118. 

Mattera D, Capuano G, Colao A, Pivonello R, Manguso F, Puzziello A and D’Agostino L: Increased IGF-I : IGFBP-3 ratio in patients with hepatocellular carcinoma. Clin Endocrinol. 59:699–706. 2003. View Article : Google Scholar : PubMed/NCBI

119. 

Moller S, Juul A, Becker U, Flyvbjerg A, Skakkebaek NE and Henriksen JH: Concentrations, release, and disposal of insulin-like growth factor (IGF)-binding proteins (IGFBP), IGF-I, and growth hormone in different vascular beds in patients with cirrhosis. J Clin Endocrinol Metab. 80:1148–1157. 1995.PubMed/NCBI

120. 

Juul A, Dalgaard P, Blum WF, et al: Serum levels of insulin-like growth factor (IGF)-binding protein-3 (IGFBP-3) in healthy infants, children, and adolescents: the relation to IGF-I, IGF-II, IGFBP-1, IGFBP-2, age, sex, body mass index, and pubertal maturation. J Clin Endocrin Metabol. 80:2534–2542. 1995.PubMed/NCBI

121. 

Raslan HM, Ezzat WM, Ahmed MM and Rasheed EA: Insulin growth factor-1 and insulin growth factor binding protein-3 in Egyptian patients with chronic hepatitis C. Arch Med Sci. 3:46–51. 2007.

122. 

Shin SY, Lee JR, Noh GW, Kim HJ, Kang WJ, Kim SH and Chung JK: Analysis of serum levels of anti-Mullerian hormone, inhibin B, insulin-like growth factor-I, insulin-like growth factor binding protein-3, and follicle-stimulating hormone with respect to age and menopausal status. J Korean Med Sci. 23:104–110. 2008. View Article : Google Scholar : PubMed/NCBI

123. 

Okabe H, Satoh S, Kato T, et al: Genome-wide analysis of gene expression in human hepatocellular carcionamas using cDNA microarray: identification of genes involved in viral carcinogenesis and tumor progression. Cancer Res. 61:2129–2137. 2001.PubMed/NCBI

124. 

Su TS, Liu WY, Han SH, Jansen M, Yang-Fen TL, P’eng FK and Chou CK: Transcripts of the insulin-like growth factors I and II in human hepatoma. Cancer Res. 49:1773–1777. 1989.PubMed/NCBI

125. 

Ng IO, Lee J MF, Srivastava G and Ng M: Expression of insulin-like growth factor II mRNA in hepatocellular carcinoma. J Gastroenterol Hepatol. 13:152–157. 1998. View Article : Google Scholar : PubMed/NCBI

126. 

Cariani E, Lasserre C, Seurin D, et al: Differential expression of insulin-like growth factor II mRNA in human primary liver cancers, benign liver tumors, and liver cirrhosis. Cancer Res. 48:6844–6849. 1988.PubMed/NCBI

127. 

Lamas E, Le Bail B, Housset C, Boucher O and Brechot C: Localization of insulin-like growth factor-II and hepatitis B virus mRNAs and proteins in human hepatocellular carcinomas. Lab Invest. 64:98–104. 1991.PubMed/NCBI

128. 

Lamas E, Zindy F, Seurin D, Guguen-Guillouzo and Brechot C: Expression of insulin-like growth factor II and receptors for insulin-like growth factor II, insulin-like growth factor I and insulin in isolated and cultured rat hepatocytes. Hepatology. 13:936–940. 1991. View Article : Google Scholar : PubMed/NCBI

129. 

D’Errico A, Grignioni WF, Fiorentino M, et al: Expression of insulin-like growth factor II (IGF-II) in human hepatocellular carcinoma: an immunohistochemical study. Pathol Int. 44:131–137. 1994.

130. 

Iizuka N, Oka M, Tamesa T, Hamamoto Y and Yamada-Okabe H: Imbalance in expression levels of insulin-like growth factor 2 and H19 transcripts linked to progression of hepatocellular carcinoma. Anticancer Res. 24:4085–4089. 2004.PubMed/NCBI

131. 

Sedlaczek N, Hasilik A, Neuhaus P, Schuppan D and Herbst H: Focal overexpression of insulin-like growth factor 2 by hepatocytes and cholangiocytes in viral liver cirrhosis. Br J Cancer. 88:733–739. 2003. View Article : Google Scholar : PubMed/NCBI

132. 

Caro JF, Poulos J, Ittoop O, Pories WJ, Flickinger EG and Sinha MK: Insulin-like growth factor binding in hepatocytes from human liver, human hepatoma, and normal, regenerating, and fetal rat liver. J Clin Invest. 81:976–981. 1988. View Article : Google Scholar : PubMed/NCBI

133. 

Verspohl EJ, Maddux BA and Goldfine ID: Insulin and insulin-like growth factor I regulate the same biological functions in HEP-G2 cells via their own specific receptors. J Clin Endocrinol Metab. 67:169–174. 1988. View Article : Google Scholar : PubMed/NCBI

134. 

Zhang YC, Wang XP, Zhang LY, Song AL, Kou ZM and Li XS: Effect of blocking IGF-I receptor on growth of human hepatocellular carcinoma cells. World J Gastroenterol. 12:3977–3982. 2006.PubMed/NCBI

135. 

Lee YI, Han YJ, Lee SY, et al: Activation of insulin-like growth factor II signaling by mutant type p53. Physiological implications for potentiation of IGF-II signaling by p53 mutant 249. Mol Cell Endocrinol. 203:51–63. 2003. View Article : Google Scholar : PubMed/NCBI

136. 

Lee YI, Lee S, Das GC, Park US and Park SM: Activation of the insulin-like growth factor II transcription by aflatoxin B1 induced p53 mutant 249 is caused by activation of transcription complexes; implications for a gain-of-function during formation of hepatocellular carcinoma. Oncogene. 19:3717–3726. 2000. View Article : Google Scholar

137. 

De Souza AT, Hankins GR, Washington MK, Orton TC and Jirtle RL: M6P/IGF2R gene is mutated in human hepatocellular carcinomas with loss of heterozygosity. Nat Genet. 11:447–449. 1995.PubMed/NCBI

138. 

Su WW, Lee KT, Yeh YT, Soon MS, Wang CL, Yu ML and Wang SN: Association of circulating insulin-like growth factor 1 with hepatocellular carcinoma: one cross-sectional correlation study. J Clin Lab Anal. 24:195–200. 2010. View Article : Google Scholar : PubMed/NCBI

139. 

Plöckinger U, Krüger D, Bergk A, Wiedenmann B and Berg T: Hepatitis-C patients have reduced growth hormone (GH) secretion which improves during long-term therapy with pegylated interferon-alpha. Am J Gastroenterol. 102:2724–2731. 2007.PubMed/NCBI

140. 

Helaly GF, Hussein NG, Refai W and Ibrahim M: Relation of serum insulin-like growth factor-1 (IGF-1) levels with hepatitis C virus infection and insulin resistance. Transl Res. 158:155–162. 2011. View Article : Google Scholar : PubMed/NCBI

141. 

Adamek A, Kasprzak A, Seraszek A, Mikos H, Bura A and Mozer-Lisewska I: Alterations of serum levels of insulin-like growth factor I (IGF-I) and estradiol in chronic hepatitis C. Onkol Wspol. 16:234–239. 2012.PubMed/NCBI

142. 

Conchillo M, Prieto J and Quiroga J: Insulin like growth factor I (IGF-I) and liver cirrhosis. Rev Esp Enferm Dig. 99:156–164. 2007.(In Spanish).

143. 

Hung CH, Wang JH, Hu TH, et al: Insulin resistance is associated with hepatocellular carcinoma in chronic hepatitis C infection. World J Gastroenterol. 16:2265–2271. 2010. View Article : Google Scholar : PubMed/NCBI

144. 

Stefano JT, Correa-Giannella ML, Ribeiro CMF, Alves VAF, Massarollo PCB, Machado MCC and Giannella-Neto D: Increased hepatic expression of insulin-like growth factor-I receptor in chronic hepatitis C. World J Gastroenterol. 28:3821–3828. 2006.PubMed/NCBI

145. 

Tanaka S, Takenaka K, Matsumata T, Mori R and Sugimachi K: Hepatitis C virus replication is associated with expression of transforming growth factor-alpha and insulin-like growth factor-II in cirrhotic livers. Dig Dis Sci. 41:208–215. 1996. View Article : Google Scholar : PubMed/NCBI

146. 

Sohda T, Oka Y, Iwata K, et al: Co-localisation of insulin-like growth factor II and the proliferation marker MIB1 in hepatocellular carcinoma cells. J Clin Pathol. 50:135–137. 1997. View Article : Google Scholar : PubMed/NCBI

147. 

Yakar S, LeRoith D and Brodt P: The role of the growth hormone/insulin-like growth factor axis in tumor growth and progression: lessons from animal models. Cytokine Growth Factor Rev. 16:407–420. 2005. View Article : Google Scholar : PubMed/NCBI

148. 

Wu Y, Brodt P, Sun H, et al: Insulin-like growth factor-I regulates the liver microenvironment in obese mice and promotes liver metastasis. Cancer Res. 70:57–67. 2010. View Article : Google Scholar : PubMed/NCBI

149. 

Lee S, Park U and Lee YI: Hepatitis C virus core protein transactivates insulin-like growth factor II gene transcription through acting concurrently on Egr1 and Sp1 sites. Virology. 283:167–177. 2001. View Article : Google Scholar : PubMed/NCBI

150. 

Kasprzak A, Adamek A, Przybyszewska W, et al: Expression of IGF-I and viral proteins (C, NS3, NS5A) in the livers of patients with chronic HCV infection. Adv Clin Exp Med. 20:263–273. 2011.

Related Articles

Journal Cover

December 2012
Volume 41 Issue 6

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Kasprzak A and Kasprzak A: The insulin-like growth factor (IGF) signaling axis and hepatitis C virus-associated carcinogenesis (Review). Int J Oncol 41: 1919-1931, 2012
APA
Kasprzak, A., & Kasprzak, A. (2012). The insulin-like growth factor (IGF) signaling axis and hepatitis C virus-associated carcinogenesis (Review). International Journal of Oncology, 41, 1919-1931. https://doi.org/10.3892/ijo.2012.1666
MLA
Kasprzak, A., Adamek, A."The insulin-like growth factor (IGF) signaling axis and hepatitis C virus-associated carcinogenesis (Review)". International Journal of Oncology 41.6 (2012): 1919-1931.
Chicago
Kasprzak, A., Adamek, A."The insulin-like growth factor (IGF) signaling axis and hepatitis C virus-associated carcinogenesis (Review)". International Journal of Oncology 41, no. 6 (2012): 1919-1931. https://doi.org/10.3892/ijo.2012.1666